ArticlePDF Available

Factors That Limit Positron Emission Tomography Imaging of P-Glycoprotein Density at the Blood-Brain Barrier

Authors:

Abstract and Figures

Efflux transporters located at the blood-brain barrier, such as P-gp and BCRP, regulate the passage of many drugs in and out of the brain. Changes in the function and density of these proteins, in particular P-gp, may play a role in several neurological disorders. Several radioligands have been developed for measuring P-gp function at the blood-brain barrier of human subjects with positron emission tomography (PET). However, attempts to measure P-gp density with radiolabeled inhibitors that bind to these proteins in vivo have not thus far provided useful, quantifiable PET signals. Herein, we argue that not only the low density of transporters in the brain as a whole but also their very high density in brain capillaries act to lower the concentration of ligand in the plasma and thereby contribute to absent or low signals in PET studies of P-gp density. Our calculations, based on published data and theoretical approximations, estimate that whole brain densities of many efflux transporters at the blood-brain barrier range from 0.04 to 5.19 nM. We conclude that the moderate affinities (> 5 nM) of currently labeled inhibitors may not allow measurement of efflux transporter density at the blood-brain barrier, and inhibitors with substantially higher affinity will be needed for density imaging of P-gp and other blood-brain barrier transporters.
Content may be subject to copyright.
Factors That Limit Positron Emission Tomography Imaging of
PGlycoprotein Density at the BloodBrain Barrier
Pavitra Kannan,
,
Victor W. Pike,
Christer Halldin,
Oliver Langer,
§,
Michael M. Gottesman,
Robert B. Innis,*
,
and Matthew D. Hall
Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, United
States
Department of Clinical Neuroscience, Karolinska Institutet, 17176 Stockholm, Sweden
§
Health and Environment Department, Biomedical Systems, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria
Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
ABSTRACT: Eux transporters located at the bloodbrain
barrier, such as P-glycoprotein (P-gp) and breast cancer
resistance protein (BCRP), regulate the passage of many drugs
in and out of the brain. Changes in the function and density of
these proteins, in particular P-gp, may play a role in several
neurological disorders. Several radioligands have been
developed for measuring P-gp function at the bloodbrain
barrier of human subjects with positron emission tomography
(PET). However, attempts to measure P-gp density with
radiolabeled inhibitors that bind to these proteins in vivo have
not thus far provided useful, quantiable PET signals. Herein,
we argue that not only the low density of transporters in the
brain as a whole but also their very high density in brain capillaries act to lower the concentration of ligand in the plasma and
thereby contribute to absent or low signals in PET studies of P-gp density. Our calculations, based on published data and
theoretical approximations, estimate that whole brain densities of many eux transporters at the bloodbrain barrier range from
0.04 to 5.19 nM. We conclude that the moderate anities (>5 nM) of currently labeled inhibitors may not allow measurement of
eux transporter density at the bloodbrain barrier, and inhibitors with substantially higher anity will be needed for density
imaging of P-gp and other bloodbrain barrier transporters.
KEYWORDS: positron emission tomography (PET), ABC transporters, imaging, bloodbrain barrier, inhibitors, density
INTRODUCTION
P-Glycoprotein (P-gp, encoded by ABCB1) and breast cancer
resistance protein (BCRP, encoded by ABCG2) are among the
eux transporters that are essential to the function of the blood
brain barrier.
1
These two transporters are apically located
(blood-facing) in the endothelial cells of brain capillaries to
prevent the entry of drugs into the brain, thereby protecting it
from exposure to toxins in the blood.
2,3
As a result, P-gp and
BCRP can also impede the entry of potential therapeutics to the
brain, and this barrier function may be exacerbated in
pathophysiological conditions. Increased function of P-gp, for
example, may contribute to drug resistance in epilepsy and may
decrease the eectiveness of treating HIV infection of the
brain.
4,5
Changes in expression (i.e., density) of these trans-
porters may also be clinically relevant because expression aects
eux capacity. For example, loss of P-gp expression would result
in net decrease in function and, consequently, in dysfunction of
the bloodbrain barrier. Because the function and density of P-
gp and BCRP in neurological disorders are not well understood
in vivo, quantifying both transporter function and density is a
signicant and important challenge.
Molecular imaging techniques such as positron emission
tomography (PET) oer the potential for in vivo measurement of
function and density of protein targets. In PET, a radiotracer is
injected at a subpharmacological dose, and biomathematical
modeling is applied to acquired data to determine output
measures related to the interaction of the radiotracer with the
receptor target. In the case of functional studies, increased or
decreased uptake of a radiotracer measures protein function.
Glucose metabolism in tissue, for example, is reected by
increased uptake of [18F]uorodeoxyglucose.
6
Eux transport
by P-gp or BCRP, on the other hand, is reected by little to no
uptake of the radiolabeled substrate in tissue, and inhibition of
eux transport results in increased substrate accumulation.
1
PET
Received: January 8, 2013
Revised: March 27, 2013
Accepted: April 18, 2013
Published: April 18, 2013
Review
pubs.acs.org/molecularpharmaceutics
© 2013 American Chemical Society 2222 dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 22222229
has been successful in using radiolabeled substrates to measure P-
gp function at the bloodbrain barrier for at least two reasons.
First, each molecule of P-gp can transport multiple substrate
molecules. Second, upon inhibition of transport, amplication of
the PET signal can occur by trapping some of the substrate in
cellular organelles.
7,8
In the case of density studies, the density of the target is
inferred from the binding potentiala parameter that is the
product of the concentration of binding sites (Bmax) and the
anity of the radioligand for the protein (1/KD, where KDis the
equilibrium dissociation constant).
9
[The term radioligand
describes a specic class of compounds that reversibly or
irreversibly bind to the protein target (e.g., radiolabeled
inhibitor), whereas the term radiotracer refers to a more general
class of compounds that includes substrates and radioligands.]
Because substrates are transported quickly when in the vicinity of
a transporter,
10
they cannot be used to measure the density of
these eux transporters by PET. Inhibitors of P-gp or of BCRP,
on the other hand, are known to directly bind to their respective
transporters
10
and might behave akin to antagonist receptor
radioligands, thereby measuring density. However, PET has been
unsuccessful at measuring P-gp density using radiolabeled
inhibitors. Unlike the signal amplication obtained by substrate
trapping, the maximal signal that can be obtained by binding is
one radiolabeled inhibitor for each transporter (assuming one
binding site per transporter). The diculty in imaging P-gp
density is also due to two previously described phenomena. First,
many of the inhibitors cross-react with P-gp and BCRP,
11
which
confounds results obtained in P-gp knockout, Bcrp knockout, or
P-gp/Bcrp knockout mice. Second, some inhibitors of P-gp are
also substrates for BCRP.
12
At very low concentrations, high
anity inhibitors might be transported by P-gp, but evidence for
this appears to be conicting.
13,14
The purpose of this perspective is to highlight two other
diculties of imaging P-gp density at the bloodbrain barrier
using PET. The rst is that the density of P-gp in the whole brain
has been variably reported in the literature but plays a critical role
in the anity of the radioligand required to have a measurable
signal from in vivo PET imaging. This may be exacerbated by the
low resolution of PET imaging. The second diculty, which has
not been discussed in the literature, is the high density of P-gp
within the local microenvironment (microcompartment) of the
capillary. We propose that the density of P-gp (and BCRP) is
high enough within the capillary compartment to substantially
aect the free concentration of radiotracer during its 12 s transit
through the capillaries. Here, we use the results from PET studies
of P-gp density to explain how these two factorslow binding
potential and high, localized transporter densitymay greatly
aect brain signal.
In the following sections, we assume for simplicity that: (1)
The brain signal measured in PET studies of P-gp density is
binding to P-gp. In reality, brain uptake of radioactivity
includes both parenchymal uptake (after passing the blood
brain barrier) and radioactivity in the vascular compartment
(about 5% of total brain volume (see below)). PET studies seek
to determine the amount of radiolabeled inhibitor bound to P-gp
in the brain capillaries. However, PET lacks the resolution to
separately measure uptake in parenchyma and that in the vascular
compartment. (2) A radiolabeled inhibitor can bind to all the P-
gp in brain endothelial cells, but the majority of P-gp is localized
at the luminal membrane. While P-gp is constantly recycled on/
othe cell surface, the majority is thought to be expressed at the
cell surface.
RESULTS FROM PET STUDIES MEASURING P-GP
DENSITY
PET studies using high-anity P-gp/BCRP inhibitors to
measure transporter density at the bloodbrain barrier have
yielded confusing results (Table 1). According to conventional
PET studies measuring receptor density, the specic binding of a
radioligand to its receptor should be displaceable.
9
If we assume
that a radiolabeled inhibitor binds only to P-gp (i.e., is specic),
then high concentrations of unlabeled inhibitor should displace
the radioligand bound to P-gp and consequently decrease the
measured brain signal. However, brain uptake of all except one of
the tested radiolabeled inhibitors was low 30 min after injection
in wild-type rats but increased at least 150% after blockade with
high doses of nonradiolabeled inhibitors; the increase was not
due to altered peripheral metabolism after blockade.
1519
Results
obtained from transgenic mice are also not consistent with
expectations for a P-gp binding molecule. For example, brain
uptake of the high-anityP-gp inhibitor [11C]tariquidar was
low in wild-type, P-gp (mdr1a//mdr1b/6), and Bcrp(abcg26)
knockout mice but was high in P-gp/Bcrp (mdr1a//mdr1b//
Table 1. Brain Concentrations of Radiolabeled P-gp and BCRP Inhibitors in Rat and Mouse Measured 30 min after Injection
radiotracer uptake in rat brain at
25 min radiotracer uptake in mouse brain at 30 min
radiolabeled inhibitor baseline blocked unit
a
wild-type P-gp knockout BCRP knockout P-gp/BCRP knockout unit reference
[11C]laniquidar 0.07 0.6 %ID/g 18
[11C]elacridar 0.05 0.15 0.05 1.0 SUV 22
0.20 1.08 SUV 0.18 0.46 0.20 1.39 SUV 16
1-[18F]uoroelacridar 0.14 1.29 SUV 0.23 1.76 SUV 17
[18F]uoroethylelacridar 0.08 0.18 0.08 0.70 SUV 21
[11C]tariquidar 0.02 0.04 0.02 0.75 SUV 20
0.19 0.54 SUV 0.15 0.31 0.18 1.39 SUV 15
[18F]uoroethyltariquidar 0.07 0.14 0.10 0.60 SUV 21
[11C]-1
b
1.2 0.90 SUV 19
[11C]-2
c
0.96 1.17 0.80 1.79 SUV 23
a
%ID/g = % injected dose/gram of tissue. SUV = standardized uptake value, which is the ratio of radioactivity concentration measured in tissue at
time t to injected dose at time of injection divided by body weight. A SUV of 1.0 is the value that would be obtained throughout the body for a
hypothetical perfectly uniform distribution.
b
1 = novel P-gp inhibitor, [11C]6,7-dimethoxy-2-{3-[4-methoxy-3,4-dihydro-2Hnaphthalene-(1E)-
ylidene]-propyl}-1,2,3,4-tetrahydro-isoquinoline
c
2 = putative BCRP inhibitor, [11C]methyl 4-((4-(2-(6,7-dimethoxy-1,2,3,4-tetrahydroisoqunolin-2-
yl)ethyl)phenyl)amino-carbonyl)-2-(quinoline-2-carbonylamino)benzoate
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292223
abcg26) triple knockout mice.
15,20
Tariquidar is a substrate for
BCRP,
12
which explains its low uptake in wild-type and P-gp
knockout mice. However, one would have expected higher
uptake in Bcrp knockout mice because P-gp is still expressed in
brain capillaries and can bind to [11C]tariquidar. Brain uptake of
other radiolabeled inhibitors followed a similar pattern in the
four strains of mice.
16,17,21,22
Although extensive work has been done on P-gp inhibitors,
only one putative BCRP inhibitor has been radiolabeled and
tested in animals. The brain uptake of this 11C-labeled compound
(chemical name [11C]methyl 4-((4-(2-(6,7-dimethoxy-1,2,3,4-
tetrahydroisoquinolin-2-yl)ethyl)phenyl)amino-carbonyl)-2-
(quinoline-2-carbonylamino)benzoate
23
) in wild-type mice was
high but was even higher in P-gp and P-gp/BCRP knockout
mice, similar to results obtained with most of the P-gp
inhibitors.
23
Among these eight reported ligands, only one inhibitor
showed a dierent uptake prole. The P-gp inhibitor [11C]-1
(chemical name 6,7-dimethoxy-2-{3-[4-methoxy-3,4-dihydro-
2Hnaphthalene-(1E)-ylidene]-propyl}-1,2,3,4-tetrahydro-iso-
quinoline
19
) had a high brain uptake (>1.0 SUV) in rats at
baseline conditions and a 30% reduced uptake after blockade of
transporters.
19
While these results are what one might expect
from a PET study measuring eux transporter density, they
should be interpreted cautiously. The inhibitor [11C]-1 is
approximately 2 orders of magnitude less potent than tariquidar
and elacridar
24
and may not, therefore, have the anity required
to specically measure the density of P-gp at the bloodbrain
barrier (see discussion below).
TWO FACTORS AFFECT BRAIN SIGNAL IN PET
STUDIES OF P-GP DENSITY
Low Binding Potential of Eux Transporters. Even after
blockade or genetic knockout of transporters, the raw values of
brain uptake for most of the radiolabeled inhibitors are still lower
(<1.8 standardized uptake value, SUV, Table 1) than those
typically achieved with PET radioligands that bind to a specic
receptor in the brain.
25
The fact that little or no brain signal was
observed in conditions where P-gp is expressed and its density
should be measurable suggests that either the density of P-gp in
the whole brain (Bmax) is not sucient to allow visualization by
PET or that the anity (1/KD) of the inhibitor is not high
enough. In other words, while an inhibitor may bind to P-gp at
the bloodbrain barrier, binding in the whole brain is not
detectable by PET because the binding potential (Bmax/KD)of
the whole brain is too low. For a radioligand to have measurable
brain uptake, the Bmax should exceed the KDof the ligand [A value
of >5 for binding potential (Bmax/KD) is generally recommended
to achieve measurable brain uptake; however, the eld lacks
consensus on this value]: if a binding site has nanomolar
concentrations in vivo, then the radioligand should ideally have
subnanomolar anity.
26
What, then, is the binding potential for
P-gp and a high-anity inhibitor (e.g., tariquidar) in the whole
brain?
Estimation of Eux Transporter Density in the Whole Brain
(Bmax). To calculate the binding potential, an estimate of P-gp
density in the whole brain (Bmax) is required. Although the
anities of radiolabeled inhibitors for P-gp have been
experimentally determined, the Bmax has not [reports have
been published on P-gp expression in cells of the brain
parenchyma expressing P-gp (particularly during neuroinam-
mation) and in inltrating lymphocytes]. However, for the
purposes of this discussion we will assume that P-gp expression in
Table 2. Calculated Values of Receptor Density for Various Eux Transporters Expressed in Human, Monkey, and Mouse Brains
a
human monkey mouse
gene protein protein conc.
(fmol/μg) calculated Bmax
(nM) protein conc.
(fmol/μg) calculated Bmax
(nM) protein conc.
(fmol/μg) calculated Bmax
(nM)
ABC Transporters
ABCA2 2.86 0.61 NR ULQ
ABCA8 1.21 0.26 NR ULQ
ABCB1 P-gp 6.06 1.28 4.71 1.00 14.1 2.99
ABCC4 Mrp4 0.195 0.04 0.286 0.06 1.59 0.34
ABCG2 Bcrp 4.07 0.86 7.10 1.51 2.20 0.47
SLC Transporters
SLC1A3 EAAT1 24.5 5.19 NR NR
SLC2A1 GLUT1 139 29.47 129 27.35 90.0 19.08
SLC2A3 GLUT3 4.40 0.93 1.22 0.26 ULQ
SLC3A2 4F2hc 3.47 0.74 NR 16.4 3.48
SLC6A12 BGT1 3.16 0.67 NR NR
SLC7A1 CAT1 1.13 0.24 NR NR
SLC7A5 LAT1 0.431 0.09 ULQ 2.19 0.46
SLC16A1 MCT1 2.27 0.48 0.834 0.18 23.7 5.02
SLC19A1 RFC 0.763 0.16 NR NR
SLC29A1 ENT1 0.568 0.12 0.541 0.11 0.985 0.21
Receptors
INSR insulin receptor 1.09 0.23 1.52 0.32 1.16 0.25
LRP1 Lrp1 1.51 0.32 1.29 0.27 1.07 0.23
Tf R1 transferrin
receptor 2.34 0.50 NR 5.84 1.24
a
ABCG2/BCRP values are half that reported in Uchida and colleagues,
29
to account for the homodimerization of protein product to form a
functional unit. ULQ = Under limit quantication. NR = Gene not reported/examined.
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292224
the brain occurs in the brain capillary endothelial cells only.
Ordinarily, in vitro studies with brain homogenate are used to
determine an approximate Bmax value,
9
but the high lipophilicity
of the P-gp inhibitors (e.g., clog Ptariquidar = 6)
27
has made it
dicult to estimate this value using this method. Instead, we use
the values published recently by Terasaki and co-workers, who
measured the expression of transporters in isolated brain
capillary endothelial cells using quantitative liquid chromatog-
raphy-tandem mass spectrometry. This method allows for
measurement of protein levels, given as fmol/μg of cellular
protein in endothelial cells isolated from brain capillaries of
mice,
28
monkeys,
29
and humans
30
(summarized in Table 2).
From these studies, we know the concentration of transporters
in human brain capillary endothelial cells, but not in the whole
brain, which is necessary to calculate Bmax. Therefore, the
calculation of the concentration of a protein expressed in
capillary endothelial cells in the whole brain is critically reliant on
knowledge of the density of brain capillary endothelial cells as a
proportion of total brain tissue. This is distinct from the total
brain capillary volume, which is much higher, as it includes blood
volume. Blood volume in the brain is reported to be
approximately 5% of total brain volume,
31,32
but the capillary
volume is signicantly lower. A three-dimensional reconstruction
of brain capillaries from frozen serial sections of feline temporo-
parietal SuprasylvianGyrus revealed an average capillary
diameter of 4.68 ±0.92 μm, total capillary length of 42.55 cm/
mm2of brain tissue, and a capillary volume of 2.8%.
33
Yet there is surprisingly little experimental data on the volume
occupied by endothelial cells in the brain. The principal values
cited come from Pardridge,
34
who stated that, Since the brain
capillary endothelial cell volume is only 1 μL/g brain, the
endothelial volume in brain is only 0.2% of the total cell volume
in brain.In an earlier report, Pardridge estimated that
endothelial cell volume is 0.8 μL per g of brain, given a capillary
volume of 1% of total brain volume, and that endothelial cells
occupy one-tenth of the total capillary volume (Figure 1).
35
Is there experimental support for these approximations? The
most useful data appears to come from Hicks and colleagues,
who assessed the cross-sectional area of capillary components
(basement membrane, endothelial cell, lumen, and pericyte) in
the hippocampus and frontal cortex of rats of 3, 9, and 24 months
of age.
36
Assessment of area was achieved by producing large
photographic copies of each electron microscope image, cutting
out the components, and weighing the pieces to calculate area.
The percentage of area composed of endothelial cells in the
frontal cortex was 12% and in the hippocampus was 11%
(average basement membrane 4%, lumen 80%, and pericyte 4%).
Given an experimental representative brain capillary volume of
2%, and an experimental representative endothelial cell volume
that is 10% of the total capillary volume, brain capillary
endothelial cell content is indeed estimated at 0.2% of brain
volume.
Using the numbers from previous studies, we estimate that the
Bmax of P-gp in the whole human brain is 1.3 nM (see Appendix A
for detailed calculations). The method employed by Terasaki and
colleagues to measure the expression of transporter protein did
not dierentiate between functional transporter at the luminal
membrane and internalized transporter; as such, Bmax at the
luminal membrane may be slightly lower (as noted earlier,
inhibitors may bind both pools of P-gp). We have determined the
Bmax values for other brain capillary transporters in a similar
fashion (Table 2). It is notable that only two solute carrier
transporters demonstrate higher expression in human brain
endothelial cells than P-gp. They are associated with energy
supply to the brain (glucose transporter 1, Glut1, SLC2A1,
calculated brain concentration 29.5 nM) and glutamate supply
(glutamate aspartate transporter, Eaat1, SLC1A3, calculated
concentration 5.2 nM). While PET functional studies of Glut1
have been reported, and it is expressed only in brain endothelial
cells,
37
no PET binding studies exist in the literature for this
transporter.
Binding Potential (Bmax/KD) of P-gp and High-Anity
Inhibitors. Using the Bmax value mentioned above for P-gp (1.3
nM) and reported KDvalues for various P-gp inhibitors, we
calculate that Bmax/KDvalues range from 0.004 to 0.470 (Table
3). These values are well below the Bmax/KDcutovalue of >5
10 recommended for a useful PET radioligand.
26
Although a
similar mathematical approach was described in the literature,
previous calculations used a vascular volume of 5% rather than a
capillary density of 0.2%,
15
concluding that the Bmax/KD(i.e., the
binding potential) for tariquidar, for instance, is 15, which would
Figure 1. Diagram of cross-section from a rat brain capillary
demonstrating that endothelial cells constitute a small percentage of
the total capillary volume. The diagram was generated from electron
micrograph published by Hicks and colleagues.
36
The scale bar
represents 1 μm.
Table 3. Calculated Values of Binding Potential (Bmax/KD)
a
for Various P-gp Inhibitors
inhibitor KD
b
(nM) species used for
cell assay reference calculated Bmax/KD
for P-gp
cyclosporin A 300 hamster 42 0.004
elacridar 2.7 human 43 0.474
tariquidar 5.1 hamster 14 0.251
valspodar 80 hamster 42 0.016
zosuquidar 55 hamster 42 0.023
a
Bmax = concentration of binding sites. KD= equilibrium dissociation
constant, where anity of the ligand for the target is dened as 1/KD.
b
The KDvalue for the P-gp inhibitor, laniquidar, has not been
reported.
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292225
allow for obtaining a suciently high in vivo signal with
radiolabeled tariquidar (subsequently corrected
38
).
There are clearly limitations to our calculations because they
are based on experimentally determined values derived from a
range of species: rabbit (brain capillary density), rat (capillary
cross sections), human (capillary transporter density and brain
protein density), and hamster (inhibitorsKDfor P-gp).
Nevertheless, it is clear that the concentration of P-gp in the
whole brain is very low (<1 nM). Given such a low Bmax for P-gp
in the whole brain, the anity of tariquidar and other high-
anityinhibitors needs to be at least an order of magnitude
higher (picomolar) to allow PET imaging. For example, based on
a calculated Bmax of 1.3 nM for P-gp in the whole brain and a
binding potential threshold of 5 for imaging, a KDof 260 pM (or
lower in value) would need to be achieved before imaging the
density of P-gp is possible.
High, Localized Transporter Density. Although conven-
tional PET studies of receptor density in the brain require brain
penetration of a radioligand,
25
PET studies of transporter density
do not, as the target (i.e., eux transporters) is located within
capillary walls. At this location, which is exposed to the plasma
compartment, eux transporters can have a direct eect on the
free concentration of radioligand. Because only free radioligand
can bind to a target, we speculate that this eect on drug
disposition is more likely to aect tracer concentrations of drug
than pharmacological doses.
We propose that the local density of P-gp in the endothelial
cells is so high that it transiently binds a high percentage of
radiolabeled inhibitor, thereby lowering the free concentration in
the capillary such that a negligible amount would enter the brain.
If all P-gp is concentrated in the capillary volume, then its
concentration (Bmax) in the capillary space is 40 nM. To illustrate
the eect of high, localized density on free ligand concentration,
Figure 2. The high, localized density of P-glycoprotein (P-gp) may transiently reduce the free plasma concentration of [11C]tariquidar in brain
capillaries, which may subsequently reduce the concentration of free radioligand in the brain. (A) Uptake of [11C]tariquidar, as measured by positron
emission tomography (PET), is low in brains of wild-type and Bcrp knockout, 2-fold higher in P-gp knockout mice, and 9.3-fold higher in P-gp/Bcrp
knockout mice. Modied with permission from data published by Langer and colleagues
15
(2010). Copyright 2010 Elsevier. (B) Scheme demonstrating
the potential interactions of radiolabeled inhibitor [11C]tariquidar (indicated by *) with transporters P-gp (red) and breast cancer resistance protein
(Bcrp, yellow). The numbers to the right of each image are representative values for the density (Bmax) of P-gp, free concentration of tariquidar inplasma
([TQR]FP), and free concentration of tariquidar in brain ([TQR]FB) for each mouse strain. The values were calculated assuming that the density of P-gp
is 40 000 pM in the capillaries, and that tariquidar has a concentration of 1 nM in the capillary, a KDof 5 nM, and a free fraction of 0.05 (meaning a free
concentration of tariquidar of 0.50 pM). In wild-type and Bcrp knockout mice, for every one molecule of tariquidar that is free, eight molecules are
bound to P-gp; this binding further decreases the free concentration of tariquidar in plasma to 0.06 pM. In P-gp knockout mice, the free concentration of
tariquidar is not further reduced by high-anity binding to P-gp, although overall parenchymal uptake is still low because the inhibitor is eciently
euxed as a substrate of Bcrp. In P-gp/Bcrp knockout mice, the radiolabeled molecule is taken up in the brain and trapped (probably by lysosomal
trapping
7
).
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292226
we used the MichaelisMenten equation to calculate the
boundfree ratio and receptor occupancy for the inhibitor
tariquidar (see Appendix B). As the concentration of free
radioligand decreases, the boundfree ratio increases to a
maximum of eight. In other words, for each free molecule of
tariquidar in the brain capillary, eight molecules are bound to P-
gp. Despite this high boundfree ratio, the receptor occupancy at
this concentration (0.001 nM) is very low (0.02%).
What would be the predicted receptor occupancy with tracer
doses of [11C]tariquidar in PET? Radioligands that are too
lipophilic are highly bound to plasma proteins, which would
dramatically reduce the free plasma concentration of radio-
ligand.
39
Like elacridar and laniquidar, tariquidar is a highly
lipophilic compound (clog P=6)
27
that is highly bound to
plasma proteins: we have observed the free plasma concentration
of [11C]tariquidar to be 0.05% (i.e., 99.95% protein binding). In
other words, if the concentration of tariquidar is 1 nM in the
vascular space, then the free plasma concentration of tariquidar is
0.5 pM. Thus, in a capillary where the local concentration of P-gp
is 40 000 pM, the total bound P-gp in this situation is 4 pM or
0.01%a negligible part of total radioactivity (1 nM).
Based on the above calculations, we hypothesize that the
binding of [11C]tariquidar to P-gp is so eective that it lowers the
concentration of free radioligand in plasma, which then passively
aects the concentration of free radioligand in the brain. One
limitation of this analysis is that it is based on theoretical data and
approximations and thus may not represent the entire in vivo
situation. However, our hypothesis that high, localized density of
P-gp contributes to the low brain signal in PET is supported by
the observation that P-gp knockout mice had slightly higher brain
signal from [11C]tariquidar than wild-type mice did
15
(Figure 2).
This result would be predicted because P-gp knockout mice lack
the receptor that transiently decreases the free concentration of
[11C]tariquidar in capillary plasma; however, parenchymal
uptake is still low because tariquidar is euxed as a substrate
of BCRP. Taken together, the above calculations and the
observations in mice support a hypothesis, not previously
discussed, that the microenvironment of the capillary aects the
low brain signal obtained from radiolabeled inhibitors in PET
studies of P-gp density.
CONCLUSIONS
Imaging the density of eux transporters, such as P-gp, in the
bloodbrain barrier using high-anity inhibitors and PET
presents a particular challenge. Based on our calculations, we
postulate that, while P-gp is highly expressed in brain capillary
endothelial cells, the endothelial cells themselves are present in a
low but relatively uniform density throughout the brain.
Compared with a small region of the brain with locally high
expression of a protein, which can be quantied by PET imaging,
the very low density of P-gp in the brain, high protein binding,
and locally reduced plasma concentrations will require an
imaging agent with a signicantly higher anity for P-gp. More
generally, locally high concentrations of transporters and
receptors in brain endothelial cells do not appear amenable to
imaging due to the vast dilution of endothelial capillary cells in
the brain. The development of radiotracers with picomolar
anity for targets will be needed to develop diagnostic tools
capable of providing insight into an ever-increasing list of
pathophysiologies associated with dysfunction at the blood
brain barrier.
APPENDIX A: CALCULATION OF BMAX (MOL/L) OF
P-GP IN THE WHOLE BRAIN
Values are derived from values found in the literature that can be
generally applied to determine the Bmax of other proteins in brain
endothelial cells. Four values are necessary:
(a) The human brain endothelial cell P-gp expression, which is
6.06 fmol/μg protein (6.06 nmol/g);
28
(b) The protein content of the human brain, which is reported
as 10% of total wet weight (i.e., 100 mg protein/g brain),
and fairly uniform across all regions;
40
(c) The density of the human brain, which is 1060 g/L;
41
and
(d) The aforementioned endothelial cell density in the brain of
0.2%.
A concentration of 6.06 nmol P-gp/g of protein (a) is 0.606
nmol P-gp/g brain (10-fold dilution of protein content in cells,
value b), which consequently gives a P-gp concentration of 642
nmol/L (using value c for density of brain). Diluting for
endothelial cells as a component of total brain (0.2%, value d)
gives a nal estimated concentration of P-gp in the brain of 1.3
nM.
APPENDIX B: CALCULATION OF BOUNDFREE
RATIO AND RECEPTOR OCCUPANCY FOR
TARIQUIDAR AND P-GP IN BRAIN CAPILLARIES
Equilibrium MichaelisMenten:
=
+
B
F
B
KF
max
D
(1)
If FKD:
==×
B
F
B
K
Baffinity
max
D
max (2)
where B= concentration of bound radiotracer; F= concentration
of free radiotracer; Bmax = concentration of receptor target; KD=
dissociation constant of radiotracer.
The equilibrium MichaelisMenten eq 1 is used to determine
the bound (B) concentration of radiotracer given that the other
variables are known. When the free (F) concentration is much
lower than the KDfor the radiotracer (FKD), Fcan be
removed (eq 2).
The receptor occupancy (RO), the percentage of receptor
bound by radiotracer, can subsequently be calculated using eq 3:
==
+
B
B
F
KF
RO
max
max D
(3)
Given that in capillaries the Bmax for P-gp and KDfor tariquidar
are known, a range of free tariquidar concentrations (F) can be
used to compute theoretical bound (B) concentration, bound
free ratio (B/F), and receptor occupancy (RO) of the radiotracer
(see Table A1).
Table A1
Bmax (nM) F(nM) B(nM) B/FRO
40 10 26.67 2.7 66.67%
1 6.67 6.7 16.67%
KD(nM) 0.1 0.78 7.8 1.96%
5 0.01 0.08 8.0 0.20%
0.001 0.01 8.0 0.02%
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292227
AUTHOR INFORMATION
Corresponding Author
*National Institute of Mental Health, 10 Center Drive, Rm
B1D43, Bethesda, Maryland 20892-1026, United States. Fax: +1
301 480 3610. Tel.: +1 301 594 1368. E-mail: robert.innis@nih.
gov.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This research was supported by the Intramural Research
Programs of the National Institute of Mental Health (Project
Nos. Z01-MH-002852-04 and MH002793-09) and the National
Cancer Institute (Project no. Z01-BC-005598). Oliver Langers
studies are funded by the Austrian Science Fund (FWF) project
‘‘Transmembrane Transporters in Health and Disease’’ (SFB
F35) and by the European Communitys Seventh Framework
Programme (FP7/2007-2013) under grant agreement number
201380 (EURIPIDES). We thank George Leiman for editorial
assistance.
REFERENCES
(1) Kannan, P.; John, C.; Zoghbi, S. S.; Halldin, C.; Gottesman, M. M.;
Innis, R. B.; Hall, M. D. Imaging the function of P-glycoprotein with
radiotracers: pharmacokinetics and in vivo applications. Clin. Pharm.
Ther. 2009,86 (4), 36877.
(2) Cooray, H. C.; Blackmore, C. G.; Maskell, L.; Barrand, M. A.
Localisation of breast cancer resistance protein in microvessel
endothelium of human brain. Neuroreport 2002,13 (16), 205963.
(3) Graff, C. L.; Pollack, G. M. Drug transport at the bloodbrain
barrier and the choroid plexus. Curr. Drug Metab. 2004,5(1), 95108.
(4) Dombrowski, S. M.; Desai, S. Y.; Marroni, M.; Cucullo, L.;
Goodrich, K.; Bingaman, W.; Mayberg, M. R.; Bengez, L.; Janigro, D.
Overexpression of multiple drug resistance genes in endothelial cells
from patients with refractory epilepsy. Epilepsia 2001,42 (12), 15016.
(5) Loscher, W.; Potschka, H. Drug resistance in brain diseases and the
role of drug efflux transporters. Nat. Rev. Neurosci. 2005,6(8), 591602.
(6) Phelps, M. Molecular Imaging and Its Biological Applications;
Springer: New York, 2004.
(7) Kannan, P.; Brimacombe, K. R.; Kreisl, W. C.; Liow, J. S.; Zoghbi, S.
S.; Telu, S.; Zhang, Y.; Pike, V. W.; Halldin, C.; Gottesman, M. M.; Innis,
R. B.; Hall, M. D. Lysosomal trapping of a radiolabeled substrate of P-
glycoprotein as a mechanism for signal amplification in PET. Proc. Natl.
Acad. Sci. U.S.A. 2011,108 (6), 25938.
(8) Piwnica-Worms, D.; Sharma, V. Probing multidrug resistance P-
glycoprotein transporter activity with SPECT radiopharmaceuticals.
Curr. Top. Med. Chem. 2010,10 (17), 183445.
(9) Innis, R. B.; Cunningham, V. J.; Delforge, J.; Fujita, M.; Gjedde, A.;
Gunn, R. N.; Holden, J.; Houle, S.; Huang, S. C.; Ichise, M.; Iida, H.; Ito,
H.; Kimura, Y.; Koeppe, R. A.; Knudsen, G. M.; Knuuti, J.; Lammertsma,
A. A.; Laruelle, M.; Logan, J.; Maguire, R. P.; Mintun, M. A.; Morris, E.
D.; Parsey, R.; Price, J. C.; Slifstein, M.; Sossi, V.; Suhara, T.; Votaw, J. R.;
Wong, D. F.; Carson, R. E. Consensus nomenclature for in vivo imaging
of reversibly binding radioligands. J. Cereb. Blood Flow Metab. 2007,27
(9), 15339.
(10) Ambudkar, S. V.; Dey, S.; Hrycyna, C. A.; Ramachandra, M.;
Pastan, I.; Gottesman, M. M. Biochemical, cellular, and pharmacological
aspects of the multidrug transporter. Annu. Rev. Pharmacol. Toxicol.
1999,39, 36198.
(11) Colabufo, N. A.; Berardi, F.; Perrone, M. G.; Capparelli, E.;
Cantore, M.; Inglese, C.; Perrone, R. Substrates, inhibitors and
activators of P-glycoprotein: candidates for radiolabeling and imaging
perspectives. Curr. Top. Med. Chem. 2010,10 (17), 170314.
(12) Kannan, P.; Telu, S.; Shukla, S.; Ambudkar, S. V.; Pike, V. W.;
Halldin, C.; Gottesman, M. M.; Innis, R. B.; Hall, M. D. The SpecificP-
Glycoprotein Inhibitor Tariquidar Is Also a Substrate and an Inhibitor
for Breast Cancer Resistance Protein (BCRP/ABCG2). ACS Chem.
Neurosci. 2011,2(2), 829.
(13) Loo, T. W.; Bartlett, M. C.; Detty, M. R.; Clarke, D. M. The
ATPase activity of the P-glycoprotein drug pump is highly activated
when the N-terminal and central regions of the nucleotide-binding
domains are linked closely together. J. Biol. Chem. 2012,287 (32),
2680616.
(14) Martin, C.; Berridge, G.; Mistry, P.; Higgins, C.; Charlton, P.;
Callaghan, R. The molecular interaction of the high affinity reversal
agent XR9576 with P-glycoprotein. Br. J. Pharmacol. 1999,128 (2),
40311.
(15) Bauer, F.; Kuntner, C.; Bankstahl, J. P.; Wanek, T.; Bankstahl, M.;
Stanek, J.; Mairinger, S.; Dorner, B.; Loscher, W.; Muller, M.; Erker, T.;
Langer, O. Synthesis and in vivo evaluation of [11C]tariquidar, a
positron emission tomography radiotracer based on a third-generation
P-glycoprotein inhibitor. Bioorg. Med. Chem. 2010,18 (15), 548997.
(16) Dorner, B.; Kuntner, C.; Bankstahl, J. P.; Bankstahl, M.; Stanek, J.;
Wanek, T.; Stundner, G.; Mairinger, S.; Loscher, W.; Muller, M.; Langer,
O.; Erker, T. Synthesis and small-animal positron emission tomography
evaluation of [11C]-elacridar as a radiotracer to assess the distribution of
P-glycoprotein at the bloodbrain barrier. J. Med. Chem. 2009,52 (19),
607382.
(17) Dorner, B.; Kuntner, C.; Bankstahl, J. P.; Wanek, T.; Bankstahl,
M.; Stanek, J.; Mullauer, J.; Bauer, F.; Mairinger, S.; Loscher, W.; Miller,
D. W.; Chiba, P.; Muller, M.; Erker, T.; Langer, O. Radiosynthesis and in
vivo evaluation of 1-[18F]fluoroelacridar as a positron emission
tomography tracer for P-glycoprotein and breast cancer resistance
protein. Bioorg. Med. Chem. 2011,19 (7), 21908.
(18) Luurtsema, G.; Schuit, R. C.; Klok, R. P.; Verbeek, J.; Leysen, J. E.;
Lammertsma, A. A.; Windhorst, A. D. Evaluation of [11C]laniquidar as a
tracer of P-glycoprotein: radiosynthesis and biodistribution in rats. Nucl.
Med. Biol. 2009,36 (6), 6439.
(19) van Waarde, A.; Ramakrishnan, N. K.; Rybczynska, A. A.; Elsinga,
P. H.; Berardi, F.; de Jong, J. R.; Kwizera, C.; Perrone, R.; Cantore, M.;
Sijbesma, J. W.; Dierckx, R. A.; Colabufo, N. A. Synthesis and preclinical
evaluation of novel PET probes for P-glycoprotein function and
expression. J. Med. Chem. 2009,52 (14), 452432.
(20) Kawamura, K.; Konno, F.; Yui, J.; Yamasaki, T.; Hatori, A.;
Yanamoto, K.; Wakizaka, H.; Takei, M.; Nengaki, N.; Fukumura, T.;
Zhang, M. R. Synthesis and evaluation of [11C]XR9576 to assess the
function of drug efflux transporters using PET. Ann. Nucl. Med. 2010,24
(5), 40312.
(21) Kawamura, K.; Yamasaki, T.; Konno, F.; Yui, J.; Hatori, A.;
Yanamoto, K.; Wakizaka, H.; Ogawa, M.; Yoshida, Y.; Nengaki, N.;
Fukumura, T.; Zhang, M. R. Synthesis and in vivo evaluation of (1)(8)F-
fluoroethyl GF120918 and XR9576 as positron emission tomography
probes for assessing the function of drug efflux transporters. Bioorg. Med.
Chem. 2011,19 (2), 86170.
(22) Kawamura, K.; Yamasaki, T.; Konno, F.; Yui, J.; Hatori, A.;
Yanamoto, K.; Wakizaka, H.; Takei, M.; Kimura, Y.; Fukumura, T.;
Zhang, M. R. Evaluation of limiting brain penetration related to P-
glycoprotein and breast cancer resistance protein using [(11)C]-
GF120918 by PET in mice. Mol. Imaging Biol. 2011,13 (1), 15260.
(23) Mairinger, S.; Langer, O.; Kuntner, C.; Wanek, T.; Bankstahl, J. P.;
Bankstahl, M.; Stanek, J.; Dorner, B.; Bauer, F.; Baumgartner, C.;
Loscher, W.; Erker, T.; Muller, M. Synthesis and in vivo evaluation of the
putative breast cancer resistance protein inhibitor [11C]methyl 4-((4-
(2-(6,7-dimethoxy-1,2,3,4-tetrahydroisoquinolin-2-yl)ethyl)phenyl)-
amino-car Bonyl)-2-(quinoline-2-carbonylamino)benzoate. Nucl. Med.
Biol. 2010,37 (5), 63744.
(24) Mairinger, S.; Wanek, T.; Kuntner, C.; Doenmez, Y.; Strommer,
S.; Stanek, J.; Capparelli, E.; Chiba, P.; Muller, M.; Colabufo, N. A.;
Langer, O. Synthesis and preclinical evaluation of the radiolabeled P-
glycoprotein inhibitor [(11)C]MC113. Nucl. Med. Biol. 2012,39 (8),
121925.
(25) Halldin, C.; Gulyas, B.; Langer, O.; Farde, L. Brain radioligands
state of the art and new trends. Q. J. Nucl. Med. 2001,45 (2), 13952.
(26) Eckelman, W. C.; Mathis, C. A. Targeting proteins in vivo: in vitro
guidelines. Nucl. Med. Biol. 2006,33 (2), 1614.
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292228
(27) Luurtsema, G.; Verbeek, G. L.; Lubberink, M.; Lammertsma, A.
A.; Dierckx, R.; Elsinga, P.; Windhorst, A. D.; van Waarde, A. Carbon-11
labeled tracers for in vivo imaging P-glycoprotein function: kinetics,
advantages and disadvantages. Curr. Top. Med. Chem. 2010,10 (17),
182033.
(28) Kamiie, J.; Ohtsuki, S.; Iwase, R.; Ohmine, K.; Katsukura, Y.;
Yanai, K.; Sekine, Y.; Uchida, Y.; Ito, S.; Terasaki, T. Quantitative atlas of
membrane transporter proteins: development and application of a
highly sensitive simultaneous LC/MS/MS method combined with
novel in-silico peptide selection criteria. Pharm. Res. 2008,25 (6),
146983.
(29) Ito, K.; Uchida, Y.; Ohtsuki, S.; Aizawa, S.; Kawakami, H.;
Katsukura, Y.; Kamiie, J.; Terasaki, T. Quantitative membrane protein
expression at the bloodbrain barrier of adult and younger cynomolgus
monkeys. J. Pharm. Sci. 2011,100 (9), 393950.
(30) Uchida, Y.; Ohtsuki, S.; Katsukura, Y.; Ikeda, C.; Suzuki, T.;
Kamiie, J.; Terasaki, T. Quantitative targeted absolute proteomics of
human bloodbrain barrier transporters and receptors. J. Neurochem.
2011,117 (2), 33345.
(31) Larsson, H. B.; Courivaud, F.; Rostrup, E.; Hansen, A. E.
Measurement of brain perfusion, blood volume, and bloodbrain
barrier permeability, using dynamic contrast-enhanced T(1)-weighted
MRI at 3 T. Magn. Reson. Med. 2009,62 (5), 127081.
(32) Phelps, M. E.; Huang, S. C.; Hoffman, E. J.; Kuhl, D. E. Validation
of tomographic measurement of cerebral blood volume with C-11-
labeled carboxyhemoglobin. J. Nucl. Med. 1979,20 (4), 32834.
(33) Wiederhold, K. H.; Bielser, W., Jr.; Schulz, U.; Veteau, M. J.;
Hunziker, O. Three-dimensional reconstruction of brain capillaries from
frozen serial sections. Microvasc. Res. 1976,11 (2), 17580.
(34) Pardridge, W. M. Blood-brain barrier biology and methodology. J.
Neurovirol. 1999,5(6), 55669.
(35) Pardridge, W. Peptide Drug Delivery to the Brain; Raven Press:
New York, 1991; p 357.
(36) Hicks, P.; Rolsten, C.; Brizzee, D.; Samorajski, T. Age-related
changes in rat brain capillaries. Neurobiol. Aging 1983,4(1), 6975.
(37) Pardridge, W. M.; Boado, R. J.; Farrell, C. R. Brain-type glucose
transporter (GLUT-1) is selectively localized to the bloodbrain
barrier. Studies with quantitative western blotting and in situ
hybridization. J. Biol. Chem. 1990,265 (29), 1803540.
(38) Mairinger, S.; Erker, T.; Muller, M.; Langer, O. PET and SPECT
radiotracers to assess function and expression of ABC transporters in
vivo. Curr. Drug Metab. 2011,12 (8), 77492.
(39) Zoghbi, S. S.; Anderson, K. B.; Jenko, K. J.; Luckenbaugh, D. A.;
Innis, R. B.; Pike, V. W. On quantitative relationships between drug-like
compound lipophilicity and plasma free fraction in monkey and human.
J. Pharmaceut. Sci. 2012,101 (3), 102839.
(40) Banay-Schwartz, M.; Kenessey, A.; DeGuzman, T.; Lajtha, A.;
Palkovits, M. Protein content of various regions of rat brain and adult
and aging human brain. Age 1992,15 (2), 5154.
(41) Witelson, S. F.; Beresh, H.; Kigar, D. L. Intelligence and brain size
in 100 postmortem brains: sex, lateralization and age factors. Brain 2006,
129 (Pt 2), 38698.
(42) Melchior, D. L.; Sharom, F. J.; Evers, R.; Wright, G. E.; Chu, J. W.;
Wright, S. E.; Chu, X.; Yabut, J. Determining P-glycoprotein-drug
interactions: evaluation of reconstituted P-glycoprotein in a liposomal
system and LLC-MDR1 polarized cell monolayers. J. Pharmacol. Toxicol.
Meth. 2012,65 (2), 6474.
(43) Ferry, D. R.; Russell, M. A.; Kerr, D. J.; Correa, I. D.; Prakash, S. R.
[3H]-GG918 (GF120918) binds with positive co-operativity to human
P-glycoprotein with a nM dissociation constant. Proc. Am. Assoc. Cancer
Res. 1996,37, 2246.
Molecular Pharmaceutics Review
dx.doi.org/10.1021/mp400011g |Mol. Pharmaceutics 2013, 10, 222222292229
... So far, however, these tracers have shown to be substrates at tracer doses despite their known inhibitory nature at pharmacological doses as determined in in vitro assays (Luurtsema et al., 2009). It has been postulated that, at these tracer doses, the concentration of these compounds is too low to achieve full inhibition of P-gp function, leading to transport of tracer and a picomolar affinity would be required to measure P-gp expression (Kannan et al., 2013). This dose dependent behaviour has been confirmed for [ 11 C]laniquidar in experiments with variable amounts of co-injected unlabelled laniquidar (Moerman et al., 2012). ...
Article
Full-text available
Background Several P-glycoprotein (P-gp) substrate tracers are available to assess P-gp function in vivo, but attempts to develop a tracer for measuring expression levels of P-gp have not been successful. Recently, (Z)-2-(5-fluoro-2-oxoindolin-3-ylidene)-N-(4-methoxyphenyl)hydrazine-carbothioamide was described as a potential selective P-gp inhibitor that is not transported by P-gp. Therefore, the purpose of this study was to radiolabel two of its analogues and to assess their potential for imaging P-gp expression using PET. Results [¹⁸F]2-(4-fluoro-2-oxoindolin-3-ylidene)-N-(4-methoxyphenyl)hydrazine-carbothioamide ([¹⁸F]5) and [¹⁸F]2-(6-fluoro-2-oxoindolin-3-ylidene)-N-(4-methoxyphenyl)hydrazine-carbothioamide ([¹⁸F]6) were synthesized and both their biodistribution and metabolism were evaluated in rats. In addition, PET scans were acquired in rats before and after tariquidar (P-gp inhibitor) administration as well as in P-gp knockout (KO) mice. Both [¹⁸F]5 and [¹⁸F]6 were synthesized in 2–3% overall yield, and showed high brain uptake in ex vivo biodistribution studies. [¹⁸F]6 appeared to be metabolically unstable in vivo, while [¹⁸F]5 showed moderate stability with limited uptake of radiolabelled metabolites in the brain. PET studies showed that transport of [¹⁸F]5 across the blood-brain barrier was not altered by pre-treatment with the P-gp inhibitor tariquidar, and uptake was significantly lower in P-gp KO than in wild-type animals and indeed transported across the BBB or bound to P-gp in endothelial cells. Conclusion In conclusion, [¹⁸F]5 and [¹⁸F]6 were successfully and reproducibly synthesized, albeit with low radiochemical yields. [¹⁸F]5 appears to be a radiotracer that binds to P-gp, as showed in P-gp knock-out animals, but is not a substrate for P-gp.
... This second approach may potentially be very useful, in particular for tissues which are protected by ABC transporters, such as the brain, for which radiolabeled transporter substrates afford very low imaging signals. However, this approach has remained unsuccessful so far, presumably due to the low density of ABC transporters, in particular in the brain (~1 nM), combined with insufficiently high binding affinities of the tested compounds (Bauer et al., 2013;Kannan et al., 2013;Müllauer et al., 2013). Based on available in vitro data with [ 3 H]Ko143 (Weidner et al., 2015), we were interested in evaluating if Ko143 can serve as a PET tracer to map the density of ABCG2. ...
Article
Ko143 is a reference inhibitor of the adenosine triphosphate-binding cassette (ABC) transporter breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) for in vitro and in vivo use. Previous in vitro data indicate that Ko143 binds specifically to ABCG2/Abcg2, suggesting a potential utility of Ko143 as a positron emission tomography (PET) tracer to assess the density (abundance) of ABCG2 in different tissues. In this work we radiolabeled Ko143 with carbon-11 (¹¹C) and performed small-animal PET experiments with [¹¹C]Ko143 in wild-type, Abcg2(−/−), Abcb1a/b(−/−) and Abcb1a/b(−/−)Abcg2(−/−) mice to assess the influence of Abcg2 and Abcb1a/b on tissue distribution and excretion of [¹¹C]Ko143. [¹¹C]Ko143 was extensively metabolized in vivo and unidentified radiolabeled metabolites were found in all investigated tissues. We detected no significant differences between wild-type and Abcg2(−/−) mice in the distribution of [¹¹C]Ko143-derived radioactivity to Abcg2-expressing organs (brain, liver and kidney). [¹¹C]Ko143 and possibly its radiolabeled metabolites were transported by Abcb1a and not by Abcg2 at the mouse blood-brain barrier. [¹¹C]Ko143-derived radioactivity underwent both hepatobiliary and urinary excretion, with Abcg2 playing a possible role in mediating the transport of radiolabeled metabolites of [¹¹C]Ko143 from the kidney into urine. Experiments in which a pharmacologic dose of unlabeled Ko143 (10 mg/kg) was co-administered with [¹¹C]Ko143 revealed pronounced effects of the vehicle used for Ko143 formulation (containing polyethylene glycol 300 and polysorbate 80) on radioactivity distribution to the brain and the liver, as well as on hepatobiliary and urinary excretion of radioactivity. Our results highlight the challenges associated with the development of PET tracers for ABC transporters and emphasize that inhibitory effects of pharmaceutical excipients on membrane transporters need to be considered when performing in vivo drug-drug interaction studies. Finally, our study illustrates the power of small-animal PET to assess the interaction of drug molecules with membrane transporters on a whole body level.
... A radiolabeled weak substrate not only overcomes the -floor effect‖ of avid substrates that have no brain uptake at baseline, but also the -spare capacity‖ shown by many efflux transporters at the blood-brain barrier. P-gp and BCRP are so highly concentrated in the endothelium of only the capillary segments of the vascular system that a small percentage (probably <10%) can completely block the uptake of avid substrates (Kannan et al., 2013). This spare capacity means that a high percentage of transporters must be blocked to increase the uptake of an avid substrate. ...
Article
Full-text available
Efflux transporters at the blood–brain barrier can decrease the entry of drugs and increase the removal of those molecules able to bypass the transporter. We previously hypothesized that 18F-FCWAY, a radioligand for the serotonin 5-HT1A receptor, is a weak substrate for permeability glycoprotein (P-gp) based on its very early peak and rapid washout from human brain. To determine whether 18F-FCWAY is a substrate for P-gp, breast cancer resistance protein (BCRP) and multidrug resistance protein (MRP1) — the three most prevalent efflux transporters at the blood–brain barrier — we performed three sets of experiments. In vitro, we conducted fluorescence-activated cell sorting (FACS) flow cytometry studies in cells over-expressing P-gp, BCRP and MRP1 treated with inhibitors specific to each transporter and with FCWAY. Ex vivo, we measured 18F-FCWAY concentration in plasma and brain homogenate of transporter knockout mice using γ-counter and radio-HPLC. In vivo, we conducted positron emission tomography (PET) studies to assess changes in humans who received 18F-FCWAY during an infusion of tariquidar (2–4 mg/kg iv), a potent and selective P-gp inhibitor. In vitro studies showed that FCWAY allowed fluorescent substrates to get into the cell by competitive inhibition of all three transporters at the cell membrane. Ex vivo measurements in knockout mice indicate that 18F-FCWAY is a substrate only for P-gp and not BCRP. In vivo, tariquidar increased 18F-FCWAY brain uptake in seven of eight subjects by 60–100% compared to each person's baseline. Tariquidar did not increase brain uptake via some peripheral mechanism, given that it did not significantly alter concentrations in plasma of the parent radioligand 18F-FCWAY or its brain-penetrant radiometabolite 18F-FC. These results show that 18F-FCWAY is a weak substrate for efflux transport at the blood–brain barrier; some radioligand can enter brain, but its removal is hastened by P-gp. Although 18F-FCWAY is not ideal for measuring 5-HT1A receptors, it demonstrates that weak substrate radioligands can be useful for measuring both increased and decreased function of efflux transporters, which is not possible with currently available radioligands such as 11C-loperamide and 11C-verapamil that are avid substrates for transporters.
... The required affinity for a PET tracer has been discussed elsewhere and as a rule of thumb is given by the formula K d < B max /5. [15] Moreover, the concentration of radiotracer available for binding to the receptor is dependent on its free fraction, which can be measured using, for example, tissue homogenates and LC-MS. It also depends on the ratio between specific and nonspecific binding, a property that can be quantified and optimized using LSE. ...
Article
Ligand efficiency indices are widely used to guide chemical optimization in drug discovery, due to their predictive value in the early steps of optimization. At later stages, however, as more complex properties become critical for success, indices relying on calculated, rather than experimental, parameters become less informative. This problem is particularly acute when developing positron emission tomography (PET) imaging agents, for which nonspecific binding (NSB) to membranes and non-target proteins is a frequent cause of failure. NSB cannot be predicted using in silico parameters. To address this gap, we explored the use of the experimentally determined chromatographic hydrophobicity index on immobilized artificial membranes, CHI(IAM), to guide the optimization of NSB. The ligand specific efficiency (LSE) index was defined as the ratio between affinity (pIC50 or pKd ) and the logarithmic value of CHI(IAM). It allows for quantification of binding affinity to the target of interest, relative to NSB. Its use was illustrated by the optimization of PET tracer candidates for the prostacyclin receptor.
... However, these compounds behaved like P-gp substrates, and were therefore not suitable for detecting up-regulation of P-gp at the BBB. Furthermore, theoretical approximations of P-gp densities at the BBB (1-3 nM) are too low to detect currently available labelled inhibitors (K D N 5 nM) [29]. It is also important to note that P-gp function and expression do not change in a linear manner. ...
Article
Introduction: 2-Isopropyl-5-[methyl-(2-phenylethyl)amino]-2-phenylpentanenitrile (emopamil; EMP) is a calcium channel blocker of the phenylalkylamine class, with weak substrate properties for P-glycoprotein (P-gp). A weak substrate for P-gp would be suitable for measuring enhanced P-gp function. This study was performed to synthesise (R)- and (S)-[(11)C]EMP and characterise their properties as P-gp tracers. Methods: We synthesised (R)- and (S)-[(11)C]EMP and compared their biodistribution, peripheral metabolism, and effects of the P-gp inhibitor cyclosporine A (CsA, 50mg/kg). We compared the brain pharmacokinetics of (R)-[(11)C]EMP and (R)-[(11)C]verapamil [(R)-[(11)C]VER] at baseline and CsA pretreatment with small animal positron emission tomography (PET). Results: (R)- and (S)-[(11)C]EMP were synthesised from (R)- and (S)-noremopamil, respectively, by methylation with [(11)C]methyl triflate in the presence of NaOH at room temperature. (R)- and (S)-[(11)C]EMP yields were ~30%, with specific activity>74GBq/μmol and radiochemical purity>99%. (R)-[(11)C]EMP showed significantly greater uptake in the mouse brain than (S)-[(11)C]EMP. Both showed homogeneous non-stereoselective regional brain distributions. (R)- and (S)-[(11)C]EMP were rapidly metabolised to hydrophilic metabolites. Unchanged plasma (S)-[(11)C]EMP level was significantly lower than that of (R)-[(11)C]EMP 15minutes post-injection, whilst>88% of radioactivity in the brain was intact at 15minutes post-injection and was non-stereoselective. CsA pretreatment increased brain activity ~3-fold in mice, but was non-stereoselective. The baseline area-under-the-curve (AUC) of brain radioactivity (0-60minutes) of (R)-[(11)C]EMP was 2-fold higher than that of (R)-[(11)C]VER, but their AUCs after CsA pretreatment were comparable. Conclusions: (R)-[(11)C]EMP is a novel tracer for imaging P-gp function with higher baseline uptake than (R)-[(11)C]VER.
Article
Alzheimer's disease is characterized by the presence of extracellular amyloid-β plaques. Positron emission tomography (PET) imaging with tracers radiolabeled with positron-emitting radionuclides that bind to amyloid-β plaques can assist in the diagnosis of Alzheimer's disease. With the goal of designing new imaging agents radiolabeled with positron-emitting copper-64 radionuclides that bind to amyloid-β plaques, a family of bis(thiosemicarbazone) ligands with appended substituted stilbenyl functional groups has been prepared. The ligands form charge-neutral and stable complexes with copper(II). The new ligands can be radiolabeled with copper-64 at room temperature. Two lead complexes were demonstrated to bind to amyloid-β plaques present in post-mortem brain tissue from subjects with clinically diagnosed Alzheimer's disease and crossed the blood-brain barrier in mice. The work presented here provides strategies to prepare compounds with radionuclides of copper that can be used for targeted brain PET imaging.
Article
Bis(thiosemicarbazonato)copper(II) complexes are of interest as potential therapeutics for cancer and neurodegenerative diseases as well as imaging agents for positron emission tomography (PET). The cellular uptake of six bis(thiosemcarbazonato)copper(II)complexes derived from glyoxal, with different functional groups, Cu(gtsx) where x = different functional groups, was investigated in SKOV-3, HEK293 and HEK293 P-gp cell lines. Treatment of the cells with the copper complexes increased intracellular copper and increased levels of p-ERK due to activation of the Ras-Raf-MEK-ERK pathway. Treatment of SKOV-3 cells with low concentrations (M) of two of the copper complexes led to trafficking of the endogenous copper transporter ATP7A from the Golgi network to the cell membrane. Experiments in HEK293 and HEK293-P-gp cells suggest that Cu(gtsm) and Cu(gtse) are substrates for the P-gp efflux protein but the complex with a pyrrolidine functional group, Cu(gtspyr), is not. A PET experiment in mice showed that [64Cu]Cu(gtspyr) has reasonable brain uptake but high liver uptake.
Chapter
The development of positron emission tomography (PET) tracers to assess transporter density at the blood-brain barrier (BBB) is complicated by the very low density of adenosine triphosphate (ATP)-binding cassette (ABC) transporters in brain. PET imaging with ABC transporter probes may allow to better understand disease-related alterations of cerebral ABC transporter function/expression, such as in therapy-refractory epilepsy or in Alzheimer's disease. PET imaging of ABC transporters is a powerful tool to elucidate the role of ABC transporters in neurological diseases and possibly to predict treatment response in individual patients. Most efforts to visualize ABC transporters at the BBB with PET have concentrated on P-glycoprotein (P-gp). Radiolabelled P-gp inhibitors, such as [11C]tariquidar, were developed to measure P-gp expression levels at the BBB, which has so far remained unsuccessful as these probes were unexpectedly recognized at tracer concentrations by P-gp and breast cancer resistance protein (BCRP) as substrates resulting in low brain PET signals.
Article
Full-text available
Since its development, tariquidar (XR9576, TQR) has been widely regarded as one of the more potent inhibitors of P-glycoprotein (P-gp), an efflux transporter of the ATP-binding cassette (ABC) transporter family. A third-generation inhibitor, TQR exhibits high affinity for P-gp, although it is also a substrate of another ABC transporter, breast cancer resistance protein (BCRP). Recently, several studies have questioned the mechanism by which TQR interfaces with P-gp, suggesting that TQR is a substrate for P-gp instead of a noncompetitive inhibitor. We investigated TQR and its interaction with human and mouse P-gp to determine if TQR is a substrate of P-gp in vitro. To address these questions, we used multiple in vitro transporter assays including cytotoxicity, flow cytometry, accumulation, ATPase, and transwell assays. A newly generated BCRP cell line was used as a positive control that demonstrates TQR-mediated transport. Based on our results, we conclude that TQR is a potent inhibitor of both human and mouse P-gp and shows no signs of being a substrate at the concentrations tested. These in vitro data further support our position that the in vivo uptake of [(11)C]TQR into the brain can be explained by its high affinity binding to P-gp and by it being a substrate of BCRP, followed by amplification of the brain signal by ionic trapping in acidic lysosomes.
Article
Full-text available
P-glycoprotein (P-gp) is a drug efflux transporter with broad substrate specificity localized in the blood-brain barrier and in several peripheral organs. In order to understand the role of P-gp in physiological and patho-physiological conditions, several carbon-11 labelled P-gp tracers have been developed and validated. This review provides an overview of the spectrum of radiopharmaceuticals that is available for this purpose. A short overview of the physiology of the blood-brain barrier in health and disease is also provided. Tracer kinetic modelling for quantitative analysis of P-gp function and expression is highlighted, and the advantages and disadvantages of the various tracers are discussed.
Article
Full-text available
Tariquidar was developed as a specific inhibitor of the efflux transporter ABCB1. Recent positron emission tomographic brain imaging studies using [(11)C]tariquidar to measure ABCB1 (P-gp, P-glycoprotein) density in mice indicate that the inhibitor may not be as specific as previously thought. We examined its selectivity as an inhibitor and a substrate for the human transporters P-gp, breast cancer resistance protein (BCRP, ABCG2), and multidrug resistance protein 1 (MRP1, ABCC1). Our results show that at low concentrations, tariquidar acts selectively as an inhibitor of P-gp and also as a substrate of BCRP. At much higher concentrations (≥100 nM), tariquidar acts as an inhibitor of both P-gp and BCRP. Thus, the in vivo specificity of tariquidar depends on concentration and the relative density and capacity of P-gp vs BCRP.
Article
Full-text available
The P-glycoprotein (P-gp, ABCB1) drug pump protects us from toxic compounds and confers multidrug resistance. Each of the homologous halves of P-gp is composed of a transmembrane domain (TMD) with 6 TM segments followed by a nucleotide-binding domain (NBD). The predicted drug- and ATP-binding sites reside at the interface between the TMDs and NBDs, respectively. Crystal structures and EM projection images suggest that the two halves of P-gp are separated by a central cavity that closes upon binding of nucleotide. Binding of drug substrates may induce further structural rearrangements because they stimulate ATPase activity. Here, we used disulfide cross-linking with short (8 Å) or long (22 Å) cross-linkers to identify domain-domain interactions that activate ATPase activity. It was found that cross-linking of cysteines that lie close to the LSGGQ (P517C) and Walker A (I1050C) sites of NBD1 and NBD2, respectively, as well as the cytoplasmic extensions of TM segments 3 (D177C or L175C) and 9 (N820C) with a short cross-linker activated ATPase activity over 10-fold. A pyrylium compound that inhibits ATPase activity blocked cross-linking at these sites. Cross-linking between the NBDs was not inhibited by tariquidar, a drug transport inhibitor that stimulates P-gp ATPase activity but is not transported. Cross-linking between extracellular cysteines (T333C/L975C) predicted to lock P-gp into a conformation that prevents close NBD association inhibited ATPase activity. The results suggest that trapping P-gp in a conformation in which the NBDs are closely associated likely mimics the structural rearrangements caused by binding of drug substrates that stimulate ATPase activity.
Article
The blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) represent the main interfaces between the central nervous system (CNS) and the peripheral circulation. Drug exposure to the CNS is dependent on a variety of factors, including the physical barrier presented by the BBB and the BCSFB and the affinity of the substrate for specific transport systems located at both of these interfaces. It is the aggregate effect of these factors that ultimately determines the total CNS exposure, and thus pharmacological efficacy, of a drug or drug candidate. This review discusses the anatomical and biochemical barriers presented to solute access to the CNS. In particular, the important role played by various efflux transporters in the overall barrier function is considered in detail, as current literature suggests that efflux transport likely represents a key determinant of overall CNS exposure for many substrates. Finally, it is important to consider not only the net delivery of the agent to the CNS, but also the ability of the agent to access the relevant target site within the CNS. Potential approaches to increasing both net CNS and target-site exposure, when such exposure is dictated by efflux transport, are considered.
Article
With the aim to develop a PET tracer to visualize P-glycoprotein (Pgp) expression levels in different organs, the Pgp inhibitor MC113 was labeled with (11)C and evaluated using small-animal PET. [(11)C]MC113 was synthesized by reaction of O-desmethyl MC113 with [(11)C]methyl triflate. Small-animal PET was performed with [(11)C]MC113 in FVB wild-type and Mdr1a/b((-/-)) mice (n=3 per group) and in a mouse model of high (EMT6Ar1.0) and low (EMT6) Pgp expressing tumor grafts (n=5). In the tumor model, PET scans were performed before and after administration of the reference Pgp inhibitor tariquidar (15mg/kg). Brain uptake of [(11)C]MC113, expressed as area under the time-activity curve from time 0 to 60min (AUC(0-60)), was moderately but not significantly increased in Mdr1a/b((-/-)) compared with wild-type mice (mean±SD AUC(0-60), Mdr1a/b((-/-)): 88±7min, wild-type: 62±6min, P=0.100, Mann Whitney test). In the tumor model, AUC(0-60) values were not significantly different between EMT6Ar1.0 and EMT6 tumors. Neither in brain nor in tumors was activity concentration significantly changed in response to tariquidar administration. Half-maximum effect concentrations (IC(50)) for inhibition of Pgp-mediated rhodamine 123 efflux from CCRFvcr1000 cells were 375±60nM for MC113 versus 8.5±2.5nM for tariquidar. [(11)C]MC113 showed higher brain uptake in mice than previously described Pgp PET tracers, suggesting that [(11)C]MC113 was only to a low extent effluxed by Pgp. However, [(11)C]MC113 was found unsuitable to visualize Pgp expression levels presumably due to insufficiently high Pgp binding affinity of MC113 in relation to Pgp densities in brain and tumors.
Article
Purpose: It has been suggested that altered drug permeability across the blood–brain barrier (BBB) may be involved in pharmacoresistance to antiepileptic drugs (AEDs). To test this hypothesis further, we measured multiple drug resistance (MDR) gene expression in endothelial cells (ECs) isolated from temporal lobe blood vessels of patients with refractory epilepsy. ECs from umbilical cord or temporal lobe vessels obtained from aneurysm surgeries were used as comparison tissue. Methods: cDNA arrays were used to determine MDR expression. MDR protein (MRP1) immunocytochemistry and Western blot analysis were used to confirm cDNA array data. Results: We found overexpression of selected MDR and significantly higher P-glycoprotein levels in “epileptic” versus “control” ECs. Specifically, MDR1, cMRP/MRP2, and MRP5 were upregulated in epileptic tissue, whereas Pgp3/MDR3 levels were comparable to those measured in comparison tissue. The gene encoding cisplatin resistance–associated protein (hCRA-α) also was overexpressed in epileptic tissue. Immunocytochemical analysis revealed that MDR1 immunoreactivity was localized primarily in ECs; MRP1 protein levels also were significantly higher in epileptic tissue. Conclusions: Complex MDR expression changes may play a role in AEDs pharmacoresistance by altering the permeability of AEDs across the BBB.
Article
The protein content of 14 rat brain regions and 44 human brain regions was assayed. With the human tissue we compared brain areas from adult with those from aged subjects. In each case tissue was obtained soon after death and was quickly frozen. Although the heterogeneous distribution of many proteins in the brain is well established, unexpectedly the content of protein per unit weight of fresh tissue showed little variation either regionally or with age. It seems that the various regional heterogeneities of proteins cancel each other, resulting in a fairly homogeneous distribution of the total protein content.
Article
Introduction: P-Glycoprotein (ABCB1, MDR1) is a multidrug efflux pump that is a member of the ATP-binding cassette (ABC) superfamily. Many drugs in common clinical use are either substrates or inhibitors of this transporter. Quantitative details of P-glycoprotein inhibition by pharmaceutical agents are essential for assessment of their pharmacokinetic behavior and prevention of negative patient reactions. Cell-based systems have been widely used for determination of drug interactions with P-glycoprotein, but they suffer from several disadvantages, and results are often widely variable between laboratories. We aimed to demonstrate that a novel liposomal system employing contemporary biochemical methodologies could measure the ability of clinically used drugs to inhibit the P-glycoprotein pump. To accomplish this we compared results with those of cell-based approaches. Methods: Purified transport-competent hamster Abcb1a P-glycoprotein was reconstituted into a unilamellar liposomal system, Fluorosome-trans-pgp, whose aqueous interior contains fluorescent drug sensors. This provides a well-defined system for measuring P-glycoprotein transport inhibition by test drugs in real time using rapid fluorescence-based technology. Results: Inhibition of ATP-driven transport by Fluorosome-trans-pgp employed a panel of 46 representative drugs. Resulting IC50 values correlated well (r2=0.80) with Kd values for drug binding to purified P-glycoprotein. They also showed a similar trend to transport inhibition data obtained using LLC-MDR1 cell monolayers. Fluorosome-trans-pgp IC50 values were in agreement with published results of digoxin drug-drug interaction studies in humans. Discussion: This novel approach using a liposomal system and fluorescence-based technology is shown to be suitable to study whether marketed drugs and drug candidates are P-glycoprotein inhibitors. The assay is rapid, allowing a 7-point IC50 determination in <6 min, and requires minimal quantities of test drug. The method is amenable to robotics and offers a cost advantage relative to conventional cell-based assays. The well-defined nature of this assay also obviates many of the inherent complications and ambiguities of cell-based systems.