SlideShare a Scribd company logo
1 of 13
Download to read offline
Section 1
Chapter
7
Principles of drug action
Drug transport and transporters
Roland J. Bainton and Evan D. Kharasch
Drug transporters and localized
pharmacokinetics
Xenobiotics and defining chemical space
Living organisms use biologic compartments to perform dis-
crete biochemical processes, to localize and control the natural
small molecules contained within [1]. Foreign small molecules,
called xenobiotics, share the same molecular weight range and
solubility characteristics as endogenous small molecules and are
thus able to penetrate biologic spaces. As xenobiotics also pos-
sess potent biointeraction (i.e., drug-like) properties they are the
source for nearly all clinically useful pharmaceuticals [2]. To
balance the cells’ needs of access to endogenous small molecules
and control of xenobiotic penetration, powerful biotransforma-
tion and bioelimination pathways have evolved to control
chemical space (Chapter 6). These same processes present chal-
lenges for delivering clinically useful therapies to particular
organ spaces, as xenobiotics have complex and unpredictable
relationships with the selectively expressed small molecule
transporters or specially tuned metabolic systems [2]. Thus
understanding how any one pharmaceutical interacts with bio-
logically defined chemical space requires a closer examination
of that drug’s susceptibility to highly localized pharmacokinetic
processes affecting chemical persistence at the site of action.
Lipid bilayers and drug localization
Lipid bilayers physically partition aqueous spaces [1] (Fig. 7.1),
creating cells and specialized internal subcellular compartments.
In addition, formation of cell–cell contacts in conjunction with
diffusion-tight junctional complexes divides the external cellular
milieu into different interstitial spaces [1] (Fig. 7.2). This bio-
physical boundary prevents passive diffusion of large molecules
such as proteins and nucleic acids between different compart-
ments and greatly curtails the movement of electrolytes, soluble
components of metabolism, and drugs. Drugs and xenobiotics
are often in equilibrium between charged and charge-neutral
states (Fig. 7.2). Charge-neutral molecules more readily transit
lipid bilayers and possess better penetration rates into cells by
orders of magnitude (passive permeability, Fig. 7.1). Cellular
penetration may be augmented (influx transport) or opposed
(efflux transport) by active cellular processes (see below), a fea-
ture that allows drugs to take multiple routes to a target (Fig. 7.2).
To control the ability of drugs to transit freely in cellular space
and still maintain chemical partition, cells organize molecular
systems to contain, modify, or move endogenous and foreign
lipophilic molecules from one space to another.
Small-molecule partition and transporters
Chemical partition is the sine qua non of cell biology, and cellular
transport systems play a vital role in chemical compartmental-
ization. An enormous variety of transmembrane protein trans-
porters are dedicated to selectively moving small molecules
across membranes [3]. Their role is so broad that by some
estimates 15–30% of all membrane proteins are transporters
[1]. Among the many substrates that transmembrane protein
transporters relocalize are glucose, anabolic intermediates, amino
acids, neurotransmitters, metabolic products, and lipids [3]. The
vast majority of transporters have high substrate specificity and
are organized by DNA sequence relatedness and known function.
They are referred to as solute carrier (SLC) proteins and include
46 different gene families with over 360 genes in humans [3].
Their enormous diversity speaks to the wide variety of intracel-
lular environments and their highly specific needs.
The precision, specificity, and ability of these transport
systems to act against concentration gradients allows small mol-
ecules to have discrete functions depending on their localization.
For example, plasma membrane transporters of the SLC6 class
specialize in the transport of monoamines from the interstitial
space to the cytoplasm. These transporters perform two key
functions for the cell: (1) providing amino acids to the cytoplasm
for metabolic purposes, and (2) reducing extracelluar concen-
trations of monoamines when unwanted [4]. However, when the
same molecule (or close derivative) is to be used as a
neurotransmitter, the SLC18 transporters highly concentrate
and package the molecule into synaptic vesicles. After release
into the extracelluar space for signaling at the postsynaptic
membrane, neurotransmitters are rapidly taken up by SLC6 class
Anesthetic Pharmacology, 2nd edition, ed. Alex S. Evers, Mervyn Maze, Evan D. Kharasch. Published by Cambridge University Press. # Cambridge University Press 2011.
90
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
transporters. Moving small molecules across cell membranes is
entropically unfavorable, and thus transporters require energy
expenditure either directly using adenosine triphosphate (ATP),
or dissipating other existing electrochemical gradients (e.g., Na/
K) by using symport or antiport mechanisms [3]. Together,
transporter systems create the chemical preconditions for cellu-
lar function, restore chemical conditions after perturbation, and
clear chemical space of unwanted metabolites. Thus transporters
are essential actors in almost every biologic function.
Localized pharmacokinetic spaces
Acting in concert with, or after the action of, phase I and
phase II metabolism (Chapter 6), to move drugs and their
metabolites across lipid bilayers, transporters are often
referred to as phase III metabolism. Therefore transporters
are often highly expressed in organs of biotransformation such
as the liver and intestine, and metabolism and transport
together create formidable pharmacokinetic barriers. Different
organ systems tune their biodefense systems by locally
applying phase I, II, and III mechanisms [5]. Thus, for a drug
to be clinically effective, it must possess high target specificity,
favorable organismal pharmacokinetics, and complementary
properties for highly localized chemical partition processes at
the cellular level. It is in this latter context that the special role
of drug transporters and their ability to control xenobiotic
chemical space in drug action are considered.
Drug transporters
Discovery of drug efflux transporters
Xenobiotic transporters were identified as a consequence of the
study of multidrug resistance to chemical cancer treatment in
the 1970s [6,7]. Highly efficient cytotoxic resistance to a broad
spectrum of cancer drugs could occur suddenly and unexpect-
edly after initiation of chemotherapy [8]. Analysis of drug-
resistant tumor cells determined that this new-found resilience
correlated with upregulation of a few transmembrane proteins,
suggesting that chemotheraputic protection systems were quite
nucleus
hydrophobic
small/uncharged
polar
large/uncharged
polar
ions
lipid bilayer
intracellular space
extracellular space
~10–7 cm/sec ~10–11 cm/sec
~10–4 cm/sec
>10–2 cm/sec
Figure 7.1. Chemical partition across bilayers.
Lipid bilayers (green lines) are formed by virtue of
the chemical characteristics of their constituent
phospholipids: a hydrophilic phosphate head group
and a hydrophobic carbon-chain tail. The head
groups associate with the aqueous environment,
while the tail groups self-associate and form the
internal, hydrophobic core of the bilayer. This layer
must be transited if nutrients as well as drugs are to
reach targets inside the cell, and movement across
this bilayer is also dictated by specific chemical
properties. Paracellular movement refers to passage
of molecules between cells, bypassing cell mem-
brane barriers. Transcellular movement requires pas-
sage across membrane barriers. More hydrophobic
chemicals and compounds can cross the barrier
with relative ease and have a high permeability
coefficient (expressed in the figure as cm s–1
)
whereas chemicals with low permeability coeffi-
cients (such as ions) cannot passively transit the lipid
bilayer and must cross via other means such as
carrier-mediated transport. All chemicals critical to
sustaining life fall along this continuum. Modified
with permission from Alberts et al. [1].
( )+/– ( )0
Transcellular
Paracellular
Drug
Transporters
(Ionized Form)+/– (Non-ionized Form)0
Junctional
Complexes
Interstitial Space
(vascular)
Interstitial Space
(organ)
Figure 7.2. Chemical partition across cellular
boundaries. Cells separate one interstitial space
from another. Cellular barriers rely upon two modes
of exclusion to maintain xenobiotic exclusion, as
small molecules can interact with cells in two ways.
Charged molecules (blue stars) are excluded by the
boundary function of lipid bilayers, very low rates of
endocytosis, and tight diffusion barriers provided by
special lateral-border junctional complexes (blue
ovals) of barrier epithelium. Small uncharged
molecules (red stars) pass easily through lipid
bilayers (green), but active efflux transporters (red)
move them back into the humoral space, creating
an active transport barrier. As drugs in aqueous
solution are in equilibrium between charged and
uncharged forms, a true xenobiotic barrier must
maintain both properties simultaneously to mani-
fest chemical exclusion.
Chapter 7: Drug transport and transporters
91
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
different from the highly specific SLC transporter systems dis-
cussed above. Ultimately these genes were identified as active
drug efflux pumps that, unlike most SLC transporters, had
surprisingly broad substrate specificity. These proteins were
found to bind and transport a wide spectrum of chemothera-
peutics back into the extracellular space, thus reducing the
effective internal concentration of cytotoxic drugs [8]. Interest-
ingly, these cells were not intrinsically more resistant to che-
motherapeutics at the target sites, but had instead coopted and
enhanced an endogenous chemical partition system to
strengthen the chemical barrier at the membrane [8].
Whole genome sequencing methods have discovered large
families of similar multidrug resistance transporter (MDR)
genes in nearly all organisms [9,10]. Most of these proteins
belong to a large and highly conserved class of transmembrane
proteins known as ATP-binding cassette (ABC) transporters
[8,11]. The archetypal ABC transporter contains two six-pass
transmembrane domains and two cytoplasmic nucleotide-
binding domains, although the proteins do vary structurally
depending on their class (Fig. 7.3). Full transporters are large
proteins with 1200–2200 amino acids, and half-transporters
have 650–800 amino acids. Half-transporters are thought to
function as dimers in the membrane, potentially generating
additional functional heterogeneity by combining different
substrate preference into one transport pocket [8].
Unfortunately, unlike other transport systems where
ligand binding is well understood, only a few specifics are
known about the mechanism of xenobiotic binding to ABC
transporters. Efficient efflux, for example, is thought to result
from conformational changes of the transporter which allow
an alternating high substrate-affinity (cell-facing)/low
substrate-affinity (extracellular-facing) organization [8]. This
permits intracellular presentation of the drug at one interface
and release (or efflux) into the extracellular milieu on the
other side. Evidence is mounting that drug binding to the
major substrate-promiscuous xenobiotic transporters occurs
in the membrane and not from the solution. Thus, these
transporters may be designed to act at a protein/lipid-mem-
brane interface [8].
The ABC gene family
The approximately 50 human ABC genes occur in seven
families (A through G), although only a subset are known
to play major roles in drug transport. The major xenobiotic
efflux transporter familes are the B, C, and G gene classes
(Table 7.1). Interestingly, while ABC gene sequence and sub-
families are highly conserved from yeast to flies to humans,
the specific roles of most gene products are unknown. ABC
transporter localization in polarized cell interfaces is essential
to chemoprotective function, as net flux of xenobiotics into or
away from various interstitial spaces is determined by the
type, quantity, efflux or influx orientation, and apical/basal
orientation at the protective interface (e.g., the vascular endo-
thelium of the blood–brain barrier, as in Figs. 7.4 and 7.5)
[8,13]. Unfortunately as the best studies of ABC transporters
in the animal are limited to a few genes (see below), it is not
known whether or how they function as a complementary
system in the biology of chemical exclusion.
Important subtypes of drug
efflux transporters
ABCB1 (P-glycoprotein)
P-glycoprotein (P-gp, ABCB1, or the multidrug resistance
transporter MDR1) was the first xenobiotic transporter
discovered in multidrug-resistant cancer cells [7] and is
Domain 2
Extracellular
Intracellular
Extracellular
Intracellular
Extracellular
Intracellular
NBD NBD
C-Terminus
N-Terminus
NBD
NBD
C-Terminus
N-Terminus
NBD
N-Terminus
Lipid Bilayer
Lipid Bilayer
Lipid Bilayer
C-Terminus
Examples
MDR1 (ABCB1)
MRP4 (ABCC4)
MRP5 (ABCC5)
MRP7 (ABCC1)
BSEP/SPGP (ABCB11)
MRP1 (ABCC1)
MRP2 (ABCC2)
MRP3 (ABCC3)
MRP6 (ABC6)
MXP/BCRP/ABC-P
(ABCG2)
Domain 1 Figure 7.3. Membrane topologies of known xeno-
biotic transporters. (Top) P-gp-like ABC transporters
have 12 transmembrane (TM) domains and two
nucleotide-binding domains (NBD). (Middle) In the
ABCC class a second membrane topology preserves
the core P-gp-like transporter with an additional five
TMs on the N-terminus. (Bottom) The half-transport-
ers of the ABCG class have six TMs and one NBD.
They are thought to dimerize to promote drug
transport function. Modified with permission from
Gottesman et al. [12].
Section 1: Principles of drug action
92
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
responsible for the most profound effects on localized xeno-
biotic protection of all the ABC transporter subfamily
proteins. P-gp is a twelve-pass transmembrane protein of
140 kD and is highly expressed at important chemoprotec-
tive interfaces (Table 7.2). It has the broadest substrate
specificity of the ABC genes, transporting a significant
number of drugs, including anticancer drugs, HIV-protease
inhibitors, antibiotics, antidepressants, antiepileptics, and
opioids such as morphine and loperamide (Table 7.2).
P-gp substrates are generally amphipathic and range in size
from 300 to 2000 Da.
P-gp also functions in a wide range of environments
including the GI tract, renal proximal tubule cells, the
canalicular surfaces of hepatocytes, the capillary endothelial
cells of testes, and the blood–brain barrier. Thus the poten-
tial for significant drug interactions is widespread [8]. For
example, oral bioavailability of the anti-inflammatory drug
cyclosporine A is greatly increased by amlodipine, a P-gp
substrate that doubles as a P-gp inhibitor [8]. Another
example is loperamide, an opioid antidiarrheal that pro-
duces no notable central nervous system (CNS) effects at
normal doses because intestinal P-gp-mediated efflux retards
systemic absorption [8]. However, when loperamide is given
with the P-gp inhibitor quinidine, significant systemic
absorption occurs, CNS delivery ensues, and side effects
such as respiratory depression arise [16]. Thus localized
transporter interactions can have substantial effects on drug
bioavailability. This is particularly pertinent in fentanyl
pharmacodynamics, based on the association between P-gp
polymorphisms and fentanyl respiratory depression [17].
In patients under spinal anesthesia given intravenous
fentanyl (2.5 mg kg–1
), there are differences in suppression
of ventilatory effort between three distinct P-gp genotypes
prominent in ethincally Korean populations. The presump-
tion is that P-gp transports fentanyl at CNS barriers, and
therefore P-gp polymorphisms significantly alter brain
concentration despite equivalent doses. Thus, a highly local-
ized drug partition phenomenon can affect drug responses,
but they are dependent on the relationship of the specific
interface (i.e., the blood–brain barrier) and the protected
localization of the drug target (i.e., neurons containing
opioid receptors).
ABCC transporters (MRPs)
Multidrug resistance proteins (MRPs) are the largest class
of ABC transporters relevant to drug resistance and drug
distribution and are ubiquitously expressed [18]. There are
12 proteins in this class with two distinct types of transmem-
brane protein topologies (Fig. 7.3). Although they contain
only 15% sequence identity to P-gp they are predicted to
posses a similar structure, with two nucleotide-binding
domains and similar membrane topology to ABCB transport-
ers (Fig. 7.3) [12]. Furthermore, like P-gp, they can extrude a
wide array of amphipathic anions. Interestingly, all members
of this particular subfamily, also called ABCC transporters,
are lipophilic anion transporters that are broadly involved in
cancer therapeutic resistance [18]. They are quite adept at
effluxing methotrexate, thereby lessening the effect of
an important chemotherapeutic in the brain [8]. MRP sub-
strate specificities show a preference for glutathione and
glucuronide conjugates, thus drug elimination could be
greatly facilitated when transporters are locally paired with
the appropriate phase II enzymes (e.g., glucuronyl trans-
ferases, Chapter 6). Nevertheless MRPs also efficiently trans-
port unconjugated compounds such as anthracyclines,
Table 7.1. ABC transporter genes are highly conserved. Pairwise
analysis using human ABC transporters versus Drosophila using NCBI
BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi) demonstrates high
conservation of transporter number per class and high identity among
classes. Similar results are obtained from comparison with other
metazoan genomes, and thus ABC genes are highly evolutionarily
conserved and likely to play fundamental roles in cellular biology. The
major xenobiotic transporters are from the B, C, and G classes, but
A-class transporters may play roles in some cases.
Subfamily Human Drosophila Identities
(%)
Positives
(%)
A 12 10 20–37 34–56
B 11 8 43–57 59–75
C 13 14 23–50 40–67
D 4 2 56–59 71–74
E 1 1 77 88
F 3 3 41–69 56–82
G 5 (þ1) 15 22–54 41–71
Total 50 53
Vascular
Space
A
VE
P
CNS
Interstitial
Space
Figure 7.4. Organization of the blood–brain barrier (BBB). While chemical
protection is focused at discrete epithelial layers, surrounding tissue can have
essential roles in molding and assisting the localized PK potential. This is
particularly true at the BBB, where properties of the vascular endothelium (VE)
are induced and controlled by surrounding tissues. The BBB VE is thought to
receive inductive cell-fate signals from the CNS parenchyma (not shown), while
astrocytes (A) and pericytes (P) assist in and regulate localized responses to
chemical and immune stress.
Chapter 7: Drug transport and transporters
93
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
epipodophyllotoxins, vinca alkaloids, and camptothecins
(Table 7.2) [18]. MRPs are known to localize to both apical
and basal interfaces of chemical protection interfaces and thus
can participate in moving drugs in both directions between
interstitial spaces (Fig. 7.5) [8,13].
ABCG2 (BCRP)
Breast cancer resistance protein (BCRP), so named for its
discovery in an especially drug-resistant breast cancer cell line
[21], is the second member of the five-member
half-transporter ABCG subfamily (Fig. 7.3) [10]. BCRP is
expressed throughout the body but most notably in the
liver, intestinal epithelium, placenta, blood–brain barrier, and
a variety of stem cells. Like P-gp, BCRP is quite promiscuous
in its choice of substrate and is powered by the hydrolysis of
ATP. BCRP substrates generally include compounds that are
relatively large in molecular weight and either positively or
negatively charged and amphipathic. Specific substrates for
BCRP-mediated efflux include antineoplastic drugs and their
metabolites (e.g., methotrexate and sulfated methotrexate),
Table 7.2. Major human drug transporters P-glycoprotein (P-gp), multidrug resistance protein (MRP), breast cancer resistance protein (BRCP), organic
anion transporting polypeptide (OATP), organic cation transporter (OCT), and organic anion transporter (OAT) are the best-studied drug transporters. They can
share substrates and inhibitors. Modified with permission from Loscher and Potschka [13], Dobson and Kell [14], Zhang et al. [15].
Transporter
gene
Aliases Localization Function Selected substrates Selected inhibitors
ABC family
ABCB1 P-glycoprotein,
MDR1
BBB, BRB, intestine, liver,
kidney, placenta, adrenal,
testis
efflux digoxin, fexofenadine, morphine,
loperamide, corticoids,
cyclosporine, paclitaxel,
verapamil, methotrexate
cyclosporine,
verapamil,
quinidine, ritonavir,
ketocoanzole,
itraconazole
ABCC1 MRP1 BBB, breast, lung, heart,
kidney, blood cells, liver,
intestines, testes, placenta
efflux glutathione or glucuronide
conjugates, leukotriene C4,
etoposide, methotrexate
probenecid,
sulfinpyrazone,
cyclosporine
ABCC2 MRP2 intestine, liver, kidney,
brain, placenta
efflux bilirubin, indinavir, cisplatin cyclosporine
ABCG2 BCRP BBB, intestine, liver,
breast, placenta
efflux anthracyclines, mitoxantrone,
prazosin, topotecan, methotrexate
elacridar (GF120918),
cyclosporine,
fumitremorgin C
SLCO family
SLCO1A2 OATP1A2 Liver, kidney, brain, lung,
colon
uptake,
efflux
fexofenadine, rocuronium,
enalapril, rosuvastatin
SLCO1B1 OATP1B1 BBB, brain, kidney, liver uptake,
efflux
organic anions, bile salts, digoxin,
fexofenadine, methotrexate,
rifampin
cyclosporine,
verapamil, rifampin,
similar to P-gp
SLC family
SLC22A1 OCT1 liver uptake metformin quinidine, quinine,
verapamil, cimetidine
SLC22A2 OCT2 kidney, brain uptake metformin, amantadine,
memantine, varenicline
cimetidine
SLC22A6 OAT1 kidney, brain uptake acyclovir, adefovir, methotrexate,
zidovudine
probenecid,
cefadroxil,
cefamandole,
cefazolin, diclofenac
SLC22A8 OAT3 kidney, brain uptake cimetidine, methotrexate,
zidovudine
probenecid,
cefadroxil,
cefamandole,
cefazolin, cimetidine
BBB, blood–brain barrier; BRB, blood–retinal barrier.
Section 1: Principles of drug action
94
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
topotecan and irinotecan (both topoisomerase I inhibitors), as
well as mitoxantrone, camptothecin and flavopiridol, xenobio-
tic toxins, food-borne carcinogens, and endogenous com-
pounds [8]. BCRP, unlike MRPs, is also adept at transporting
glutamated folates and folate antagonists [8]. Therefore, like
P-gp, BCRP is a major factor in conferring resistance to a wide
range of therapeutics and is central to pharmacokinetics and
toxicity considerations [8].
Other drug transporters
SLC21 transporters (organic anion
transporter proteins)
Despite the relative abundance of SLC transporters at protect-
ive barrier interfaces, few are known to participate directly in
drug transport. One exception is the organic anion transporter
proteins (OATPs), which are not structurally related to ABC
transporters, but belong to the solute carrier transporters class
21 (SLC21/SLCO) [3]. OATPs primarily use passive transport
mechanisms driven by electrochemical gradients or symport/
antiport mechanisms, but interestingly show other functional
similarities to P-gp. The best-characterized OATP xenobiotic
transporters are OATP1A2, OATP1B1, OATP1B3, and
OATP2B1 [14,15]. They are generally efflux transporters,
found at chemoprotective interfaces, prominently in brain
endothelial cells, and also in liver, lung, kidney, and testes.
In-situ mRNA hybridization also demonstrates broad expres-
sion in central brain tissue, suggesting a role in general cellular
protection as well as barrier function. In-vitro experiments
suggest that OATPs can transport many structurally divergent
molecules, reminiscent of P-gp. Examples include bile acids,
Intestine
MCT1 OATP-B OCTN2 MDR1 MRP2
Blood-Cerebrospinal
Fluid Barrier
Blood
Blood-Brain Barrier
others?
MCT1
MRP1/2//4/5/6
OAT3
OCTN1/2
MDR1
BCRP
PEPT1/2
OAT4
NPT1
URAT1
OCTN1/2
MDR1
MRP2/4
Kidney
others?
OCT2
OAT1/3
MRP3/6
Liver
NTCP
OATP-B/C/8
OAT2
NPT1
MRP1/3/6
others?
others?
MRP2
BCRP
MDR1
OCT3
MRP1/3
Placenta
PEPT1 OCT3 ASBT
MRP3
MCT1
OATP-A
OST α/β
others?
BCRP
?
MDR1
BSEP
BCRP
MRP2
PEPT2 OCT2
others?
MRP1
Figure 7.5. Membrane localization of drug transporters at the blood–brain barrier. Organ spaces subject to specific chemical constraints usually organize the
tightest control of chemical space at one cellular layer. These chemical barriers use tight junctions and transporter content, type, and apical/basal localization to
modulate the protected interstitial space to the purpose and susceptibilities of the underlying tissue [19,20]. The vascular endothelium of the BBB is one of the
most potent localized pharmacokinetic interfaces and has a vast array of drug efflux and influx transporters at both vascular and CNS interfaces. Similar
organization is repeated at the blood–cerebrospinal fluid barrier, renal epithelium of the kidney, intestinal epithelium, hepatocyte/bile-duct interfaces, and
placenta; thus drug action at an individual organ can depend greatly on the chemoprotective efficiency of a particular exposure interface. For transcellular
transport, drugs need to cross two different membranes on the basal and apical sides. In the intestine, drugs are absorbed from the luminal side (brush border
membrane) and excreted into the portal blood across the basolateral membrane. In the liver, drugs are taken up into hepatocytes across the sinusoidal membrane
and excreted into the bile. In the kidney, drugs undergo secretion (urinary excretion) or reabsorption. In the case of secretion, drugs are taken up from the
basolateral side and excreted in the urine across the apical side. However, in the case of reabsorption, drugs are taken up from the urine and excreted in the
circulating blood. Reproduced with permission from Tsuji [19].
Chapter 7: Drug transport and transporters
95
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
steroids, estrogen, many anionic compounds, and glutathione
or glucuronide conjugates [22]. Certain drug–food inter-
actions (e.g., grapefruit juice) [23] may be attributable to
inhibition of OATPs, but further characterization of the in-
vivo role of OATPs in drug interactions is needed. Interest-
ingly, unlike ABC transporters, some OATPs can also function
as drug influx transporters (Fig. 7.4). Bidirectional transport
and/or the counteracting of efflux transport occurs at numer-
ous tissue interfaces including the gut, liver, kidney, and
blood–brain barrier [19,20]. How these competing transporter
processes are regulated, coordinated, and potentially circum-
vented or controlled will require a better understanding of
drug–transporter interactions at cellular interfaces and
methods for analyzing net localized pharmacokinetic dispos-
ition of drugs.
Specially protected anatomic
spaces
Specialized cellular boundary systems have evolved to limit
access to sensitive biologic spaces [12,24]. Transporters are
critical to the processes of drug uptake, distribution, metabol-
ism, and excretion and may play multiple roles simultaneously
depending on their localization, expression levels, and activity.
Transporters are especially important in highly sensitive
organs such as the CNS, where they are important determin-
ants of drug pharmacodynamics [25].
CNS drug barriers
Blood–brain barrier
The blood–brain barrier (BBB) is the largest pharmacokinetic
interfacewithintheCNS(about20m2
)[26],andthustheprimary
focusofthe manyefforts to understand and controldrugaccess to
the brain [27]. The BBB evolved due to the brain’s unique chem-
ical sensitivity and requirement for the highest level of chemical
protection. BBB drug transporters can profoundly influence the
clinical effectiveness of drugs acting in the brain [13].
The BBB has a complex cellular makeup that is functionally
integrated with surrounding brain tissue as part of the neuro-
vascular unit [11,28]. The complete neurovascular unit com-
prises the capillary vascular endothelium, pericytes, a basement
membrane, closely associated astrocytic glia, and nearby
neurons [29] (Fig. 7.4). The highly polarized vascular endothe-
lium is the primary site of chemical exclusion. It possesses very
tight lateral border junctions [30] and a diverse array of apically
(i.e., vascular) facing efflux drug transporters, including P-gp,
MRP1, and BCRP (Fig. 7.5). The functional importance of
these transporters to drug partition in the brain has been
studied using gene-specific genetic null mice (knockouts)
[31–35]. In single transporter loss-of-function experiments,
two- to ten-fold increases in CNS penetration of specific ABC
transporter substrates including chemotherapeutics can result
after IV administration [36]. That ABC transporters can so
profoundly affect brain drug concentration by their presence
or absence in a single endothelial layer suggests a plausible
mechanism for increased drug sensitivity of the CNS in trans-
porter-associated pharmacogenomic studies [17] (see P-gp,
above). Furthermore these studies promote the idea of specif-
ically targeting xenobiotic transporter function to more readily
concentrate drugs in the brain for treating CNS illnesses such as
brain cancer, multiple sclerosis, and Alzheimer’s disease [27].
Astrocytic glia form a continuous circumferential and close
association with the basement membrane, further isolating the
vascular and CNS interstitial spaces, but their role in chemical
exclusion is unclear. The astroglial cell layer is less than a
micron thick and lacks very tight junctions at cell–cell contacts,
and xenobiotic transporters should therefore not directly affect
CNS glial cell chemopartition. Indeed, genomic data show a
much lower level of xenobiotic transporter and junctional pro-
tein expression in the astroglia compared to vascular endothe-
lium (R. Daneman and B. Barres, personal communication and
[37]). Thus, while astrocytes are essential for BBB maintenance
and function, their role in chemical protection may be to sense
or integrate chemical, metabolic, and inflammatory stresses
which must be coordinated to maintain proper physiologic
balance for neurons [11]. Substantial efforts are under way to
develop model systems and new methods to identify regulatory
controls and chemoresponsive physiologies in the hope of
specifically modulating drug barrier function at the BBB and
ultimately improving clinical treatment options [27].
Blood-retinal barrier (BRB)
Transport of molecules between the vitreous/retina and sys-
temic circulation is restricted by the blood-retinal barrier
(BRB, also called the retinal BBB or RBBB), composed of
retinal pigment epithelium (RPE) and endothelial cells of the
retinal blood vessels. The RPE maintains a tight barrier to
diffusion to the interstitial fluid of the surrounding connective
tissue of the eye. Vascular access to the retina is primarily
through the retinal blood vessels, which penetrate with the
optic nerve and run parallel to the retinal surface. The capillary
bed of the retinal vascular endothelium maintains tight junc-
tions and apically facing transporters morphologically similar
to the BBB in the central brain. A unique feature of the eye
is the ability to bypass the barrier by topical application or
injection into the vitreous humor. However, while drug
delivery to the CNS is uniquely possible in the eye, transport
processes resident at the RBBB promote very rapid removal of
small molecules into the systemic circulation. For example,
common topical ophthalmic drugs such as erythromycin, dex-
amethasone, and cyclosporine are substrates for prominent
RBBB transporters and can be found in the systemic circula-
tion after ocular administration [38]. These transporters
include MDR1, MRP1, BRCP, and numerous OATPs [38].
Localized pharmacokinetic methods for measuring efflux
across the BRB are limited; therefore reliable quantitative
Section 1: Principles of drug action
96
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
estimation of the roles of inner and outer BRB has not been
well established in humans. In rabbits, however, MDR1 pre-
sent at the RBBB can be selectively modulated using traditional
inhibitors such as quinidine and cyclosporine A [39]. Further-
more, diffusion barrier studies have been carried out in mice,
where labeled mannitol is used as a biomarker for retinal
diffusion barrier integrity [40] and has shown diffusion-tight
function of the BRB. Thus experimental models identify the
BRB as an effective barrier similar to the BBB.
Blood–cerebrospinal fluid barrier and choroid plexus
The blood–cerebrospinal fluid barrier (B-CSFB) is derived
from a special confluence of the pia mater and a single layer
of CSF-facing glial cells (the ependyma). This meeting of two
barrier layers is known as the choroid plexus and is the site of
CSF production. Throughout the rest of the CNS, ependymal
cells become diffusion-tight, utilizing the same modes of sep-
aration discussed at the capillary–brain interface above. Here,
therefore, chemical exclusivity of the CNS is maintained by an
anatomically simpler system than at the BBB. The CSF pro-
duction cycle is completed by rapid uptake at arachnoid granu-
lations. Together, the CSF production and brain barrier system
is thought to perform cleansing functions, further augmenting
the chemical protection provided by the BBB [41].
Because the diffusion of polar substances at the B-CSFB is
less hindered than at the BBB [41], drug administration into
CSF has been proposed to circumvent the BBB and gain access
to the CNS. However, potent chemical exclusion properties are
present at the B-CSFB (i.e., ependymal cells), as at the choroid
plexus [42]. Specifically, transporters OAT, OATP1, OATP2,
MRP1, and MDR1 are present in high levels, suggesting an
explanation for why the CSF has not proven to be a depend-
able access point for the administration of chemotherapy or
other drugs into the CNS. Nevertheless, certain drugs can
overcome these barrier processes by a combination of favor-
able penetration chemistry and poor transportability. For
example, sodium-channel blockers such as bupivicaine are
used for subarachnoid block.
Gastrointestinal tract
Intestinal drug uptake usually occurs by mass action and
passive permeability when high concentrations are presented
to the microvillus absorption surface. An empirical set of rules,
known as Lipinski’s Rule of Fives, describes how small mol-
ecules passively cross the lipid bilayer. Oral drugs can violate
only one of the following criteria: (1) not more than five
hydrogen bond donors, (2) not more than 10 hydrogen bond
acceptors, (3) molecular weight under 500 Da, and (4) an
octanol/water partition coefficient log P less than 5. Pharma-
ceuticals that possess these properties are better at passing the
chemoprotective interface of the gut, but these constraints do
not allow small molecules to bypass the action of transporters.
Xenobiotic transporters are abundantly represented
throughout the enteral space (Fig. 7.5) [43,44]. P-gp is apically
localized in microvilli throughout the gut epithelium, found in
highest amounts in the ileum and jejunum, and counteracts
the passive or active absorption of xenobiotics [25]. For
instance, mouse P-gp knockouts demonstrate increased
absorption and bioavailability of many oral xenobiotics [45],
and uptake of some drugs is markedly increased by small
molecules that inhibit P-gp function [16]. BCRP is the trans-
porter expressed in highest abundance throughout the human
small intestine [46]. In addition to countering the absorption
of some orally administered drugs, certain transporter systems,
particularly ABCC proteins, counter the absorption of drug
glucuronides that are excreted in bile. Intestinal efflux trans-
porters act in concert with intestinal phase I metabolism to
limit drug bioavailability.
Liver
Hepatic drug elimination depends on passive or active hepatic
uptake, intracellular metabolism, excretion of metabolites or
parent drug into bile, and/or excretion of metabolites or parent
drug into sinusoidal blood. Transporters are involved in
uptake, biliary excretion, and sinusoidal efflux (Fig. 7.5) [23].
SLC-type transporters in the basolateral (sinusoidal) mem-
brane (OATPs, OATs, OCTs) mediate the uptake of organic
cations, anions, bilirubin, and bile salts. ABC transporters
(P-gp, BCRP, MRPs) at the canalicular membrane mediate
the efflux into bile of drugs, metabolites, and bile salts. MRPs
may also mediate drug and metabolite efflux back into the
blood. The balance of vectorial uptake and efflux drug trans-
port together with intracellular metabolism, determines drug
elimination. Hepatic drug transporters are subject to pharma-
cogenetic variability, and drug–drug interactions.
In murine models, transporter disruption can effect the
distribution of important anesthetic drugs such as opiates.
Morphine-3-glucuronide (M3G), the most abundant metab-
olite of morphine, is transported in vitro by human MRP2
(ABCC2) that is present in the apical membrane of hepato-
cytes. Loss of biliary M3G secretion in MRP2(-/-) mice results
in its increased sinusoidal transport, which can be attributed
to MRP3. Combined loss of MRP2 and MRP3 leads to a
substantial accumulation of M3G in the liver, resulting in
low rates of sinusoidal membrane transport and the pro-
longed presence of M3G in plasma [47]. These results suggest
that in vertebrates MRP2 and MRP3 transporters provide
alternative routes for the excretion of a glucuronidated sub-
strate from the liver in vivo. This illustrates the potential
complexity of interactions between transporters and modify-
ing enzymes intrinsic to particular chemoprotective
environments.
Kidney
Transporters mediate active tubular secretion and reabsorp-
tion in the kidney, which, together with glomerular filtration,
determine drug elimination (Fig. 7.5). The primary focus of
renal excretion is the proximal tubule, which actively takes up
organic anions and cations at the basolateral membrane and
Chapter 7: Drug transport and transporters
97
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
extrudes them across the apical brush border membrane into
the urine; hence transepithelial flux requires two distinct sets
of transporters. Many drugs (weak acids) and their phase II
conjugates are organic anions. OATs 1 and 3 are the primary
organic anion uptake transporters at the basolateral mem-
brane (although they may also interact with neutral and
cationic compounds), while efflux into the tubular lumen is
mediated primarily by the ABC subfamily C transporters
MRP2, MRP4, and BCRP [48]. Organic cations are typically
transported into renal tubular cells at the basolateral mem-
brane by OCTs 1 and 2, and effluxed into urine at the apical
membrane by P-gp, OCT3, and novel organic cation trans-
porters OCTN1 and OCTN2. Selective expression and spe-
cific localization of transporters at different apical and basal
interfaces in the tubule, pharmacogenetic variability in their
activity, and drug interactions play a determinative role in
drug elimination.
Transporter-based drug interactions can occur in the
kidney by competing for elimination. For example, probencid
inhibits OATP-mediated excretion of antibiotics [49]. In add-
ition, methotrexate has strong potential interactions with non-
steroidal anti-inflammatory drugs (NSAIDs), particularly after
prolonged high-dose chemotheraputic use [49].
Placenta
The placenta is the sole medium of metabolic exchange
between fetus and mother; hence it protects the fetus by
providing a major obstacle to chemical penetration. The
important chemical properties determining placental transfer
by passive diffusion are similar to other organs: molecular
weight, pKa of drug, lipid solubility, and plasma protein
binding. The transport barrier in placenta is found in the
syncytiotrophoblast, a multinucleated single cell layer that
performs nutrient and waste exchange between mother and
fetus [50]. This layer is further elaborated into two distinct
interfaces: a maternal-facing brush border and a fetal-facing
basal membrane [50].
It is in these two interfaces that a variety of membrane
transporters perform directional fluxing of metabolites and
drugs. Specifically, the brush border membrane contains all
three major classes of ABC transporters (i.e., P-gp, MRP1–3,
and BCRP) as well as specialized transporters such as the
serotonin, norepinephrine, and carnitine transporters (SERT,
NET, OCTN2) [50]. On the other side of the divide, the fetal-
facing basal membrane contains transporters such as the
organic anion transporters OAT4 and OATP-B in addition
to the reduced folate transporter RFT-1 [50]. Combinations
of genetic loss of P-gp function with specific inhibitors of
BCRP markedly increase drug penetration of topetecan to the
fetus, which is not seen in the genetically sensitized back-
ground alone. Thus the placenta is an essential chemical filter
for the fetal protected space and exemplifies the potential for
overlapping and redundant function of chemoprotective
mechanisms [51].
Transporter biology: special topics
Drug transporter methods
The field of transporter biology is relatively new, and a brief
description of experimental methods used to identify the role
of transporters in drug disposition may be helpful. Accurate
physical measurement of localized drug concentration can be
difficult in very compact biologic spaces like the BBB/CNS
interface, and consequently methods of measurement require
some explanation [52].
In-vitro methods are most commonly used to study human
transporter activity and identify candidate transporters for a
given substrate. Cell culture systems amenable to transfection
of influx or efflux transporter genes, or direct injection of
transporter mRNA into Xenopus oocytes, express and localize
transporter proteins to the cell surface. Isolated membrane
vesicles can also be prepared. Exposure to drug on one side
of a cell layer, then measurement on both sides, to determine
the ratio of substrate penetration to negative controls or by
using transporter inhibitors, allows individual assessment of
luminal and abluminal transporters and their mechanisms.
Unfortunately, heterologous systems have several problems
when attributing a specific physiologic function to a trans-
porter, because it is specifically overexpressed. Studies using
membranes of confluent primary human cells can provide
more information, particularly on the contribution of relevant
transporters for a specific drug. Ex-vivo assays, however, lack
functional context since organ-specific gross anatomy and
transporter interface conditions do not mimic the animal. In
addition, the transporter must function away from its normal
molecular constituents, and thus potential modulators of sub-
strate preference and functional compensations are lost.
In animals (and occasionally in humans) ex-vivo or in-situ
organ perfusion allows direct measurement of drug influx and
efflux concentrations. Advantages of this method are a close
control of the time course of drug administration and concen-
tration. Furthermore, drug goes directly from perfusate to the
brain (or other organ studied), bypassing systemic biotrans-
formation. Although this experimental methodology is more
precise, it still requires bulk tissue concentration measurement,
and hence does not provide data about discrete localization in
the brain (or other organ). Calculation of the brain efflux
index can be used as an indirect measure of chemical partition
at the BBB. A drug is injected directly into the brain with an
impermanent tracer. The ratio of test solute to the reference
tracer determines the brain efflux activity. An improved
method is tissue (often intracerebral) microdialysis. This
method directly samples small-molecule solutes in brain inter-
stitial fluid through a semiporous membrane. Microdialysis is
difficult and costly to perform, however, and may have its own
artifacts. Transgenic rodent models using knockouts or knock-
ins of transporters are a common approach to identifying
transporter function with respect to drug disposition and
Section 1: Principles of drug action
98
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
effect. However, there is still a great deal of argument sur-
rounding the similarity of physiologic function even between
highly homologous vertebrate systems.
In humans, pharmacogenomic approaches often attempt to
identify transporter polymorphisms which may alter pharma-
cokinetics or pharmacodynamics. The use of drug probes to
selectively inhibit a transporter’s function, and assess the effect
on pharmacokinetics or pharmacodynamics, is limited by the
lack of transporter-selective inhibitor probes. This is unlike the
methods used to profile cytochrome P450s in drug metabol-
ism, where P450-selective inhibitor drugs are available.
Drug–drug interactions and transporters
Drugs themselves can act to modulate xenobotic transport, and
any new pharmaceutical is a potential effector of chemical
partition. While the number of current examples is small, as
new therapeutics are developed the opportunity for drug–drug
interactions through transporter inhibition becomes increas-
ingly problematic. This issue is further exacerbated by the wide
tissue distribution of transporters like P-gp, which makes it
difficult to predict the potential for affecting drug distribution
and efficacy in a site-dependent fashion. As noted above, P-gp
loss of function can affect both brain and gut uptake
depending on the drug in question [36,45]. This is not just a
theoretical problem but occurs with many commonly used
medications. For instance, verapamil and the cyclosporine
analog PSC833 lead to increased brain concentration of anti-
epileptics such as carbamazapine, phenytoin, and phenobarbi-
tol [13,53]. The combination of quercetin, a P-gp substrate,
and digoxin also has demonstrated toxicity [53]. Lastly,
b-blockers such as talinolol exhibit increased oral bioavailability
when intestinal P-gp activity is reduced in both animal models
and human observations [53]. Unfortunately there is no set of
chemoprotective rules that can warn clinicians of potential inter-
actions. Currently all drug–drug interactions are determined
empirically, and hence the need for a more complete analysis of
the organization of chemical protection in the animal.
Modulating transporter function
in vivo for clinical purposes
The BBB is a dynamic structure regulated by many competing
inputs [11]. In vertebrates, transcellular diffusion can be modi-
fied by adjusting either the level or the activity of transporter
proteins [54]. Transcription of a number of efflux transporters
including P-gp, MRP2, MRP3, and OATP2 is regulated in part
by the pregnane X receptor (PXR), a nuclear receptor that is
activated by naturally occurring steroids as well as a wide
variety of xenobiotics [55–57]. Higher levels of transporter
expression result in more efficient xenobiotic efflux and a
tighter BBB [58]. The activity of some transporters can also
be influenced by cell-signaling pathways. For example, binding
of endothelin 1 to the ETB receptor rapidly and reversibly
inhibits transport activity of P-gp via nitric oxide synthase
and protein kinase-C mediated pathways [59].
ABC transporter chemopartition function can also be
modulated by drugs. In this case one drug acts as competitive
substrate antagonist against more favored transport molecules
(Table 7.1). This explains why many drugs are listed as both
substrate and antagonist in substrate/inhibitor tables. Several
clinical scenarios have attempted to introduce drugs like
“selective” transport inhibitors at the BBB interface to improve
penetration of chemotherapeutics, but no clinical benefit has
yet been obtained [13,60]. Nevertheless the search for more
selective competitive inhibitors or allosteric modulators of
ABC transporters is quite active in both academic and indus-
trial research [8].
Future directions in the study
of chemical protection
The ability of an organism to protect itself against environ-
mental threats, whether predation or chemical insult, is as
essential as the ability to convert food into energy. Chemical
transporters of the variety discussed in this chapter are present
across evolutionary time, with the essential structure of pro-
teins such as members of the ABC superfamily changing very
little across phyla. For example, there is increasing evidence
that species as distant as fruit flies and humans use very similar
means to exclude xenobiotics from privileged compartments
such as the CNS [61]. The decoding of the fruit fly Drosophila
melanogaster genome has enabled investigators to identify
genes and proteins that are conserved across species, including
analogs of many of the ABC transporters relevant to human
physiology and pathology (Table 7.1) [10]. While the compon-
ent parts of chemoprotective systems are just coming to light
(i.e., transporters and metabolic enzymes that are chemically
organized), there is little understanding of the regulation and
adaptations such a system requires. Model systems using
human cell culture, animal models, and the tools of pharma-
cogenomics will play a large role in furthering the understanding
of chemical protection. Once there are better understandings of
localized cellular or subcellular drug concentrations and identifi-
cation of novel modulators of chemical partition, better methods
of therapeutic drug delivery can be developed to improve clinical
outcomes.
Summary
Biophysical boundaries prevent passive diffusion of large mol-
ecules, such as proteins and nucleic acids, between different
compartments and greatly curtail the movement of electro-
lytes, soluble components of metabolism, and drugs. An enor-
mous variety of transmembrane protein transporters are
dedicated to selectively moving small molecules across mem-
branes. Transporters are essential actors in almost every bio-
logic function, as transporter systems create the chemical
preconditions for cellular function, restore chemical condi-
tions after perturbation, and clear chemical space of unwanted
metabolites. The precision, specificity, and ability of these
Chapter 7: Drug transport and transporters
99
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
transport systems to act against concentration gradients allow
small molecules to have discrete functions depending on their
localization. Drugs share the same molecular weight range and
solubility characteristics as many endogenous small molecules
and are subject to the same physiologic processes, and the
same transporters, as endogenous molecules.
Drug (xenobiotic) transporters were initially identified
during studies of multidrug cancer resistance. These proteins
were efflux pumps with broad substrate specificity, capable of
binding and transporting a wide spectrum of chemotherapeu-
tics back into the extracellular space, thereby reducing the
effective intracellular concentration of cytotoxic drugs. Cancer
cells had coopted and enhanced an endogenous chemical par-
tition system to strengthen the cell-membrane chemical barrier
and confer drug resistance.
Cell-surface transporters are now known to be ubiquitous,
present across evolutionary time with relatively conserved
functions across species, and capable of influx and/or efflux
transport of numerous drugs. P-glycoprotein (abbreviated
P-gp, and also termed ABCB1 or the multidrug resistance
transporter, MDR1) was the first xenobiotic transporter dis-
covered in multidrug-resistant cancer cells. P-gp also functions
in a wide range of normal tissues such as the gastrointestinal
tract, renal proximal tubule cells, the canalicular surfaces of
hepatocytes, the capillary endothelial cells of testes, and the
blood–brain barrier. Transporters may influence drug absorp-
tion, distribution, tissue penetration, metabolism, and excre-
tion. There is also the potential for significant drug
interactions.
How these competing transporter processes are regulated,
coordinated, and potentially circumvented or controlled will
require a better understanding of drug–transporter inter-
actions at cellular interfaces and methods for analyzing net
localized pharmacokinetic disposition of drugs. For a drug to
be clinically effective, it must possess high target specificity,
favorable organismal pharmacokinetics, and complementary
properties for highly localized chemical partition processes at
the cellular level. Drugs themselves can act to modulate xeno-
botic transport, and any new pharmaceutical is a potential
effector of chemical partition.
References
1. Alberts B, Johnson A, Lewis J, et al.
Molecular Biology of the Cell. New York,
NY:Garland, 2007.
2. van de Waterbeemd H, Gifford E.
ADMET in silico modelling: towards
prediction paradise? Nat Rev Drug Discov
2003; 2: 192–204.
3. Hediger MA, Romero MF, Peng JB, et al.
The ABCs of solute carriers:
physiological, pathological and
therapeutic implications of human
membrane transport proteins
Introduction. Pflugers Arch 2004; 447:
465–8.
4. Torres GE, Gainetdinov RR, Caron MG.
Plasma membrane monoamine
transporters: structure, regulation and
function. Nat Rev Neurosci 2003; 4:
13–25.
5. Torrie LS, Radford JC, Southall TD, et al.
Resolution of the insect ouabain paradox.
Proc Natl Acad Sci U S A 2004; 101:
13689–93.
6. Dano K. Active outward transport of
daunomycin in resistant Ehrlich ascites
tumor cells. Biochim Biophys Acta 1973;
323: 466–83.
7. Juliano RL, Ling V. A surface
glycoprotein modulating drug
permeability in Chinese hamster ovary
cell mutants. Biochim Biophys Acta 1976;
455: 152–62.
8. Sarkadi B, Homolya L, Szakacs G, Varadi
A. Human multidrug resistance ABCB
and ABCG transporters: participation in
a chemoimmunity defense system.
Physiol Rev 2006; 86: 1179–236.
9. Annilo T, Chen ZQ, Shulenin S, et al.
Evolution of the vertebrate ABC gene
family: analysis of gene birth and death.
Genomics 2006; 88: 1–11.
10. Dean M, Annilo T. Evolution of the ATP-
binding cassette (ABC) transporter
superfamily in vertebrates. Annu Rev
Genomics Hum Genet 2005; 6: 123–42.
11. Zlokovic BV. The blood–brain barrier
in health and chronic
neurodegenerative disorders. Neuron
2008; 57: 178–201.
12. Gottesman MM, Fojo T, Bates SE.
Multidrug resistance in cancer: role of
ATP-dependent transporters. Nat Rev
Cancer 2002; 2: 48–58.
13. Loscher W, Potschka H. Blood–brain
barrier active efflux transporters: ATP-
binding cassette gene family. NeuroRx
2005; 2: 86–98.
14. Dobson PD, Kell DB. Carrier-mediated
cellular uptake of pharmaceutical drugs:
an exception or the rule? Nat Rev Drug
Discov 2008; 7: 205–20.
15. Zhang L, Zhang YD, Strong JM, Reynolds
KS, Huang SM. A regulatory viewpoint on
transporter-based drug interactions.
Xenobiotica 2008; 38: 709–24.
16. Sadeque AJ, Wandel C, He H, Shah S,
Wood AJ. Increased drug delivery to the
brain by P-glycoprotein inhibition. Clin
Pharmacol Ther 2000; 68: 231–7.
17. Park HJ, Shinn HK, Ryu SH, et al.
Genetic polymorphisms in the ABCB1
gene and the effects of fentanyl in
Koreans. Clin Pharmacol Ther 2007; 81:
539–46.
18. Kruh GD, Belinsky MG. The MRP
family of drug efflux pumps. Oncogene
2003; 22: 7537–52.
19. Tsuji A. Impact of transporter-mediated
drug absorption, distribution,
elimination and drug interactions in
antimicrobial chemotherapy. J Infect
Chemother 2006; 12: 241–50.
20. Shitara Y, Horie T, Sugiyama Y.
Transporters as a determinant of drug
clearance and tissue distribution. Eur
J Pharm Sci 2006; 27: 425–46.
21. Doyle LA, Yang W, Abruzzo LV, et al.
A multidrug resistance transporter from
human MCF-7 breast cancer cells. Proc
Natl Acad Sci U S A 1998; 95: 15665–70.
22. Petrovic V, Teng S, Piquette-Miller M.
Regulation of drug transporters during
infection and inflammation. Mol Interv
2007; 7: 99–111.
23. Kim RB. Organic anion-transporting
polypeptide (OATP) transporter family
and drug disposition. Eur J Clin
Invest2003; 33 (Suppl 2): 1–5.
Section 1: Principles of drug action
100
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
24. Abbott NJ. Dynamics of CNS barriers:
evolution, differentiation, and
modulation. Cell Mol Neurobiol 2005;
25: 5–23.
25. Schinkel AH, Jonker JW. Mammalian
drug efflux transporters of the ATP
binding cassette (ABC) family: an
overview. Adv Drug Deliv Rev 2003; 55:
3–29.
26. Begley DJ, Brightman MW. Structural
and functional aspects of the blood–
brain barrier. Prog Drug Res 2003; 61:
39–78.
27. Neuwelt E, Abbott NJ, Abrey L, et al.
Strategies to advance translational
research into brain barriers. Lancet
Neurol
2008; 7: 84–96.
28. McCarty JH. Cell biology of the
neurovascular unit: implications for
drug delivery across the blood-brain
barrier. Assay Drug Dev Technol 2005; 3:
89–95.
29. Ballabh P, Braun A, Nedergaard M. The
blood–brain barrier: an overview:
structure, regulation, and clinical
implications. Neurobiol Dis 2004;
16: 1–13.
30. Reese TS, Karnovsky MJ. Fine structural
localization of a blood-brain barrier to
exogenous peroxidase. J Cell Biol 1967;
34: 207–17.
31. Wijnholds J, Evers R, van Leusden MR,
et al. Increased sensitivity to anticancer
drugs and decreased inflammatory
response in mice lacking the multidrug
resistance-associated protein. Nat Med
1997; 3: 1275–9.
32. Lorico A, Rappa G, Flavell RA,
Sartorelli AC. Double knockout of the
MRP gene leads to increased drug
sensitivity in vitro. Cancer Res 1996; 56:
5351–5.
33. Schinkel AH, Smit JJ, van Tellingen O,
et al. Disruption of the mouse mdr1a
P-glycoprotein gene leads to a
deficiency in the blood-brain barrier
and to increased sensitivity to drugs.
Cell 1994; 77: 491–502.
34. Vlaming ML, Lagas JS, Schinkel AH.
Physiological and pharmacological roles
of ABCG2 (BCRP): recent findings in
Abcg2 knockout mice. Adv Drug Deliv
Rev 2009; 61: 14–25.
35. Enokizono J, Kusuhara H, Ose A,
Schinkel AH, Sugiyama Y. Quantitative
investigation of the role of breast cancer
resistance protein (Bcrp/Abcg2) in
limiting brain and testis penetration of
xenobiotic compounds. Drug Metab
Dispos 2008; 36: 995–1002.
36. Schinkel AH. Pharmacological insights
from P-glycoprotein knockout mice. Int
J Clin Pharmacol Ther 1998; 36: 9–13.
37. Cahoy JD, Emery B, Kaushal A, et al.
A transcriptome database for astrocytes,
neurons, and oligodendrocytes: a new
resource for understanding brain
development and function. J Neurosci
2008; 28: 264–78.
38. Mannermaa E, Vellonen KS, Urtti A.
Drug transport in corneal epithelium
and blood–retina barrier: emerging role
of transporters in ocular
pharmacokinetics. Adv Drug Deliv Rev
2006; 58: 1136–63.
39. Kajikawa T, Mishima HK, Murakami T,
Takano M. Role of P-glycoprotein in
distribution of rhodamine 123 into
aqueous humor in rabbits. Curr Eye Res
1999; 18: 240–6.
40. Derevjanik NL, Vinores SA, Xiao WH,
et al. Quantitative assessment of the
integrity of the blood-retinal barrier in
mice. Invest Ophthalmol Vis Sci 2002;
43: 2462–7.
41. Johanson CE, Duncan JA, Stopa EG,
Baird A. Enhanced prospects for drug
delivery and brain targeting by the
choroid plexus-CSF route. Pharm Res
2005; 22: 1011–37.
42. Emerich DF, Skinner SJ, Borlongan CV,
Vasconcellos AV, Thanos CG. The
choroid plexus in the rise, fall and repair
of the brain. Bioessays 2005; 27:
262–74.
43. Dietrich CG, Geier A, Wasmuth HE,
et al. Influence of biliary cirrhosis on the
detoxification and elimination of a food
derived carcinogen. Gut 2004; 53:
1850–5.
44. Kusuhara H, Sugiyama Y. Role of
transporters in the tissue-selective
distribution and elimination of drugs:
transporters in the liver, small intestine,
brain and kidney. J Control Release
2002; 78: 43–54.
45. Lagas JS, Sparidans RW, van
Waterschoot RA, et al. P-glycoprotein
limits oral availability, brain
penetration, and toxicity of an anionic
drug, the antibiotic salinomycin.
Antimicrob Agents Chemother 2008;
52: 1034–9.
46. Englund G, Rorsman F, Rönnblom A,
et al. Regional levels of drug
transporters along the human intestinal
tract: co-expression of ABC and SLC
transporters and comparison with
Caco-2 cells. Eur J Pharm Sci 2006; 29:
269–77.
47. van de Wetering K, Zelcer N, Kuil A,
et al. Multidrug resistance proteins 2
and 3 provide alternative routes for
hepatic excretion of morphine-
glucuronides. Mol Pharmacol 2007; 72:
387–94.
48. El-Sheikh AA, Masereeuw R, Russel FG.
Mechanisms of renal anionic drug
transport. Eur J Pharmacol 2008; 585:
245–55.
49. Inui KI, Masuda S, Saito H. Cellular and
molecular aspects of drug transport in
the kidney. Kidney Int 2000; 58: 944–58.
50. Ganapathy V, Prasad PD. Role of
transporters in placental transfer of
drugs. Toxicol Appl Pharmacol 2005;
207: 381–7.
51. Jonker JW, Smit JW, Brinkhuis RF,
et al. Role of breast cancer resistance
protein in the bioavailability and fetal
penetration of topotecan. J Natl Cancer
Inst 2000; 92: 1651–6.
52. Smith QR. A review of blood–brain
barrier transport techniques. In: Nag S,
ed., Methods in Molecular Medicine.
Totowa, NJ:Humana Press, 2003:
193–208.
53. Balayssac D, Authier N, Cayre A,
Coudore F. Does inhibition of P-
glycoprotein lead to drug–drug
interactions? Toxicol Lett 2005; 156:
319–29.
54. Miller DS, Bauer B, Hartz AM.
Modulation of P-glycoprotein at the
blood-brain barrier: opportunities to
improve central nervous system
pharmacotherapy. Pharmacol Rev 2008;
60: 196–209.
55. Kliewer SA, Goodwin B, Willson TM.
The nuclear pregnane X receptor: a key
regulator of xenobiotic metabolism.
Endocr Rev 2002; 23: 687–702.
56. Kliewer SA, Moore JT, Wade L, et al. An
orphan nuclear receptor activated by
pregnanes defines a novel steroid
signaling pathway. Cell 1998; 92:
73–82.
Chapter 7: Drug transport and transporters
101
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
57. Bauer B, Yang X, Hartz AM, et al.
In vivo activation of human pregnane
X receptor tightens the blood–brain
barrier to methadone through
P-glycoprotein up-regulation.
Mol Pharmacol 2006; 70:
1212–19.
58. Perez-Tomas R. Multidrug resistance:
retrospect and prospects in anti-cancer
drug treatment. Curr Med Chem 2006;
13: 1859–76.
59. Hartz AM, Bauer B, Fricker G, Miller
DS. Rapid regulation of P-glycoprotein
at the blood–brain barrier by
endothelin-1. Mol Pharmacol 2004; 66:
387–94.
60. Kemper EM, Boogerd W, Thuis I,
Beijnen JH, van Tellingen O.
Modulation of the blood-brain barrier
in oncology: therapeutic opportunities
for the treatment of brain tumours?
Cancer Treat Rev 2004; 30: 415–23.
61. Mayer F, Mayer N, Chinn L, et al.
Evolutionary conservation of vertebrate
blood–brain barrier chemoprotective
mechanisms in Drosophila. J Neurosci
2009; 29: 3538–50.
Section 1: Principles of drug action
102
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008
Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at

More Related Content

What's hot

Abc transporters family
Abc transporters familyAbc transporters family
Abc transporters familyStella Evelyn
 
Membrane transporters
Membrane transportersMembrane transporters
Membrane transportersPavaniSSLD
 
Transporters as targets for drugs
Transporters as targets for drugsTransporters as targets for drugs
Transporters as targets for drugsshikha singh
 
Active transporter
Active transporter Active transporter
Active transporter SAKSHI YADAV
 
Membrane transporters and drug response
Membrane transporters and drug responseMembrane transporters and drug response
Membrane transporters and drug responsebhagyamohod90
 
Membrane transporters and drug response
Membrane transporters and drug responseMembrane transporters and drug response
Membrane transporters and drug responseSoujania Singh
 
Genetic variation in drug transporters
Genetic variation in drug transportersGenetic variation in drug transporters
Genetic variation in drug transportersDeepak Kumar
 
Structure and function of multidrug transporter proteins and mechanism of res...
Structure and function of multidrug transporter proteins and mechanism of res...Structure and function of multidrug transporter proteins and mechanism of res...
Structure and function of multidrug transporter proteins and mechanism of res...Ravinder Bhardwaj
 
Liposomal gene delivery system
Liposomal gene delivery systemLiposomal gene delivery system
Liposomal gene delivery systemDurga Bhavani
 
P glycoprotein
P glycoprotein P glycoprotein
P glycoprotein khaterehz
 
Liposomsl gene delivery SYSTEM
Liposomsl gene delivery SYSTEMLiposomsl gene delivery SYSTEM
Liposomsl gene delivery SYSTEMSUJITHA MARY
 

What's hot (20)

Drug transport
Drug transportDrug transport
Drug transport
 
Abc transporters family
Abc transporters familyAbc transporters family
Abc transporters family
 
Membrane transporters
Membrane transportersMembrane transporters
Membrane transporters
 
Transporters as targets for drugs
Transporters as targets for drugsTransporters as targets for drugs
Transporters as targets for drugs
 
p-glycoprotein
p-glycoproteinp-glycoprotein
p-glycoprotein
 
Active transporter
Active transporter Active transporter
Active transporter
 
Drug transporters
Drug transportersDrug transporters
Drug transporters
 
Membrane transporters and drug response
Membrane transporters and drug responseMembrane transporters and drug response
Membrane transporters and drug response
 
Memb transporter
Memb transporterMemb transporter
Memb transporter
 
Membrane transporters and drug response
Membrane transporters and drug responseMembrane transporters and drug response
Membrane transporters and drug response
 
Genetic variation in drug transporters
Genetic variation in drug transportersGenetic variation in drug transporters
Genetic variation in drug transporters
 
bd
bdbd
bd
 
Membrane transporters
Membrane transportersMembrane transporters
Membrane transporters
 
Pgp
PgpPgp
Pgp
 
Drug interactions linked to transporters
Drug interactions linked to transportersDrug interactions linked to transporters
Drug interactions linked to transporters
 
Structure and function of multidrug transporter proteins and mechanism of res...
Structure and function of multidrug transporter proteins and mechanism of res...Structure and function of multidrug transporter proteins and mechanism of res...
Structure and function of multidrug transporter proteins and mechanism of res...
 
Liposomal gene delivery system
Liposomal gene delivery systemLiposomal gene delivery system
Liposomal gene delivery system
 
P glycoprotein
P glycoprotein P glycoprotein
P glycoprotein
 
Liposomsl gene delivery SYSTEM
Liposomsl gene delivery SYSTEMLiposomsl gene delivery SYSTEM
Liposomsl gene delivery SYSTEM
 
24 rajesh z. mujoriya 8
24 rajesh z. mujoriya 824 rajesh z. mujoriya 8
24 rajesh z. mujoriya 8
 

Similar to CBO9780511781933.008.pdf

04.pharmacokinetics(1)
04.pharmacokinetics(1)04.pharmacokinetics(1)
04.pharmacokinetics(1)sisy10
 
Medicinal Chemistrt Unit -1.pptx
Medicinal Chemistrt Unit -1.pptxMedicinal Chemistrt Unit -1.pptx
Medicinal Chemistrt Unit -1.pptxNikita Gupta
 
Drug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal TractDrug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal TractFeba Elsa Mathew
 
TDDS targeted drug delivery system
TDDS targeted drug delivery systemTDDS targeted drug delivery system
TDDS targeted drug delivery systemNaveen Reddy
 
Mechanism of absorption(biopharma)
Mechanism of absorption(biopharma)Mechanism of absorption(biopharma)
Mechanism of absorption(biopharma)MdIrfanUddin2
 
Pharmacokinetics: Absorption (Transport mechanism)
Pharmacokinetics: Absorption (Transport mechanism)Pharmacokinetics: Absorption (Transport mechanism)
Pharmacokinetics: Absorption (Transport mechanism)Vijay Kevlani
 
Pharmacology 1
Pharmacology 1Pharmacology 1
Pharmacology 1axix
 
2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materialsrossbiology
 
targeting methods.pptx
targeting methods.pptxtargeting methods.pptx
targeting methods.pptxMit Bhatt
 
distribution.pdf
distribution.pdfdistribution.pdf
distribution.pdfEducation
 
Pharmacokinetics study
Pharmacokinetics studyPharmacokinetics study
Pharmacokinetics studyMaheshThakur42
 
Biopharmaceutics lecture 2(2)
Biopharmaceutics lecture 2(2)Biopharmaceutics lecture 2(2)
Biopharmaceutics lecture 2(2)homebwoi
 
Biopharmaceutics lecture 2
Biopharmaceutics lecture 2Biopharmaceutics lecture 2
Biopharmaceutics lecture 2homebwoi
 
Cell Membrane Structure-1.pptx cell outer layer
Cell Membrane Structure-1.pptx cell outer layerCell Membrane Structure-1.pptx cell outer layer
Cell Membrane Structure-1.pptx cell outer layerSalsabeelArif
 
Cell Communication, Cell Junction and Cell Signaling.pptx
Cell Communication, Cell Junction and Cell Signaling.pptxCell Communication, Cell Junction and Cell Signaling.pptx
Cell Communication, Cell Junction and Cell Signaling.pptxSheetal Patil
 
2) PHARMACOKINETICS.pptx
2) PHARMACOKINETICS.pptx2) PHARMACOKINETICS.pptx
2) PHARMACOKINETICS.pptxVarshaPatel72
 

Similar to CBO9780511781933.008.pdf (20)

04.pharmacokinetics(1)
04.pharmacokinetics(1)04.pharmacokinetics(1)
04.pharmacokinetics(1)
 
Medicinal Chemistrt Unit -1.pptx
Medicinal Chemistrt Unit -1.pptxMedicinal Chemistrt Unit -1.pptx
Medicinal Chemistrt Unit -1.pptx
 
Drug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal TractDrug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal Tract
 
TDDS targeted drug delivery system
TDDS targeted drug delivery systemTDDS targeted drug delivery system
TDDS targeted drug delivery system
 
Mechanism of absorption(biopharma)
Mechanism of absorption(biopharma)Mechanism of absorption(biopharma)
Mechanism of absorption(biopharma)
 
Pharmacokinetics: Absorption (Transport mechanism)
Pharmacokinetics: Absorption (Transport mechanism)Pharmacokinetics: Absorption (Transport mechanism)
Pharmacokinetics: Absorption (Transport mechanism)
 
Overview of movement of drug molecules across cell membrane.pptx
Overview of movement of drug molecules across cell membrane.pptxOverview of movement of drug molecules across cell membrane.pptx
Overview of movement of drug molecules across cell membrane.pptx
 
Pharmacology 1
Pharmacology 1Pharmacology 1
Pharmacology 1
 
2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials
 
targeting methods.pptx
targeting methods.pptxtargeting methods.pptx
targeting methods.pptx
 
distribution.pdf
distribution.pdfdistribution.pdf
distribution.pdf
 
Pharmacokinetics study
Pharmacokinetics studyPharmacokinetics study
Pharmacokinetics study
 
Biopharmaceutics lecture 2(2)
Biopharmaceutics lecture 2(2)Biopharmaceutics lecture 2(2)
Biopharmaceutics lecture 2(2)
 
Biopharmaceutics lecture 2
Biopharmaceutics lecture 2Biopharmaceutics lecture 2
Biopharmaceutics lecture 2
 
Cell Membrane.pptx
Cell Membrane.pptxCell Membrane.pptx
Cell Membrane.pptx
 
Cell Membrane Structure-1.pptx cell outer layer
Cell Membrane Structure-1.pptx cell outer layerCell Membrane Structure-1.pptx cell outer layer
Cell Membrane Structure-1.pptx cell outer layer
 
Cell Communication, Cell Junction and Cell Signaling.pptx
Cell Communication, Cell Junction and Cell Signaling.pptxCell Communication, Cell Junction and Cell Signaling.pptx
Cell Communication, Cell Junction and Cell Signaling.pptx
 
Pharmacokinetics
PharmacokineticsPharmacokinetics
Pharmacokinetics
 
Drug absorption naresh
Drug absorption   nareshDrug absorption   naresh
Drug absorption naresh
 
2) PHARMACOKINETICS.pptx
2) PHARMACOKINETICS.pptx2) PHARMACOKINETICS.pptx
2) PHARMACOKINETICS.pptx
 

Recently uploaded

Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentInMediaRes1
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeThiyagu K
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Sapana Sha
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxiammrhaywood
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
Concept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfConcept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfUmakantAnnand
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...EduSkills OECD
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactdawncurless
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfsanyamsingh5019
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Educationpboyjonauth
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingTechSoup
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Celine George
 
The basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxThe basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxheathfieldcps1
 
Science 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsScience 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsKarinaGenton
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxpboyjonauth
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 

Recently uploaded (20)

Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
Alper Gobel In Media Res Media Component
Alper Gobel In Media Res Media ComponentAlper Gobel In Media Res Media Component
Alper Gobel In Media Res Media Component
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and Mode
 
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111Call Girls in Dwarka Mor Delhi Contact Us 9654467111
Call Girls in Dwarka Mor Delhi Contact Us 9654467111
 
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptxSOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
SOCIAL AND HISTORICAL CONTEXT - LFTVD.pptx
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
Concept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.CompdfConcept of Vouching. B.Com(Hons) /B.Compdf
Concept of Vouching. B.Com(Hons) /B.Compdf
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
 
Accessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impactAccessible design: Minimum effort, maximum impact
Accessible design: Minimum effort, maximum impact
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdf
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Education
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy Consulting
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
The basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxThe basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptx
 
Science 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsScience 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its Characteristics
 
Introduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptxIntroduction to AI in Higher Education_draft.pptx
Introduction to AI in Higher Education_draft.pptx
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 

CBO9780511781933.008.pdf

  • 1. Section 1 Chapter 7 Principles of drug action Drug transport and transporters Roland J. Bainton and Evan D. Kharasch Drug transporters and localized pharmacokinetics Xenobiotics and defining chemical space Living organisms use biologic compartments to perform dis- crete biochemical processes, to localize and control the natural small molecules contained within [1]. Foreign small molecules, called xenobiotics, share the same molecular weight range and solubility characteristics as endogenous small molecules and are thus able to penetrate biologic spaces. As xenobiotics also pos- sess potent biointeraction (i.e., drug-like) properties they are the source for nearly all clinically useful pharmaceuticals [2]. To balance the cells’ needs of access to endogenous small molecules and control of xenobiotic penetration, powerful biotransforma- tion and bioelimination pathways have evolved to control chemical space (Chapter 6). These same processes present chal- lenges for delivering clinically useful therapies to particular organ spaces, as xenobiotics have complex and unpredictable relationships with the selectively expressed small molecule transporters or specially tuned metabolic systems [2]. Thus understanding how any one pharmaceutical interacts with bio- logically defined chemical space requires a closer examination of that drug’s susceptibility to highly localized pharmacokinetic processes affecting chemical persistence at the site of action. Lipid bilayers and drug localization Lipid bilayers physically partition aqueous spaces [1] (Fig. 7.1), creating cells and specialized internal subcellular compartments. In addition, formation of cell–cell contacts in conjunction with diffusion-tight junctional complexes divides the external cellular milieu into different interstitial spaces [1] (Fig. 7.2). This bio- physical boundary prevents passive diffusion of large molecules such as proteins and nucleic acids between different compart- ments and greatly curtails the movement of electrolytes, soluble components of metabolism, and drugs. Drugs and xenobiotics are often in equilibrium between charged and charge-neutral states (Fig. 7.2). Charge-neutral molecules more readily transit lipid bilayers and possess better penetration rates into cells by orders of magnitude (passive permeability, Fig. 7.1). Cellular penetration may be augmented (influx transport) or opposed (efflux transport) by active cellular processes (see below), a fea- ture that allows drugs to take multiple routes to a target (Fig. 7.2). To control the ability of drugs to transit freely in cellular space and still maintain chemical partition, cells organize molecular systems to contain, modify, or move endogenous and foreign lipophilic molecules from one space to another. Small-molecule partition and transporters Chemical partition is the sine qua non of cell biology, and cellular transport systems play a vital role in chemical compartmental- ization. An enormous variety of transmembrane protein trans- porters are dedicated to selectively moving small molecules across membranes [3]. Their role is so broad that by some estimates 15–30% of all membrane proteins are transporters [1]. Among the many substrates that transmembrane protein transporters relocalize are glucose, anabolic intermediates, amino acids, neurotransmitters, metabolic products, and lipids [3]. The vast majority of transporters have high substrate specificity and are organized by DNA sequence relatedness and known function. They are referred to as solute carrier (SLC) proteins and include 46 different gene families with over 360 genes in humans [3]. Their enormous diversity speaks to the wide variety of intracel- lular environments and their highly specific needs. The precision, specificity, and ability of these transport systems to act against concentration gradients allows small mol- ecules to have discrete functions depending on their localization. For example, plasma membrane transporters of the SLC6 class specialize in the transport of monoamines from the interstitial space to the cytoplasm. These transporters perform two key functions for the cell: (1) providing amino acids to the cytoplasm for metabolic purposes, and (2) reducing extracelluar concen- trations of monoamines when unwanted [4]. However, when the same molecule (or close derivative) is to be used as a neurotransmitter, the SLC18 transporters highly concentrate and package the molecule into synaptic vesicles. After release into the extracelluar space for signaling at the postsynaptic membrane, neurotransmitters are rapidly taken up by SLC6 class Anesthetic Pharmacology, 2nd edition, ed. Alex S. Evers, Mervyn Maze, Evan D. Kharasch. Published by Cambridge University Press. # Cambridge University Press 2011. 90 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 2. transporters. Moving small molecules across cell membranes is entropically unfavorable, and thus transporters require energy expenditure either directly using adenosine triphosphate (ATP), or dissipating other existing electrochemical gradients (e.g., Na/ K) by using symport or antiport mechanisms [3]. Together, transporter systems create the chemical preconditions for cellu- lar function, restore chemical conditions after perturbation, and clear chemical space of unwanted metabolites. Thus transporters are essential actors in almost every biologic function. Localized pharmacokinetic spaces Acting in concert with, or after the action of, phase I and phase II metabolism (Chapter 6), to move drugs and their metabolites across lipid bilayers, transporters are often referred to as phase III metabolism. Therefore transporters are often highly expressed in organs of biotransformation such as the liver and intestine, and metabolism and transport together create formidable pharmacokinetic barriers. Different organ systems tune their biodefense systems by locally applying phase I, II, and III mechanisms [5]. Thus, for a drug to be clinically effective, it must possess high target specificity, favorable organismal pharmacokinetics, and complementary properties for highly localized chemical partition processes at the cellular level. It is in this latter context that the special role of drug transporters and their ability to control xenobiotic chemical space in drug action are considered. Drug transporters Discovery of drug efflux transporters Xenobiotic transporters were identified as a consequence of the study of multidrug resistance to chemical cancer treatment in the 1970s [6,7]. Highly efficient cytotoxic resistance to a broad spectrum of cancer drugs could occur suddenly and unexpect- edly after initiation of chemotherapy [8]. Analysis of drug- resistant tumor cells determined that this new-found resilience correlated with upregulation of a few transmembrane proteins, suggesting that chemotheraputic protection systems were quite nucleus hydrophobic small/uncharged polar large/uncharged polar ions lipid bilayer intracellular space extracellular space ~10–7 cm/sec ~10–11 cm/sec ~10–4 cm/sec >10–2 cm/sec Figure 7.1. Chemical partition across bilayers. Lipid bilayers (green lines) are formed by virtue of the chemical characteristics of their constituent phospholipids: a hydrophilic phosphate head group and a hydrophobic carbon-chain tail. The head groups associate with the aqueous environment, while the tail groups self-associate and form the internal, hydrophobic core of the bilayer. This layer must be transited if nutrients as well as drugs are to reach targets inside the cell, and movement across this bilayer is also dictated by specific chemical properties. Paracellular movement refers to passage of molecules between cells, bypassing cell mem- brane barriers. Transcellular movement requires pas- sage across membrane barriers. More hydrophobic chemicals and compounds can cross the barrier with relative ease and have a high permeability coefficient (expressed in the figure as cm s–1 ) whereas chemicals with low permeability coeffi- cients (such as ions) cannot passively transit the lipid bilayer and must cross via other means such as carrier-mediated transport. All chemicals critical to sustaining life fall along this continuum. Modified with permission from Alberts et al. [1]. ( )+/– ( )0 Transcellular Paracellular Drug Transporters (Ionized Form)+/– (Non-ionized Form)0 Junctional Complexes Interstitial Space (vascular) Interstitial Space (organ) Figure 7.2. Chemical partition across cellular boundaries. Cells separate one interstitial space from another. Cellular barriers rely upon two modes of exclusion to maintain xenobiotic exclusion, as small molecules can interact with cells in two ways. Charged molecules (blue stars) are excluded by the boundary function of lipid bilayers, very low rates of endocytosis, and tight diffusion barriers provided by special lateral-border junctional complexes (blue ovals) of barrier epithelium. Small uncharged molecules (red stars) pass easily through lipid bilayers (green), but active efflux transporters (red) move them back into the humoral space, creating an active transport barrier. As drugs in aqueous solution are in equilibrium between charged and uncharged forms, a true xenobiotic barrier must maintain both properties simultaneously to mani- fest chemical exclusion. Chapter 7: Drug transport and transporters 91 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 3. different from the highly specific SLC transporter systems dis- cussed above. Ultimately these genes were identified as active drug efflux pumps that, unlike most SLC transporters, had surprisingly broad substrate specificity. These proteins were found to bind and transport a wide spectrum of chemothera- peutics back into the extracellular space, thus reducing the effective internal concentration of cytotoxic drugs [8]. Interest- ingly, these cells were not intrinsically more resistant to che- motherapeutics at the target sites, but had instead coopted and enhanced an endogenous chemical partition system to strengthen the chemical barrier at the membrane [8]. Whole genome sequencing methods have discovered large families of similar multidrug resistance transporter (MDR) genes in nearly all organisms [9,10]. Most of these proteins belong to a large and highly conserved class of transmembrane proteins known as ATP-binding cassette (ABC) transporters [8,11]. The archetypal ABC transporter contains two six-pass transmembrane domains and two cytoplasmic nucleotide- binding domains, although the proteins do vary structurally depending on their class (Fig. 7.3). Full transporters are large proteins with 1200–2200 amino acids, and half-transporters have 650–800 amino acids. Half-transporters are thought to function as dimers in the membrane, potentially generating additional functional heterogeneity by combining different substrate preference into one transport pocket [8]. Unfortunately, unlike other transport systems where ligand binding is well understood, only a few specifics are known about the mechanism of xenobiotic binding to ABC transporters. Efficient efflux, for example, is thought to result from conformational changes of the transporter which allow an alternating high substrate-affinity (cell-facing)/low substrate-affinity (extracellular-facing) organization [8]. This permits intracellular presentation of the drug at one interface and release (or efflux) into the extracellular milieu on the other side. Evidence is mounting that drug binding to the major substrate-promiscuous xenobiotic transporters occurs in the membrane and not from the solution. Thus, these transporters may be designed to act at a protein/lipid-mem- brane interface [8]. The ABC gene family The approximately 50 human ABC genes occur in seven families (A through G), although only a subset are known to play major roles in drug transport. The major xenobiotic efflux transporter familes are the B, C, and G gene classes (Table 7.1). Interestingly, while ABC gene sequence and sub- families are highly conserved from yeast to flies to humans, the specific roles of most gene products are unknown. ABC transporter localization in polarized cell interfaces is essential to chemoprotective function, as net flux of xenobiotics into or away from various interstitial spaces is determined by the type, quantity, efflux or influx orientation, and apical/basal orientation at the protective interface (e.g., the vascular endo- thelium of the blood–brain barrier, as in Figs. 7.4 and 7.5) [8,13]. Unfortunately as the best studies of ABC transporters in the animal are limited to a few genes (see below), it is not known whether or how they function as a complementary system in the biology of chemical exclusion. Important subtypes of drug efflux transporters ABCB1 (P-glycoprotein) P-glycoprotein (P-gp, ABCB1, or the multidrug resistance transporter MDR1) was the first xenobiotic transporter discovered in multidrug-resistant cancer cells [7] and is Domain 2 Extracellular Intracellular Extracellular Intracellular Extracellular Intracellular NBD NBD C-Terminus N-Terminus NBD NBD C-Terminus N-Terminus NBD N-Terminus Lipid Bilayer Lipid Bilayer Lipid Bilayer C-Terminus Examples MDR1 (ABCB1) MRP4 (ABCC4) MRP5 (ABCC5) MRP7 (ABCC1) BSEP/SPGP (ABCB11) MRP1 (ABCC1) MRP2 (ABCC2) MRP3 (ABCC3) MRP6 (ABC6) MXP/BCRP/ABC-P (ABCG2) Domain 1 Figure 7.3. Membrane topologies of known xeno- biotic transporters. (Top) P-gp-like ABC transporters have 12 transmembrane (TM) domains and two nucleotide-binding domains (NBD). (Middle) In the ABCC class a second membrane topology preserves the core P-gp-like transporter with an additional five TMs on the N-terminus. (Bottom) The half-transport- ers of the ABCG class have six TMs and one NBD. They are thought to dimerize to promote drug transport function. Modified with permission from Gottesman et al. [12]. Section 1: Principles of drug action 92 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 4. responsible for the most profound effects on localized xeno- biotic protection of all the ABC transporter subfamily proteins. P-gp is a twelve-pass transmembrane protein of 140 kD and is highly expressed at important chemoprotec- tive interfaces (Table 7.2). It has the broadest substrate specificity of the ABC genes, transporting a significant number of drugs, including anticancer drugs, HIV-protease inhibitors, antibiotics, antidepressants, antiepileptics, and opioids such as morphine and loperamide (Table 7.2). P-gp substrates are generally amphipathic and range in size from 300 to 2000 Da. P-gp also functions in a wide range of environments including the GI tract, renal proximal tubule cells, the canalicular surfaces of hepatocytes, the capillary endothelial cells of testes, and the blood–brain barrier. Thus the poten- tial for significant drug interactions is widespread [8]. For example, oral bioavailability of the anti-inflammatory drug cyclosporine A is greatly increased by amlodipine, a P-gp substrate that doubles as a P-gp inhibitor [8]. Another example is loperamide, an opioid antidiarrheal that pro- duces no notable central nervous system (CNS) effects at normal doses because intestinal P-gp-mediated efflux retards systemic absorption [8]. However, when loperamide is given with the P-gp inhibitor quinidine, significant systemic absorption occurs, CNS delivery ensues, and side effects such as respiratory depression arise [16]. Thus localized transporter interactions can have substantial effects on drug bioavailability. This is particularly pertinent in fentanyl pharmacodynamics, based on the association between P-gp polymorphisms and fentanyl respiratory depression [17]. In patients under spinal anesthesia given intravenous fentanyl (2.5 mg kg–1 ), there are differences in suppression of ventilatory effort between three distinct P-gp genotypes prominent in ethincally Korean populations. The presump- tion is that P-gp transports fentanyl at CNS barriers, and therefore P-gp polymorphisms significantly alter brain concentration despite equivalent doses. Thus, a highly local- ized drug partition phenomenon can affect drug responses, but they are dependent on the relationship of the specific interface (i.e., the blood–brain barrier) and the protected localization of the drug target (i.e., neurons containing opioid receptors). ABCC transporters (MRPs) Multidrug resistance proteins (MRPs) are the largest class of ABC transporters relevant to drug resistance and drug distribution and are ubiquitously expressed [18]. There are 12 proteins in this class with two distinct types of transmem- brane protein topologies (Fig. 7.3). Although they contain only 15% sequence identity to P-gp they are predicted to posses a similar structure, with two nucleotide-binding domains and similar membrane topology to ABCB transport- ers (Fig. 7.3) [12]. Furthermore, like P-gp, they can extrude a wide array of amphipathic anions. Interestingly, all members of this particular subfamily, also called ABCC transporters, are lipophilic anion transporters that are broadly involved in cancer therapeutic resistance [18]. They are quite adept at effluxing methotrexate, thereby lessening the effect of an important chemotherapeutic in the brain [8]. MRP sub- strate specificities show a preference for glutathione and glucuronide conjugates, thus drug elimination could be greatly facilitated when transporters are locally paired with the appropriate phase II enzymes (e.g., glucuronyl trans- ferases, Chapter 6). Nevertheless MRPs also efficiently trans- port unconjugated compounds such as anthracyclines, Table 7.1. ABC transporter genes are highly conserved. Pairwise analysis using human ABC transporters versus Drosophila using NCBI BLAST (http://blast.ncbi.nlm.nih.gov/Blast.cgi) demonstrates high conservation of transporter number per class and high identity among classes. Similar results are obtained from comparison with other metazoan genomes, and thus ABC genes are highly evolutionarily conserved and likely to play fundamental roles in cellular biology. The major xenobiotic transporters are from the B, C, and G classes, but A-class transporters may play roles in some cases. Subfamily Human Drosophila Identities (%) Positives (%) A 12 10 20–37 34–56 B 11 8 43–57 59–75 C 13 14 23–50 40–67 D 4 2 56–59 71–74 E 1 1 77 88 F 3 3 41–69 56–82 G 5 (þ1) 15 22–54 41–71 Total 50 53 Vascular Space A VE P CNS Interstitial Space Figure 7.4. Organization of the blood–brain barrier (BBB). While chemical protection is focused at discrete epithelial layers, surrounding tissue can have essential roles in molding and assisting the localized PK potential. This is particularly true at the BBB, where properties of the vascular endothelium (VE) are induced and controlled by surrounding tissues. The BBB VE is thought to receive inductive cell-fate signals from the CNS parenchyma (not shown), while astrocytes (A) and pericytes (P) assist in and regulate localized responses to chemical and immune stress. Chapter 7: Drug transport and transporters 93 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 5. epipodophyllotoxins, vinca alkaloids, and camptothecins (Table 7.2) [18]. MRPs are known to localize to both apical and basal interfaces of chemical protection interfaces and thus can participate in moving drugs in both directions between interstitial spaces (Fig. 7.5) [8,13]. ABCG2 (BCRP) Breast cancer resistance protein (BCRP), so named for its discovery in an especially drug-resistant breast cancer cell line [21], is the second member of the five-member half-transporter ABCG subfamily (Fig. 7.3) [10]. BCRP is expressed throughout the body but most notably in the liver, intestinal epithelium, placenta, blood–brain barrier, and a variety of stem cells. Like P-gp, BCRP is quite promiscuous in its choice of substrate and is powered by the hydrolysis of ATP. BCRP substrates generally include compounds that are relatively large in molecular weight and either positively or negatively charged and amphipathic. Specific substrates for BCRP-mediated efflux include antineoplastic drugs and their metabolites (e.g., methotrexate and sulfated methotrexate), Table 7.2. Major human drug transporters P-glycoprotein (P-gp), multidrug resistance protein (MRP), breast cancer resistance protein (BRCP), organic anion transporting polypeptide (OATP), organic cation transporter (OCT), and organic anion transporter (OAT) are the best-studied drug transporters. They can share substrates and inhibitors. Modified with permission from Loscher and Potschka [13], Dobson and Kell [14], Zhang et al. [15]. Transporter gene Aliases Localization Function Selected substrates Selected inhibitors ABC family ABCB1 P-glycoprotein, MDR1 BBB, BRB, intestine, liver, kidney, placenta, adrenal, testis efflux digoxin, fexofenadine, morphine, loperamide, corticoids, cyclosporine, paclitaxel, verapamil, methotrexate cyclosporine, verapamil, quinidine, ritonavir, ketocoanzole, itraconazole ABCC1 MRP1 BBB, breast, lung, heart, kidney, blood cells, liver, intestines, testes, placenta efflux glutathione or glucuronide conjugates, leukotriene C4, etoposide, methotrexate probenecid, sulfinpyrazone, cyclosporine ABCC2 MRP2 intestine, liver, kidney, brain, placenta efflux bilirubin, indinavir, cisplatin cyclosporine ABCG2 BCRP BBB, intestine, liver, breast, placenta efflux anthracyclines, mitoxantrone, prazosin, topotecan, methotrexate elacridar (GF120918), cyclosporine, fumitremorgin C SLCO family SLCO1A2 OATP1A2 Liver, kidney, brain, lung, colon uptake, efflux fexofenadine, rocuronium, enalapril, rosuvastatin SLCO1B1 OATP1B1 BBB, brain, kidney, liver uptake, efflux organic anions, bile salts, digoxin, fexofenadine, methotrexate, rifampin cyclosporine, verapamil, rifampin, similar to P-gp SLC family SLC22A1 OCT1 liver uptake metformin quinidine, quinine, verapamil, cimetidine SLC22A2 OCT2 kidney, brain uptake metformin, amantadine, memantine, varenicline cimetidine SLC22A6 OAT1 kidney, brain uptake acyclovir, adefovir, methotrexate, zidovudine probenecid, cefadroxil, cefamandole, cefazolin, diclofenac SLC22A8 OAT3 kidney, brain uptake cimetidine, methotrexate, zidovudine probenecid, cefadroxil, cefamandole, cefazolin, cimetidine BBB, blood–brain barrier; BRB, blood–retinal barrier. Section 1: Principles of drug action 94 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 6. topotecan and irinotecan (both topoisomerase I inhibitors), as well as mitoxantrone, camptothecin and flavopiridol, xenobio- tic toxins, food-borne carcinogens, and endogenous com- pounds [8]. BCRP, unlike MRPs, is also adept at transporting glutamated folates and folate antagonists [8]. Therefore, like P-gp, BCRP is a major factor in conferring resistance to a wide range of therapeutics and is central to pharmacokinetics and toxicity considerations [8]. Other drug transporters SLC21 transporters (organic anion transporter proteins) Despite the relative abundance of SLC transporters at protect- ive barrier interfaces, few are known to participate directly in drug transport. One exception is the organic anion transporter proteins (OATPs), which are not structurally related to ABC transporters, but belong to the solute carrier transporters class 21 (SLC21/SLCO) [3]. OATPs primarily use passive transport mechanisms driven by electrochemical gradients or symport/ antiport mechanisms, but interestingly show other functional similarities to P-gp. The best-characterized OATP xenobiotic transporters are OATP1A2, OATP1B1, OATP1B3, and OATP2B1 [14,15]. They are generally efflux transporters, found at chemoprotective interfaces, prominently in brain endothelial cells, and also in liver, lung, kidney, and testes. In-situ mRNA hybridization also demonstrates broad expres- sion in central brain tissue, suggesting a role in general cellular protection as well as barrier function. In-vitro experiments suggest that OATPs can transport many structurally divergent molecules, reminiscent of P-gp. Examples include bile acids, Intestine MCT1 OATP-B OCTN2 MDR1 MRP2 Blood-Cerebrospinal Fluid Barrier Blood Blood-Brain Barrier others? MCT1 MRP1/2//4/5/6 OAT3 OCTN1/2 MDR1 BCRP PEPT1/2 OAT4 NPT1 URAT1 OCTN1/2 MDR1 MRP2/4 Kidney others? OCT2 OAT1/3 MRP3/6 Liver NTCP OATP-B/C/8 OAT2 NPT1 MRP1/3/6 others? others? MRP2 BCRP MDR1 OCT3 MRP1/3 Placenta PEPT1 OCT3 ASBT MRP3 MCT1 OATP-A OST α/β others? BCRP ? MDR1 BSEP BCRP MRP2 PEPT2 OCT2 others? MRP1 Figure 7.5. Membrane localization of drug transporters at the blood–brain barrier. Organ spaces subject to specific chemical constraints usually organize the tightest control of chemical space at one cellular layer. These chemical barriers use tight junctions and transporter content, type, and apical/basal localization to modulate the protected interstitial space to the purpose and susceptibilities of the underlying tissue [19,20]. The vascular endothelium of the BBB is one of the most potent localized pharmacokinetic interfaces and has a vast array of drug efflux and influx transporters at both vascular and CNS interfaces. Similar organization is repeated at the blood–cerebrospinal fluid barrier, renal epithelium of the kidney, intestinal epithelium, hepatocyte/bile-duct interfaces, and placenta; thus drug action at an individual organ can depend greatly on the chemoprotective efficiency of a particular exposure interface. For transcellular transport, drugs need to cross two different membranes on the basal and apical sides. In the intestine, drugs are absorbed from the luminal side (brush border membrane) and excreted into the portal blood across the basolateral membrane. In the liver, drugs are taken up into hepatocytes across the sinusoidal membrane and excreted into the bile. In the kidney, drugs undergo secretion (urinary excretion) or reabsorption. In the case of secretion, drugs are taken up from the basolateral side and excreted in the urine across the apical side. However, in the case of reabsorption, drugs are taken up from the urine and excreted in the circulating blood. Reproduced with permission from Tsuji [19]. Chapter 7: Drug transport and transporters 95 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 7. steroids, estrogen, many anionic compounds, and glutathione or glucuronide conjugates [22]. Certain drug–food inter- actions (e.g., grapefruit juice) [23] may be attributable to inhibition of OATPs, but further characterization of the in- vivo role of OATPs in drug interactions is needed. Interest- ingly, unlike ABC transporters, some OATPs can also function as drug influx transporters (Fig. 7.4). Bidirectional transport and/or the counteracting of efflux transport occurs at numer- ous tissue interfaces including the gut, liver, kidney, and blood–brain barrier [19,20]. How these competing transporter processes are regulated, coordinated, and potentially circum- vented or controlled will require a better understanding of drug–transporter interactions at cellular interfaces and methods for analyzing net localized pharmacokinetic dispos- ition of drugs. Specially protected anatomic spaces Specialized cellular boundary systems have evolved to limit access to sensitive biologic spaces [12,24]. Transporters are critical to the processes of drug uptake, distribution, metabol- ism, and excretion and may play multiple roles simultaneously depending on their localization, expression levels, and activity. Transporters are especially important in highly sensitive organs such as the CNS, where they are important determin- ants of drug pharmacodynamics [25]. CNS drug barriers Blood–brain barrier The blood–brain barrier (BBB) is the largest pharmacokinetic interfacewithintheCNS(about20m2 )[26],andthustheprimary focusofthe manyefforts to understand and controldrugaccess to the brain [27]. The BBB evolved due to the brain’s unique chem- ical sensitivity and requirement for the highest level of chemical protection. BBB drug transporters can profoundly influence the clinical effectiveness of drugs acting in the brain [13]. The BBB has a complex cellular makeup that is functionally integrated with surrounding brain tissue as part of the neuro- vascular unit [11,28]. The complete neurovascular unit com- prises the capillary vascular endothelium, pericytes, a basement membrane, closely associated astrocytic glia, and nearby neurons [29] (Fig. 7.4). The highly polarized vascular endothe- lium is the primary site of chemical exclusion. It possesses very tight lateral border junctions [30] and a diverse array of apically (i.e., vascular) facing efflux drug transporters, including P-gp, MRP1, and BCRP (Fig. 7.5). The functional importance of these transporters to drug partition in the brain has been studied using gene-specific genetic null mice (knockouts) [31–35]. In single transporter loss-of-function experiments, two- to ten-fold increases in CNS penetration of specific ABC transporter substrates including chemotherapeutics can result after IV administration [36]. That ABC transporters can so profoundly affect brain drug concentration by their presence or absence in a single endothelial layer suggests a plausible mechanism for increased drug sensitivity of the CNS in trans- porter-associated pharmacogenomic studies [17] (see P-gp, above). Furthermore these studies promote the idea of specif- ically targeting xenobiotic transporter function to more readily concentrate drugs in the brain for treating CNS illnesses such as brain cancer, multiple sclerosis, and Alzheimer’s disease [27]. Astrocytic glia form a continuous circumferential and close association with the basement membrane, further isolating the vascular and CNS interstitial spaces, but their role in chemical exclusion is unclear. The astroglial cell layer is less than a micron thick and lacks very tight junctions at cell–cell contacts, and xenobiotic transporters should therefore not directly affect CNS glial cell chemopartition. Indeed, genomic data show a much lower level of xenobiotic transporter and junctional pro- tein expression in the astroglia compared to vascular endothe- lium (R. Daneman and B. Barres, personal communication and [37]). Thus, while astrocytes are essential for BBB maintenance and function, their role in chemical protection may be to sense or integrate chemical, metabolic, and inflammatory stresses which must be coordinated to maintain proper physiologic balance for neurons [11]. Substantial efforts are under way to develop model systems and new methods to identify regulatory controls and chemoresponsive physiologies in the hope of specifically modulating drug barrier function at the BBB and ultimately improving clinical treatment options [27]. Blood-retinal barrier (BRB) Transport of molecules between the vitreous/retina and sys- temic circulation is restricted by the blood-retinal barrier (BRB, also called the retinal BBB or RBBB), composed of retinal pigment epithelium (RPE) and endothelial cells of the retinal blood vessels. The RPE maintains a tight barrier to diffusion to the interstitial fluid of the surrounding connective tissue of the eye. Vascular access to the retina is primarily through the retinal blood vessels, which penetrate with the optic nerve and run parallel to the retinal surface. The capillary bed of the retinal vascular endothelium maintains tight junc- tions and apically facing transporters morphologically similar to the BBB in the central brain. A unique feature of the eye is the ability to bypass the barrier by topical application or injection into the vitreous humor. However, while drug delivery to the CNS is uniquely possible in the eye, transport processes resident at the RBBB promote very rapid removal of small molecules into the systemic circulation. For example, common topical ophthalmic drugs such as erythromycin, dex- amethasone, and cyclosporine are substrates for prominent RBBB transporters and can be found in the systemic circula- tion after ocular administration [38]. These transporters include MDR1, MRP1, BRCP, and numerous OATPs [38]. Localized pharmacokinetic methods for measuring efflux across the BRB are limited; therefore reliable quantitative Section 1: Principles of drug action 96 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 8. estimation of the roles of inner and outer BRB has not been well established in humans. In rabbits, however, MDR1 pre- sent at the RBBB can be selectively modulated using traditional inhibitors such as quinidine and cyclosporine A [39]. Further- more, diffusion barrier studies have been carried out in mice, where labeled mannitol is used as a biomarker for retinal diffusion barrier integrity [40] and has shown diffusion-tight function of the BRB. Thus experimental models identify the BRB as an effective barrier similar to the BBB. Blood–cerebrospinal fluid barrier and choroid plexus The blood–cerebrospinal fluid barrier (B-CSFB) is derived from a special confluence of the pia mater and a single layer of CSF-facing glial cells (the ependyma). This meeting of two barrier layers is known as the choroid plexus and is the site of CSF production. Throughout the rest of the CNS, ependymal cells become diffusion-tight, utilizing the same modes of sep- aration discussed at the capillary–brain interface above. Here, therefore, chemical exclusivity of the CNS is maintained by an anatomically simpler system than at the BBB. The CSF pro- duction cycle is completed by rapid uptake at arachnoid granu- lations. Together, the CSF production and brain barrier system is thought to perform cleansing functions, further augmenting the chemical protection provided by the BBB [41]. Because the diffusion of polar substances at the B-CSFB is less hindered than at the BBB [41], drug administration into CSF has been proposed to circumvent the BBB and gain access to the CNS. However, potent chemical exclusion properties are present at the B-CSFB (i.e., ependymal cells), as at the choroid plexus [42]. Specifically, transporters OAT, OATP1, OATP2, MRP1, and MDR1 are present in high levels, suggesting an explanation for why the CSF has not proven to be a depend- able access point for the administration of chemotherapy or other drugs into the CNS. Nevertheless, certain drugs can overcome these barrier processes by a combination of favor- able penetration chemistry and poor transportability. For example, sodium-channel blockers such as bupivicaine are used for subarachnoid block. Gastrointestinal tract Intestinal drug uptake usually occurs by mass action and passive permeability when high concentrations are presented to the microvillus absorption surface. An empirical set of rules, known as Lipinski’s Rule of Fives, describes how small mol- ecules passively cross the lipid bilayer. Oral drugs can violate only one of the following criteria: (1) not more than five hydrogen bond donors, (2) not more than 10 hydrogen bond acceptors, (3) molecular weight under 500 Da, and (4) an octanol/water partition coefficient log P less than 5. Pharma- ceuticals that possess these properties are better at passing the chemoprotective interface of the gut, but these constraints do not allow small molecules to bypass the action of transporters. Xenobiotic transporters are abundantly represented throughout the enteral space (Fig. 7.5) [43,44]. P-gp is apically localized in microvilli throughout the gut epithelium, found in highest amounts in the ileum and jejunum, and counteracts the passive or active absorption of xenobiotics [25]. For instance, mouse P-gp knockouts demonstrate increased absorption and bioavailability of many oral xenobiotics [45], and uptake of some drugs is markedly increased by small molecules that inhibit P-gp function [16]. BCRP is the trans- porter expressed in highest abundance throughout the human small intestine [46]. In addition to countering the absorption of some orally administered drugs, certain transporter systems, particularly ABCC proteins, counter the absorption of drug glucuronides that are excreted in bile. Intestinal efflux trans- porters act in concert with intestinal phase I metabolism to limit drug bioavailability. Liver Hepatic drug elimination depends on passive or active hepatic uptake, intracellular metabolism, excretion of metabolites or parent drug into bile, and/or excretion of metabolites or parent drug into sinusoidal blood. Transporters are involved in uptake, biliary excretion, and sinusoidal efflux (Fig. 7.5) [23]. SLC-type transporters in the basolateral (sinusoidal) mem- brane (OATPs, OATs, OCTs) mediate the uptake of organic cations, anions, bilirubin, and bile salts. ABC transporters (P-gp, BCRP, MRPs) at the canalicular membrane mediate the efflux into bile of drugs, metabolites, and bile salts. MRPs may also mediate drug and metabolite efflux back into the blood. The balance of vectorial uptake and efflux drug trans- port together with intracellular metabolism, determines drug elimination. Hepatic drug transporters are subject to pharma- cogenetic variability, and drug–drug interactions. In murine models, transporter disruption can effect the distribution of important anesthetic drugs such as opiates. Morphine-3-glucuronide (M3G), the most abundant metab- olite of morphine, is transported in vitro by human MRP2 (ABCC2) that is present in the apical membrane of hepato- cytes. Loss of biliary M3G secretion in MRP2(-/-) mice results in its increased sinusoidal transport, which can be attributed to MRP3. Combined loss of MRP2 and MRP3 leads to a substantial accumulation of M3G in the liver, resulting in low rates of sinusoidal membrane transport and the pro- longed presence of M3G in plasma [47]. These results suggest that in vertebrates MRP2 and MRP3 transporters provide alternative routes for the excretion of a glucuronidated sub- strate from the liver in vivo. This illustrates the potential complexity of interactions between transporters and modify- ing enzymes intrinsic to particular chemoprotective environments. Kidney Transporters mediate active tubular secretion and reabsorp- tion in the kidney, which, together with glomerular filtration, determine drug elimination (Fig. 7.5). The primary focus of renal excretion is the proximal tubule, which actively takes up organic anions and cations at the basolateral membrane and Chapter 7: Drug transport and transporters 97 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 9. extrudes them across the apical brush border membrane into the urine; hence transepithelial flux requires two distinct sets of transporters. Many drugs (weak acids) and their phase II conjugates are organic anions. OATs 1 and 3 are the primary organic anion uptake transporters at the basolateral mem- brane (although they may also interact with neutral and cationic compounds), while efflux into the tubular lumen is mediated primarily by the ABC subfamily C transporters MRP2, MRP4, and BCRP [48]. Organic cations are typically transported into renal tubular cells at the basolateral mem- brane by OCTs 1 and 2, and effluxed into urine at the apical membrane by P-gp, OCT3, and novel organic cation trans- porters OCTN1 and OCTN2. Selective expression and spe- cific localization of transporters at different apical and basal interfaces in the tubule, pharmacogenetic variability in their activity, and drug interactions play a determinative role in drug elimination. Transporter-based drug interactions can occur in the kidney by competing for elimination. For example, probencid inhibits OATP-mediated excretion of antibiotics [49]. In add- ition, methotrexate has strong potential interactions with non- steroidal anti-inflammatory drugs (NSAIDs), particularly after prolonged high-dose chemotheraputic use [49]. Placenta The placenta is the sole medium of metabolic exchange between fetus and mother; hence it protects the fetus by providing a major obstacle to chemical penetration. The important chemical properties determining placental transfer by passive diffusion are similar to other organs: molecular weight, pKa of drug, lipid solubility, and plasma protein binding. The transport barrier in placenta is found in the syncytiotrophoblast, a multinucleated single cell layer that performs nutrient and waste exchange between mother and fetus [50]. This layer is further elaborated into two distinct interfaces: a maternal-facing brush border and a fetal-facing basal membrane [50]. It is in these two interfaces that a variety of membrane transporters perform directional fluxing of metabolites and drugs. Specifically, the brush border membrane contains all three major classes of ABC transporters (i.e., P-gp, MRP1–3, and BCRP) as well as specialized transporters such as the serotonin, norepinephrine, and carnitine transporters (SERT, NET, OCTN2) [50]. On the other side of the divide, the fetal- facing basal membrane contains transporters such as the organic anion transporters OAT4 and OATP-B in addition to the reduced folate transporter RFT-1 [50]. Combinations of genetic loss of P-gp function with specific inhibitors of BCRP markedly increase drug penetration of topetecan to the fetus, which is not seen in the genetically sensitized back- ground alone. Thus the placenta is an essential chemical filter for the fetal protected space and exemplifies the potential for overlapping and redundant function of chemoprotective mechanisms [51]. Transporter biology: special topics Drug transporter methods The field of transporter biology is relatively new, and a brief description of experimental methods used to identify the role of transporters in drug disposition may be helpful. Accurate physical measurement of localized drug concentration can be difficult in very compact biologic spaces like the BBB/CNS interface, and consequently methods of measurement require some explanation [52]. In-vitro methods are most commonly used to study human transporter activity and identify candidate transporters for a given substrate. Cell culture systems amenable to transfection of influx or efflux transporter genes, or direct injection of transporter mRNA into Xenopus oocytes, express and localize transporter proteins to the cell surface. Isolated membrane vesicles can also be prepared. Exposure to drug on one side of a cell layer, then measurement on both sides, to determine the ratio of substrate penetration to negative controls or by using transporter inhibitors, allows individual assessment of luminal and abluminal transporters and their mechanisms. Unfortunately, heterologous systems have several problems when attributing a specific physiologic function to a trans- porter, because it is specifically overexpressed. Studies using membranes of confluent primary human cells can provide more information, particularly on the contribution of relevant transporters for a specific drug. Ex-vivo assays, however, lack functional context since organ-specific gross anatomy and transporter interface conditions do not mimic the animal. In addition, the transporter must function away from its normal molecular constituents, and thus potential modulators of sub- strate preference and functional compensations are lost. In animals (and occasionally in humans) ex-vivo or in-situ organ perfusion allows direct measurement of drug influx and efflux concentrations. Advantages of this method are a close control of the time course of drug administration and concen- tration. Furthermore, drug goes directly from perfusate to the brain (or other organ studied), bypassing systemic biotrans- formation. Although this experimental methodology is more precise, it still requires bulk tissue concentration measurement, and hence does not provide data about discrete localization in the brain (or other organ). Calculation of the brain efflux index can be used as an indirect measure of chemical partition at the BBB. A drug is injected directly into the brain with an impermanent tracer. The ratio of test solute to the reference tracer determines the brain efflux activity. An improved method is tissue (often intracerebral) microdialysis. This method directly samples small-molecule solutes in brain inter- stitial fluid through a semiporous membrane. Microdialysis is difficult and costly to perform, however, and may have its own artifacts. Transgenic rodent models using knockouts or knock- ins of transporters are a common approach to identifying transporter function with respect to drug disposition and Section 1: Principles of drug action 98 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 10. effect. However, there is still a great deal of argument sur- rounding the similarity of physiologic function even between highly homologous vertebrate systems. In humans, pharmacogenomic approaches often attempt to identify transporter polymorphisms which may alter pharma- cokinetics or pharmacodynamics. The use of drug probes to selectively inhibit a transporter’s function, and assess the effect on pharmacokinetics or pharmacodynamics, is limited by the lack of transporter-selective inhibitor probes. This is unlike the methods used to profile cytochrome P450s in drug metabol- ism, where P450-selective inhibitor drugs are available. Drug–drug interactions and transporters Drugs themselves can act to modulate xenobotic transport, and any new pharmaceutical is a potential effector of chemical partition. While the number of current examples is small, as new therapeutics are developed the opportunity for drug–drug interactions through transporter inhibition becomes increas- ingly problematic. This issue is further exacerbated by the wide tissue distribution of transporters like P-gp, which makes it difficult to predict the potential for affecting drug distribution and efficacy in a site-dependent fashion. As noted above, P-gp loss of function can affect both brain and gut uptake depending on the drug in question [36,45]. This is not just a theoretical problem but occurs with many commonly used medications. For instance, verapamil and the cyclosporine analog PSC833 lead to increased brain concentration of anti- epileptics such as carbamazapine, phenytoin, and phenobarbi- tol [13,53]. The combination of quercetin, a P-gp substrate, and digoxin also has demonstrated toxicity [53]. Lastly, b-blockers such as talinolol exhibit increased oral bioavailability when intestinal P-gp activity is reduced in both animal models and human observations [53]. Unfortunately there is no set of chemoprotective rules that can warn clinicians of potential inter- actions. Currently all drug–drug interactions are determined empirically, and hence the need for a more complete analysis of the organization of chemical protection in the animal. Modulating transporter function in vivo for clinical purposes The BBB is a dynamic structure regulated by many competing inputs [11]. In vertebrates, transcellular diffusion can be modi- fied by adjusting either the level or the activity of transporter proteins [54]. Transcription of a number of efflux transporters including P-gp, MRP2, MRP3, and OATP2 is regulated in part by the pregnane X receptor (PXR), a nuclear receptor that is activated by naturally occurring steroids as well as a wide variety of xenobiotics [55–57]. Higher levels of transporter expression result in more efficient xenobiotic efflux and a tighter BBB [58]. The activity of some transporters can also be influenced by cell-signaling pathways. For example, binding of endothelin 1 to the ETB receptor rapidly and reversibly inhibits transport activity of P-gp via nitric oxide synthase and protein kinase-C mediated pathways [59]. ABC transporter chemopartition function can also be modulated by drugs. In this case one drug acts as competitive substrate antagonist against more favored transport molecules (Table 7.1). This explains why many drugs are listed as both substrate and antagonist in substrate/inhibitor tables. Several clinical scenarios have attempted to introduce drugs like “selective” transport inhibitors at the BBB interface to improve penetration of chemotherapeutics, but no clinical benefit has yet been obtained [13,60]. Nevertheless the search for more selective competitive inhibitors or allosteric modulators of ABC transporters is quite active in both academic and indus- trial research [8]. Future directions in the study of chemical protection The ability of an organism to protect itself against environ- mental threats, whether predation or chemical insult, is as essential as the ability to convert food into energy. Chemical transporters of the variety discussed in this chapter are present across evolutionary time, with the essential structure of pro- teins such as members of the ABC superfamily changing very little across phyla. For example, there is increasing evidence that species as distant as fruit flies and humans use very similar means to exclude xenobiotics from privileged compartments such as the CNS [61]. The decoding of the fruit fly Drosophila melanogaster genome has enabled investigators to identify genes and proteins that are conserved across species, including analogs of many of the ABC transporters relevant to human physiology and pathology (Table 7.1) [10]. While the compon- ent parts of chemoprotective systems are just coming to light (i.e., transporters and metabolic enzymes that are chemically organized), there is little understanding of the regulation and adaptations such a system requires. Model systems using human cell culture, animal models, and the tools of pharma- cogenomics will play a large role in furthering the understanding of chemical protection. Once there are better understandings of localized cellular or subcellular drug concentrations and identifi- cation of novel modulators of chemical partition, better methods of therapeutic drug delivery can be developed to improve clinical outcomes. Summary Biophysical boundaries prevent passive diffusion of large mol- ecules, such as proteins and nucleic acids, between different compartments and greatly curtail the movement of electro- lytes, soluble components of metabolism, and drugs. An enor- mous variety of transmembrane protein transporters are dedicated to selectively moving small molecules across mem- branes. Transporters are essential actors in almost every bio- logic function, as transporter systems create the chemical preconditions for cellular function, restore chemical condi- tions after perturbation, and clear chemical space of unwanted metabolites. The precision, specificity, and ability of these Chapter 7: Drug transport and transporters 99 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 11. transport systems to act against concentration gradients allow small molecules to have discrete functions depending on their localization. Drugs share the same molecular weight range and solubility characteristics as many endogenous small molecules and are subject to the same physiologic processes, and the same transporters, as endogenous molecules. Drug (xenobiotic) transporters were initially identified during studies of multidrug cancer resistance. These proteins were efflux pumps with broad substrate specificity, capable of binding and transporting a wide spectrum of chemotherapeu- tics back into the extracellular space, thereby reducing the effective intracellular concentration of cytotoxic drugs. Cancer cells had coopted and enhanced an endogenous chemical par- tition system to strengthen the cell-membrane chemical barrier and confer drug resistance. Cell-surface transporters are now known to be ubiquitous, present across evolutionary time with relatively conserved functions across species, and capable of influx and/or efflux transport of numerous drugs. P-glycoprotein (abbreviated P-gp, and also termed ABCB1 or the multidrug resistance transporter, MDR1) was the first xenobiotic transporter dis- covered in multidrug-resistant cancer cells. P-gp also functions in a wide range of normal tissues such as the gastrointestinal tract, renal proximal tubule cells, the canalicular surfaces of hepatocytes, the capillary endothelial cells of testes, and the blood–brain barrier. Transporters may influence drug absorp- tion, distribution, tissue penetration, metabolism, and excre- tion. There is also the potential for significant drug interactions. How these competing transporter processes are regulated, coordinated, and potentially circumvented or controlled will require a better understanding of drug–transporter inter- actions at cellular interfaces and methods for analyzing net localized pharmacokinetic disposition of drugs. For a drug to be clinically effective, it must possess high target specificity, favorable organismal pharmacokinetics, and complementary properties for highly localized chemical partition processes at the cellular level. Drugs themselves can act to modulate xeno- botic transport, and any new pharmaceutical is a potential effector of chemical partition. References 1. Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the Cell. New York, NY:Garland, 2007. 2. van de Waterbeemd H, Gifford E. ADMET in silico modelling: towards prediction paradise? Nat Rev Drug Discov 2003; 2: 192–204. 3. Hediger MA, Romero MF, Peng JB, et al. The ABCs of solute carriers: physiological, pathological and therapeutic implications of human membrane transport proteins Introduction. Pflugers Arch 2004; 447: 465–8. 4. Torres GE, Gainetdinov RR, Caron MG. Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci 2003; 4: 13–25. 5. Torrie LS, Radford JC, Southall TD, et al. Resolution of the insect ouabain paradox. Proc Natl Acad Sci U S A 2004; 101: 13689–93. 6. Dano K. Active outward transport of daunomycin in resistant Ehrlich ascites tumor cells. Biochim Biophys Acta 1973; 323: 466–83. 7. Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta 1976; 455: 152–62. 8. Sarkadi B, Homolya L, Szakacs G, Varadi A. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiol Rev 2006; 86: 1179–236. 9. Annilo T, Chen ZQ, Shulenin S, et al. Evolution of the vertebrate ABC gene family: analysis of gene birth and death. Genomics 2006; 88: 1–11. 10. Dean M, Annilo T. Evolution of the ATP- binding cassette (ABC) transporter superfamily in vertebrates. Annu Rev Genomics Hum Genet 2005; 6: 123–42. 11. Zlokovic BV. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron 2008; 57: 178–201. 12. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002; 2: 48–58. 13. Loscher W, Potschka H. Blood–brain barrier active efflux transporters: ATP- binding cassette gene family. NeuroRx 2005; 2: 86–98. 14. Dobson PD, Kell DB. Carrier-mediated cellular uptake of pharmaceutical drugs: an exception or the rule? Nat Rev Drug Discov 2008; 7: 205–20. 15. Zhang L, Zhang YD, Strong JM, Reynolds KS, Huang SM. A regulatory viewpoint on transporter-based drug interactions. Xenobiotica 2008; 38: 709–24. 16. Sadeque AJ, Wandel C, He H, Shah S, Wood AJ. Increased drug delivery to the brain by P-glycoprotein inhibition. Clin Pharmacol Ther 2000; 68: 231–7. 17. Park HJ, Shinn HK, Ryu SH, et al. Genetic polymorphisms in the ABCB1 gene and the effects of fentanyl in Koreans. Clin Pharmacol Ther 2007; 81: 539–46. 18. Kruh GD, Belinsky MG. The MRP family of drug efflux pumps. Oncogene 2003; 22: 7537–52. 19. Tsuji A. Impact of transporter-mediated drug absorption, distribution, elimination and drug interactions in antimicrobial chemotherapy. J Infect Chemother 2006; 12: 241–50. 20. Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci 2006; 27: 425–46. 21. Doyle LA, Yang W, Abruzzo LV, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 1998; 95: 15665–70. 22. Petrovic V, Teng S, Piquette-Miller M. Regulation of drug transporters during infection and inflammation. Mol Interv 2007; 7: 99–111. 23. Kim RB. Organic anion-transporting polypeptide (OATP) transporter family and drug disposition. Eur J Clin Invest2003; 33 (Suppl 2): 1–5. Section 1: Principles of drug action 100 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 12. 24. Abbott NJ. Dynamics of CNS barriers: evolution, differentiation, and modulation. Cell Mol Neurobiol 2005; 25: 5–23. 25. Schinkel AH, Jonker JW. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview. Adv Drug Deliv Rev 2003; 55: 3–29. 26. Begley DJ, Brightman MW. Structural and functional aspects of the blood– brain barrier. Prog Drug Res 2003; 61: 39–78. 27. Neuwelt E, Abbott NJ, Abrey L, et al. Strategies to advance translational research into brain barriers. Lancet Neurol 2008; 7: 84–96. 28. McCarty JH. Cell biology of the neurovascular unit: implications for drug delivery across the blood-brain barrier. Assay Drug Dev Technol 2005; 3: 89–95. 29. Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis 2004; 16: 1–13. 30. Reese TS, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J Cell Biol 1967; 34: 207–17. 31. Wijnholds J, Evers R, van Leusden MR, et al. Increased sensitivity to anticancer drugs and decreased inflammatory response in mice lacking the multidrug resistance-associated protein. Nat Med 1997; 3: 1275–9. 32. Lorico A, Rappa G, Flavell RA, Sartorelli AC. Double knockout of the MRP gene leads to increased drug sensitivity in vitro. Cancer Res 1996; 56: 5351–5. 33. Schinkel AH, Smit JJ, van Tellingen O, et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 1994; 77: 491–502. 34. Vlaming ML, Lagas JS, Schinkel AH. Physiological and pharmacological roles of ABCG2 (BCRP): recent findings in Abcg2 knockout mice. Adv Drug Deliv Rev 2009; 61: 14–25. 35. Enokizono J, Kusuhara H, Ose A, Schinkel AH, Sugiyama Y. Quantitative investigation of the role of breast cancer resistance protein (Bcrp/Abcg2) in limiting brain and testis penetration of xenobiotic compounds. Drug Metab Dispos 2008; 36: 995–1002. 36. Schinkel AH. Pharmacological insights from P-glycoprotein knockout mice. Int J Clin Pharmacol Ther 1998; 36: 9–13. 37. Cahoy JD, Emery B, Kaushal A, et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 2008; 28: 264–78. 38. Mannermaa E, Vellonen KS, Urtti A. Drug transport in corneal epithelium and blood–retina barrier: emerging role of transporters in ocular pharmacokinetics. Adv Drug Deliv Rev 2006; 58: 1136–63. 39. Kajikawa T, Mishima HK, Murakami T, Takano M. Role of P-glycoprotein in distribution of rhodamine 123 into aqueous humor in rabbits. Curr Eye Res 1999; 18: 240–6. 40. Derevjanik NL, Vinores SA, Xiao WH, et al. Quantitative assessment of the integrity of the blood-retinal barrier in mice. Invest Ophthalmol Vis Sci 2002; 43: 2462–7. 41. Johanson CE, Duncan JA, Stopa EG, Baird A. Enhanced prospects for drug delivery and brain targeting by the choroid plexus-CSF route. Pharm Res 2005; 22: 1011–37. 42. Emerich DF, Skinner SJ, Borlongan CV, Vasconcellos AV, Thanos CG. The choroid plexus in the rise, fall and repair of the brain. Bioessays 2005; 27: 262–74. 43. Dietrich CG, Geier A, Wasmuth HE, et al. Influence of biliary cirrhosis on the detoxification and elimination of a food derived carcinogen. Gut 2004; 53: 1850–5. 44. Kusuhara H, Sugiyama Y. Role of transporters in the tissue-selective distribution and elimination of drugs: transporters in the liver, small intestine, brain and kidney. J Control Release 2002; 78: 43–54. 45. Lagas JS, Sparidans RW, van Waterschoot RA, et al. P-glycoprotein limits oral availability, brain penetration, and toxicity of an anionic drug, the antibiotic salinomycin. Antimicrob Agents Chemother 2008; 52: 1034–9. 46. Englund G, Rorsman F, Rönnblom A, et al. Regional levels of drug transporters along the human intestinal tract: co-expression of ABC and SLC transporters and comparison with Caco-2 cells. Eur J Pharm Sci 2006; 29: 269–77. 47. van de Wetering K, Zelcer N, Kuil A, et al. Multidrug resistance proteins 2 and 3 provide alternative routes for hepatic excretion of morphine- glucuronides. Mol Pharmacol 2007; 72: 387–94. 48. El-Sheikh AA, Masereeuw R, Russel FG. Mechanisms of renal anionic drug transport. Eur J Pharmacol 2008; 585: 245–55. 49. Inui KI, Masuda S, Saito H. Cellular and molecular aspects of drug transport in the kidney. Kidney Int 2000; 58: 944–58. 50. Ganapathy V, Prasad PD. Role of transporters in placental transfer of drugs. Toxicol Appl Pharmacol 2005; 207: 381–7. 51. Jonker JW, Smit JW, Brinkhuis RF, et al. Role of breast cancer resistance protein in the bioavailability and fetal penetration of topotecan. J Natl Cancer Inst 2000; 92: 1651–6. 52. Smith QR. A review of blood–brain barrier transport techniques. In: Nag S, ed., Methods in Molecular Medicine. Totowa, NJ:Humana Press, 2003: 193–208. 53. Balayssac D, Authier N, Cayre A, Coudore F. Does inhibition of P- glycoprotein lead to drug–drug interactions? Toxicol Lett 2005; 156: 319–29. 54. Miller DS, Bauer B, Hartz AM. Modulation of P-glycoprotein at the blood-brain barrier: opportunities to improve central nervous system pharmacotherapy. Pharmacol Rev 2008; 60: 196–209. 55. Kliewer SA, Goodwin B, Willson TM. The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocr Rev 2002; 23: 687–702. 56. Kliewer SA, Moore JT, Wade L, et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 1998; 92: 73–82. Chapter 7: Drug transport and transporters 101 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at
  • 13. 57. Bauer B, Yang X, Hartz AM, et al. In vivo activation of human pregnane X receptor tightens the blood–brain barrier to methadone through P-glycoprotein up-regulation. Mol Pharmacol 2006; 70: 1212–19. 58. Perez-Tomas R. Multidrug resistance: retrospect and prospects in anti-cancer drug treatment. Curr Med Chem 2006; 13: 1859–76. 59. Hartz AM, Bauer B, Fricker G, Miller DS. Rapid regulation of P-glycoprotein at the blood–brain barrier by endothelin-1. Mol Pharmacol 2004; 66: 387–94. 60. Kemper EM, Boogerd W, Thuis I, Beijnen JH, van Tellingen O. Modulation of the blood-brain barrier in oncology: therapeutic opportunities for the treatment of brain tumours? Cancer Treat Rev 2004; 30: 415–23. 61. Mayer F, Mayer N, Chinn L, et al. Evolutionary conservation of vertebrate blood–brain barrier chemoprotective mechanisms in Drosophila. J Neurosci 2009; 29: 3538–50. Section 1: Principles of drug action 102 https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9780511781933.008 Downloaded from https://www.cambridge.org/core. Kaohsiung Medical University, on 08 Mar 2018 at 17:20:55, subject to the Cambridge Core terms of use, available at