You are on page 1of 16

1392

Send Orders for Reprints to reprints@benthamscience.ae


Current Medicinal Chemistry, 2016, 23, 1392-1407
eISSN: 1875-533X ISSN: 0929-8673

Current
Impact
Factor:
3.85
Medicinal
Chemistry

Selecting Good ‘Drug-Like’ Properties to Optimize Small


The
International
Journal for
Timely In-depth
Reviews
in Medicinal

Molecule Blood-Brain Barrier Penetration Chemistry

BENTHAM
SCIENCE

Paul C. Trippier*

Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health


Sciences Center, Amarillo, TX, USA and Center for Chemical Biology, Department of Chem-
istry and Biochemistry, Texas Tech University, Lubbock, TX, USA
Please provide
corresponding author(s)
Abstract: The success rate to achieve clinical approval of drugs developed to treat diseases photograph

of the central nervous system (CNS) is the lowest of all disease indications. A large contribu-
tor to this poor success rate is failure of small molecules to pass through the blood-brain bar-
rier (BBB), a barrier composed of capillary endothelial cells connected by tight junctions
that functions to extrude xenobiotics from the brain. Designing small molecules to be BBB
penetrant has been the subject of intensive research and has resulted in a series of guidelines to attain the best
 
  

possible chances of BBB penetration. This review will analyze the current state of thinking in ranking the im-
portance of various physicochemical properties required to select BBB penetrant molecules, describe model
systems to determine BBB penetration, summarize data analysis methods and provide an outlook on further
developments in the field.
Keywords: Blood-brain barrier, Drug-likeness, Central Nervous System, Neuropharmacology, Physicochemical
Properties, Drug Design.
Received: April 04, 2015 Revised: March 22, 2016 Accepted: April 04, 2016

INTRODUCTION packed monolayer lining all of the surfaces of the cap-


illaries supplying essential nutrients to the brain [4].
The development of new therapeutics to treat cen-
CNS targeting NCEs must penetrate through this bar-
tral nervous system (CNS) disorders and diseases suf-
rier before they can enter the brain and interact with
fers from the lowest statistical chance of success of all
their therapeutic target. This transport is predominantly
drug categories. The length of time required to bring a
by transcellular passive diffusion, a process that is in-
CNS indication drug to market is estimated at 12-16
fluenced by the same physicochemical properties as for
years, compared to 10-12 years for other disease states
transport across cellular membranes and within the gas-
[1]. Off the total number of new chemical entities
trointestinal (GI) tract, albeit, with much stricter toler-
(NCEs) that enter phase I clinical trial for CNS indica-
ances.
tions only 8% proceed to clinical approval [2]. In order
for such NCEs to penetrate the brain and engage their The existence within the BBB of efflux transporters
target they must pass through the blood-brain barrier serves to further limit total brain penetration for many
(BBB), a feat estimated to be unachievable by 98% of compounds [5]. Of all the active efflux transporters
potential drug molecules [3]. present in the BBB P-glycoprotein (P-gp) is perhaps
the most adversarial in extruding NCEs from the brain
The BBB is more limiting to compound permeabil-
before they can engage their target of action [6-8]. This
ity than most other barriers within the body as it is re-
has led to many efforts to design predictive models to
sponsible for ensuring homeostasis of the brain by pro-
assess substrates of P-gp, prior to the synthesis of com-
tecting against the incursion of toxic substances. It is
pounds for biological evaluation [9]. However, recent
composed of endothelial cells that form a tightly
years have seen the identification of additional active
*Address correspondence to this author at the School of Pharmacy,
efflux transporters at the BBB. While these transporters
Texas Tech University Health Sciences Center, Department of are not as limiting to brain penetration as P-gp they do
Pharmaceutical Sciences, 1300 S. Coulter, Amarillo, TX 79106, represent a non-trivial effect on overall penetration to
USA; Tel: 806-414-9245; Fax: 806-356-4034; the brain [10].
E-mail: paul.trippier@ttuhsc.edu

1875-533X/16 $58.00+.00 © 2016 Bentham Science Publishers


Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1393

While efflux transporters at the BBB represent a change the terms permeability, penetration, crossing,
significant challenge for penetration, influx transporters transport across, etc. Is this correct or does each term
offer an opportunity to enhance penetration. These have subtle differences?
transporters facilitate the uptake of essential molecules
(such as amino acids and peptides) from the blood that BBB Permeability (Papp)
would otherwise be excluded from the brain [11]. The This term is defined as the rate at which the com-
design of CNS drugs to act as substrates of influx pound in question crosses the BBB [22]. When used as
transporters is extremely difficult with most successful a sole definer for evaluation of a compound, care
substrates discovered through serendipity; almost all should be exercised and the nature of the disease/target
are amino acid-like structures. However, the large evaluated. If rapid permeability is required for thera-
amino acid transporter 1 has been shown to be suscep- peutic onset, a low Papp represents a legitimate concern
tible to recognizing small molecules by use of a pro- about the potential efficacy of the NCE. However, if a
drug strategy, indicating that it may one day be possi- compound is designed to counter a disease that requires
ble to target influx transporters as a mechanism to cross a steady release of the drug into the brain (e.g. depres-
the BBB [12]. Currently, influx transporters at the BBB sion) a low Papp is acceptable, albeit not too low as to
[13], such as the organic anion transporting polypep- render insufficient levels of the drug into the brain. As
tides (OATP) play a role in BBB penetration of BBB a general measure of potential for a compound to cross
permeable therapeutics [14]. For a comprehensive re- the BBB, without considering physiological effects,
view on the subject of transporters at the BBB the Papp is an excellent parameter allowing direct compari-
reader is directed to a recent article [15]. son between NCEs.
Another factor (albeit a contentious one) to consider
in penetrating the BBB is plasma protein binding where Brain Distribution
opinion of the importance of this facet of drug design is This term can be defined as the concentration of un-
split [16-18]. Structural and physicochemical properties bound drug in the brain (Cb,u) versus the concentration
that make small molecules effective drugs can also of unbound drug in the plasma (Cp,u), Cb,u/Cp,u or
make them effective protein binders. NCEs that dem- Kp,uu,[23] (also called Kp,free) [24]. A compound of in-
onstrate reduced penetration over predicted values may terest will distribute between the plasma and brain at a
be exhibiting high levels of plasma protein binding in rate determined by its permeability. Compounds with
the circulation. high plasma protein binding may experience low rates
A critical consideration in designing compounds to of permeability and result in poor BBB penetration no
penetrate the BBB is that its function can be effected mater how high their Papp [16]. However, other theories
by disease pathology [19]. Changes that can occur at posit that high plasma protein binding will have no ef-
the BBB in several CNS conditions include altered ex- fect on BBB penetration [17]. The BBB distribution is
pression levels of transporters in the endothial cells in effect a measure of quantity (or extent) of compound
which comprise the BBB, and increased ‘leakage’ crossing the BBB. A Kp,uu close to 1 represents an equi-
through tight junctions [18, 20, 21]. This changing librium of the compound between plasma and brain. By
facet of the BBB makes prediction of penetration even measuring plasma concentration Kp,uu takes into ac-
more fraught, especially when one considers an aging count the effect of permeability and efflux transporters
population and the often complicated multiple health [23, 25]. One way to increase the unbound drug con-
conditions that this population displays. centration in the brain is to increase the dose of com-
pound given, which raises unbound concentration in
This review article will address the current state of the plasma.
the field in what constitutes good ‘drug-like’ properties
to enable BBB penetration. Physicochemical properties BBB Penetration
will be examined as to their relationship to BBB pene-
tration. Model systems for predicting penetration, both A term encompassing both distribution rates and
physical and virtual, will be summarized and the suc- permeability representing an overview of how well a
cess of these models and guidelines will be assessed, compound will cross the BBB. There is no overall dif-
with future directions addressed. ference between BBB penetration, BBB permeability,
or brain distribution.
At the outset of this review it is critical to define the
terms ‘permeability’ ‘distribution’ and ‘penetration’. It Many additional parameters have been defined to
is often the case that many literature reports inter- help the drug discovery process predict and assess BBB
1394 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

penetration such as total brain to plasma ratio (B/P), discussed later, current trends are moving away from
efflux ratio (ER) and fraction of drug unbound (fu,p or this classical view [32].
fu,b). The reader is directed to an excellent review that A revised set of rules [33] for the design of success-
discusses the relative benefits and reliability of these ful BBB permeable compounds stated that compounds
factors to CNS drug discovery [22]. In this review we should contain:
will consider mainly those factors affecting BBB per-
meability and their implications to overall BBB pene- • <8-10 functional groups capable of forming
tration. hydrogen bonds
• A total molecular weight limit of 500 with
PHYSICOCHEMICAL PROPERTIES INFLU- <400 preferred
ENCING BBB PENETRATION
• No acidic functional groups
The physicochemical properties of a molecule
are directly related to its ability to cross the BBB [26]. Over time these rules have been refined as more ac-
To the medicinal chemist this represents both a signifi- curate model systems for assessing BBB penetration
cant opportunity to tailor the structure of compounds to have become available [32, 34], greater numbers of
increase the chances of constructing a successful CNS successful CNS drugs have been clinically approved,
therapeutic, and a considerable challenge to ensure a and their structural features revealed. This information
NCE retains ability to engage its target. has led to a general consensus that hydrogen bond do-
nors are more limiting to BBB permeability than hy-
The ‘standard’ predictor of ‘drug-likeness’ for drogen bond acceptors. When one considers the chemi-
membrane permeation and absorption are Lipinski’s cal composition of phospholipids that comprise the
rule of five that apply only to orally administered drugs BBB (Fig. 1) it is not surprising that hydrogen bond
[27]. These rules were derived after analysis of >2000 donors limit compound penetration. Phospholipids
clinically approved and late stage clinical trial drug have a polar head group comprised of nitrogen, oxygen
candidates and apply only to drugs that absorb by pas- and phosphorous moieties that are hydrophilic and two
sive diffusion through cell membranes. Those com- hydrophobic hydrocarbon tails. Any NCE with avail-
pounds that are substrates for transporters are excep- able hydrogen bond donors that comes into proximity
tions to the rule. The rule states that to cross a cell with the polar head group will interact and form hydro-
membrane a compound should meet defined criteria in gen bonds with the multiple electronegative atoms (hy-
molecular weight, lipophilicity, number of hydrogen drogen bond acceptors), thereby slowing or halting its
bond donors, and number of hydrogen bond acceptors. progress through the BBB. As the phospholipid con-
As noted above, however, the BBB is not just any cell tains no hydrogen bond donors, a compound with only
membrane and it was soon discovered that Lipinski’s hydrogen bond acceptors will not interact and will pass
original rules required modification to be applicable to by the polar head at a faster rate.
BBB penetration.
To account for these observations a new set of rules
Analysis of clinically effective CNS drugs revealed for penetrating the BBB where proposed [35, 36]. In
that compared to other indication drugs the CNS drugs order to penetrate the BBB a compound should have
possessed lower molecular weight, lower polar surface the following characteristics:
area (PSA, a measure of overall charge on the mole-
cule), fewer hydrogen bond donors, higher lipophilic- • Total number of nitrogen and oxygen atoms
ity, and were overall more rigid [26, 28]. Of these (N+O) <6
properties it was classically regarded that lipophilicity • Polar surface area <60-70 Å2
was key. A general linear relationship between lipo-
• Molecular weight <450
philicity and BBB penetration was believed to exist in
that the higher the lipophilicity of a compound the • Log D (distribution coefficient, a measure of
greater its penetration through the BBB [29-31]. Lipo- total lipophilicity of ionized and unionized
philic regions of molecules do not contain functionali- compound) = 1-3
ties capable of forming hydrogen bonds, hence they are • Clog P (partition coefficient, a measure of
not soluble in water. This relationship between hydro- lipophilicity of the unionized compound) –
gen bonding groups and lipophilic groups has a long- (N+O) = >0
standing importance in CNS drug design. However, as
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1395

Fig. (1). A): Structure of the representative BBB phospholipid phosphatidylcholine illustrating the polar head and hydrophobic
tails. Top: chemical structure. Bottom: Ball and Stick model. B): Drug molecules with few or no hydrogen bond donors, only
hydrogen bond acceptors will not bind to the phospholipid and will pass through the BBB at a faster rate. C): Drug molecules
with large numbers of hydrogen bond donors will form hydrogen bonds (dashed lines) to the hydrogen bond acceptors on the
phospholipids of the BBB and will be slowed or stopped from passing through the BBB.

An amendment to Lipinski’s original rules was pro- 19% are neutral and only 6% are acidic [39]. Basic
posed by Veber et. al. after analyzing 1100 orally drugs at physiological pH are largely cationic and can
bioavailable drugs [37]. They concluded that reduction interact with the charged phospholipid heads, facilitat-
of PSA correlates better to increased cell penetration ing passive diffusion [40]. This is a rather fortuitous
than does lipophilicty. The defining factor in reducing happenstance as the majority of CNS drugs rely on ba-
PSA was attributed to the number of rotatable bonds in sic functionality in their pharmacophore to exhibit ac-
the molecule – the more rotatable bonds, the higher the tivity, similar to the endogenous monamine neuro-
PSA value and the lower the penetration rate. It was transmitters, a popular scaffold for CNS drug discov-
established that >10 rotatable bonds reduces oral ery. The same functionality it would appear, also as-
bioavailability. Penetration through cellular membranes sists in BBB penetration.
is analogous with BBB penetration (albeit the BBB is It is evident that all of these parameters define, to
exceedingly more difficult to penetrate) and most CNS some extent, the overall lipophilicity of a drug candi-
drugs have significantly higher rigidity than other date. Electronegative (nitrogen and oxygen) and ionic
classes [26], for example the Alzheimer’s Disease drug species (defined to an extent by PSA) are capable of
memantine compared with the antiviral Acyclovir (Fig. forming hydrogen bonds and therefore are hydrophilic
2). Most centrally acting drug compounds have less (lipophobic) [41]. Optimum levels of lipophilicity for
than five rotatable bonds [38]. BBB penetration have been reported to be representa-
tive of a logD = 2-3 [42]. Is lipophilicity therefore the
most important aspect for the medicinal chemist to
consider when designing a new CNS therapeutic? Re-
cent studies based on new experimental methods to
determine Kp,uu (see model section) have revealed that
unbound drug concentration in the brain does not corre-
late to lipophilicity (as was suggested by a previous
Fig. (2). Chemical structures of the highly rigid clinically study measuring total brain-to-plasma ratio) [32]. A
available Alzheimer’s Disease drug Memantine and the clear dependence of BBB penetration on hydrogen
flexible clinical antiviral drug Acyclovir. bonding was demonstrated. This was attributed to the
In addition to these rules it was noted that acidic ‘additive effect of reducing passive permeability while
functional groups correlated with poor BBB penetra- increasing the possibility of interactions with efflux
tion. Analysis of the structure of clinically successful transporters’. The study demonstrates that the addition
CNS drugs reveals that approximately 75% are basic, of two hydrogen-bond acceptors to a CNS active drug
results in a doubling of its therapeutic window. This
1396 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

again reiterates the importance of avoiding hydrogen- dictive in silico model was used to determine that
bond donors to enhance BBB penetration. BBB+ compounds have a N+O value of <5 and ClogP
– (N+O) should be positive, which were incorporated
IN SILICO MODELS OF BBB PENETRATION into the revised rules for BBB penetration presented
With the advent of more powerful and accurate above [53]. A key consideration when evaluating the
computer software for simulating (bio)chemical proc- usefulness of computer simulation software for assess-
esses significant effort has been invested in predictive ing BBB penetration is the training set of molecules
models of BBB penetration [43-46]. Prediction of BBB used to establish a baseline for BBB penetration. If the
penetration is a complex and challenging area when molecule under consideration varies significantly in its
one considers the multitude of factors requiring simula- structure from the training set, prediction reliability and
tion – uptake, plasma protein binding, efflux pumps, accuracy is impeded. Currently in silico models have
influx transporters, brain tissue binding, first-pass approximately 80% predictive accuracy [54].
metabolic clearance, etc. However accurate prediction One of the most difficult challenges to in silico pre-
software is, the drug molecule under analysis must ul- diction software is accounting for the effects of P-gp
timately be synthesized and tested for BBB penetration (and other transporter) substrates. It has been calculated
in in vivo and/or in vitro model systems. The use of in that approx. 70% of clinical CNS drugs are highly
silico prediction provides a relatively inexpensive permeable and lack P-gp efflux, however, approx. 20%
(compared with animal models) and rapid initial screen of clinical CNS drugs show both low permeability and
for BBB penetration capability of screening libraries. are P-gp substrate specific [48]. No explanations have
Perhaps the first and most widely used in silico pre- yet accounted for this observation. As a result of this
diction of BBB penetration is that of lipophilicity cal- situation and the lack of a known ‘pharmacophore’ for
culation – calculated logP (ClogP) [47]. While calcula- P-gp substrate specificity (Fig. 3) few predictive mod-
tion of this parameter is now a standard feature in a els take into account the effects of P-gp efflux. In an
variety of widely used chemical drawing packages its attempt to address this situation the Biopharmaceuticals
value is limited. Lipophilicity is only one physico- Drug Disposition Classification System (BDDCS),
chemical aspect of a compound that influences BBB which has been used to predict drug disposition in or-
penetration and, as discussed above, is now considered gans throughout the human body was utilized as a
less of a priority than hydrogen bonding when consid- baseline dataset for predicting BBB penetration [55].
ering structural modifications to drug candidates. A Upon combining BDDCS with in vitro and in silico P-
recent analysis of 119 marketed CNS drugs and Pfizer gp substrate data it was found that >90% prediction
CNS preclinical candidates indicated that higher lipo- accuracy could be achieved. Compounds that are pre-
philicity (ClogP >3) is detrimental to BBB penetration dicted as class I drugs in BDDCS should be prioritized
and reduces toxicity potential [48]. for CNS indications [56].

The same group developed a CNS molecular pa- While recent efforts are making advances in sub-
rameter optimization (MPO) algorithm that may be strate predictability they are hampered by the very na-
applicable to identifying drug candidates with an en- ture of the efflux transporter including the existence of
hanced ability to become clinically approved CNS multiple binding sites dictating a much more promis-
drugs. The MPO approach considers six physicochemi- cuous range of substrate possibilities [57]. An in silico
cal properties of a compound under analysis; ClogP, dynamic trans-membrane model of P-gp has recently
ClogD, molecular weight, topological PSA, number of been reported that successfully predicted the substrate
hydrogen bond donors, and most basic center (pKa). binding residues of anticancer agents. The model was
Each parameter is assigned a desirability score ranging constructed using homology modeling of human P-gp
from 0-1 with weighting for lipophilicity and pKa. The based on the structure of C. elgans P-gp, molecular
algorithm was applied to a set of 119 marketed CNS docking, molecular dynamics and binding free energy
drugs as a test set. Results showed that 74% of these calculations [58]. Pharmacophore mapping of P-gp
successful CNS drugs had a high MPO score of ≥4 substrates and non-substrates has led to the identifica-
suggesting the value of this method in predicting suc- tion of pharmacophoric moieties for substrate activity,
cessful CNS drugs [49]. namely hydrogen bond acceptor, positive ionizable,
aromatic ring and hydrophobic groups [59]. These
Many models have been developed that attempt to pharmacophores were confirmed when docking was
classify compounds as either BBB+ (BBB permeable) performed into the P-gp binding cavity [60] of a newly
or BBB- (not BBB permeable) [50-52]. Indeed, a pre-
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1397

Fig. (3). Illustration of the wide chemical space occupied by three substrates of P-gp. The rigid cyclic structure of the H1 His-
tamine antagonist Loratidine, the flexible L-type calcium channel blocker Verapamil, and the steroidal glycoside cardiac drug
Digoxin. No pharmacophore for predicting P-gp substrate specificity has yet been determined.

resolved X-ray structure of murine P-gp [61]. Sepa-


IN VITRO MEASUREMENT OF BBB PENETRA-
rately a data set composed of 423 P-gp substrates and TION
399 non-substrates confirmed these findings. Compari-
son of eight physicochemical properties resulted in mo- Two concepts are important to the measurement of
lecular weight and solubility being identifed as critical brain penetration; rate and extent. The initial rate of
for predicting P-gp substrates or non-substrates. This brain uptake is a measure of BBB permeability ex-
training set lead to a predictive accuracy of 91.2%, pressed, most often, as the permeability surface area
with a predictive accuracy of 83.5% for a test set of (PS) [70]. This measure has several advantages to
200 compounds. Hydrogen bond acceptors in particular measure BBB penetration as it is not affected by me-
spatial patterns along with flexibility were found to be tabolism of the parent compound nor plasma protein
essential to distinguish P-gp substrate likeness [62]. binding [71]. The extent of brain exposure is most of-
Further, a set of 15 favorable and 15 unfavorable frag- ten expressed as the B/P ratio (brain concentration
ments for the design of P-gp inhibitors has been identi- (AUC) / plasma concentration (AUC)) and is deter-
fied using a training set of 973 compounds [63]. mined by the compound of interest’s preferential bind-
ing between brain tissue and blood [70]. Most clini-
The process of quantitative structure-activity rela-
cally available CNS drugs have a B/P of >0.3, that is
tionship (QSAR) has been used extensively for design-
the compound concentration in the brain is 30% of the
ing CNS active drug candidates [64]. Traditionally this
total compound concentration in the plasma [7].
process uses the classical Hansch algorithms to corre-
late observed biological activity to physicochemical The most commonly used methods to predict BBB
properties. Generally QSAR model systems have been penetration rate in cellular models utilize either Caco-2
shown to have an average predictive performance of [72] or Madin-Darby canine kidney (MDCK) cells [73,
within 0.4 log unit [51]. More recent models have ap- 74]. Caco-2 cells are human intestinal cells whose
plied BBB permeability-surface area coefficient data membranes demonstrate many of the same transporters
combined with lipophilicty and hydrogen bonding pre- and tight junctions as the BBB. However, despite these
dictors [65, 66] as well as statistical approaches using similarities Caco-2 cell permeability is not a reliable
molecular energy related discriptors combined with predictor for in vivo BBB penetration [75-77]. The ap-
lipophilicity [67]. For a more detailed review of QSAR plicability of MDCK cells has also been questioned due
in CNS drug discovery the reader is directed to several to their origin from canine kidney capillaries not hu-
excellent reviews [26, 68, 69]. Computer prediction man brain capillaries. A modified MDCK cell line
software can narrow down a large library of com- transfected with the human MDR1 gene (which en-
pounds to those most likely to penetrate the BBB but codes for P-gp) has however, been shown to be a good
ultimately the compound must be synthesized and its predictor of CNS drug effects on 93 clinical CNS com-
ability to penetrate the BBB measured. pounds, including P-gp substrate assessment [78]. As a
result of this transfection MDR-MDCK cells are the
1398 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

most often employed in vitro predictor of P-gp sub- IN VIVO MEASUREMENT OF BBB PENETRA-
strate potential [79]. TION
Much effort focuses on improving microvessel en- Intravenous techniques performed in animal models
dothelial cell permeability assays. These are based on represent the ‘gold standard’ of BBB penetration
isolated brain microvessels from a variety of species measurement as they take into account full physiologi-
and the resulting culture of endothelial cells [80]. cal conditions [94]. Early intravenous techniques relied
When cultured and plated they form monolayers with upon the brain uptake index (BUI) method which used
tight junctions [81]. There are several drawbacks to this radiolabelled drug injected into the animal with triti-
method not least of which is that the cells are used as ated water [95]. The tritiated water is used as an inter-
primary cultures. This presents the need for a supply of nal standard to allow calculation of the amount of
fresh brain, rapid preparation and use, and a specialized compound in the brain. This method is now mostly ob-
skillset. More importantly the expression of transport- solete with newer methods developed to address the
ers varies with each preparation leading to substantial limitations of BUI, namely low sensitivity and short
reproducibility and predictability issues [82, 83]. Addi- measurement time [94].
tionally the brain capillary endothelial cell monolayers The in situ perfusion method addresses the limita-
display a transendothelial electric resistance (TEER) tions of BUI [96]. In this method the direct catheriza-
lower than that of the BBB resulting in a much ‘leak- tion of the common carotid artery of an anesthetized
ier’ model system [84]. subject animal is performed. Blood flow is stopped and
Human pluripotent stem cells (hPSCs) are an area of the perfusate (drug compound, oxygen, etc.) substitutes
much concentration in developing cellular models of for fluid flow to the brain. The integrity of the BBB has
the BBB. The hPSCs can be specified to express many been shown to be maintained during this experiment
properties of the BBB including tight junctions, trans- which can be performed on a timescale of several sec-
porters, and efflux transporters [85]. For further infor- onds to several minutes. The brain is removed from the
mation on this emerging area of BBB modeling the animal and the compound concentration in the brain is
reader is directed to several excellent reviews on the measured (it should be noted that this method only per-
subject [86, 87]. fuses one half of the brain – the half that receives blood
A frequently utilized and high-throughput method flow from the common carotid artery). Perfusion times
of BBB permeability rate prediction is the PAMPA- can be extended up to one hour by addition of oxygen
BBB method. This method uses porcine brain lipid in carriers [97]. This technique has been shown to be
dodecane as a model blood-brain barrier [88]. PAMPA- around twice as sensitive as the BUI method, attributed
BBB successfully predicted the ability of 30 commer- to the enhanced exposure time of the compound to the
cial drugs to either permeate or not permeate the BBB BBB. The method can be extended to assess the trans-
when effects from active uptake/efflux transporters porter dependence of compounds, with the comparison
were discounted [30]. The method was also used to of wild-type animals to transgenic animals that lack a
show significant correlation to in situ brain perfusion of certain transporter e.g. P-gp deficient mice [98]. The in
tested compounds leading to the PAMPA-BBB method situ perfusion method provides high-quality data but
being a reliable, accurate, and low cost model for sim- requires considerable investment in time, surgery tech-
ple BBB-penetration prediction [70]. However, the nique, and animals.
PAMPA-BBB model is a passive transport model, that The mouse brain uptake assay is a rapid and rela-
is without efflux transporters, making this model of tively inexpensive in vivo measure of BBB permeabil-
limited use to predict BBB transport in vivo if used in ity and brain distribution [99]. This method involves
isolation [89]. injection into the tail vain of two mice. A mouse is sac-
Several other methods have, and are, being devel- rificed at five minutes and 60 minutes and the brain
oped for BBB penetration prediction including immo- removed. At five minutes there is minimum nonspe-
bilized lipid HPLC columns [90] and immortalized mi- cific binding to brain tissue allowing accurate analysis
crovessel endothelial cell lines such as TR-BBB [91]. of BBB permeability. At 60 minutes it can be deter-
For more detailed information concerning in vitro mined if the compound is cleared from the brain at the
methods of BBB permeability prediction the reader is same rate as the plasma, i.e. rapid equilibration across
directed to recent reviews [92, 93]. the BBB. The two data points provide insights into ex-
act mechanisms of BBB permeability.
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1399

Brain microdialysis involves the implantation of a throughout the brain. However, access to radionuclei
stereotactic probe directly into the brain [100, 101]. labeled analogues of compounds for these studies re-
Once the probe is implanted, multiple data points can quires specialized skillsets and relevant licenses.
be obtained over the course of many days in animals It is obvious that in vivo methods to determine BBB
that are freely moving. Moreover, the method has been penetration are the most accurate, however the cost and
used in humans to sample the concentration of drugs at specialized skillsets required to use these methods pre-
various time points. The data provided by this method clude their application to early stage drug discovery.
can provide a ‘real time’ picture of BBB penetration. The in vivo models discussed above are all low
However, the surgery is traumatic, time consuming and throughput and resource intensive, making them un-
the probe can actually disrupt the BBB, engendering suitable for early stage drug discovery where hundreds,
leakage. In addition, highly lipophilic compounds have thousands, or tens of thousands of molecules are to be
been noted to absorb onto the membrane within the screened. The use of an ex vivo insect brain may pro-
probe. vide a viable alternative for higher throughput screen-
Cerebrospinal fluid (CSF) measurement as an in ing while retaining many of the advantages of an in
vivo method to assess BBB penetration is non-ideal vivo model. An invertebrate ex vivo brain has been em-
[71, 92]. The blood-CSF barrier (BCSFB) which regu- ployed to screen approximately six molecules a day for
lates compound permeability into the CSF is only about BBB permeability. Species differences seem to be
1/5000th of the surface area of the BBB; compounds comparable, with the insect BBB containing the tight
detected in the CSF may have entered by the BCSFB junctions, lipophilic characteristics and transporters
rather than the BBB [102]. In addition, the CSF is re- encountered in the mammalian BBB [112]. Further de-
freshed every five hours providing rapid turnover. The tails on all highlighted models and additional tech-
prevailing consensus is that drug distribution to the niques can be obtained from recent comprehensive re-
brain and CSF must not be considered identical [103]. views [99, 113-115].
Radiolabelling of compounds to predict BBB pene-
DATA ANALYSIS
tration originated from quantitative radiography meth-
ods [104]. Intravenous injection of the radiolabelled No matter which method is used to assess BBB
compounds into an animal model and subsequent har- penetration, data analysis is paramount. When compar-
vesting of the brain can provide BBB penetration data ing sets of data for a library of drug candidates it is ob-
by measuring radioactivity. The method is particularly viously essential that all data is analyzed in the same
suited for situations when assessment of localization is way using the same calculations to arrive at a defined
critical, i.e. in brain tumors [105]. Recent advances parameter. It is equally essential to ensure that com-
have resulted in the development of radiolabeled P- parative data for known CNS active drugs is available
glycoprotein substrates as positron emission tomogra- and what parameter has been used to define ‘good’
phy (PET) tracers allowing pharmacokinetic assess- BBB penetration. It is therefore especially important to
ment of P-gp substrate specificity [106, 107]. account for both compound penetration through the
BBB and active transport through (or out of) the BBB.
The use of animal-dedicated positron emission to-
mography (microPET) employing 11C radiolabelled The ratio of brain to plasma concentration (Kp) is,
compounds of interest (which has the same chemical historically, the most widely used parameter to quantify
properties as 14C but is detectable using PET) has es- brain penetration, however its relevance has been ques-
tablished itself as a superior technique for in vivo tioned [71]. The range of values accepted as defining
evaluation of BBB penetration [108]. The technique ‘good’ BBB penetration is particularly broad, with a
has been employed both in animals and human volun- variability of up to 2000-fold [25]. In a study of the 32
teers to trace the distribution of radiolabelled drug can- most prescribed CNS drugs, most displayed a range of
didates [109]. An additional advantage of the PET Kp = 0.1-24 in mice, suggesting Kp alone is not an ef-
technique for neuroimaging is that it is not confined to fective determinant of BBB penetration [7]. Protein
the 11C isotope. Fluorine is commonly encountered in binding in both brain and plasma exerts an influence on
many drug molecules and may be replaced by the 18F BBB penetration as was discussed above and a clear
isotope which is a positron emitter and detectable by understanding of this factor is required to assess BBB
PET [110, 111]. penetration. Indeed, it has been shown that different
stereoisomers bind with different rates to plasma pro-
Use of radiolabelled compounds provides very de-
teins [116]. The two enantiomers of cetirizine (R and S)
tailed and accurate information on distribution
1400 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

show distinct values for Kp of 0.04 and 0.22 respec- OUTLOOK


tively (Fig. 4). These values would suggest, at face Unbound-Drug Brain Exposure Assessment
value, that S-cetirizine would penetrate the BBB better
than the R-isomer. However, when BBB permeability It is widely agreed upon that a concentration of un-
was evaluated by using the unbound concentration ratio bound-drug brain exposure at, or exceeding, pharmaco-
(Kp,uu) both stereoisomers were shown to possess iden- logically relevant levels is a critical requirement for
tical penetration. NCEs to show therapeutic benefit in CNS disorders and
may very well be the most relevant measure of BBB
penetration [25]. The high-throughput equilibrium di-
alysis-based assay for estimating the fraction of un-
bound drug in the brain and measuring complete brain
concentration of a compound of interest, promised to
usher in a new era in predicting successful CNS drugs
[123]. However, this assay utilized brain homogenate
that by its very nature changes brain tissue-binding
properties that introduce errors into datasets from this
technique [124]. Estimates place the error within 3-fold
when used as a surrogate to predict brain concentration
Fig. (4). Chemical structures of Cetirizine. The R- and S- [125]. As such, while a useful model, its accuracy has
isomers show distinct values for ratio of brain to plasma fallen short of expectations.
concentration yet penetrate the BBB in equal ammounts. Recently the brain slice method has been applied to
To avoid such confusion and false-positives/ nega- assess drug distribution in the CNS [126]. This has the
tives in the future, the drug discovery community is substantial benefit of retaining close to normal physio-
now moving to a definition of extent of BBB penetra- logical conditions in terms of pH gradients, cell-cell
tion that is defined as the ratio of free brain concentra- interactions and transport systems, providing an in vitro
tion and free plasma concentration at equilibrium (Kp, tissue model very similar to an in vivo brain. The
free or Kp, uu) [16, 23, 117]. method allows a simple, high-throughput, and cost ef-
fective technique to determine unbound volume of dis-
Kp,uu can be derived at steady state from the three-
tribution in the brain [127]. There is a great deal of an-
compartment model:
ticipation that this method which measures binding and
distribution of a compound within the brain, when
combined with other techniques to measure BBB
transport, will be highly successful in predicting BBB
Where PS is the permeability surface area product; penetration of NCEs [115].
CLin is the active uptake into the brain; CLout is the ac- The exact relationship between physicochemical
tive efflux out of the brain; CLbulk is clearance due to properties of drugs and unbound-drug CNS exposure
brain interstitial fluid bulk flow; CLmetabolism is brain has yet to be defined [32]. A recent study of 43 struc-
metabolic clearance [113]. turally diverse CNS drugs was analyzed for unbound-
Relevant in vivo estimations of drug delivery to the drug concentration in the brain [32]. The study found
brain can be described by three parameters: a) CLin to that hydrogen bonding unequivocally influenced un-
account for active transport into the brain, b) Kp,uu to bound-drug concentration in the brain, while lipophilic-
account for the ratio of unbound drug in the brain (and ity showed no effects – the level of unbound brain ex-
thereby free to engage its target providing therapeutic posure is influenced by structure. Use of the brain slice
effect [118]) to that in the blood and c) Vu,brain to ac- method to define structural characteristics that influ-
count for intra-brain distribution [25]. It is recom- ence unbound-drug concentration in the brain is ex-
mended that a combination of these three parameters pected to result in new insights for prioritizing phys-
are measured for each compound in the drug discovery icochemical properties to ensure NCEs remain un-
program, as a single method will fail to take into ac- bound.
count all variables. Among known CNS drugs Kp,uu can In silico methods to predict unbound-drug concen-
vary by up to 150-fold, and Kp can vary by up to 2000- tration in the brain are under development and are
fold. becoming increasingly more reliable. A 89% prediction
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1401

rate with one model was observed on a test set of 111 the potential to provide a novel solution to penetrate
marketed CNS drugs [128]. A computer model based the BBB [139, 140].
on lipid binding and pH partitioning simulations has Physical methods to temporarily open the BBB are
recently been described to predict brain exposure based approaching clinical trial in humans. Microbubbles can
solely on chemical structure [129]. be made to vibrate by high-intensity focused ultrasound
that result in the endothelial cells that form the BBB
P-glycoprotein Substrate Prediction
being pushed apart to allow the penetration of drugs.
Predictive methodologies for many aspects affecting This method has been successfully applied in murine
BBB penetration are at, or approaching an advanced models with no adverse effects [141]. Should this tech-
stage. The area that is being left behind however, is nique prove successful and widely applicable in hu-
arguably the most important – that of BBB influx and mans, the need to design small molecule clinical candi-
efflux transporter substrate prediction. While predic- dates to fit specific physicochemical properties for
tions of P-gp substrate specificity are improving, the BBB penetration may become an artifact of the past.
large binding site of P-gp, which allows multiple bind-
It would seem that the field of CNS drug discovery
ing modes across a wide range of chemical space re-
is at a pivotal moment when considering the definition
mains a significant challenge [130]. In addition, several
of good physicochemical properties for BBB penetra-
studies have reported that the breast cancer resistance
tion. Previous work by many individuals has focused
protein (BCRP) is more highly expressed at the BBB
on optimizing penetration through optimization of
than P-gp [131, 132]. However, significant expression
lipophilicity and removal of acidic functionality. How-
level difference has been observed across species
ever, this would appear to be only part of the equation
[133]. This protein functions to remove exogenous
to ensure BBB penetration. The medicinal chemist has
compounds from the BBB in the same way as P-gp,
developed many ways to penetrate the BBB, yet failure
thus its high expression level represents a new problem
in CNS drug discovery projects continues. This can in
for BBB penetration prediction. This is especially so
no small part be attributed to efflux transporters at the
when it is considered that modeling systems for this
BBB removing substantial amounts of drug compound
transporter are under developed compared with those
from the brain, coupled with less than effective levels
for P-gp [134].
of unbound drug concentration within the brain.
The success encountered in QSAR predictions on
The ultimate aim of medicinal chemists is the de-
substrate specificity for two BBB transporters - the glu-
sign of compounds that can penetrate the BBB them-
cose transporter (GLUT-1) and the large-neutral amino
selves in sufficient unbound brain concentration to
acid transporter (LAT1), where in silico prediction
elicit a therapeutic effect.
models give good reliability, points to the possibility
that P-gp, BCRP and other BBB transporters will OPTIMUM BBB PENETRATION – A FINE BAL-
succumb to accurate modeling predictions [135, 136]. ANCING ACT
Other Developments There have been several attempts at defining ranges
of parameters to increase the probability of good BBB
Cell penetrating peptides as drug delivery technolo-
penetration to several degrees of success. Lipinski’s
gies have been known since 2000 and are a promising
modified rules for BBB penetration are more accu-
area for enhancing BBB penetration of CNS therapeu-
rately guidelines and have served the medicinal chem-
tics [137]. The guanidinium group was determined to
ist well. However the traditional priority towards lipo-
be the active constituent for BBB and cellular mem-
philicty is now shifting more toward hydrogen bonding
brane permeation and a wide variety of guanidinylated
as the major influence on a compound’s BBB penetra-
scaffolds have been utilized from peptides to carbohy-
tion ability. The ratio of brain to plasma concentration
drates [138]. These molecular transporters have been
has been shown to have too broad of a range for pre-
used to deliver small molecules and proteins across
dicting which compound will penetrate the BBB. The
biochemical barriers. As a drug delivery method these
ratio of unbound drug in the brain is widely accepted as
and other methods for penetrating the BBB are valu-
the best parameter for predicting BBB penetration [32].
able assets for CNS therapeutic development however,
they currently suffer from a lack of generality in the However, the very basic attributes that control BBB
molecules they can transport. This will be an area of penetration are all related to the physicochemical prop-
continued and exciting progress in the near future with erties of the NCE in question (Mw, logP, logD, PSA,
1402 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

HBA, HBD). While these can be manipulated to en- the optimized value for a particular compound. If the
hance or reduce BBB penetration there is a self- drug in question is designed to act peripherally, the op-
regulating upper limit of design. Almost all drugs in timum BBB penetration would be zero as no undesir-
clinical use are small molecules, while the exact mo- able CNS side effects would be possible if the com-
lecular weight range for successfully penetrating the pound cannot penetrate the BBB.
BBB is debatable (<400, <500) it is undeniable that a After all the BBB penetration design parameters
finite amount of space is available on a drug molecule have been applied and predictive models performed
to optimize physicochemical properties. Within this there is one more aspect of BBB penetration that can-
space three essential properties need to be tailored – not be ignored – serendipity – a compound may simply
potency, solubility, and BBB penetration. ‘get lucky’ and be a substrate for active transporters at
The intent in designing CNS active NCEs is that the BBB, or ‘unlucky’ if that transporter is P-gp.
they are effective therapeutics in treating the disease
they are designed to combat. To carry out this function CONCLUSION
they must possess a pharmacophore – an exact spacial Prediction of good drug like properties for BBB
arrangement of functionalities that display hydrogen penetration has evolved over the past decades to take
bonding suitable for binding the biological target of into account several distinct parameters – brain distri-
action [119]. The pharmacophore represents an essen- bution (including unbound-drug concentration in the
tial component of the molecule optimized over many brain), permeability, and efflux transporters. It appears
generations of compound analogues to be the most ef- that for each of these parameters, successful guidelines
fective arrangement of functionalities. While it is hy- for the development of a CNS drug have been devel-
drogen bonds that control BBB penetration they also oped with the exception of predicting BBB transporter
control potency and even a reduction of a single hydro- substrate specificity. These guidelines can be collated
gen bonding functional group can inactivate a com- into a priority table of physicochemical characteristics
pound [120, 121]. There is therefore a minimum that if followed will represent the highest chance of
amount of hydrogen bond acceptors and donors re- successes in achieving BBB penetration. However,
quired for a drug compound to be active. This number caution is required so as to ensure that the pharma-
varies widely with target and compound structure. cophore of the drug candidate is not impeded. Success-
In turn, solubility in aqueous media for delivery is fully penetrating the BBB with an inactive compound
determined by hydrogen bonding and lipophilicity. A is just as bad as not penetrating the BBB with the most
compound’s ability to hydrogen bond to water allows it active compound.
to be soluble, a lack of hydrogen bonding interactions From the preceding review it is apparent that an up-
leads to lipophilicity. A balance is required that does dated set of priorities can be constructed to enhance the
not infringe on the pharmacophore if the drug is to chances of maximum BBB penetration of NCEs di-
reach its target and be active. While different delivery rected to the CNS:
methods can counter high water solubility the patients
quality of life and compliance is ultimately the driving 1. Minimize the number of hydrogen bond donors (in-
force behind oral delivery. creases rate of penetration and unbound-drug brain
concentration)
It has been commented on that the majority of CNS
drugs contain amine functionalities. Simply adding 2. Exclude acid functionality (increases potential to
amines to a NCEs structure is not an effective strategy penetrate the BBB as a hydrogen bond donor and
to improve BBB penetration. Addition of an amine to a ionizable group is removed)
phenyl ring will provide an aniline – a know toxi- 3. Higher lipophilicity (increases rate of penetration,
cophore that forms a variety of active species that bind no effect on unbound-drug brain concentration)
proteins [122]. Care also needs to be exercised in
4. Small molecular weight molecules (<500 Da, in-
avoiding intra-molecular hydrogen bonding that can
creases ability to penetrate the BBB)
render a compound inactive [120].
The traditional guidelines of higher lipophilicity
It is therefore a careful balancing act between po-
equals better BBB penetration are called into question.
tency, solubility, toxicity, and BBB penetration that is
Certainly limiting hydrogen bond donors in the NCE
required to design an effective CNS therapeutic. What
will impact both penetration and unbound-drug brain
is the optimum BBB penetration? This is unique to
concentration thereby enhancing overall BBB perme-
each individual molecule and can only be defined as
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1403

ability. It is noteworthy that for a medicinal chemist the NCE = New chemical entity
question of higher lipophilicity or lower number of hy- P-gp = P-glycoprotein
drogen bond donors may be a matter of philosophy.
Lipophilic groups on molecules are lipophilic in nature PS = permeability surface area
due to their inability to form hydrogen bonds with wa- PSA = Polar surface area
ter. It is therefore possible that reducing the number of
QSAR = Quantitative structure-activity relation-
hydrogen bond donors or increasing lipophilicty result
ship
in the same net gain in BBB penetration. Other descrip-
tors for good drug like properties include PSA and TEER = Transendothelial electric resistance
logD, both of which are heavily influenced by the hy-
drogen bonding potential of the NCE in question. CONFLICT OF INTEREST
Perhaps the correct priority assignment is a matter The author confirms that this article content has no
of semantics and relies on the use of hydrogen bond conflict of interest.
acceptors – increases rate of BBB penetration, has
minimal effect on unbound-drug brain fraction and re- ACKNOWLEDGEMENTS
duces lipophilicity. A quick survey of the structures of We are grateful to Texas Tech University Health
clinically approved CNS drugs shows that most contain Sciences Center for funding.
several purely hydrogen bond acceptor functionalities
(ketones, ethers, etc.). REFERENCES
The use of new molecular modeling software and [1] Alavijeh, M.S.; Chishty, M.; Qaiser, M.Z.; Palmer, A.M.
Drug metabolism and pharmacokinetics, the blood-brain
high-throughput models for determining brain distribu- barrier, and central nervous system drug discovery. Neu-
tion as described herein are expected to clarify the roRx,, 2005, 2, 554-571.
guidelines on good drug like properties for BBB pene- [2] Kola, I.; Landis, J. Can the pharmaceutical industry reduce
attrition rates? Nat. Rev. Drug Discov., 2004, 3, 711-715.
tration. No matter how good our predictive models be- [3] Pardridge, W.M. Crossing the blood-brain barrier: are we
come there is however, at present, no substitute for getting it right? Drug Discov. Today, 2001, 6, 1-2.
synthesizing NCEs and testing BBB penetration in [4] Ballabh, P.; Braun, A.; Nedergaard, M. The blood-brain
vivo. barrier: an overview - Structure, regulation, and clinical im-
plications. Neurobiol. Dis., 2004, 16, 1-13.
[5] Loscher, W.; Potschka, H. Blood-brain barrier active efflux
LIST OF ABBREVIATIONS transporters: ATP-binding cassette gene family. NeuroRx,
2005, 2, 86-98.
AUC = Area under the curve [6] Schinkel, A.H. P-Glycoprotein, a gatekeeper in the blood-
BBB = Blood-brain barrier brain barrier. Adv. Drug Deliv. Rev., 1999, 36, 179-194.
[7] Doran, A.; Obach, R.S.; Smith, B.J.; Hosea, N.A.; Becker,
BCSF = Blood-cerebrospinal fluid barrier S.; Callegari, E.; Chen, C.; Chen, X.; Choo, E.; Cianfrogna,
J.; Cox, L.M.; Gibbs, J.P.; Gibbs, M.A.; Hatch, H.; Hop,
BDDCS = Biopharmaceutical drug disposition C.E.; Kasman, I.N.; Laperle, J.; Liu, J.; Liu, X.; Logman,
classification system M.; Maclin, D.; Nedza, F.M.; Nelson, F.; Olson, E.; Rahe-
matpura, S.; Raunig, D.; Rogers, S.; Schmidt, K.; Spracklin,
BUI = Brain uptake index D.K.; Szewc, M.; Troutman, M.; Tseng, E.; Tu, M.; Van
Deusen, J.W.; Venkatakrishnan, K.; Walens, G.; Wang,
Clog P = partition coefficient E.Q.; Wong, D.; Yasgar, A.S.; Zhang, C. The impact of P-
glycoprotein on the disposition of drugs targeted for indica-
CNS = Central nervous system tions of the central nervous system: evaluation using the
MDR1A/1B knockout mouse model. Drug Metab. Dispos.,
CSF = Cerebrospinal fluid 2005, 33, 165-174.
HBA = Hydrogen bond acceptor [8] Cordon-Cardo, C.; O'Brien, J.P.; Casals, D.; Rittman-
Grauer, L.; Biedler, J.L.; Melamed, M.R.; Bertino, J.R.
HBD = Hydrogen bond donor Multidrug-resistance gene (P-glycoprotein) is expressed by
endothelial cells at blood-brain barrier sites. Proc. Natl.
HPLC = High pressure liquid chromatography Acad. Sci. U. S. A., 1989, 86, 695-698.
[9] Ha, S.N.; Hochman, J.; Sheridan, R.P. Mini review on mo-
Log D = Distribution coefficient lecular modeling of P-glycoprotein (Pgp). Curr. Top. Med.
Chem., 2007, 7, 1525-1529.
MDCK = Madin-Darby canine Kidney [10] Maurer, T.S.; DeBartolo, D.B.; Tess, D.A.; Scott, D.O.
MPO = molecular parameter optimization Relationship between exposure and nonspecific binding of
thirty-three central nervous system drugs in mice. Drug Me-
Mw = Molecular weight tab. Dispos., 2005, 33, 175-181.
1404 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

[11] Hawkins, R.A.; O'Kane, R.L.; Simpson, I.A.; Vina, J.R. [30] Liu, X.; Tu, M.; Kelly, R.S.; Chen, C.; Smith, B.J. Devel-
Structure of the blood-brain barrier and its role in the trans- opment of a computational approach to predict blood-brain
port of amino acids. J. Nutr., 2006, 136, 218S-226S. barrier permeability. Drug Metab. Dispos., 2004, 32, 132-
[12] Peura, L.; Malmioja, K.; Laine, K.; Leppanen, J.; Gynther, 139.
M.; Isotalo, A.; Rautio, J. Large amino acid transporter 1 [31] Summerfield, S.G.; Read, K.; Begley, D.J.; Obradovic, T.;
(LAT1) prodrugs of valproic acid: new prodrug design Hidalgo, I.J.; Coggon, S.; Lewis, A.V.; Porter, R.A.; Jef-
ideas for central nervous system delivery. Mol. Pharm., frey, P. Central nervous system drug disposition: the rela-
2011, 8, 1857-1866. tionship between in situ brain permeability and brain free
[13] Tsuji, A. Small molecular drug transfer across the blood- fraction. J. Pharmacol. Exp. Ther., 2007, 322, 205-213.
brain barrier via carrier-mediated transport systems. Neu- [32] Friden, M.; Winiwarter, S.; Jerndal, G.; Bengtsson, O.;
roRx, 2005, 2, 54-62. Wan, H.; Bredberg, U.; Hammarlund-Udenaes, M.; Antons-
[14] Kalliokoski, A.; Niemi, M. Impact of OATP transporters on son, M. Structure-brain exposure relationships in rat and
pharmacokinetics. Br. J. Pharmacol., 2009, 158, 693-705. human using a novel data set of unbound drug concentra-
[15] Pardridge, W.M. Drug transport across the blood-brain tions in brain interstitial and cerebrospinal fluids. J. Med.
barrier. J. Cereb. Blood Flow Metab., 2012, 32, 1959-1972. Chem., 2009, 52, 6233-6243.
[16] Liu, X.R.; Chen, C.P.; Hop, C.E.C.A. Do we need to opti- [33] Pardridge, W.M. CNS drug design based on principles of
mize plasma protein and tissue binding in drug discovery? blood-brain barrier transport. J. Neurochem., 1998, 70,
Curr. Top. Med. Chem., 2011, 11, 450-466. 1781-1792.
[17] Smith, D.A.; Di, L.; Kerns, E.H. The effect of plasma pro- [34] Naik, P.; Cucullo, L. In vitro blood-brain barrier models:
tein binding on in vivo efficacy: misconceptions in drug current and perspective technologies. J. Pharm. Sci., 2012,
discovery. Nat. Rev. Drug Discov., 2010, 9, 929-939. 101, 1337-1354.
[18] Abbott, N.J.; Friedman, A. Overview and introduction: the [35] Lobell, M.; Molnar, L.; Keseru, G.M. Recent advances in
blood-brain barrier in health and disease. Epilepsia, 2012, the prediction of blood-brain partitioning from molecular
53(Suppl 6), 1-6. structure. J. Pharm. Sci., 2003, 92, 360-370.
[19] Hawkins, B.T.; Davis, T.P. The blood-brain bar- [36] Clark, D.E. In silico prediction of blood-brain barrier per-
rier/neurovascular unit in health and disease. Pharmacol. meation. Drug Discov. Today, 2003, 8, 927- 933.
Rev., 2005, 57, 173-185. [37] Veber, D.F.; Johnson, S.R.; Cheng, H.Y.; Smith, B.R.;
[20] Potschka, H. Transporter hypothesis of drug-resistant epi- Ward, K.W.; Kopple, K.D. Molecular properties that influ-
lepsy: challenges for pharmacogenetic approaches. Phar- ence the oral bioavailability of drug candidates. J. Med.
macogenomics, 2010, 11, 1427-1438. Chem., 2002, 45, 2615-2623.
[21] Drouin-Ouellet, J.; Sawiak, S.J.; Cisbani, G.; Lagace, M.; [38] Leeson, P.D.; Davis, A.M. Time-related differences in the
Kuan, W.L.; Saint-Pierre, M.; Dury, R.J.; Alata, W.; St- physical property profiles of oral drugs. J. Med. Chem.,
Amour, I.; Mason, S.L.; Calon, F.; Lacroix, S.; Gowland, 2004, 47, 6338-6348.
P.A.; Francis, S.T.; Barker, R.A.; Cicchetti, F. Cerebrovas- [39] Kearns, E.H.; Di, L. Drug-Like Properties: Concepts,
cular and blood-brain barrier impairments in Huntington's Structure Design and Methods. Elsevier: London, UK,
disease: Potential implications for its pathophysiology. Ann. 2008.
Neurol., 2015, 78, 160-177. [40] Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof,
[22] Di, L.; Rong, H.J.; Feng, B. Demystifying brain penetration S.R.; Begley, D.J. Structure and function of the blood-brain
in central nervous system drug discovery. J. Med. Chem., barrier. Neurobiol. Dis., 2010, 37, 13-25.
2013, 56, 2-12. [41] Kelder, J.; Grootenhuis, P.D.; Bayada, D.M.; Delbressine,
[23] Gupta, A.; Chatelain, P.; Massingham, R.; Jonsson, E.N.; L.P.; Ploemen, J.P. Polar molecular surface as a dominating
Hammarlund-Udenaes, M. Brain distribution of cetirizine determinant for oral absorption and brain penetration of
enantiomers: comparison of three different tissue-to-plasma drugs. Pharm. Res., 1999, 16, 1514-1519.
partition coefficients: K(p), K(p,u), and K(p,uu). Drug Me- [42] van de Waterbeemd, H.; Smith, D.A.; Jones, B.C. Lipophil-
tab. Dispos., 2006, 34, 318-323. icity in PK design: methyl, ethyl, futile. J. Comput. Aided
[24] Liu, X.R.; Chen, C.P. Strategies to optimize brain penetra- Mol. Des., 2001, 15, 273-286.
tion in drug discovery. Curr. Opin. Drug. Disc., 2005, 8, [43] Vastag, M.; Keseru, G.M. Current in vitro and in silico
505-512. models of blood-brain barrier penetration: a practical view.
[25] Hammarlund-Udenaes, M.; Friden, M.; Syvanen, S.; Gupta, Curr. Opin Drug. Discov. Devel., 2009, 12, 115-124.
A. On the rate and extent of drug delivery to the brain. [44] Goodwin, J.T.; Clark, D.E. In silico predictions of blood-
Pharm. Res., 2008, 25, 1737-1750. brain barrier penetration: Considerations to "keep in mind".
[26] Pajouhesh, H.; Lenz, G.R. Medicinal chemical properties of J. Pharmacol. Exp. Ther., 2005, 315, 477-483.
successful central nervous system drugs. NeuroRx, 2005, 2, [45] Hou, T.; Xu, X. ADME evaluation in drug discovery. 1.
541-553. Applications of genetic algorithms to the prediction of
[27] Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. blood-brain partitioning of a large set of drugs. J. Mol.
Experimental and computational approaches to estimate Model., 2002, 8, 337-349.
solubility and permeability in drug discovery and develop- [46] Hou, T.J.; Xu, X.J. ADME evaluation in drug discovery. 3.
ment settings. Adv. Drug Deliv. Rev., 1997, 23, 3-25. Modeling blood-brain barrier partitioning using simple mo-
[28] Doan, K.M.M.; Humphreys, J.E.; Webster, L.O.; Wring, lecular descriptors. J. Chem. Inf, Comput. Sci., 2003, 43,
S.A.; Shampine, L.J.; Serabjit-Singh, C.J.; Adkison, K.K.; 2137-2152.
Polli, J.W. Passive permeability and P-glycoprotein- [47] Leo, A.J. Calculating log P(Oct) from structures. Chem.
mediated efflux differentiate central nervous system (CNS) Rev., 1993, 93, 1281-1306.
and non-CNS marketed drugs. J. Pharmacol. Exp. Ther., [48] Wager, T.T.; Chandrasekaran, R.Y.; Hou, X.; Troutman,
2002, 303, 1029-1037. M.D.; Verhoest, P.R.; Villalobos, A.; Will, Y. Defining de-
[29] Levin, V.A. Relationship of octanol/water partition coeffi- sirable central nervous system drug space through the
cient and molecular weight to rat brain capillary permeabil- alignment of molecular properties, in vitro ADME, and
ity. J. Med. Chem., 1980, 23, 682-684. safety attributes. ACS Chem. Neurosci., 2010, 1, 420-434.
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1405

[49] Wager, T.T.; Hou, X.; Verhoest, P.R.; Villalobos, A. Mov- [67] Bujak, R.; Struck-Lewicka, W.; Kaliszan, M.; Kaliszan, R.;
ing beyond rules: the development of a central nervous sys- Markuszewski, M.J. Blood-brain barrier permeability
tem multiparameter optimization (CNS MPO) approach to mechanisms in view of quantitative structure-activity rela-
enable alignment of druglike properties. ACS Chem. Neuro- tionships (QSAR). J. Pharmaceut. Biomed., 2015, 108, 29-
sci., 2010, 1, 435-449. 37.
[50] Crivori, P.; Cruciani, G.; Carrupt, P.A.; Testa, B. Predicting [68] Mehdipour, A.R.; Hamidi, M. Brain drug targeting: a com-
blood-brain barrier permeation from three- dimensional mo- putational approach for overcoming blood- brain barrier.
lecular structure. J. Med. Chem., 2000, 43, 2204-2216. Drug Discov. Today, 2009, 14, 1030-1036.
[51] Clark, D.E. Computational prediction of blood-brain barrier [69] Quignot, N. Modeling bioavailability to organs protected by
permeation. Annu. Rep. Med. Chem., 2005, 40, 403-415. biological barriers. In silico Pharmacol., 2013, 1, 8.
[52] Cruciani, C.; Crivori, P.; Carrupt, P.A.; Testa, B. Molecular [70] Di, L.; Kerns, E.H.; Bezar, I.F.; Petusky, S.L.; Huang, Y.
fields in quantitative structure-permeation relationships: the Comparison of blood-brain barrier permeability assays: in
VolSurf approach. J. Mol. Struc-Theochem., 2000, 503, 17- situ brain perfusion, MDR1-MDCKII and PAMPA-BBB. J.
30. Pharm. Sci., 2009, 98, 1980-1991.
[53] Norinder, U.; Haeberlein, M. Computational approaches to [71] Pardridge, W.M. Log(BB), PS products and in silico mod-
the prediction of the blood-brain distribution. Adv. Drug els of drug brain penetration. Drug Discov. Today, 2004, 9,
Deliv. Rev., 2002, 54, 291-313. 392-393.
[54] Ecker, G.F.; Noe, C.R. In silico prediction models for [72] Camenisch, G.; Alsenz, J.; van de Waterbeemd, H.; Folkers,
blood-brain barrier permeation. Curr. Med. Chem., 2004, G. Estimation of permeability by passive diffusion through
11, 1617-1628. Caco-2 cell monolayers using the drugs' lipophilicity and
[55] Broccatelli, F.; Cruciani, G.; Benet, L.Z.; Oprea, T.I. molecular weight. Eur. J. Pharm. Sci., 1998, 6, 317-324.
BDDCS class prediction for new molecular entities. Mol. [73] Irvine, J.D.; Takahashi, L.; Lockhart, K.; Cheong, J.; Tolan,
Pharm., 2012, 9, 570-580. J.W.; Selick, H.E.; Grove, J.R. MDCK (Madin-Darby ca-
[56] Broccatelli, F.; Larregieu, C.A.; Cruciani, G.; Oprea, T.I.; nine kidney) cells: A tool for membrane permeability
Benet, L.Z. Improving the prediction of the brain disposi- screening. J. Pharm. Sci., 1999, 88, 28-33.
tion for orally administered drugs using BDDCS. Adv. Drug [74] Hellinger, E.; Veszelka, S.; Toth, A.E.; Walter, F.; Kittel,
Deliv. Rev., 2012, 64, 95-109. A.; Bakk, M.L.; Tihanyi, K.; Hada, V.; Nakagawa, S.; Duy,
[57] Liu, M.; Hou, T.; Feng, Z.; Li, Y. The flexibility of P- T.D.; Niwa, M.; Deli, M.A.; Vastag, M. Comparison of
glycoprotein for its poly-specific drug binding from mo- brain capillary endothelial cell-based and epithelial
lecular dynamics simulations. J. Biomol. Struct. Dyn., 2013, (MDCK-MDR1, Caco-2, and VB-Caco-2) cell-based surro-
31, 612-629. gate blood-brain barrier penetration models. Eur. J. Pharm.
[58] Prajapati, R.; Singh, U.; Patil, A.; Khomane, K.S.; Bagul, Sci., 2012, 82, 340-351.
P.; Bansal, A.K.; Sangamwar, A.T. In silico model for P- [75] Faassen, F.; Vogel, G.; Spanings, H.; Vromans, H. Caco-2
glycoprotein substrate prediction: insights from molecular permeability, P-glycoprotein transport ratios and brain
dynamics and in vitro studies. J. Comput. Aided Mol. Des., penetration of heterocyclic drugs. Intl. J. Pharm., 2003,
2013, 27, 347-363. 263, 113-122.
[59] Li, W.X.; Li, L.; Eksterowicz, J.; Ling, X.B.; Cardozo, M. [76] Lundquist, S.; Renftel, M.; Brillault, J.; Fenart, L.; Cec-
Significance analysis and multiple pharmacophore models chelli, R.; Dehouck, M.P. Prediction of drug transport
for differentiating P-glycoprotein substrates. J. Chem. Inf. through the blood-brain barrier in vivo: a comparison be-
Model., 2007, 47, 2429-2438. tween two in vitro cell models. Pharm. Res., 2002, 19, 976-
[60] Pajeva, I.K.; Globisch, C.; Wiese, M. Combined pharma- 981.
cophore modeling, docking, and 3D QSAR studies of [77] Eigenmann, D.E.; Xue, G.; Kim, K.S.; Moses, A.V.; Ham-
ABCB1 and ABCC1 transporter inhibitors. ChemMed- burger, M.; Oufir, M. Comparative study of four immortal-
Chem, 2009, 4, 1883-1896. ized human brain capillary endothelial cell lines,
[61] Aller, S.G.; Yu, J.; Ward, A.; Weng, Y.; Chittaboina, S.; hCMEC/D3, hBMEC, TY10, and BB19, and optimization
Zhuo, R.; Harrell, P.M.; Trinh, Y.T.; Zhang, Q.; Urbatsch, of culture conditions, for an in vitro blood-brain barrier
I.L.; Chang, G. Structure of P-glycoprotein reveals a mo- model for drug permeability studies. Fluids Barriers CNS,
lecular basis for poly-specific drug binding. Science, 2009, 2013, 10, 33.
323, 1718-1722. [78] Wang, Q.; Rager, J.D.; Weinstein, K.; Kardos, P.S.; Dob-
[62] Li, D.; Chen, L.; Li, Y.; Tian, S.; Sun, H.; Hou, T. ADMET son, G.L.; Li, J.; Hidalgo, I.J. Evaluation of the MDR-
evaluation in drug discovery. 13. Development of in silico MDCK cell line as a permeability screen for the blood-brain
prediction models for P-glycoprotein substrates. Mol. barrier. Intl. J. Pharm., 2005, 288, 349-359.
Pharm., 2014, 11, 716- 726. [79] Dolghih, E.; Jacobson, M.P. Predicting efflux ratios and
[63] Chen, L.; Li, Y.; Zhao, Q.; Peng, H.; Hou, T. ADME blood-brain barrier penetration from chemical structure:
evaluation in drug discovery. 10. Predictions of P- glyco- combining passive permeability with active efflux by P-
protein inhibitors using recursive partitioning and naive glycoprotein. ACS Chem. Neurosci., 2013, 4, 361-367.
Bayesian classification techniques. Mol. Pharm., 2011, 8, [80] Gumbleton, M.; Audus, K.L. Progress and limitations in the
889-900. use of in vitro cell cultures to serve as a permeability screen
[64] Gupta, S.P. QSAR Studies on Drugs Acting at the Central for the blood-brain barrier. J. Pharm. Sci., 2001, 90, 1681-
Nervous System. Chem. Rev., 1989, 89, 1765-1800. 1698.
[65] Abraham, M.H. The factors that influence permeation [81] Zhang, Y.; Li, C.S.; Ye, Y.; Johnson, K.; Poe, J.; Johnson,
across the blood-brain barrier. Eur. J. Med. Chem., 2004, S.; Bobrowski, W.; Garrido, R.; Madhu, C. Porcine brain
39, 235-240. microvessel endothelial cells as an in vitro model to predict
[66] Wang, W.; Kim, M.T.; Sedykh, A.; Zhu, H. Developing in vivo blood-brain barrier permeability. Drug Metab. Dis-
enhanced blood-brain barrier permeability models: integrat- pos., 2006, 34, 1935-1943.
ing external bio-assay data in QSAR modeling. Pharm. [82] Kramer, S.D. Absorption prediction from physicochemical
Res., 2015, 32, 3055-3065. parameters. Pharm. Sci. Technol. Today, 1999, 2, 373-380.
1406 Current Medicinal Chemistry, 2016, Vol. 23, No. 14 Paul C. Trippier

[83] Reichel, A.; Begley, D.J.; Abbott, N.J. An overview of in evolving new concept. Chem. Biodivers., 2009, 6, 2030-
vitro techniques for blood-brain barrier studies. Methods 2049.
Mol. Med., 2003, 89, 307-324. [103] Collins, J.M.; Dedrick, R.L. Distributed model for drug
[84] Butt, A.M.; Jones, H.C.; Abbott, N.J. Electrical resistance delivery to CSF and brain tissue. Am. J. Physiol., 1983, 245,
across the blood-brain barrier in anaesthetized rats: a devel- R303-310.
opmental study. J. Physiol., 1990, 429, 47-62. [104] Sakurada, O.; Kennedy, C.; Jehle, J.; Brown, J.D.; Carbin,
[85] Lippmann, E.S.; Azarin, S.M.; Kay, J.E.; Nessler, R.A.; G.L.; Sokoloff, L. Measurement of local cerebral blood
Wilson, H.K.; Al-Ahmad, A.; Palecek, S.P.; Shusta, E.V. flow with iodo [14C] antipyrine. Am. J. Physiol., 1978,
Derivation of blood-brain barrier endothelial cells from 234, H59-66.
human pluripotent stem cells. Nat. Biotechnol., 2012, 30, [105] Asotra, K.; Ningaraj, N.; Black, K.L. Measurement of
783-791. blood-brain and blood-tumor barrier permeabilities with
[86] Lippmann, E.S.; Al-Ahmad, A.; Palecek, S.P.; Shusta, E.V. [14C]-labeled tracers. Methods Mol. Med., 2003, 89, 177-
Modeling the blood-brain barrier using stem cell sources. 190.
Fluids Barriers CNS, 2013, 10, 2. [106] Elsinga, P.H.; Hendrikse, N.H.; Bart, J.; Vaalburg, W.; van
[87] He, Y.; Yao, Y.; Tsirka, S.E.; Cao, Y. Cell-culture models Waarde, A. PET Studies on P-glycoprotein function in the
of the blood-brain barrier. Stroke, 2014, 45, 2514-2526. blood-brain barrier: how it affects uptake and binding of
[88] Di, L.; Kerns, E.H.; Fan, K.; McConnell, O.J.; Carter, G.T. drugs within the CNS. Curr. Pharm. Des., 2004, 10, 1493-
High throughput artificial membrane permeability assay for 1503.
blood-brain barrier. Eur. J. Med. Chem., 2003, 38, 223-232. [107] Colabufo, N.A.; Berardi, F.; Perrone, M.G.; Capparelli, E.;
[89] Tsaioun, K.; Bottlaender, M.; Mabondzo, A.; Alzheimer's Cantore, M.; Inglese, C.; Perrone, R. Substrates, inhibitors
Drug Discovery, F. ADDME--Avoiding Drug Development and activators of P-glycoprotein: candidates for radiolabel-
Mistakes Early: central nervous system drug discovery per- ing and imaging perspectives. Curr. Top. Med. Chem.,
spective. BMC Neurol., 2009, 9 Suppl 1, S1. 2010, 10, 1703-1714.
[90] Lazaro, E.; Rafols, C.; Abraham, M.H.; Roses, M. Chroma- [108] Lancelot, S.; Zimmer, L. Small-animal positron emission
tographic estimation of drug disposition properties by tomography as a tool for neuropharmacology. Trends
means of immobilized artificial membranes (IAM) and C18 Pharmacol. Sci., 2010, 31, 411-417.
columns. J. Med. Chem., 2006, 49, 4861-4870. [109] Brooks, D.J. Positron emission tomography and single-
[91] Terasaki, T.; Ohtsuki, S.; Hori, S.; Takanaga, H.; Naka- photon emission computed tomography in central nervous
shima, E.; Hosoya, K. New approaches to in vitro models of system drug development. NeuroRx, 2005, 2, 226-236.
blood-brain barrier drug transport. Drug Discov. Today, [110] Pike, V.W. PET radiotracers: crossing the blood-brain bar-
2003, 8, 944-954. rier and surviving metabolism. Trends Pharmacol. Sci.,
[92] Passeleu-Le Bourdonnec, C.; Carrupt, P.A.; Scherrmann, 2009, 30, 431-440.
J.M.; Martel, S. Methodologies to assess drug permeation [111] Hamill, T.G.; Eng, W.; Jennings, A.; Lewis, R.; Thomas,
through the blood-brain barrier for pharmaceutical research. S.; Wood, S.; Street, L.; Wisnoski, D.; Wolkenberg, S.;
Pharm. Res., 2013, 30, 2729- 2756. Lindsley, C.; Sanabria-Bohorquez, S.M.; Patel, S.; Riffel,
[93] Wilhelm, I.; Fazakas, C.; Krizbai, I.A. In vitro models of K.; Ryan, C.; Cook, J.; Sur, C.; Burns, H.D.; Hargreaves, R.
the blood-brain barrier. Acta Neurobiol. Exp., 2011, 71, The synthesis and preclinical evaluation in rhesus monkey
113-128. of [(1)(8)F]MK-6577 and [(1)(1)C]CMPyPB glycine trans-
[94] Bickel, U. How to measure drug transport across the blood- porter 1 positron emission tomography radiotracers. Syn-
brain barrier. NeuroRx, 2005, 2, 15-26. apse, 2011, 65, 261-270.
[95] Oldendorf, W.H. Measurement of brain uptake of radiola- [112] Al-Qadi, S.; Schiott, M.; Hansen, S.H.; Nielsen, P.A.; Ba-
beled substances using a tritiated water internal standard. dolo, L. An invertebrate model for CNS drug discovery:
Brain Res., 1970, 24, 372-376. Transcriptomic and functional analysis of a mammalian P-
[96] Takasato, Y.; Rapoport, S.I.; Smith, Q.R. An in situ brain glycoprotein ortholog. Biochim. Biophy. Acta, 2015, 1850,
perfusion technique to study cerebrovascular transport in 2439-2451.
the rat. Am. J. Physiol., 1984, 247, H484-493. [113] Liu, X.; Chen, C.; Smith, B.J. Progress in brain penetration
[97] Smith, Q.R.; Allen, D.D. In situ brain perfusion technique. evaluation in drug discovery and development. J. Pharma-
Methods Mol. Med., 2003, 89, 209-218. col. Exp. Ther., 2008, 325, 349-356.
[98] Dagenais, C.; Rousselle, C.; Pollack, G.M.; Scherrmann, [114] Boje, K.M. In vivo measurement of blood-brain barrier
J.M. Development of an in situ mouse brain perfusion permeability. Curr. Protoc. Neurosci., 2001, Chapter 7,
model and its application to mdr1a P-glycoprotein-deficient Unit 7 19.
mice. J. Cereb. Blood Metab., 2000, 20, 381-386. [115] Loryan, I.; Sinha, V.; Mackie, C.; Van Peer, A.; Drinken-
[99] Garberg, P.; Ball, M.; Borg, N.; Cecchelli, R.; Fenart, L.; burg, W.; Vermeulen, A.; Morrison, D.; Monshouwer, M.;
Hurst, R.D.; Lindmark, T.; Mabondzo, A.; Nilsson, J.E.; Heald, D.; Hammarlund-Udenaes, M. Mechanistic under-
Raub, T.J.; Stanimirovic, D.; Terasaki, T.; Oberg, J.O.; Os- standing of brain drug disposition to optimize the selection
terberg, T. In vitro models for the blood-brain barrier. Toxi- of potential neurotherapeutics in drug discovery. Pharm.
col. In Vitro, 2005, 19, 299-334. Res., 2014, 31, 2203-2219.
[100] Deguchi, Y.; Morimoto, K. Application of an in vivo brain [116] Gupta, A.; Hammarlund-Udenaes, M.; Chatelain, P.; Mass-
microdialysis technique to studies of drug transport across ingham, R.; Jonsson, E.N. Stereoselective pharmacokinetics
the blood-brain barrier. Curr. Drug Metab., 2001, 2, 411- of cetirizine in the guinea pig: role of protein binding. Bio-
423. pharm. Drug Dispos., 2006, 27, 291-297.
[101] Darvesh, A.S.; Carroll, R.T.; Geldenhuys, W.J.; Gudelsky, [117] Syvanen, S.; Xie, R.; Sahin, S.; Hammarlund-Udenaes, M.
G.A.; Klein, J.; Meshul, C.K.; Van der Schyf, C.J. In vivo Pharmacokinetic consequences of active drug efflux at the
brain microdialysis: advances in neuropsychopharmacology blood-brain barrier. Pharm. Res., 2006, 23, 705-717.
and drug discovery. Expert Opin. Drug Discov., 2011, 6, [118] Liu, X.; Vilenski, O.; Kwan, J.; Apparsundaram, S.; Weik-
109-127. ert, R. Unbound brain concentration determines receptor
[102] Reichel, A. Addressing central nervous system (CNS) pene- occupancy: a correlation of drug concentration and brain se-
tration in drug discovery: basics and implications of the rotonin and dopamine reuptake transporter occupancy for
Selecting Good ‘Drug-Like’ Properties to Optimize Small Molecule Current Medicinal Chemistry, 2016, Vol. 23, No. 14 1407

eighteen compounds in rats. Drug Metab. Dispos., 2009, [130] Chen, L.; Li, Y.; Yu, H.; Zhang, L.; Hou, T. Computational
37, 1548- 1556. models for predicting substrates or inhibitors of P-
[119] Mason, J.S.; Cheney, D.L. Library design and virtual glycoprotein. Drug Discov. Today, 2012, 17, 343-351.
screening using multiple 4-point pharmacophore finger- [131] Eisenblatter, T.; Huwel, S.; Galla, H.J. Characterisation of
prints. Pac. Symp. Biocomput., 2000, pp. 576-587. the brain multidrug resistance protein (BMDP/ABCG2/
[120] Trippier, P.C.; Benmohammed, R.; Kirsch, D.R.; Silver- BCRP) expressed at the blood-brain barrier. Brain Res.,
man, R.B. Substituted pyrazolones require N2 hydrogen 2003, 971, 221-231.
bond donating ability to protect against cytotoxicity from [132] Shawahna, R.; Uchida, Y.; Decleves, X.; Ohtsuki, S.;
protein aggregation of mutant superoxide dismutase 1. Yousif, S.; Dauchy, S.; Jacob, A.; Chassoux, F.; Daumas-
Bioorg. Med. Chem. Lett., 2012, 22, 6647-6650. Duport, C.; Couraud, P.O.; Terasaki, T.; Scherrmann, J.M.
[121] Trippier, P.C.; Zhao, K.T.; Fox, S.G.; Schiefer, I.T.; Ben- Transcriptomic and quantitative proteomic analysis of
mohamed, R.; Moran, J.; Kirsch, D.R.; Morimoto, R.I.; Sil- transporters and drug metabolizing enzymes in freshly iso-
verman, R.B. Proteasome activation is a mechanism for lated human brain microvessels. Mol. Pharm., 2011, 8,
pyrazolone small molecules displaying therapeutic potential 1332-1341.
in amyotrophic lateral sclerosis. ACS Chem. Neurosci., [133] Ito, K.; Uchida, Y.; Ohtsuki, S.; Aizawa, S.; Kawakami, H.;
2014, 5, 823- 829. Katsukura, Y.; Kamiie, J.; Terasaki, T. Quantitative mem-
[122] Attia, S.M. Deleterious effects of reactive metabolites. brane protein expression at the blood-brain barrier of adult
Oxid. Med. Cell Longev., 2010, 3, 238-253. and younger cynomolgus monkeys. J. Pharm. Sci., 2011,
[123] Kalvass, J.C.; Maurer, T.S. Influence of nonspecific brain 100, 3939-3950.
and plasma binding on CNS exposure: implications for ra- [134] Ishikawa, T.; Saito, H.; Hirano, H.; Inoue, Y.; Ikegami, Y.
tional drug discovery. Biopharm. Drug Dispos., 2002, 23, Human ABC transporter ABCG2 in cancer chemotherapy:
327-338. drug molecular design to circumvent multidrug resistance.
[124] Friden, M.; Bergstrom, F.; Wan, H.; Rehngren, M.; Ahlin, Methods Mol. Biol., 2012, 910, 267-278.
G.; Hammarlund-Udenaes, M.; Bredberg, U. Measurement [135] Abbott, N.J. Blood-brain barrier structure and function and
of unbound drug exposure in brain: modeling of pH parti- the challenges for CNS drug delivery. J. Inherit. Metab.
tioning explains diverging results between the brain slice Dis., 2013, 36, 437-449.
and brain homogenate methods. Drug Metab. Dispos., [136] Montanari, F.; Ecker, G.F. Prediction of drug-ABC-
2011, 39, 353-362. transporter interaction--Recent advances and future chal-
[125] Liu, X.; Van Natta, K.; Yeo, H.; Vilenski, O.; Weller, P.E.; lenges. Adv. Drug Deliv. Rev., 2015, 86, 17-26.
Worboys, P.D.; Monshouwer, M. Unbound drug concentra- [137] Zou, L.L.; Ma, J.L.; Wang, T.; Yang, T.B.; Liu, C.B. Cell-
tion in brain homogenate and cerebral spinal fluid at steady penetrating Peptide-mediated therapeutic molecule delivery
state as a surrogate for unbound concentration in brain in- into the central nervous system. Curr. Neuropharmacol.,
terstitial fluid. Drug Metab. Dispos., 2009, 37, 787-793. 2013, 11, 197-208.
[126] Loryan, I.; Friden, M.; Hammarlund-Udenaes, M. The brain [138] Wender, P.A.; Mitchell, D.J.; Pattabiraman, K.; Pelkey,
slice method for studying drug distribution in the CNS. Flu- E.T.; Steinman, L.; Rothbard, J.B. The design, synthesis,
ids Barriers CNS, 2013, 10, 6. and evaluation of molecules that enable or enhance cellular
[127] Friden, M.; Ducrozet, F.; Middleton, B.; Antonsson, M.; uptake: peptoid molecular transporters. Proc. Natl. Acad.
Bredberg, U.; Hammarlund-Udenaes, M. Development of a Sci. U. S. A., 2000, 97, 13003-13008.
high-throughput brain slice method for studying drug distri- [139] Stanzl, E.G.; Trantow, B.M.; Vargas, J.R.; Wender, P.A.
bution in the central nervous system. Drug Metab. Dispos., Fifteen Years of Cell-Penetrating, Guanidinium-Rich Mo-
2009, 37, 1226-1233. lecular Transporters: Basic Science, Research Tools, and
[128] Chen, H.; Winiwarter, S.; Friden, M.; Antonsson, M.; Eng- Clinical Applications. Acc. Chem. Res., 2013, 46, 2944-
kvist, O. In silico prediction of unbound brain-to-plasma 2954.
concentration ratio using machine learning algorithms. J. [140] Lalatsa, A.; Schatzlein, A.G.; Uchegbu, I.F. Strategies to
Mol. Grpah. Model., 2011, 29, 985-995. deliver peptide drugs to the brain. Mol. Pharm., 2014, 11,
[129] Spreafico, M.; Jacobson, M.P. In silico prediction of brain 1081-1093.
exposure: drug free fraction, unbound brain to plasma con- [141] Arvanitis, C.D.; Livingstone, M.S.; Vykhodtseva, N.;
centration ratio and equilibrium half-life. Curr. Top. Med. McDannold, N. Controlled ultrasound-induced blood-brain
Chem., 2013, 13, 813-820. barrier disruption using passive acoustic emissions monitor-
ing. PLoS One, 2012, 7, e45783.

You might also like