SlideShare a Scribd company logo
1 of 14
Download to read offline
REVIEW ARTICLE
Drug transport across the blood–brain barrier
William M Pardridge
The blood–brain barrier (BBB) prevents the brain uptake of most pharmaceuticals. This property arises from the epithelial-like tight
junctions within the brain capillary endothelium. The BBB is anatomically and functionally distinct from the blood–cerebrospinal
fluid barrier at the choroid plexus. Certain small molecule drugs may cross the BBB via lipid-mediated free diffusion, providing the
drug has a molecular weight o400 Da and forms o8 hydrogen bonds. These chemical properties are lacking in the majority of
small molecule drugs, and all large molecule drugs. Nevertheless, drugs can be reengineered for BBB transport, based on
the knowledge of the endogenous transport systems within the BBB. Small molecule drugs can be synthesized that access carrier-
mediated transport (CMT) systems within the BBB. Large molecule drugs can be reengineered with molecular Trojan horse delivery
systems to access receptor-mediated transport (RMT) systems within the BBB. Peptide and antisense radiopharmaceuticals are
made brain-penetrating with the combined use of RMT-based delivery systems and avidin–biotin technology. Knowledge on
the endogenous CMT and RMT systems expressed at the BBB enable new solutions to the problem of BBB drug transport.
Journal of Cerebral Blood Flow & Metabolism (2012) 32, 1959–1972; doi:10.1038/jcbfm.2012.126; published online 29 August 2012
Keywords: blood–brain barrier; cerebrospinal fluid; endothelium; receptors; vascular biology
INTRODUCTION
The blood–brain barrier (BBB) prevents entry into the brain of
most drugs from the blood. The presence of the BBB makes
difficult the development of new treatments of brain diseases, or
new radiopharmaceuticals for neuroimaging of brain. All of the
products of biotechnology are large molecule drugs that do not
cross the BBB. While it is assumed that small molecules are freely
transported across the BBB, B98% of all small molecules are not
transported across the BBB.1
The lack of small molecule transport
across the BBB is illustrated by the whole body autoradiogram in
Figure 1A. The mouse was euthanized 5 minutes after the
intravenous (IV) injection of [14
C]-histamine, a small molecule of
only 111 Da. Histamine rapidly enters the postvascular space of all
organs of the body, with the exception of the brain and spinal
cord. The anatomical site of the BBB is endothelial wall of the
microvasculature in the brain.2
The density in the brain of the
microvessels is shown at the light microscopic level for rat brain in
Figure 1B, and at the electron microscopic level for the human
brain in Figure 1C. The BBB is comprised of two membranes in
series, the luminal and abluminal membranes of the brain capillary
endothelium, which are separated by about 200 nm of endothelial
cytoplasm. Owing to the presence of epithelial-like, high
resistance tight junctions within the brain capillary endothelium,
the intercellular pores that exist in the endothelial barriers in
peripheral organs are absent in the endothelial barrier in the brain.
In addition, there is minimal fluid-phase pinocytosis in brain
capillary endothelium.2
The absence of paracellular or transcellular
channels within the BBB means that molecules in the circulation
gain access to brain ISF (interstitial fluid) via only one of the two
mechanisms: (1) lipid-mediated free diffusion through the BBB or
(2) carrier- or receptor-mediated transport (RMT) through the BBB.
The BBB is the principal interface between blood and the ISF
that bathes synaptic connections within the parenchyma of the
brain. A separate barrier system in the brain is localized to the
epithelial cells of the choroid plexus, which form the blood–
cerebrospinal fluid (CSF) barrier (BCSFB). The choroid plexus is the
principal interface between the blood and CSF that bathes the
surface of the brain.
The BBB is the fundamental problem blocking progress in the
development of new therapeutics for brain disorders, or the
development of new radiopharmaceuticals for imaging brain. This
review will discuss both the principals of BBB drug transport, and
strategies for the reengineering of drugs to enable BBB transport.
The classical approaches to drug delivery to the brain, transcranial
drug delivery or small molecules, are discussed, and the limita-
tions of these strategies are reviewed. The endogenous carrier-
mediated transport (CMT) and RMT systems within the BBB are
discussed, and strategies are reviewed for the reengineering of
pharmaceuticals that penetrate the brain from blood via access of
the CMT and RMT systems within the BBB.
HISTORICAL ORIGINS OF MODERN CONCEPTS ON BRAIN DRUG
TRANSPORT
Drug distribution into CSF is frequently used as an indicator of BBB
transport. This is a misconception that stems from the lumping of
the BBB and BCSFB systems as a single brain barrier. In fact, the
BCSFB is leaky compared with the BBB, and all molecules in blood
enter the CSF at a rate inversely related to molecular weight
(MW).3
The finding of drug penetration into CSF is expected for
any molecule, and does not provide information on rates of drug
penetration across the BBB at the brain capillary endothelium.
Thus, drug distribution in CSF is not an index of BBB transport, but
rather is simply a measure of transport across the choroid plexus
at the BCSFB. The idea that CSF composition reflects BBB transport
is 100 years old, and finds its origins in Goldman’s vital dye
experiments published in 1913.4
Before 1913, it was known that
certain acidic vital dyes, such as trypan blue, are excluded from
Department of Medicine, UCLA, Los Angeles, California, USA. Correspondence: Dr WM Pardridge, Department of Medicine, UCLA, Warren Hall 13-164, 900 Veteran Avenue,
Los Angeles, CA 90024, USA.
E-mail: wpardridge@mednet.ucla.edu
Received 15 June 2012; revised 4 August 2012; accepted 9 August 2012; published online 29 August 2012
Journal of Cerebral Blood Flow & Metabolism (2012) 32, 1959–1972
& 2012 ISCBFM All rights reserved 0271-678X/12 $32.00
www.jcbfm.com
entry into the brain following peripheral administration, and this
observation was explained, not within the context of a barrier
between blood and brain, but rather because the brain lacked the
‘receptors’ that bind the dye and sequester the molecule within
the tissue. Goldman injected the trypan blue into the lumbar sac,
or subdural space, of rabbits and observed vital dye staining of
spinal cord and brain tissue, respectively.4
Thus, the brain did
express vital dye binding sites. The failure to stain the brain
following peripheral administration of the dye was hypothesized
to be due to a ‘blut-gehirn schranke’ or BBB. However, in 1913, it
was believed that the pathway of nutrient flux from blood to brain
took place across the choroid plexus. The anatomical site of the
BBB was erroneously placed at the choroid plexus.4
The idea that
nutrient flux from blood to brain occurred across the capillaries
within the parenchyma of brain was not accepted in 1913.
By the 1930s, studies with brain-penetrating basic vital dyes
showed that the dye entered the brain from blood across the
walls of capillaries perfusing brain parenchyma.5
Broman6
and
Friedemann5
clearly observed that the BBB was localized to the
capillary wall in brain, and that drug entry into the CSF across the
choroid plexus was an entirely separate problem from drug
transport across the BBB. Indeed, in his 1942 review on the BBB,
Friedemann5
stated that, ‘distribution between blood and CSF is
an entirely different problem and remains outside the scope of
this review.’ Nevertheless, the idea that the BBB and BCSFB are
functionally equivalent, and that the CSF and ISF are functionally
equivalent, is perpetuated in neuroscience text books.7
These
ideas form the basis for bypassing the BBB with intrathecal drug
delivery to the brain. In 2013, or 100 years after the publication
of Goldman’s experiment defining the BBB, there are multiple
ongoing clinical trials of intrathecal delivery to the human brain of
drugs that do not cross the BBB.
BRAIN DRUG TRANSPORT AND CEREBROSPINAL FLUID
The brain has no lymphatic system. The CSF of the brain is
produced at the choroid plexi of the ventricles, moves over the
surface of the brain, and is absorbed into the general circulation
across the arachnoid villi into the superior sagittal sinus of the
venous bloodstream.8,9
In the human brain, there is about 100 to
140 mL of CSF, and this entire volume is turned over completely
every 4 to 5 hours, or 4 to 5 times per day.10
In the mouse brain,
there is about 40 mL of CSF, and the entire volume is turned over
every 2 hours, or about 12 times per day.11
Cerebrospinal fluid is a
fluid compartment in rapid equilibrium with the blood, which is
due to the rapid rate of bulk flow (convection) of CSF from brain to
blood. Conversely, drug movement into brain tissue from the CSF
flow tracts occurs via diffusion. The differential rates of convection
and diffusion create the paradox that drug injected into the CSF
distributes easily to blood and poorly to brain beyond the
ependymal surface.
Poor Drug Transport from Cerebrospinal Fluid to Brain
The poor distribution of drug into brain parenchyma is shown by
the autoradiogram of rat brain prepared 20 hours after a single
injection of [125
I]-BDNF (brain-derived neurotrophic factor) into a
lateral ventricle.12
The drug distributes only to the ependymal
surface of brain ipsilateral to the ventricular injection (Figure 2A).
From the lateral ventricle, the drug moves via bulk flow to
the third ventricle (3 V), then to the fourth ventricle, then over the
surface of the brain, where the drug undergoes exodus from the
CSF space to the venous blood. In contrast to the relative rapid
rate of bulk flow of CSF out of brain, the transport of drug from the
CSF to brain tissue is slow, and limited by diffusion. Diffusion
decreases with the square of the distance. The limited distribution
into brain from the CSF is also observed for small molecules. The
concentration of water-soluble small molecules in brain tissue at
increasing distances from the CSF surface was measured in the
primate brain following the intracerebroventricular (ICV) injection
of the drug.13
Small molecule drug concentration in the brain
decreased logarithmically with each mm of distance from the CSF
surface. There was an B10-fold decrease in drug concentration of
small molecule with each mm of distance removed from the brain
surface. In the case of a lipid soluble small molecule, which can
diffuse into brain cells, there is a 10-fold decrease in drug
concentration with each 500 mm of distance into the brain.14
The
logarithmic decrease in drug concentration in the brain removed
from the CSF surface is even steeper for a large molecule drug,
which has a lower diffusion coefficient. To achieve a therapeutic
drug level in the brain at 5 mm removed from the CSF surface, it is
necessary to inject into the CSF a dose of drug that generates at
least a 5-log increase in CSF drug concentration. This approach
Figure 1. (A) Whole body autoradiogram of a mouse euthanized
5 minutes after the intravenous injection of [14
C]-histamine.124
(B)
India ink visualization of complexity of the microvasculature of the
cortex of rat brain.125
(C) Vascular cast of the microvasculature of the
human cerebellar cortex. The distance between capillaries in brain is
about 40 mm.126
Blood–brain barrier drug delivery
WM Pardridge
1960
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
may cover drug distribution in the mouse brain, where all parts of
the brain are within 3 to 5 mm of the CSF surface. However, in the
human brain, the distance between brain and the CSF surface is
up to 50 mm, which precludes significant drug distribution into
brain parenchyma from the CSF space. A side effect of the ICV
injection of very high doses of drug is the exposure of the
ependymal surface of the brain to potentially toxic concentrations
of drug. The ICV injection of nerve growth factor in rats or
monkeys15
or basic fibroblast growth factor in rats16
causes
leptomeningeal changes with axonal sprouting and astrogliosis at
the ependymal surface of the brain.
In addition to the macrocirculation of CSF through the
ventricular system, there is a CSF microcirculation through the
Virchow-Robin compartment, which is formed by the perivascular
space around penetrating cortical precapillary arterioles, which
emanate from the pial vessels on the surface of the brain.17
However, the fluid flow via the microcirculation is quantitatively
small compared with the CSF macrocirculation. The rate of flow of
the CSF macrocirculation is 2 mL/min in the rat,8
whereas the rate
of flow of the Virchow-Robin microcirculation is only 5% of the
CSF flow rate, or 0.1 mL/min in the rat.18
Rapid Drug Transport from Cerebrospinal Fluid to Blood
Fishman and Christy19
observed nearly 50 years ago that an
intrathecal injection of drug ‘is, in effect, equivalent to no more
than a prolonged IV injection.’ The rapid movement of CSF to
blood means that drug injected into the CSF compartment will
also rapidly move to the general bloodstream. This creates the
paradox that drugs injected into the CSF may have a pharma-
cological effect within brain parenchyma, but only after transport
of the drug from CSF to blood and then to brain across the BBB
(Figure 2B). Working with barbiturates in dogs, Aird20
observed
that the anesthesia was maintained at the same dose of drug,
0.2 mg per kg per minute, whether the barbiturate was infused
by either the IV or the ICV route of administration. Drug injected
into the CSF is rapidly distributed to blood, followed by entry into
brain tissue from the blood via transport across the BBB. Drug
concentration in the dialysate of an intracerebral dialysis fiber was
measured following the ICV injection of drug; drug entry into the
blood compartment was an obligatory intermediate step between
the drug injection into the CSF and drug entry into the dialysis
fiber placed within brain parenchyma.21
A secondary effect of the
rapid movement of drug from CSF to blood is the finding of a
pharmacological response following ICV injection, owing to the
pharmacological activity of the drug in peripheral tissues. The
effect of the neuropeptide, cholecystokinin, on feeding following
ICV injection of the peptide is mediated via cholecystokinin
transfer from CSF to blood, which enables activation of chole-
cystokinin receptors in the stomach.22
Transnasal Drug Delivery to Brain
The olfactory region of the submucous space of the nose is
contiguous with the CSF flow tracts around the olfactory lobe.
Therefore, intranasal drug administration could lead to direct
delivery into the CSF providing (1) the drug is transportable across
the nasal epithelium and (2) the drug is transportable across the
arachnoid membrane separating the submucous space of the
nose and the olfactory CSF compartment.23
The properties
governing drug distribution from the nose to the olfactory CSF
are similar to the properties determining drug transport across the
BBB. A lipid soluble small molecule introduced into the nose
would be expected to have some distribution into the olfactory
CSF. However, a large molecule, or a water-soluble small molecule,
would not enter olfactory CSF from the nose in the absence of
local injury to the nasal epithelial barriers. Injury is induced in the
nose following the intranasal instillation of volumes 4100 mL/
nares in humans.24
The volume that induces local injury in the
mouse or rat nose is likely o10 mL/nares, and most studies of
intranasal drug delivery to the brain in rodents use larger volumes,
which may create nasal membrane disruption. There are species
differences between humans and rodents with respect to the part
of the nasal mucosa that corresponds to the olfactory region.
Whereas the olfactory region of the nasal epithelium constitutes
50% of the nasal mucosa in the rat or mouse, the olfactory
region is only 3% to 5% of the nasal mucosa in humans.25,26
Consequently, it is difficult to detect drug movement into CSF
following intranasal drug delivery in humans. When vitamin B12 or
melatonin was administered to humans with small volumes, for
example, 70 mL/nares, no drug distribution to CSF was observed
following intranasal drug administration.24
Convection-Enhanced Diffusion
Drug delivery to the brain via the CSF is limited by the slow
diffusion of drug from the ependymal surface of the brain
(Figure 2A). So as to replace diffusion with bulk flow, drug delivery
to the brain has been attempted with CED (convection-enhanced
diffusion). In this approach, drug is placed in a peripheral reservoir
connected to a pump, which is in line with a transcranial catheter.
In theory, drug will penetrate the brain tissue by bulk flow, which
should produce high drug levels in the brain at sites remote from
the intracerebral catheter. In practice, drug concentrations in the
brain decrease logarithmically with each mm of distance removed
from the tip of the CED catheter in the brain. A logarithmic
Figure 2. (A) Film autoradiogram of rat brain section 20 hours after
the injection of [125
I]-BDNF (brain-derived neurotrophic factor) into
the lateral ventricle (LV).12
(B) Solute distribution into brain
parenchyma from the cerebrospinal fluid (CSF) compartment is
limited by the slow rate of solute diffusion from the ependymal
surface, relative to the rapid rate of bulk flow (convection) of CSF
from the ventricles to the systemic circulation across the arachnoid
villi. (C) Logarithmic decrease in the brain concentration of glial-
derived neurotrophic factor (GDNF) in the brain of the Rhesus
monkey relative to the distance from the intracerebral catheter used
for brain drug delivery with convection-enhanced diffusion.127
Blood–brain barrier drug delivery
WM Pardridge
1961
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
decrease in the brain concentration of GDNF (glial-derived neuro-
trophic factor) is observed following the CED infusion of the
neurotrophin in the monkey brain,27
and these data are plotted in
Figure 2C. Instead of a broad distribution of the drug into brain
tissue beyond the catheter, which would be expected for a
convection or bulk flow process, there is a logarithmic decrease in
drug distribution in the brain, which is typical of a diffusion
process. It would appear that the resistance of brain tissue reduces
convection within the brain beyond the catheter tip. Similar to
intrathecal drug delivery to the brain, the CED approach requires
exposure of parts of brain proximal to the catheter to very high
drug levels, to achieve therapeutic levels of drug at sites of brain
distal to the catheter (Figure 2C).
Intrathecal or intracerebral drug delivery to the brain is limited
by diffusion, which causes logarithmic gradients of drug concen-
tration. In contrast, diffusion plays no role in the transvascular
route of drug delivery to the brain. The distance between capil-
laries in brain is about 40 mm (Figure 1C). Even a large molecule
antibody drug diffuses 40 mm within a second. Therefore, once a
drug is delivered across the BBB from blood, there is instanta-
neous equilibration of drug throughout the entire brain volume.
There are three mechanisms of trans-BBB transport of drug:
(1) lipid-mediated transport of lipid soluble small molecules;
(2) CMT transport of water-soluble small molecules that have an
affinity for an endogenous BBB CMT system; and (3) RMT transport
of large molecules that have a high affinity for an endogenous BBB
RMT system.
LIPID-MEDIATED TRANSPORT OF SMALL MOLECULES
Almost all drugs for the brain presently in clinical practice are lipid
soluble small molecules with an MW o400 Da. These drugs fit the
dual criteria for lipid-mediated free diffusion across the BBB, which
are (1) MW o400 Da threshold and (2) high lipid solubility, which
is equivalent to low hydrogen bonding. In practice, very few small
molecule drug candidates for the brain fit these dual criteria, and
do not cross the BBB. Of 46,000 drugs in the Comprehensive
Medicinal Chemistry database, only 6% are active in the brain, and
these drugs treat only a select group of brain diseases, comprised
of affective disorders and insomnia.28
In another study, only
12% of all drugs were CNS active, but if affective disorders are
excluded, only 1% of all drugs are active in the CNS.29
CNS active
drugs form r7 hydrogen bonds with solvent water.30
These
considerations indicate that the BBB transport of a small molecule
drug candidate can be predicted based on the chemical structure
of the drug, which allows for computation of (1) MW and
(2) hydrogen bonding.
Molecular Weight Threshold
Small molecule drug permeation through lipid membranes
demonstrates an MW threshold, which is not predicted by the
classical Overton rules for solute diffusion in water.31,32
Unlike
diffusion through water, solute diffusion through lipid membranes
is dependent on the molecular volume of the solute, which is
generally proportional to solute MW or solute surface area. That is,
when the size of the drug exceeds the volume threshold, then
membrane permeation is less than predicted on the basis of the
lipid solubility of the drug. The MW threshold of BBB drug
transport of small molecules has been demonstrated previously
in studies of drug penetration into the brain.33,34
Blood–brain
barrier permeation decreases 100-fold when the size of the drug is
increased from an MW of 300 Da, which corresponds to a surface
area of 50 square angstroms, to an MW of 450 Da, which cor-
responds to a surface area of 100 square angstroms.35
The
molecular mechanism of the MW threshold phenomenon is
consistent with the observation that transient water pores form
in biological membranes, which are caused by the transient
fluctuations in the conformation of the fatty acyl side chains of
membrane lipids.36–38
Hydrogen Bonding
The lipid solubility of a drug may be determined by measurement
of the 1-octanol lipid partition coefficient. However, an important
parameter, which can be determined just from an inspection of
the drug structure, is the hydrogen bonding of the drug and
solvent water, which is 55 molar. Diamond and Wright39
reviewed
the rules for determination of the number (N) of hydrogen bonds
a solute forms in water, for example, N ¼ 2 for each hydroxyl, N ¼ 3
for each primary amine, and N ¼ 4 for each amide group. Stein40
observed that there is a 10-fold decrease in membrane per-
meation with each pair of hydrogen bonds on the solute, and this
rule has been generally confirmed in studies of in vivo BBB transport
of steroid hormones41
and oligopeptides.42
Consideration of the
rules for hydrogen bonding show that even a simple dipeptide,
glycylglycine, which forms eight hydrogen bonds with water,
would have a very low BBB permeation. Conversely, if the
glycylglycine was cyclized, the number of hydrogen bonds of the
cyclic glycylglycine would be reduced to 4, and BBB transport
would be predicted to increase by 100-fold for the cyclic form
of the glycylglycine dipeptide, because glycine has no polar
side chains.
When confronted with the problem of a water-soluble drug that
does not penetrate the BBB, the medicinal chemist is asked to fix
the problem by lipidizing the drug. This can be done by blocking
Table 1. BBB carrier-mediated transporters
Carrier Substrate Genea
product Kmb
(uM) Vmax b,c
(nmol/
min/g)
Ccapd
(pmol/
mgp)
Vmax:Cape
(molecules
per second)
Glucose D-glucose GLUT1
(SLC2A1)
11,000±1,400 1,420±140 139±46 600
Monocarboxylic
acid
L-lactic acid MCT1
(SLC16A1)
1,800±600 91±35 2.3±0.8 2,300
Neutral amino
acid
L-phenyl-
alanine
LAT1 (SLC7A5) 26±6 22±4 0.43±0.09 3,000
Basic amino acid L-arginine CAT1
(SLC7A1)
40±24 5±3 1.1±0.2 270
Purine nucleoside Adenosine CNT2
(SLC28A2)
25±3 0.75±0.08 o0.14 ND
BBB, blood–brain barrier. a
Solute carrier (SLC) group of transporter genes. b
From Pardridge.47 c
Per gram wet brain in the rat. d
From Uchida et al.48
Data
reported per mg isolated human brain capillary protein. e
Vmax/Ccap ratio is the transporter turnover number, which is the number of substrate molecules
transported per second by a single transporter molecule, and is based on 0.14 mg capillary protein per gram wet brain; it is assumed that 50% of the total
capillary transporter is expressed on the endothelial luminal membrane; nd ¼ not determined.
Blood–brain barrier drug delivery
WM Pardridge
1962
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
hydrogen bond forming functional groups on the drug. For example,
when the two hydroxyl groups of morphine are acetylated, to
form heroin, the BBB penetration rate increases 100-fold.43
However, apart from the morphine/heroin example, there have
been few successes in developing new drugs for human brain
disorders by lipidization of a water-soluble drug. This is because
the hydrogen blocking functional groups are rapidly hydrolyzed
in vivo. If the hydrogen bond is blocked with a noncleavable group,
then drug activity may be reduced. An alternative approach to the
medicinal chemical modification of non-BBB-penetrating drugs is
to focus on the endogenous CMT systems within the BBB. Instead
of blocking hydrogen bonds, medicinal chemistry can be used to
modify the structure of the drug to increase drug affinity for one
of several endogenous carrier-mediated BBB transporters.
CARRIER-MEDIATED TRANSPORT OF SMALL MOLECULES
Discovery of Blood–Brain Barrier Carrier-Mediated Transport
Systems
Physiology of blood–brain barrier carrier-mediated transport. The
first evidence of saturable or CMT of a solute across the BBB was
demonstrated by Crone44
for D-glucose using the indicator dilution
technique, which is a venous sampling-carotid artery injection
method. Subsequently, the Brain Uptake Index technique, a tissue
sampling-carotid artery injection method developed by Oldendorf45
,
was used to characterize the saturable kinetics of BBB transport of
multiple water-soluble metabolic substrates. The Michelis–Menten
kinetics of BBB CMT for the major metabolic substrates were
determined,46
and this analysis showed a wide range of affinity
(Km) and maximal transport activity (Vmax) for the BBB nutrient
transporters (Table 1).
Molecular biology of blood–brain barrier carrier-mediated transport.
The methods of molecular biology were employed to identify the
principal gene product responsible for BBB transport of a given
class of metabolic substrates. The BBB glucose carrier, which also
transports other hexoses, including mannose, galactose, deox-
yglucose, or 3-0-methyl glucose,49
is the glucose transporter type
1 or GLUT1.50
The BBB phenylalanine carrier, which also transports
over 10 other large neutral amino acids,51
and to a lesser extent
small neutral amino acids, was identified as the large neutral
amino-acid transporter type 1 or LAT1.52
The BBB arginine carrier,
which also transports other cationic amino acids (lysine,
ornithine),45
was shown to be the cationic amino-acid trans-
porter type 1 or CAT1.53
The BBB lactate carrier, which also
transports other monocarboxylic acids (pyruvate, and the ketone
bodies, acetoacetate, and b-hydroxybutryyrate),54
was identified
as the monocarboxylic acid transporter type 1 or MCT1.55
The BBB
adenosine transporter, which also transports other purine nucle-
osides (guanosine, inosine), as well as the pyrimidine nucleoside,
uridine,56
is the concentrative nucleoside transporter type 2 or
CNT2 in the rat.57
Quantitation of blood–brain barrier transporter protein expression.
Terasaki and colleagues58
determined the mass of transporters in
isolated brain capillaries using the methods of LC–MS (liquid
chromatography mass spectrometry). Capillaries were isolated
from brain followed by digestion with trypsin. The tryptic peptides
were separated and identified by LC–MS. Transporter-specific
peptides were identified and quantitated by solid-phase synthesis
of synthetic peptide standards. These peptide standards, which
enable the quantification of the transporter expression, could be
synthesized based on the known amino-acid sequences of the
cloned transporters, as well as standard programs for predicting
the sequence of tryptic peptides based on known amino-acid
sequence of the target protein. The transporter concentrations in
isolated human brain capillaries, Ccap, are listed in Table 2 for
human GLUT1, MCT1, LAT1, and CAT1, and are compared with the
transporter Vmax values determined in vivo in the rat. The
comparison between rat Vmax values and human Ccap values was
made, because the Ccap values are generally comparable for
human and rodent brain capillaries.48
Transporter turnover number. The Vmax values are expressed per
gram wet brain, whereas the Ccap values are expressed per mg of
brain capillary protein. It is estimated that there is 0.17 mg
capillary protein per gram brain, since there is 100 mg protein per
gram brain, and the intracellular water volume of the brain
capillary endothelial space, 1 mL/g,61
is 1/700 parts of the total
water volume of brain. Based on this conversion, and assuming
50% of the capillary transporters are expressed on the luminal
endothelial membrane, the Vmax/Ccap ratio can be computed,
which is the transporter turnover number, or molecules of
substrate transported by the carrier per second (Table 1). The
estimated turnover numbers for BBB CMT systems range from 270
to 3,000 molecules/s. These calculations are approximations given
the divergent in vitro and in vivo methodologies that were
performed. Nevertheless, the estimated turnover rates approx-
imate the turnover rate for other transporters, for example, the
turnover number for the sucrose transporter (SUT1) is 500 mole-
cules/s.62
To design drugs that access the CMT systems listed in
Table 1, it is necessary to define the exact cellular location of the
transporter at the brain microvasculature. A transporter identified
in an isolated brain capillary preparation could be selectively
expressed on the capillary endothelium, on the capillary pericyte,
on smooth muscle cells of precapillary arterioles, or even on
astrocyte endfeet, which remain adhered to the basement
membrane of the isolated capillary. Following localization of the
transporter to the capillary endothelium, it is necessary to
determine whether the transporter is expressed on the luminal
Table 2. Reengineering of protein drugs for BBB penetration as IgG Trojan horse fusion proteins
Therapeutic class Protein therapeutic Target brain disease Animal models
Lysosomal enzymes Iduronidase (IDUA) MPS-I (Hurler’s syndrome) Rhesus monkey, Hurler mouse
Iduronate 2-sulfatase (IDS) MPS-II (Hunter’s syndrome) Rhesus monkey
Decoy receptors Type II TNFa decoy receptor (TNFR) Stroke neurodegeneration Rhesus monkey, Mouse PD, Mouse stroke
Neurotrophins Erythropoietin (EPO) Stroke Neurodegeneration Rhesus monkey, Mouse PD
Glial-derived neurotrophic factor (GDNF) Neurodegeneration Stroke Rhesus monkey, Mouse PD
Therapeutic antibodies Antiamyloid antibody (AAA) Alzheimer’s disease Rhesus monkey, Mouse AD
BBB, blood–brain barrier.
Primary references are given in Pardridge and Boado59
and Sumbria et al.60
Blood–brain barrier drug delivery
WM Pardridge
1963
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
membrane, the abluminal membrane, or both. The cellular and
subcellular expression of a BBB transporter is illustrated in the case
of GLUT1 expression at the BBB.
Expression of GLUT1 in Brain Is Localized to the Blood–Brain
Barrier
There are over 10 members of the sodium-independent GLUT
glucose transporter gene family, and the first transporter identi-
fied, GLUT1, was cloned from human HepG2 liver cells using an
antibody to the human erythrocyte glucose transporter.63
Cloning
of the cDNA allowed for prediction of the amino-acid sequence
and a two-dimensional representation of the transporter struc-
ture,63
which is shown in Figure 3A for the GLUT1 glucose
transporter cloned from a bovine BBB cDNA library.64
Shortly after
cloning from human liver cells, the same GLUT1 glucose trans-
porter cDNA was cloned from a rat brain cDNA library, and the
transporter was originally known as the brain-type glucose
transporter.65
The GLUT1 glucose transporter was observed to
be enriched at the brain microvasculature.66
However, studies of
GLUT1 glucose transporter mRNA expression in the brain with the
capillary depletion method showed that the GLUT1 glucose
transporter was expressed in the brain only at the BBB, with little,
if any, detection of GLUT1 transcript in mRNA devoid of capillary-
derived mRNA.67
The GLUT1 Northern blot for capillary-depleted
brain is illustrated in Figure 3B. The 2.9-kb GLUT1 glucose
transporter mRNA was detected in Northern blotting of non-
fractionated RNA from the brain (lane 1, left panel, Figure 3B).
However, the GLUT1 glucose transporter mRNA was 495%
removed with the capillary depletion method, which had no
effect on the abundance of a common transcript, the 2.0-kb actin
mRNA (Figure 3B). This finding indicated that GLUT1-mediated
glucose transport at the BBB, but not at brain cells. The selective
expression of the GLUT1 isoform at the BBB in the brain was
subsequently confirmed with in situ hybridization, quantitative
Western blotting,50
and quantitative immunogold electron micro-
scopy.68
The GLUT1 glucose transporter is also expressed in other
barrier structures in the brain, including the basolateral membrane
of choroid plexus epithelium, and the apical membrane of certain
regions of the ependymal epithelium.69
The localization of the immunoreactive GLUT1 glucose trans-
porter at the human BBB was demonstrated with electron micro-
scopy using an antibody directed against the human erythrocyte
GLUT1 glucose transporter and a secondary antibody conjugated
with 10 nm gold particles.70
GLUT1 glucose transporters are
observed on the endothelial plasma membranes, but not within
the neuropil of human brain (Figure 3C). The expression of the
GLUT1 glucose transporter on both luminal and abluminal mem-
branes at the human BBB illustrates how these two endothelial
membranes operate in series to regulate BBB transport (Figure 3C).
GLUT1 is also detected on the plasma membrane of the human
erythrocyte (Figure 3C). The abundance of GLUT1 within the
human erythrocyte plasma membrane is said to be highest of all
transporters. Yet, the abundance of GLUT1 glucose transporter,
expressed as transporter molecules per micron of membrane length,
is 2.6-fold higher on the luminal membrane of the human brain
capillary endothelium relative to the human red cell membrane.70
The abundance of the GLUT1 glucose transporter, Ccap, at the
microvasculature of human brain is high, relative to other
transporters, 139±46 pmol/mg protein, as determined by LC–
MS (Table 1). The Ccap value determined with LC–MS conforms to
a previous estimate of the Ccap of GLUT1, 102±25 pmol/mg
protein, determined with a cytochalasin B radioreceptor assay in
isolated brain capillaries.71
The Ccap of the GLUT1 glucose
transporter is nearly 100-fold higher than the Ccap for other
transporters (Table 1), which is attributed to the sole derivation of
Figure 3. (A) Predicted secondary structure of the bovine blood–brain barrier (BBB) GLUT1 glucose transporter is shown, which is formed by
12 transmembrane domains, a glycosylated extracellular loop between transmembrane domains 1 and 2, and intracellular amino and carboxyl
termini.128
(B) Northern blot of mRNA derived from either total rabbit brain (lane 1) or capillary-depleted rabbit brain (lane 2) for either the
GLUT1 glucose transporter or actin.71
(C) Electron microscopic immunogold study of human brain with a primary antiserum against the
purified human erythrocyte glucose transporter, and a secondary antibody conjugated with 10 nm gold particles, shows abundant expression
of immunoreactive GLUT1 glucose transporter on the luminal (L) and abluminal (AL) membranes of the capillary endothelium in human
brain.70
Blood–brain barrier drug delivery
WM Pardridge
1964
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
energy in the brain from the combustion of glucose. Cerebral rates
of glycolysis, 500 to 1,000 nmol per minute per gram, are over 100-
fold greater than rates of amino-acid incorporation into brain
proteins.46
Glucose supply in the brain is mediated solely via the
activity of the BBB GLUT1 glucose transporter.
Drug Transport into Brain via Blood–Brain Barrier Carrier-Mediated
Transporters
The characterization of the BBB GLUT1 glucose transporter
exemplifies the extensive knowledge base on the molecular
characteristics of a given BBB CMT system, and these systems have
been recently cataloged with respect to the transporter abun-
dance at the BBB.48
The cDNAs of almost all of the BBB CMT
systems are available, and cell lines can be stably transfected with
the transporter gene to enable high-throughput screening of
drugs that have an acceptable affinity for a given BBB transporter.
This knowledge base is not being exploited by the pharmaceutical
industry for the discovery of drugs that cross the BBB on a given
CMT system. However, there are historical examples of drugs that
have an unexpected affinity for a BBB CMT system that may, or
may not, be predicted on the basis of the drug structure. For
example, L-DOPA is an effective treatment for PD (Parkinson’s
disease), because this dopamine precursor is a large neutral amino
acid that is transported across the BBB via the LAT1 neutral amino-
acid carrier.51
L-DOPA is a straightforward example of a large neutral
amino-acid drug that would be predicted to have an affinity for
LAT1. However, there are examples of other drugs that have an
affinity for LAT1 that are unexpected. Gabapentin is a water-soluble
drug that is active in the CNS because the drug crosses the BBB on
the large neutral amino-acid transporter,72
which is LAT1. The
affinity of gabapentin for LAT1 is unexpected, because gabapentin
is a g-amino acid, and LAT1 has an affinity only for a-amino acids.
However, gabapentin is a cyclic form of a g-amino acid, which
places the amino and carboxyl groups in a conformation that
mimics a-amino acids. Paraquat is N,N0
-dimethyl-4,40
-bipyridinium, a
widely used, highly water-soluble herbicide, which should not cross
the BBB. However, paraquat is neurotoxic, which suggests that this
double quaternary ammonium salt is BBB penetrating. Paraquat
crosses the BBB via a process that is inhibited by valine, a large
neutral amino acid,73
which suggests that paraquat is a substrate
for LAT1. Melphalan is a polar alkylating agent used to treat brain
cancer. Melphalan, or phenylalanine mustard, crosses the BBB via
transport on the BBB large neutral amino-acid carrier.74
L-DOPA, gabapentin, paraquat, and melphalan are examples of
BBB delivery via LAT1 of drugs that have structures that mimic the
endogenous substrate, neutral amino acids. An alternative
approach is to conjugate a drug, which normally does not cross
the BBB, with a CMT substrate, for example, glucose, that does
cross the BBB. This was attempted with oligopeptides, which were
conjugated with glucose. However, the glucose moiety of the
glycopeptide no longer was transported via GLUT1.75
Instead, the
structure of the CMT substrate must be retained following conju-
gation of the nontransportable drug. This is possible with ingenious
uses of medicinal chemistry as applied to BBB structure–transport
relationships. A novel example is the case where a nontranspor-
table drug is coupled to the thiol group of L-cysteine.76
Cysteine is
a small neutral amino acid, which has a low affinity for LAT1.45
However, when the drug is conjugated to the thiol group of
cysteine, the small neutral amino acid is converted into a large
neutral amino acid, and significant BBB transport via LAT1 was
observed.76
This case shows that a CMT substrate can be used as a
drug carrier, providing the original structural characteristic of the
substrate class for the targeted CMT system is retained.
Active Efflux Transporters and Enzymatic Blood–Brain Barrier
The active efflux transporters (AET) serve to mediate the
asymmetric efflux of metabolites and drugs from the brain to
blood. The classical AET system within the BBB is p-glycoprotein,
which is a product of the MDR (multidrug resistance) gene, MDR1,
also named the ABC (ATP-binding cassette) subfamily B member 1
(ABCB1). Apart from p-glycoprotein, there are numerous other
members of the ABC gene family of transporters that are
expressed at the BBB, and these have been quantified by Terasaki
and coworkers.48
The AET systems may work in concert with
enzymes expressed within the BBB, such as P450 enzymes.77
Both
the AET systems, and the enzymatic barriers are subject to
modulation. Blood–brain barrier p-glycoprotein activity is sup-
pressed by vascular endothelial growth factor,78
and the CYP1A1
and CYP1B1 P450 enzymes are upregulated by environmental
toxins, such as dioxin derivatives, which activate the aryl
hydrocarbon receptor at the BBB.79
In addition to environmental
toxins, or growth factors, BBB permeability may be altered by
disease states, including aging.80
RECEPTOR-MEDIATED TRANSPORT OF PROTEIN
THERAPEUTICS
Discovery of Blood–Brain Barrier Receptor-Mediated Transport
Systems
Insulin is present in the brain, but there is no insulin mRNA in the
brain.81,82
Therefore, insulin in the brain is of peripheral origin and
derived from the blood. Insulin in the blood is transported across
the human BBB via RMT on the endogenous BBB insulin
receptor.83
Similarly, transferrin (Tf) in blood is transported via
RMT across the BBB on the endogenous human BBB Tf receptor
(TfR).84
There are separate BBB receptors for the insulin-like
growth factors85
and leptin.86
The BBB peptide receptors on either
the animal or human BBB were identified using isolated brain
capillaries and radioreceptor methodology. The brain capillaries
are purified from the brain with a mechanical homogenization
procedure and capillaries freshly isolated from bovine brain are
shown in Figure 4A. The BBB peptide receptors may mediate
functionally different processes, including (1) transcytosis of ligand
from blood to brain; (2) reverse transcytosis from brain to blood; or
(3) only endocytosis into the brain capillary endothelium without
net transport across the endothelial cell (Figure 4B). Insulin RMT
across the BBB from blood to brain was demonstrated with carotid
artery infusion and thaw mount autoradiography of the brain.87
Transferrin transport from the blood to brain was similarly
shown by internal carotid artery infusion and autoradiography.88
Transferrin efflux from the brain to blood was demonstrated with
the Brain Efflux Index method,89
as applied to large molecules.90
IgG molecules are asymmetrically transcytosed only in the brain to
blood direction via a Fc receptor (FcR) on the BBB.91
Confocal
microscopy of brain shows the principal FcR on the brain capillary
is the neonatal FcR or FcRn.92
The BBB FcR does not mediate the
transport of IgG molecules in the blood to brain direction. Some
BBB receptors do not mediate transcytosis across the endothelial
barrier, but only mediate endocytosis into the endothelial cell.
Acetylated LDL (low-density lipoprotein) is a ligand for the
scavenger receptor (SR) on the BBB, and this system mediates
the endocytosis of acetyl LDL from blood, but not the net
transcytosis across the BBB into the brain.93
Scavenger receptor-
related receptors, such as the LDL-related protein receptor (LRP)
type 1, similarly mediate only endocytosis of ligands into cells, and
not transcytosis.94
Proteins targeting LRP1 are taken up by brain at
reduced rates compared with proteins that target the trans-
cytosing receptor systems. The brain uptake of melanotransferrin
(p97) or angiopep-2, ligands that target LRP1, is only 0.2% to 0.3%
of injected dose (ID)/g in the mouse,95,96
which is 10-fold lower
than the brain uptake of a monoclonal antibody (MAb) that
targets the BBB TfR in the mouse.97
The BBB TfR mediates reverse transcytosis of apotransferrin in
the brain to blood direction,90
in addition to influx of holo-
transferrin from blood to brain.88
The reverse transcytosis is
Blood–brain barrier drug delivery
WM Pardridge
1965
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
enabled, because the TfR is also expressed on the abluminal
membrane of the capillary endothelium in brain. In one study, the
abluminal TfR was not detected with preembedding immune
labeling methods.98
However, abluminal receptors on the
endothelium in the brain cannot be detected with preem-
bedding methods.99
Using confocal microscopy of isolated rat
brain capillaries, and the OX26 rat MAb against the rat TfR, it was
possible to identify the TfR on both luminal and abluminal
membranes of the brain capillary endothelium (Figure 4C).100
The
pathway of transcytosis through the BBB via the TfR was examined
with electron microscopy. The OX26 TfRMAb was conjugated with
5 nm gold and infused in the carotid artery of rats for 10 minutes
followed by saline clearance of the brain vasculature and
perfusion fixation with glutaraldehyde.101
At the light
microscopic level, the TfRMAb entrapped within the capillary
endothelium was detected with a silver enhancement technique
as shown in Figure 4D. There is no visible reaction product in the
brain extravascular space (Figure 4D), owing to the differences in
intraendothelial and postendothelial volumes in the brain. The
intraendothelial volume is 1 mL/g brain and the postendothelial
volume is nearly 3-log orders higher, 700 mL/g brain. Therefore, as
the TfRMAb exits the intraendothelial volume, the antibody
concentration is diluted 700-fold, and the postvascular antibody
in the brain cannot be detected with immune labeling methods.
Figure 4. (A) Light micrograph of freshly isolated bovine brain capillaries stained with trypan blue, which stains nuclei blue and luminal red
blood cells yellow. The capillaries are isolated free of adjoining brain tissue. (B) Schematic diagram of different brain endothelial receptors,
including the insulin receptor, the transferrin (Tf) receptor, the scavenger receptor (SR), which mediates only the endocytosis from blood into
the endothelial compartment for ligands such as acetylated low density lipoprotein; and the neonatal Fc receptor (FcRn), which mediates the
asymmetric transcytosis of IgG molecules selectively from brain to blood, but not blood to brain. (C) Confocal microscopy of isolated rat brain
capillaries showing the expression of the Tf receptor on both luminal (L) and abluminal (AL) membranes of the endothelium.100
(D) Light
micrograph of rat brain removed after a 10-minute carotid artery infusion of a conjugate of 5 nm gold and the mouse OX26 monoclonal
antibody (MAb) against the rat TfR. The staining of the capillary compartment represents TfRMAb present in the intraendothelial
compartment of the brain.101
(E) Electron microscopy of rat brain after perfusion with the conjugate of gold and TfRMAb shows the MAb
concentrated in intraendothelial vesicles (intracellular arrow), as well as MAb molecules undergoing exocytosis from the endothelial
compartment to the brain interstitial space (extracellular arrow).101
Blood–brain barrier drug delivery
WM Pardridge
1966
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
The rapid postvascular distribution into the brain via transport on
the BBB TfR was demonstrated with the capillary depletion
method and autoradiography.88
At the electron microscopic level,
the OX26 TfRMAb is detected in endosomes within the capillary
endothelium (intracellular arrow, Figure 4E), and antibody is
observed exocytosing into the ISF on the brain side of the BBB
(extracellular arrow, Figure 4E).
Blood–Brain Barrier Penetrating Genetically Engineered IgG Fusion
Proteins
The presence of endogenous RMT systems within the BBB, such as
the insulin receptor or the TfR, enables the reengineering of
recombinant proteins as BBB-penetrating pharmaceuticals with
the use of molecular Trojan horse (MTH) technology.59
An MTH is
an endogenous peptide, or a peptidomimetic MAb, which traverses
the BBB on a specific RMT system. Insulin, per se, as an MTH is not
preferred, since insulin causes hypoglycemia. Tf, per se, is not a
preferred MTH, because the plasma concentration of endogenous
Tf, 25 mmol/L, is so high that the Tf-binding site on the BBB TfR is
fully saturated by endogenous Tf. Alternatively, RMT-specific MTHs
may be peptidomimetic MAbs that bind exofacial epitopes on the
BBB receptor, which are spatially removed from the endogenous
ligand binding site. Once bound to the RMT system on the BBB,
the MTH undergoes RMT across the BBB (Figure 4). An MAb
against the human insulin receptor, designated the HIRMAb,
undergoes rapid transport across the BBB of the Rhesus monkey
in vivo, and the brain uptake is 2% to 3% of ID/brain.102
The
HIRMAb crossreacts with the insulin receptor of Old World primates,
but does not react with the insulin receptor of New World primates,
such as the squirrel monkey, or lower animals such as rodents. The
murine HIRMAb has been genetically engineered as either a
chimeric or humanized antibody.103
There is no known MAb
against the mouse insulin receptor that can be used as a BBB MTH.
For BBB delivery in the mouse, a genetically engineered chimeric
MAb against the mouse TfR, designated the cTfRMAb, has been
produced.104
With the genes encoding the heavy chain and the
light chain of either the engineered HIRMAb or the cTfRMAb, IgG
fusion proteins may be genetically engineered as bifunctional
molecules, which both cross the BBB via RMT and exert pharm-
acological effects in brain, following transport into the brain.
An IgG-type MTH is also preferred for genetic engineering of
MTH fusion proteins, because the protein therapeutic can be fused
to the carboxyl terminus of the heavy chain of the IgG, as shown
in Figure 5A. It is essential that the bifunctionality of the IgG fusion
protein be retained, and that the ‘head’ of the molecule retains
high affinity (low nmol/L KD) binding to the BBB receptor, and that
the ‘tail’ of the IgG fusion protein retains the pharmacological
activity of the original protein therapeutic. If the protein
therapeutic is a neurotrophin, there must be retention of high
affinity binding of the IgG-neurotrophin fusion protein to the
cognate neurotrophin receptor in brain. If an IgG-decoy receptor
fusion protein is engineered, there must be retention of high
affinity binding to the target cytokine in the brain. If an IgG-
lysosomal enzyme fusion protein is produced, the enzyme activity
of the IgG-enzyme fusion protein must be comparable to the
native enzyme. If the therapeutic molecule is also an antibody,
then the problem is the genetic engineering of a bispecific
antibody, where there is retention of high affinity binding for both
the BBB receptor and the target antigen. This was accomplished
by fusion of an ScFv (single chain Fv) form of the therapeutic
antibody to the carboxyl terminus of the heavy chain of the IgG.105
This molecular design places the monomeric ScFv antibody in a
dimeric configuration, which restores high avidity binding to the
target antigen of a bivalent therapeutic antibody.
If the high affinity for the target BBB receptor is retained
following engineering of the IgG fusion protein, then the rate of
Figure 5. (A) Structure of an IgG fusion protein formed by fusion of a therapeutic protein to the carboxyl terminus of the heavy chain of a
blood–brain barrier (BBB) targeting monoclonal antibody (MAb). (B) Brain uptake in the mouse, expressed as % injected dose (ID)/g brain, is
shown for diazepam,110
a cTfRMAb–EPO fusion protein,59
a cTfRMAb–GDNF fusion protein,59
a cTfRMAb–TNFR fusion protein,59
and the
cationic import peptide, tat.129
(C) Brain uptake in the Rhesus monkey is expressed as % ID/100 g brain, since the size of the primate brain is
100 g. Brain uptake is shown for fallypride,111
a HIRMAb–TNFR fusion protein,109
a HIRMAb–EPO fusion protein,107
the TNFR alone,109
EPO
alone,107
and a human IgG1k isotype control antibody (hIgG1k), which is confined to the vascular compartment.108
(D) Film autoradiogram of
Rhesus monkey brain removed 2 hours after the intravenous (IV) injection of [125
I]-HIRMAb–IDUA fusion protein, showing global distribution
in brain with higher uptake in gray matter relative to white matter.130
Blood–brain barrier drug delivery
WM Pardridge
1967
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
transport of the fusion protein across the BBB will be comparable
to the rate of transport of the MTH without a fusion partner. For
the mouse, several cTfRMAb fusion proteins were engineered,
expressed, and tested for in vivo brain uptake activity. The brain
uptake of the cTfRMAb fusion protein of EPO (erythropoietin),
GDNF, or the type II TNF-a (tumor necrosis factor-a) decoy
receptor (TNFR), is high, 2% to 3% ID/g brain at 60 minutes after an
IV injection (Figure 5B). In contrast, the brain uptake of a cationic
import peptide, tat, is low, 0.1% ID/g (Figure 5B), and this low level
of brain uptake is comparable to the brain uptake of an IgG that
does not cross the BBB in the mouse.106
Without fusion to the
MTH, there is no BBB transport of the EPO alone,107
the GDNF
alone,108
or the TNFR alone.109
The brain uptake of the cTfRMAb
fusion proteins in the mouse is comparable to the brain uptake of
a lipid soluble small molecule such as diazepam (Figure 5B), which
is 1.8% ID/g at 10 minutes after an IV injection.110
Therefore,
the brain uptake of the MTH fusion proteins in the mouse is
comparable to the brain uptake of lipid soluble small molecules.
Similar findings are made in the primate. In the Rhesus monkey,
the brain uptake of the HIRMAb fusion proteins is 2% to 3% ID/
100 g brain (Figure 5C), which is comparable to the peak brain
uptake in the Rhesus monkey of fallypride, a lipid soluble small
molecule.111
The brain uptake in the Rhesus monkey is expressed
per 100 g brain, because the weight of the brain in this species is
100 g. The brain uptake in the Rhesus monkey of the TNFR alone,
or EPO alone, is low, about 0.2% ID/100 g brain. This level of brain
uptake does not represent a low level of BBB penetration, but
rather sequestration of the protein within the vascular com-
partment of brain. The primate brain uptake of an isotype IgG1
control antibody that does not cross the BBB is also 0.2% ID/100 g
at 60 minutes after an IV injection (Figure 5C). The IgG fusion
proteins undergo transcytosis through the BBB, and penetrate
brain parenchyma. The global distribution within the brain of the
fusion protein is shown by the brain scan of the Rhesus monkey at
2 hours after the IV injection of a fusion protein of the HIRMAb,
and the human lysosomal enzyme, iduronidase (IDUA) (Figure 5D).
The higher uptake in gray matter, as compared with white matter,
is due to the higher vascular density in gray matter. The finding of
transcytosis of the fusion proteins through the BBB and into brain
parenchyma is corroborated by the in vivo CNS pharmacological
effects of IV administration of the fusion proteins in mouse models
of brain disease, including Hurler’s syndrome, PD, Alzheimer’s
disease (AD), and stroke (Table 2).
BLOOD–BRAIN BARRIER TRANSPORT OF PEPTIDE
RADIOPHARMACEUTICALS FOR NEUROIMAGING
The number of potential peptide radiopharmaceuticals that could
be used to image brain disorders is large. However, peptide
radiopharmaceuticals for the brain are not developed, because
these large molecules do not cross the BBB. Peptide radio-
pharmaceuticals can be reengineered for BBB transport with the
combined use of MTH technology, avidin–biotin technology, and
radiopharmaceutical technology. The peptide radiopharmaceuti-
cal can be conjugated to the IgG MTH via a high affinity avidin–
biotin linker, as shown in Figure 6A. An MTH–avidin fusion protein
has been genetically engineered both for the HIRMAb, for primate
or human applications, and for the cTfRMAb, for mouse
studies.112,113
The IgG–avidin fusion protein is formulated in one
vial. The monobiotinylated peptide radiopharmaceutical is for-
mulated in a second vial. The two vials are mixed before IV
administration. Owing to the very high affinity of avidin binding of
biotin, there is instantaneous capture of the peptide radio-
pharmaceutical by the MTH (Figure 6A). The efficacy of this
approach has been demonstrated in imaging models in both
primates and rodents, using either the Ab1–40
amyloid peptide of
AD or epidermal growth factor (EGF) as the model peptide
radiopharmaceutical.
The Ab1–40
amyloid peptide is a potential peptide radio-
pharmaceutical for the neuroimaging of brain amyloid in AD, as
this peptide avidly binds amyloid plaque in sections of AD autopsy
brain.114
However, the Ab1–40
amyloid peptide alone does not
cross the BBB,113,115
and the absence of BBB transport prevents
the development of this peptide as a radiopharmaceutical for
brain imaging in AD. The [N-biotinyl]-Ab1–40
was radioiodinated
and injected alone IV in the Rhesus monkey.115
No measureable
brain uptake of the peptide was observed by brain scanning
(Figure 6B). However, in a second Rhesus monkey, the [N-biotinyl]-
Ab1–40
was first conjugated to the HIRMAb via a streptavidin linker
before IV injection of the HIRMAb/Ab complex. Now, the peptide
radiopharmaceutical rapidly penetrated the BBB in the primate
(Figure 6C). Following reengineering with the MTH technology,
the brain uptake of the Ab1–40
amyloid peptide is comparable to
the brain uptake of lipid soluble small molecule amyloid imaging
agents. The brain uptake of the [N-biotinyl]-Ab1–40
alone is very
low in the mouse, 0.1% ID/g, but the brain uptake of the
peptide increases to 2.1% ID/g following conjugation to the
Figure 6. (A) Conjugate of an IgG–avidin fusion protein and a
monobiotinylated peptide radiopharmaceutical. The biotin group
on the peptide binds to the avidin domain of the fusion protein to
form a high affinity linkage between the IgG molecular Trojan horse
and the peptide radiopharmaceutical. (B, C) Scans of Rhesus
monkey brain at 3 hours after the intravenous (IV) injection of
either [125
I, N-biotinyl]-Ab1–40
alone (B), or [125
I, N-biotinyl]-Ab1–40
conjugated to the HIRMAb (monoclonal antibody) molecular Trojan
horse via a streptavidin linker (C).115
(D) A rat brain tumor model was
produced by the intracerebral injection of human U87 glioma cells
in nude rats. Immunocytochemistry (ICC) of brain removed 16 days
after tumor implantation with an antibody to the human EGFR
shows abundant expression of the EGFR in the brain tumor.119
(E) Scan of rat brain removed 2 hours after the IV injection of [111
In-DTPA, Lys-biotinyl]–EGF conjugated to the TfRMAb mole-
cular Trojan horse via a streptavidin linker. The section used
for the film autoradiography was parallel to the section used for
the ICC study in (D).119
Blood–brain barrier drug delivery
WM Pardridge
1968
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
cTfRMAb–avidin fusion protein.113
This level of brain uptake of a
peptide radiopharmaceutical is comparable to the brain uptake of
a lipid soluble small molecule brain amyloid imaging agent, which
is 2% to 3% ID/g in the mouse.116
The complex of the HIRMAb–
avidin fusion protein and [N-biotinyl]-Ab1–40
retains high affinity
binding to the amyloid plaques in autopsy sections of AD brain.117
The [N-biotinyl]-Ab1–40
can be radiolabeled with 124-iodine for
positron emission tomography imaging of the brain. Alternatively,
the [N-biotinyl]-Ab1–40
can be conjugated with a chelator moiety
and labeled with 111-indium118
for single photon emission
computed tomography.
Epidermal growth factor is a potential peptide radiopharma-
ceutical for imaging brain cancers that overexpress the EGF
receptor (EGFR). Epidermal growth factor was monobiotinylated
and conjugated with diethylenediaminepentaacetic acid (DTPA)
and radiolabeled with 111-indium. A rat model of human brain
cancer was developed with the intracranial injection of human
U87 glioma cells in nude rats.119
At autopsy, immunocytoche-
mistry with an antibody against the human EGFR showed selective
expression of the EGFR in the brain tumor (Figure 6D). IV injection
of the [111
In-DTPA, biotinyl]-EGF alone enabled no imaging of the
brain tumor in vivo, because EGF does not cross the BBB.120
However, when the [111
In-DTPA, biotinyl]-EGF was conjugated to
the TfRMAb with a streptavidin linker, there was robust imaging
of the EGFR-positive brain cancer following IV injection of the
BBB-targeted peptide radiopharmaceutical (Figure 6E).
BLOOD–BRAIN BARRIER TRANSPORT OF ANTISENSE
RADIOPHARMACEUTICALS FOR NEUROIMAGING
Antisense radiopharmaceuticals are potential large molecule
agents for imaging in brain the gene expression of sequence
specific mRNA. However, antisense agents do not cross the BBB.121
An ideal antisense imaging agent is the peptide nucleic acid
(PNA). Peptide nucleic acids are resistant to nucleases, as
compared with phosphodiester-oligodeoxynucleotides (ODN),
and have less nonspecific binding than phosphorothioate-ODNs.
Peptide nucleic acid binding to target mRNAs does not trigger
RNase H activity as is observed with ODNs.122
To evaluate BBB-
targeted PNAs as antisense imaging agents for the brain, an
intracranial RG-2 brain cancer model in rats was developed.123
Gene expression within the tumor was assessed for glial fibrillary
acidic protein (GFAP) and caveolin-1a (CAV). The sequence of the
GFAP-targeted and CAV-targeted PNAs is shown in Figure 7A. The
amino terminus of the PNA was conjugated with DTPA for
radiolabelling with111
In (Figure 7A). The amide backbone of the
PNA contained a carboxyl terminal lysine (Lys) residue and the
e-amino group of the Lys residue was monobiotinylated
(Figure 7A). Confocal microscopy showed the intracranial brain
tumor underexpressed the GFAP protein and overexpressed the
CAV protein (Figure 7B). Northern blot analysis showed the level of
the GFAP and CAV mRNAs in the tumor paralleled the protein, as
the GFAP mRNA was underexpressed, and the CAV mRNA was
overexpressed.123
The biotin-GFAP-PNA or the biotin-CAV-PNA
was conjugated to the TfRMAb via a streptavidin linker.
Conjugation of the PNA to the MTH did not affect binding of
the PNA to the target transcript. This was demonstrated with
Northern blotting using the labeled PNA as the probe, and cloned
GFAP or CAV RNA as the target.123
Following IV injection of either
the [111
In-DTPA, biotinyl]-GFAP PNA alone, or the [111
In-DTPA,
biotinyl]-CAV PNA alone, there was no imaging of brain, as these
molecules do not cross the BBB. Following the IV injection of the
[111
In-DTPA, biotinyl]-GFAP PNA/TfRMAb conjugate, the nontumor
brain was imaged, but there was no signal over the tumor
Figure 7. (A) Structures are shown for model peptide nucleic acids (PNAs) with sequences that target either the rat glial fibrillary acidic protein
(GFAP) mRNA or the rat caveolin-1a (CAV) mRNA.123
The methionine initiation codon sequence is underlined. (B) Confocal microscopy of brain
from a rat with an intracranial RG-2 glioma is shown after double labeling with an antibody against GFAP (green channel) and an antibody
against CAV (red channel).123
(C) Scan of brain removed from a rat with an intracranial RG-2 glioma at 1 hour after the intravenous (IV)
injection of the [111
In-DTPA, Lys-biotinyl]-GFAP PNA conjugated to the TfRMAb (monoclonal antibody) via a streptavidin linkage. The image
over the tumor is of low intensity, as the RG-2 tumor does not express the GFAP mRNA.123
(D) Scan of brain removed from a rat with an
intracranial RG-2 glioma at 6 hours after the IV injection of the [111
In-DTPA, Lys-biotinyl]-CAV PNA conjugated to the TfRMAb via a streptavidin
linkage. The image over the tumor is of high intensity, as the RG-2 tumor expresses abundant CAV mRNA.123
Blood–brain barrier drug delivery
WM Pardridge
1969
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
(Figure 7C). Therefore, the in vivo brain scan with the targeted
GFAP-PNA antisense radiopharmaceutical confirmed the in vitro
finding of minimal expression of the GFAP mRNA in the glial tumor.
Following the IV injection of the [111
In-DTPA, biotinyl]–CAV PNA/
TfRMAb conjugate, the tumor showed an intense signal, owing to
overexpression of the CAV mRNA in the tumor (Figure 7D). The
in vivo brain scans (Figures 7C and 7D) of the GFAP or CAV mRNAs
with BBB-targeted antisense radiopharmaceuticals paralleled the
immune detection of GFAP or CAV proteins in autopsy sections
(Figure 7B), and the levels of the GFAP or CAV mRNAs in the
tumor.123
Peptide and antisense radiopharmaceuticals have the
potential to greatly expand the diagnostic capabilities of modern
neuroimaging. However, these molecules need to be reformulated
for BBB transport, and this is possible with the combined use of
MTH and avidin–biotin technologies.
In summary, small molecule drugs may be reengineered to
enable transport across the BBB via the CMT systems expressed in
the brain capillary endothelium. Large molecule drugs, including
recombinant proteins, and peptide and antisense radiopharma-
ceuticals may be delivered across the BBB with MTHs that target
the endogenous RMT systems expressed within the brain capillary
endothelium. The reengineering of pharmaceuticals for BBB
transport is enabled with knowledge on the endogenous CMT
and RMT systems within the BBB.
DISCLOSURE/CONFLICT OF INTEREST
The author is a consultant to ArmaGen Technologies.
REFERENCES
1 Pardridge WM. The blood-brain barrier: bottleneck in brain drug development.
NeuroRx 2005; 2: 3–14.
2 Brightman MW, Reese TS. Junctions between intimately apposed cell mem-
branes in the vertebrate brain. J Cell Biol 1969; 40: 648–677.
3 Reiber H, Felgenhauer K. Protein transfer at the blood cerebrospinal fluid barrier
and the quantitation of the humoral immune response within the central ner-
vous system. Clin Chim Acta 1987; 163: 319–328.
4 Mott FW. The late Professor Edwin Goldmann’s investigations on the central
nervous system by vital staining. Br Med J 1913; 2: 871–873.
5 Friedemann U. Blood-brain barrier. Physiol Rev 1942; 22: 125–145.
6 Broman T. The possibilities of the passage of substances from the blood to the
central nervous system. Acta Psych Neurol 1941; 16: 1–16.
7 Rowland LP. Blood-brain barrier, cerebrospinal fluid, brain edema, and hydro-
cephalus. In: Kandel ER, Schwartz JH (eds) Principles of Neural Science. 2nd edn
(Elsevier Science Publishing: New York, NY, 1985, pp 837–844.
8 Davson H. The cerebrospinal fluid. Handbook of Neurochemistry 1969; 2: 23–48.
9 Oldendorf WH. Cerebrospinal fluid formation and circulation. Prog Nucl Med
1972; 1: 336–358.
10 Cutler RW, Page L, Galicich J, Watters GV. Formation and absorption of
cerebrospinal fluid in man. Brain 1968; 91: 707–720.
11 Rudick RA, Zirretta DK, Herndon RM. Clearance of albumin from mouse sub-
arachnoid space: a measure of CSF bulk flow. J Neurosci Methods 1982; 6: 253–259.
12 Yan Q, Matheson C, Sun J, Radeke MJ, Feinstein SC, Miller JA. Distribution
of intracerebral ventricularly administered neurotrophins in rat brain and its
correlation with trk receptor expression. Exp Neurol 1994; 127: 23–36.
13 Blasberg RG, Patlak C, Fenstermacher JD. Intrathecal chemotherapy: brain
tissue profiles after ventriculocisternal perfusion. J Pharmacol Exp Ther 1975; 195:
73–83.
14 Fung LK, Shin M, Tyler B, Brem H, Saltzman WM. Chemotherapeutic drugs
released from polymers: distribution of 1,3-bis(2-chloroethyl)-1-nitrosourea in
the rat brain. Pharm Res 1996; 13: 671–682.
15 Day-Lollini PA, Stewart GR, Taylor MJ, Johnson RM, Chellman GJ. Hyperplastic
changes within the leptomeninges of the rat and monkey in response to chronic
intracerebroventricular infusion of nerve growth factor. Exp Neurol 1997; 145:
24–37.
16 Yamada K, Kinoshita A, Kohmura E, Sakaguchi T, Taguchi J, Kataoka K et al. Basic
fibroblast growth factor prevents thalamic degeneration after cortical infarction.
J Cereb Blood Flow Metab 1991; 11: 472–478.
17 Marin-Padilla M, Knopman DS. Developmental aspects of the intracerebral
microvasculature and perivascular spaces: insights into brain response to late-
life diseases. J Neuropathol Exp Neurol 2011; 70: 1060–1069.
18 Szentistvanyi I, Patlak CS, Ellis RA, Cserr HF. Drainage of interstitial fluid from
different regions of rat brain. Am J Physiol 1984; 246: F835–F844.
19 Fishman RA, Christy NP. Fate of adrenal cortical steroids following intrathecal
injection. Neurology 1965; 15: 1–6.
20 Aird RB. A study of intrathecal, cerebrospinal fluid-to-brain exchange. Exp Neurol
1984; 86: 342–358.
21 de Lange EC, Danhof M, de Boer AG, Breimer DD. Critical factors of intracerebral
microdialysis as a technique to determine the pharmacokinetics of drugs in rat
brain. Brain Res 1994; 666: 1–8.
22 Crawley JN, Fiske SM, Durieux C, Derrien M, Roques BP. Centrally administered
cholecystokinin suppresses feeding through a peripheral-type receptor
mechanism. J Pharmacol Exp Ther 1991; 257: 1076–1080.
23 Kristensson K, Olsson Y. Uptake of exogenous proteins in mouse olfactory cells.
Acta Neuropathol 1971; 19: 145–154.
24 Merkus P, Guchelaar HJ, Bosch DA, Merkus FW. Direct access of drugs to the
human brain after intranasal drug administration? Neurology 2003; 60: 1669–1671.
25 Westin U, Piras E, Jansson B, Bergstrom U, Dahlin M, Brittebo E et al. Transfer of
morphine along the olfactory pathway to the central nervous system after nasal
administration to rodents. Eur J Pharm Sci 2005; 24: 565–573.
26 Graff CL, Pollack GM. Nasal drug administration: potential for targeted central
nervous system delivery. J Pharm Sci 2005; 94: 1187–1195.
27 Salvatore MF, Ai Y, Fischer B, Zhang AM, Grondin RC, Zhang Z et al. Point source
concentration of GDNF may explain failure of phase II clinical trial. Exp Neurol
2006; 202: 497–505.
28 Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in
designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A
qualitative and quantitative characterization of known drug databases. J Comb
Chem 1999; 1: 55–68.
29 Lipinski CA. Drug-like properties and the causes of poor solubility and poor
permeability. J Pharmacol Toxicol Methods 2000; 44: 235–249.
30 Ajay Bemis GW, Murcko MA. Designing libraries with CNS activity. J Med Chem
1999; 42: 4942–4951.
31 Cohen BE, Bangham AD. Diffusion of small non-electrolytes across liposome
membranes. Nature 1972; 236: 173–174.
32 Lieb WR, Stein WD. Non-Stokesian nature of transverse diffusion within human
red cell membranes. J Membr Biol 1986; 92: 111–119.
33 Levin VA. Relationship of octanol/water partition coefficient and molecular
weight to rat brain capillary permeability. J Med Chem 1980; 23: 682–684.
34 van de Waterbeemd H, Camenisch G, Folkers G, Chretien JR, Raevsky OA. Esti-
mation of blood-brain barrier crossing of drugs using molecular size and shape,
and H-bonding descriptors. J Drug Target 1998; 6: 151–165.
35 Fischer H, Gottschlich R, Seelig A. Blood-brain barrier permeation: molecular
parameters governing passive diffusion. J Membr Biol 1998; 165: 201–211.
36 Trauble H. The movement of molecules across lipid membranes: a molecular
theory. J Membrane Biol 1971; 4: 193–208.
37 Hingson DJ, Diamond JM. Comparison of nonelectrolyte permeability patterns in
several epithelia. J Membr Biol 1972; 10: 93–135.
38 Marrink SJ, Jahnig F, Berendsen HJ. Proton transport across transient single-file
water pores in a lipid membrane studied by molecular dynamics simulations.
Biophys J 1996; 71: 632–647.
39 Diamond JM, Wright EM. Molecular forces governing non-electrolyte permeation
through cell membranes. Proc R Soc Lond B Biol Sci 1969; 171: 273–316.
40 Stein WD. The Molecular Basis of Diffusion Across Cell Membranes. Academic Press:
New York, NY, 1967, pp 66–125.
41 Pardridge WM, Mietus LJ. Transport of steroid hormones through the rat blood-
brain barrier. J Clin Invest 1979; 64: 145–154.
42 Chikhale EG, Ng KY, Burton PS, Borchardt RT. Hydrogen bonding potential as a
determinant of the in vitro and in situ blood-brain barrier permeability of
peptides. Pharm Res 1994; 11: 412–419.
43 Oldendorf WH, Hyman S, Braun L, Oldendorf SZ. Blood-brain barrier: penetration
of morphine, codeine, heroin, and methadone after carotid injection. Science
1972; 178: 984–986.
44 Crone C. Facilitated transfer of glucose from blood into brain tissue. J Physiol
1965; 181: 103–113.
45 Oldendorf WH. Brain uptake of radiolabeled amino acids, amines, and hexoses
after arterial injection. Am J Physiol 1971; 221: 1629–1639.
46 Pardridge WM, Oldendorf WH. Transport of metabolic substrates through the
blood-brain barrier. J Neurochem 1977; 28: 5–12.
47 Pardridge WM. Recent advances in blood-brain barrier transport. Annu Rev
Pharmacol Toxicol 1988; 28: 25–39.
48 Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie J et al. Quantitative
targeted absolute proteomics of human blood-brain barrier transporters and
receptors. J Neurochem 2011; 117: 333–345.
49 Pardridge WM, Oldendorf WH. Kinetics of blood-brain transport of hexoses.
Biochim Biophys Acta 1975; 382: 377–392.
Blood–brain barrier drug delivery
WM Pardridge
1970
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
50 Pardridge WM, Boado RJ, Farrell CR. Brain-type glucose transporter (GLUT-1) is
selectively localized to the blood-brain barrier. Studies with quantitative western
blotting and in situ hybridization. J Biol Chem 1990; 265: 18035–18040.
51 Pardridge WM, Oldendorf WH. Kinetic analysis of blood-brain barrier transport of
amino acids. Biochim Biophys Acta 1975; 401: 128–136.
52 Boado RJ, Li JY, Nagaya M, Zhang C, Pardridge WM. Selective expression of the
large neutral amino acid transporter at the blood-brain barrier. Proc Natl Acad Sci
USA 1999; 96: 12079–12084.
53 Stoll J, Wadhwani KC, Smith QR. Identification of the cationic amino acid
transporter (System y þ ) of the rat blood-brain barrier. J Neurochem 1993; 60:
1956–1959.
54 Cremer JE, Cunningham VJ, Pardridge WM, Braun LD, Oldendorf WH. Kinetics of
blood-brain barrier transport of pyruvate, lactate and glucose in suckling,
weanling and adult rats. J Neurochem 1979; 33: 439–445.
55 Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR. Expression of
monocarboxylate transporter MCT1 by brain endothelium and glia in adult and
suckling rats. Am J Physiol 1997; 273: E207–E213.
56 Cornford EM, Oldendorf WH. Independent blood-brain barrier transport systems
for nucleic acid precursors. Biochim Biophys Acta 1975; 394: 211–219.
57 Li JY, Boado RJ, Pardridge WM. Cloned blood-brain barrier adenosine transporter
is identical to the rat concentrative Na þ nucleoside cotransporter CNT2. J Cereb
Blood Flow Metab 2001; 21: 929–936.
58 Ohtsuki S, Uchida Y, Kubo Y, Terasaki T. Quantitative targeted absolute pro-
teomics-based ADME research as a new path to drug discovery and develop-
ment: methodology, advantages, strategy, and prospects. J Pharm Sci 2011; 100:
3547–3559.
59 Pardridge WM, Boado RJ. Reengineering biopharmaceuticals for targeted
delivery across the blood-brain barrier. Methods Enzymol 2012; 503:
269–292.
60 Sumbria RK, Boado RJ, Pardridge WM. Brain protection from stroke with intra-
venous TNFa decoy receptor-Trojan horse fusion protein. J Cereb Blood Flow
Metab 2012, in press.
61 Gjedde A, Christensen O. Estimates of Michaelis-Menten constants for the
two membranes of the brain endothelium. J Cereb Blood Flow Metab 1984; 4:
241–249.
62 Schulz A, Beyhl D, Marten I, Wormit A, Neuhaus E, Poschet G et al. Proton-driven
sucrose symport and antiport are provided by the vacuolar transporters SUC4
and TMT1/2. Plant J 2011; 68: 129–136.
63 Mueckler M, Caruso C, Baldwin SA, Panico M, Blench I, Morris HR et al. Sequence
and structure of a human glucose transporter. Science 1985; 229: 941–945.
64 Boado RJ, Pardridge WM. Molecular cloning of the bovine blood-brain barrier
glucose transporter cDNA and demonstration of phylogenetic conservation of
the 50
-untranslated region. Mol Cell Neurosci 1990; 1: 224–232.
65 Birnbaum MJ, Haspel HC, Rosen OM. Cloning and characterization of a cDNA
encoding the rat brain glucose-transporter protein. Proc Natl Acad Sci USA 1986;
83: 5784–5788.
66 Flier JS, Mueckler M, McCall AL, Lodish HF. Distribution of glucose transporter
messenger RNA transcripts in tissues of rat and man. J Clin Invest 1987; 79:
657–661.
67 Boado RJ, Pardridge WM. The brain-type glucose transporter mRNA is specifically
expressed at the blood-brain barrier. Biochem Biophys Res Commun 1990; 166:
174–179.
68 Farrell CL, Pardridge WM. Blood-brain barrier glucose transporter is asymme-
trically distributed on brain capillary endothelial lumenal and ablumenal mem-
branes: an electron microscopic immunogold study. Proc Natl Acad Sci USA 1991;
88: 5779–5783.
69 Farrell CL, Yang J, Pardridge WM. GLUT1 glucose transporter is present within
apical and basolateral membranes of brain epithelial interfaces and in micro-
vascular endothelia with and without tight junctions. J Histochem Cytochem
1992; 40: 193–199.
70 Cornford EM, Hyman S, Swartz BE. The human brain GLUT1 glucose transporter:
ultrastructural localization to the blood-brain barrier endothelia. J Cereb Blood
Flow Metab 1994; 14: 106–112.
71 Dwyer KJ, Pardridge WM. Developmental modulation of blood-brain barrier and
choroid plexus GLUT1 glucose transporter messenger ribonucleic acid and
immunoreactive protein in rabbits. Endocrinology 1993; 132: 558–565.
72 Wang Y, Welty DF. The simultaneous estimation of the influx and efflux blood-
brain barrier permeabilities of gabapentin using a microdialysis-pharmacokinetic
approach. Pharm Res 1996; 13: 398–403.
73 Shimizu K, Ohtaki K, Matsubara K, Aoyama K, Uezono T, Saito O et al. Carrier-
mediated processes in blood--brain barrier penetration and neural uptake of
paraquat. Brain Res 2001; 906: 135–142.
74 Cornford EM, Young D, Paxton JW, Finlay GJ, Wilson WR, Pardridge WM. Mel-
phalan penetration of the blood-brain barrier via the neutral amino acid trans-
porter in tumor-bearing brain. Cancer Res 1992; 52: 138–143.
75 Witt KA, Gillespie TJ, Huber JD, Egleton RD, Davis TP. Peptide drug modifications
to enhance bioavailability and blood-brain barrier permeability. Peptides 2001;
22: 2329–2343.
76 Killian DM, Hermeling S, Chikhale PJ. Targeting the cerebrovascular large neutral
amino acid transporter (LAT1) isoform using a novel disulfide-based brain drug
delivery system. Drug Deliv 2007; 14: 25–31.
77 Ghosh C, Gonzalez-Martinez J, Hossain M, Cucullo L, Fazio V, Janigro D et al.
Pattern of P450 expression at the human blood-brain barrier: roles of epileptic
condition and laminar flow. Epilepsia 2010; 51: 1408–1417.
78 Hawkins BT, Sykes DB, Rapid Miller DS. reversible modulation of blood-brain
barrier P-glycoprotein transport activity by vascular endothelial growth factor.
J Neurosci 2010; 30: 1417–1425.
79 Dauchy S, Miller F, Couraud PO, Weaver RJ, Weksler B, Romero IA et al.
Expression and transcriptional regulation of ABC transporters and cytochromes
P450 in hCMEC/D3 human cerebral microvascular endothelial cells. Biochem
Pharmacol 2009; 77: 897–909.
80 Zeevi N, Pachter J, McCullough LD, Wolfson L, Kuchel GA. The blood-brain
barrier: geriatric relevance of a critical brain-body interface. J Am Geriatr Soc
2010; 58: 1749–1757.
81 Coker III GT , Studelska D, Harmon S, Burke W, O’Malley KL. Analysis of tyrosine
hydroxylase and insulin transcripts in human neuroendocrine tissues. Brain Res
Mol Brain Res 1990; 8: 93–98.
82 Kojima H, Fujimiya M, Matsumura K, Nakahara T, Hara M, Chan L. Extrapancreatic
insulin-producing cells in multiple organs in diabetes. Proc Natl Acad Sci USA
2004; 101: 2458–2463.
83 Pardridge WM, Eisenberg J, Yang J. Human blood-brain barrier insulin receptor.
J Neurochem 1985; 44: 1771–1778.
84 Pardridge WM, Eisenberg J, Yang J. Human blood-brain barrier transferrin
receptor. Metabolism 1987; 36: 892–895.
85 Duffy KR, Pardridge WM, Rosenfeld RG. Human blood-brain barrier insulin-like
growth factor receptor. Metabolism 1988; 37: 136–140.
86 Golden PL, Maccagnan TJ, Pardridge WM. Human blood-brain barrier leptin
receptor. Binding and endocytosis in isolated human brain microvessels. J Clin
Invest 1997; 99: 14–18.
87 Duffy KR, Pardridge WM. Blood-brain barrier transcytosis of insulin in developing
rabbits. Brain Res 1987; 420: 32–38.
88 Skarlatos S, Yoshikawa T, Pardridge WM. Transport of [125I]transferrin through
the rat blood-brain barrier. Brain Res 1995; 683: 164–171.
89 Kakee A, Terasaki T, Sugiyama Y. Brain efflux index as a novel method of ana-
lyzing efflux transport at the blood-brain barrier. J Pharmacol Exp Ther 1996; 277:
1550–1559.
90 Zhang Y, Pardridge WM. Rapid transferrin efflux from brain to blood across the
blood-brain barrier. J Neurochem 2001; 76: 1597–1600.
91 Zhang Y, Pardridge WM. Mediated efflux of IgG molecules from brain to blood
across the blood-brain barrier. J Neuroimmunol 2001; 114: 168–172.
92 Schlachetzki F, Zhu C, Pardridge WM. Expression of the neonatal Fc receptor
(FcRn) at the blood-brain barrier. J Neurochem 2002; 81: 203–206.
93 Triguero D, Buciak J, Pardridge WM. Capillary depletion method for quantifica-
tion of blood-brain barrier transport of circulating peptides and plasma proteins.
J Neurochem 1990; 54: 1882–1888.
94 Nazer B, Hong S, Selkoe DJ. LRP promotes endocytosis and degradation, but not
transcytosis, of the amyloid-beta peptide in a blood-brain barrier in vitro model.
Neurobiol Dis 2008; 30: 94–102.
95 Karkan D, Pfeifer C, Vitalis TZ, Arthur G, Ujiie M, Chen Q et al. A unique carrier for
delivery of therapeutic compounds beyond the blood-brain barrier. PLoS One
2008; 3: e2469.
96 Che C, Yang G, Thiot C, Lacoste MC, Currie JC, Demeule M et al. New Angiopep-
modified doxorubicin (ANG1007) and etoposide (ANG1009) chemotherapeutics
with increased brain penetration. J Med Chem 2010; 53: 2814–2824.
97 Zhou QH, Boado RJ, Lu JZ, Hui EK-W, Pardridge WM. Monoclonal antibody-glial-
derived neurotrophic factor fusion protein penetrates the blood-brain barrier in
the mouse. Drug Metab Dispos 2010; 38: 566–572.
98 Roberts RL, Fine RE, Sandra A. Receptor-mediated endocytosis of transferrin at
the blood-brain barrier. J Cell Sci 1993; 104(Pt 2): 521–532.
99 Vorbrodt AW. Ultracytochemical characterization of anionic sites in the wall of
brain capillaries. J Neurocytol 1989; 18: 359–368.
100 Huwyler J, Pardridge WM. Examination of blood-brain barrier transferrin receptor
by confocal fluorescent microscopy of unfixed isolated rat brain capillaries.
J Neurochem 1998; 70: 883–886.
101 Bickel U, Kang YS, Yoshikawa T, Pardridge WM. In vivo demonstration of sub-
cellular localization of anti-transferrin receptor monoclonal antibody-colloidal
gold conjugate in brain capillary endothelium. J Histochem Cytochem 1994; 42:
1493–1497.
102 Pardridge WM, Kang YS, Buciak JL, Yang J. Human insulin receptor monoclonal
antibody undergoes high affinity binding to human brain capillaries in vitro and
Blood–brain barrier drug delivery
WM Pardridge
1971
& 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
rapid transcytosis through the blood-brain barrier in vivo in the primate. Pharm
Res 1995; 12: 807–816.
103 Boado RJ, Zhang YF, Zhang Y, Pardridge WM. Humanization of anti-human
insulin receptor antibody for drug targeting across the human blood-brain
barrier. Biotechnol Bioeng 2007; 96: 381–391.
104 Boado RJ, Zhang Y, Wang Y, Pardridge WM. Engineering and expression of a
chimeric transferrin receptor monoclonal antibody for blood-brain barrier
delivery in the mouse. Biotechnol Bioeng 2009; 102: 1251–1258.
105 Boado RJ, Zhang YF, Zhang Y, Xia CF, Pardridge WM. Fusion antibody for Alz-
heimer’s disease with bidirectional transport across the blood-brain barrier and
abeta fibril disaggregation. Bioconjug Chem 2007; 18: 447–455.
106 Lee HJ, Engelhardt B, Lesley J, Bickel U, Pardridge WM. Targeting rat anti-mouse
transferrin receptor monoclonal antibodies through blood-brain barrier in
mouse. J Pharmacol Exp Ther 2000; 292: 1048–1052.
107 Boado RJ, Hui EK, Lu JZ, Pardridge WM. Drug targeting of erythropoietin across
the primate blood-brain barrier with an IgG molecular Trojan horse. J Pharmacol
Exp Ther 2010; 333: 961–969.
108 Boado RJ, Pardridge WM. Comparison of blood-brain barrier transport of glial-
derived neurotrophic factor (GDNF) and an IgG-GDNF fusion protein in the
rhesus monkey. Drug Metab Dispos 2009; 37: 2299–2304.
109 Boado RJ, Hui EK, Lu JZ, Zhou QH, Pardridge WM. Selective targeting of a TNFR
decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG
fusion protein. J Biotechnol 2010; 146: 84–91.
110 Greenblatt DJ, Sethy VH. Benzodiazepine concentrations in brain directly reflect
receptor occupancy: studies of diazepam, lorezepam, and oxazepam. Psycho-
pharmacology (Berl) 1990; 102: 373–378.
111 Christian BT, Vandehey NT, Fox AS, Murali D, Oakes TR. Converse AK, et al. The
distribution of D2/D3 receptor binding in the adolescent rhesus monkey using
small animal PET imaging. Neuroimage 2009; 44: 1334–1344.
112 Boado RJ, Zhang Y, Xia CF, Wang Y, Pardridge WM. Genetic engineering,
expression, and activity of a chimeric monoclonal antibody-avidin fusion protein
for receptor-mediated delivery of biotinylated drugs in humans. Bioconjug Chem
2008; 19: 731–739.
113 Zhou QH, Lu JZ, Hui EK, Boado RJ, Pardridge WM. Delivery of a peptide radio-
pharmaceutical to brain with an IgG-avidin fusion protein. Bioconjug Chem 2011;
22: 1611–1618.
114 Maggio JE, Stimson ER, Ghilardi JR, Allen CJ, Dahl CE, Whitcomb DC et al.
Reversible in vitro growth of Alzheimer disease beta-amyloid plaques
by deposition of labeled amyloid peptide. Proc Natl Acad Sci USA 1992; 89:
5462–5466.
115 Wu D, Yang J, Pardridge WM. Drug targeting of a peptide radiopharmaceutical
through the primate blood-brain barrier in vivo with a monoclonal antibody to
the human insulin receptor. J Clin Invest 1997; 100: 1804–1812.
116 Ono M, Cheng Y, Kimura H, Cui M, Kagawa S, Nishi R et al. Novel 18F-labeled
benzofuran derivatives with improved properties for positron emission tomo-
graphy (PET) imaging of beta-amyloid plaques in Alzheimer’s brains. J Med Chem
2011; 54: 2971–2979.
117 Sumbria RK, Boado RJ, Pardridge WM. Imaging amyloid plaque in Alzheimer’s
disease brain with a biotinylated Abeta peptide radiopharmaceutical conjugated
to an IgG-avidin fusion protein. Bioconjug Chem 2012; 23: 1318–1321.
118 Kurihara A, Pardridge WM. Abeta(1-40) peptide radiopharmaceuticals for brain
amyloid imaging: (111)In chelation, conjugation to poly(ethylene glycol)-biotin
linkers, and autoradiography with Alzheimer’s disease brain sections. Bioconjug
Chem 2000; 11: 380–386.
119 Kurihara A, Pardridge WM. Imaging brain tumors by targeting peptide radio-
pharmaceuticals through the blood-brain barrier. Canc Res 1999; 54: 6159–6163.
120 Kurihara A, Deguchi Y, Pardridge WM. Epidermal growth factor radio-
pharmaceuticals: 111In chelation, conjugation to a blood-brain barrier delivery
vector via a biotin-polyethylene linker, pharmacokinetics, and in vivo imaging of
experimental brain tumors. Bioconjug Chem 1999; 10: 502–511.
121 Pardridge WM, Boado RJ, Kang YS. Vector-mediated delivery of a polyamide
(‘peptide’) nucleic acid analogue through the blood-brain barrier in vivo. Proc
Natl Acad Sci USA 1995; 92: 5592–5596.
122 Knudsen H, Nielsen PE. Antisense properties of duplex- and triplex-forming
PNAs. Nucleic Acids Res 1996; 24: 494–500.
123 Suzuki T, Wu D, Schlachetzki F, Li JY, Boado RJ, Pardridge WM. Imaging endo-
genous gene expression in brain cancer in vivo with 111In-peptide nucleic acid
antisense radiopharmaceuticals and brain drug-targeting technology. J Nucl Med
2004; 45: 1766–1775.
124 Pardridge WM. Misconceptions about the blood-brain barrier. In: Pardridge WM
moderator. Blood-brain barrier: interface between internal medicine and brain
Ann Int Med, 1986; 105: 82–95.
125 Bar T. The vascular system of the cerebral cortex. Adv Anat Embryol Cell Biol 1980;
59: I-VI, 1–62.
126 Duvernoy H, Delon S, Vannson JL. The vascularization of the human cerebellar
cortex. Brain Res Bull 1983; 11: 419–480.
127 Pardridge WM. Biopharmaceutical drug targeting to the brain. J Drug Target
2010; 18: 157–167.
128 Pardridge WM, Boado RJ. Molecular cloning and regulation of gene expression
of blood-brain barrier glucose transporter. In: Pardridge WM (ed The Blood-Brain
Barrier: Cellular and Molecular Biology. Raven Press: New York, 1993, pp 347–362.
129 Sarko D, Beijer B, Garcia Boy R, Nothelfer EM, Leotta K, Eisenhut M et al. The
pharmacokinetics of cell-penetrating peptides. Mol Pharm 2010; 7: 2224–2231.
130 Boado RJ, Zhang Y, Xia CF, Wang Y, Pardridge WM. Genetic engineering of a
lysosomal enzyme fusion protein for targeted delivery across the human blood-
brain barrier. Biotechnol Bioeng 2008; 99: 475–484.
Blood–brain barrier drug delivery
WM Pardridge
1972
Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM

More Related Content

What's hot

effect of toxic element on blood brain barrier
effect of toxic element on blood brain barriereffect of toxic element on blood brain barrier
effect of toxic element on blood brain barrierPavan Barot
 
Blood brain barrier
Blood brain barrierBlood brain barrier
Blood brain barrierIrfan malik
 
Presentation 3
Presentation 3Presentation 3
Presentation 3Amir Fahmi
 
Blood brain barrier ppt.
Blood brain barrier ppt.Blood brain barrier ppt.
Blood brain barrier ppt.Sagar Chawan
 
Blood brain barrier
Blood brain barrierBlood brain barrier
Blood brain barrierRajesh Goit
 
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)Sameep Koshti
 
The 'Blood-Brain Barrier' for beginners
The 'Blood-Brain Barrier' for beginnersThe 'Blood-Brain Barrier' for beginners
The 'Blood-Brain Barrier' for beginnersTeam Consulting Ltd
 
Applications of biotechnology in neurology
Applications of biotechnology in neurologyApplications of biotechnology in neurology
Applications of biotechnology in neurologySpringer
 
Tissue engineering of nervous system
Tissue engineering of nervous systemTissue engineering of nervous system
Tissue engineering of nervous systemshashank chetty
 
Cerebrospinal fluid and blood brain barrier
Cerebrospinal fluid and blood brain barrierCerebrospinal fluid and blood brain barrier
Cerebrospinal fluid and blood brain barrierRati Tandon
 
Cardiovascular stem cell therapy
Cardiovascular stem cell therapyCardiovascular stem cell therapy
Cardiovascular stem cell therapyRashedul Islam
 
Cerebrospinal fluid and intracranial pressure
Cerebrospinal fluid and intracranial pressureCerebrospinal fluid and intracranial pressure
Cerebrospinal fluid and intracranial pressureMuhammad Saim
 
The nano skeleton in the closet
The nano skeleton in the closetThe nano skeleton in the closet
The nano skeleton in the closetAdonis Sfera, MD
 
Meninges, bbb, csf, icp, brain edema, hydrocephalus
Meninges, bbb, csf, icp, brain edema, hydrocephalusMeninges, bbb, csf, icp, brain edema, hydrocephalus
Meninges, bbb, csf, icp, brain edema, hydrocephalusJoyce Mwatonoka
 

What's hot (20)

BLOOD BRAIN BARRIER
BLOOD BRAIN BARRIERBLOOD BRAIN BARRIER
BLOOD BRAIN BARRIER
 
effect of toxic element on blood brain barrier
effect of toxic element on blood brain barriereffect of toxic element on blood brain barrier
effect of toxic element on blood brain barrier
 
Blood brain barrier
Blood brain barrierBlood brain barrier
Blood brain barrier
 
Presentation 3
Presentation 3Presentation 3
Presentation 3
 
Blood brain barrier ppt.
Blood brain barrier ppt.Blood brain barrier ppt.
Blood brain barrier ppt.
 
Blood brain barrier
Blood brain barrierBlood brain barrier
Blood brain barrier
 
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)
Blood brain barrier - Dr Sameep Koshti (Consultant Neurosurgeon)
 
The 'Blood-Brain Barrier' for beginners
The 'Blood-Brain Barrier' for beginnersThe 'Blood-Brain Barrier' for beginners
The 'Blood-Brain Barrier' for beginners
 
Cns 11
Cns 11Cns 11
Cns 11
 
Applications of biotechnology in neurology
Applications of biotechnology in neurologyApplications of biotechnology in neurology
Applications of biotechnology in neurology
 
Tissue engineering of nervous system
Tissue engineering of nervous systemTissue engineering of nervous system
Tissue engineering of nervous system
 
Cerebrospinal fluid and blood brain barrier
Cerebrospinal fluid and blood brain barrierCerebrospinal fluid and blood brain barrier
Cerebrospinal fluid and blood brain barrier
 
Jovin2008 1 13-8-end
Jovin2008 1 13-8-endJovin2008 1 13-8-end
Jovin2008 1 13-8-end
 
Cardiovascular stem cell therapy
Cardiovascular stem cell therapyCardiovascular stem cell therapy
Cardiovascular stem cell therapy
 
nuclear pore complex and function nucleus
nuclear pore complex and function nucleusnuclear pore complex and function nucleus
nuclear pore complex and function nucleus
 
Cerebrospinal fluid and intracranial pressure
Cerebrospinal fluid and intracranial pressureCerebrospinal fluid and intracranial pressure
Cerebrospinal fluid and intracranial pressure
 
Stem cell
Stem cellStem cell
Stem cell
 
Stem cells in cardiac care
Stem cells in cardiac careStem cells in cardiac care
Stem cells in cardiac care
 
The nano skeleton in the closet
The nano skeleton in the closetThe nano skeleton in the closet
The nano skeleton in the closet
 
Meninges, bbb, csf, icp, brain edema, hydrocephalus
Meninges, bbb, csf, icp, brain edema, hydrocephalusMeninges, bbb, csf, icp, brain edema, hydrocephalus
Meninges, bbb, csf, icp, brain edema, hydrocephalus
 

Similar to Brain drug delivery

DRUG DELIVERY TO THE BRAIN
DRUG DELIVERY TO THE BRAINDRUG DELIVERY TO THE BRAIN
DRUG DELIVERY TO THE BRAINN Anusha
 
Cerebral Blood flow Chapter 3 in CNS.pptx
Cerebral Blood flow Chapter 3 in CNS.pptxCerebral Blood flow Chapter 3 in CNS.pptx
Cerebral Blood flow Chapter 3 in CNS.pptxAnanua1
 
BRAIN SPECIFIC DELIVERY 1.pptx
BRAIN SPECIFIC DELIVERY 1.pptxBRAIN SPECIFIC DELIVERY 1.pptx
BRAIN SPECIFIC DELIVERY 1.pptxKeerthanaN20
 
Seminar on brain targeted drug delivery system
Seminar on brain targeted drug delivery systemSeminar on brain targeted drug delivery system
Seminar on brain targeted drug delivery systemMayur Wagh
 
Several methods making drugs overcome blood-brain barier obstacle .
Several methods making drugs overcome blood-brain barier obstacle .Several methods making drugs overcome blood-brain barier obstacle .
Several methods making drugs overcome blood-brain barier obstacle .Abeer Abd Elrahman
 
Brain specific delivery
Brain specific deliveryBrain specific delivery
Brain specific deliveryKirantengse
 
ChowetalTINSreview.pdf
ChowetalTINSreview.pdfChowetalTINSreview.pdf
ChowetalTINSreview.pdfalaaaltaee3
 
blood - brain barrier
blood - brain barrierblood - brain barrier
blood - brain barrieralaaaltaee3
 
Blood Brain Barrrier vishal singh mch neurosurgery
Blood Brain Barrrier vishal singh mch neurosurgeryBlood Brain Barrrier vishal singh mch neurosurgery
Blood Brain Barrrier vishal singh mch neurosurgeryvishalsingh1756
 
Drug distribution
Drug distribution Drug distribution
Drug distribution Faisal Shah
 
Drug distribution
Drug distribution Drug distribution
Drug distribution Faisal Shah
 
Brain specific drug targetting stratergies
Brain specific drug targetting stratergiesBrain specific drug targetting stratergies
Brain specific drug targetting stratergieskoorishma
 
1.BRAIN SPECIFIC DRUG DELIVERY.pptx
1.BRAIN SPECIFIC DRUG DELIVERY.pptx1.BRAIN SPECIFIC DRUG DELIVERY.pptx
1.BRAIN SPECIFIC DRUG DELIVERY.pptxHarishreddyPaladgu
 
Distribution of Drug
Distribution of DrugDistribution of Drug
Distribution of DrugSHUBHAMGWAGH
 
Brain specific delivery system
Brain specific delivery systemBrain specific delivery system
Brain specific delivery systemTushar Chavan
 
Brain targeted drug delivery system seminar.pptx
Brain targeted drug delivery system seminar.pptxBrain targeted drug delivery system seminar.pptx
Brain targeted drug delivery system seminar.pptxShamsElfalah
 
Drug distribution and protein binding
Drug distribution and protein bindingDrug distribution and protein binding
Drug distribution and protein bindingAnjali9410
 
Factors affecting distribution of drug
Factors affecting distribution of drugFactors affecting distribution of drug
Factors affecting distribution of drugDr. SHUBHRAJIT MANTRY
 

Similar to Brain drug delivery (20)

DRUG DELIVERY TO THE BRAIN
DRUG DELIVERY TO THE BRAINDRUG DELIVERY TO THE BRAIN
DRUG DELIVERY TO THE BRAIN
 
Cerebral Blood flow Chapter 3 in CNS.pptx
Cerebral Blood flow Chapter 3 in CNS.pptxCerebral Blood flow Chapter 3 in CNS.pptx
Cerebral Blood flow Chapter 3 in CNS.pptx
 
BRAIN SPECIFIC DELIVERY 1.pptx
BRAIN SPECIFIC DELIVERY 1.pptxBRAIN SPECIFIC DELIVERY 1.pptx
BRAIN SPECIFIC DELIVERY 1.pptx
 
Seminar on brain targeted drug delivery system
Seminar on brain targeted drug delivery systemSeminar on brain targeted drug delivery system
Seminar on brain targeted drug delivery system
 
Several methods making drugs overcome blood-brain barier obstacle .
Several methods making drugs overcome blood-brain barier obstacle .Several methods making drugs overcome blood-brain barier obstacle .
Several methods making drugs overcome blood-brain barier obstacle .
 
Brain specific delivery
Brain specific deliveryBrain specific delivery
Brain specific delivery
 
ChowetalTINSreview.pdf
ChowetalTINSreview.pdfChowetalTINSreview.pdf
ChowetalTINSreview.pdf
 
blood - brain barrier
blood - brain barrierblood - brain barrier
blood - brain barrier
 
Blood Brain Barrrier vishal singh mch neurosurgery
Blood Brain Barrrier vishal singh mch neurosurgeryBlood Brain Barrrier vishal singh mch neurosurgery
Blood Brain Barrrier vishal singh mch neurosurgery
 
Drug distribution
Drug distribution Drug distribution
Drug distribution
 
Drug distribution
Drug distribution Drug distribution
Drug distribution
 
Drug Distribution
Drug DistributionDrug Distribution
Drug Distribution
 
Brain specific drug targetting stratergies
Brain specific drug targetting stratergiesBrain specific drug targetting stratergies
Brain specific drug targetting stratergies
 
1.BRAIN SPECIFIC DRUG DELIVERY.pptx
1.BRAIN SPECIFIC DRUG DELIVERY.pptx1.BRAIN SPECIFIC DRUG DELIVERY.pptx
1.BRAIN SPECIFIC DRUG DELIVERY.pptx
 
CSF.pptx
CSF.pptxCSF.pptx
CSF.pptx
 
Distribution of Drug
Distribution of DrugDistribution of Drug
Distribution of Drug
 
Brain specific delivery system
Brain specific delivery systemBrain specific delivery system
Brain specific delivery system
 
Brain targeted drug delivery system seminar.pptx
Brain targeted drug delivery system seminar.pptxBrain targeted drug delivery system seminar.pptx
Brain targeted drug delivery system seminar.pptx
 
Drug distribution and protein binding
Drug distribution and protein bindingDrug distribution and protein binding
Drug distribution and protein binding
 
Factors affecting distribution of drug
Factors affecting distribution of drugFactors affecting distribution of drug
Factors affecting distribution of drug
 

More from Smita Shukla

Chemotherapeutic agent
Chemotherapeutic agentChemotherapeutic agent
Chemotherapeutic agentSmita Shukla
 
Pulmonary drug delivery
Pulmonary drug deliveryPulmonary drug delivery
Pulmonary drug deliverySmita Shukla
 
Drug delivery fact sheet
Drug delivery fact sheetDrug delivery fact sheet
Drug delivery fact sheetSmita Shukla
 
Beta lactam antibiotics
Beta lactam antibioticsBeta lactam antibiotics
Beta lactam antibioticsSmita Shukla
 
Agents affecting immune system
Agents affecting immune systemAgents affecting immune system
Agents affecting immune systemSmita Shukla
 
Female reproductive hormones
Female reproductive hormonesFemale reproductive hormones
Female reproductive hormonesSmita Shukla
 
Synthetic antibacterial agents
Synthetic antibacterial agentsSynthetic antibacterial agents
Synthetic antibacterial agentsSmita Shukla
 
Signal transduction
Signal transductionSignal transduction
Signal transductionSmita Shukla
 
Promoters in gene expression
Promoters in gene expressionPromoters in gene expression
Promoters in gene expressionSmita Shukla
 
Enzymes used in Genetic Engineering
Enzymes used in Genetic EngineeringEnzymes used in Genetic Engineering
Enzymes used in Genetic EngineeringSmita Shukla
 
Unit 1 genetic engineering
Unit 1 genetic engineeringUnit 1 genetic engineering
Unit 1 genetic engineeringSmita Shukla
 
Bioresource and waste management
Bioresource and waste managementBioresource and waste management
Bioresource and waste managementSmita Shukla
 

More from Smita Shukla (20)

Anti-viral Agents
Anti-viral AgentsAnti-viral Agents
Anti-viral Agents
 
Chemotherapeutic agent
Chemotherapeutic agentChemotherapeutic agent
Chemotherapeutic agent
 
Pulmonary review
Pulmonary reviewPulmonary review
Pulmonary review
 
Pulmonary drug delivery
Pulmonary drug deliveryPulmonary drug delivery
Pulmonary drug delivery
 
Drug delivery fact sheet
Drug delivery fact sheetDrug delivery fact sheet
Drug delivery fact sheet
 
Beta lactam antibiotics
Beta lactam antibioticsBeta lactam antibiotics
Beta lactam antibiotics
 
Antiamoebic drugs
Antiamoebic drugsAntiamoebic drugs
Antiamoebic drugs
 
Anthelmintics
AnthelminticsAnthelmintics
Anthelmintics
 
Agents affecting immune system
Agents affecting immune systemAgents affecting immune system
Agents affecting immune system
 
Drug targeting
Drug targetingDrug targeting
Drug targeting
 
Female reproductive hormones
Female reproductive hormonesFemale reproductive hormones
Female reproductive hormones
 
Synthetic antibacterial agents
Synthetic antibacterial agentsSynthetic antibacterial agents
Synthetic antibacterial agents
 
Cloning
CloningCloning
Cloning
 
Signal transduction
Signal transductionSignal transduction
Signal transduction
 
Promoters in gene expression
Promoters in gene expressionPromoters in gene expression
Promoters in gene expression
 
Enzymes used in Genetic Engineering
Enzymes used in Genetic EngineeringEnzymes used in Genetic Engineering
Enzymes used in Genetic Engineering
 
Unit 1 genetic engineering
Unit 1 genetic engineeringUnit 1 genetic engineering
Unit 1 genetic engineering
 
Unit 4
Unit 4Unit 4
Unit 4
 
Bioresource and waste management
Bioresource and waste managementBioresource and waste management
Bioresource and waste management
 
Transgenics
TransgenicsTransgenics
Transgenics
 

Recently uploaded

The Black hole shadow in Modified Gravity
The Black hole shadow in Modified GravityThe Black hole shadow in Modified Gravity
The Black hole shadow in Modified GravitySubhadipsau21168
 
Work, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE PhysicsWork, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE Physicsvishikhakeshava1
 
Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Jshifa
 
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCE
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCESTERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCE
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCEPRINCE C P
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsssuserddc89b
 
Luciferase in rDNA technology (biotechnology).pptx
Luciferase in rDNA technology (biotechnology).pptxLuciferase in rDNA technology (biotechnology).pptx
Luciferase in rDNA technology (biotechnology).pptxAleenaTreesaSaji
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )aarthirajkumar25
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real timeSatoshi NAKAHIRA
 
Animal Communication- Auditory and Visual.pptx
Animal Communication- Auditory and Visual.pptxAnimal Communication- Auditory and Visual.pptx
Animal Communication- Auditory and Visual.pptxUmerFayaz5
 
Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Patrick Diehl
 
Physiochemical properties of nanomaterials and its nanotoxicity.pptx
Physiochemical properties of nanomaterials and its nanotoxicity.pptxPhysiochemical properties of nanomaterials and its nanotoxicity.pptx
Physiochemical properties of nanomaterials and its nanotoxicity.pptxAArockiyaNisha
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡anilsa9823
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxyaramohamed343013
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfSELF-EXPLANATORY
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsSérgio Sacani
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝soniya singh
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfSwapnil Therkar
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |aasikanpl
 
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...Sérgio Sacani
 

Recently uploaded (20)

9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
9953056974 Young Call Girls In Mahavir enclave Indian Quality Escort service
 
The Black hole shadow in Modified Gravity
The Black hole shadow in Modified GravityThe Black hole shadow in Modified Gravity
The Black hole shadow in Modified Gravity
 
Work, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE PhysicsWork, Energy and Power for class 10 ICSE Physics
Work, Energy and Power for class 10 ICSE Physics
 
Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)Recombination DNA Technology (Microinjection)
Recombination DNA Technology (Microinjection)
 
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCE
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCESTERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCE
STERILITY TESTING OF PHARMACEUTICALS ppt by DR.C.P.PRINCE
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physics
 
Luciferase in rDNA technology (biotechnology).pptx
Luciferase in rDNA technology (biotechnology).pptxLuciferase in rDNA technology (biotechnology).pptx
Luciferase in rDNA technology (biotechnology).pptx
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real time
 
Animal Communication- Auditory and Visual.pptx
Animal Communication- Auditory and Visual.pptxAnimal Communication- Auditory and Visual.pptx
Animal Communication- Auditory and Visual.pptx
 
Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?
 
Physiochemical properties of nanomaterials and its nanotoxicity.pptx
Physiochemical properties of nanomaterials and its nanotoxicity.pptxPhysiochemical properties of nanomaterials and its nanotoxicity.pptx
Physiochemical properties of nanomaterials and its nanotoxicity.pptx
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
 
Scheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docxScheme-of-Work-Science-Stage-4 cambridge science.docx
Scheme-of-Work-Science-Stage-4 cambridge science.docx
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
 
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
Call Us ≽ 9953322196 ≼ Call Girls In Mukherjee Nagar(Delhi) |
 
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
All-domain Anomaly Resolution Office U.S. Department of Defense (U) Case: “Eg...
 

Brain drug delivery

  • 1. REVIEW ARTICLE Drug transport across the blood–brain barrier William M Pardridge The blood–brain barrier (BBB) prevents the brain uptake of most pharmaceuticals. This property arises from the epithelial-like tight junctions within the brain capillary endothelium. The BBB is anatomically and functionally distinct from the blood–cerebrospinal fluid barrier at the choroid plexus. Certain small molecule drugs may cross the BBB via lipid-mediated free diffusion, providing the drug has a molecular weight o400 Da and forms o8 hydrogen bonds. These chemical properties are lacking in the majority of small molecule drugs, and all large molecule drugs. Nevertheless, drugs can be reengineered for BBB transport, based on the knowledge of the endogenous transport systems within the BBB. Small molecule drugs can be synthesized that access carrier- mediated transport (CMT) systems within the BBB. Large molecule drugs can be reengineered with molecular Trojan horse delivery systems to access receptor-mediated transport (RMT) systems within the BBB. Peptide and antisense radiopharmaceuticals are made brain-penetrating with the combined use of RMT-based delivery systems and avidin–biotin technology. Knowledge on the endogenous CMT and RMT systems expressed at the BBB enable new solutions to the problem of BBB drug transport. Journal of Cerebral Blood Flow & Metabolism (2012) 32, 1959–1972; doi:10.1038/jcbfm.2012.126; published online 29 August 2012 Keywords: blood–brain barrier; cerebrospinal fluid; endothelium; receptors; vascular biology INTRODUCTION The blood–brain barrier (BBB) prevents entry into the brain of most drugs from the blood. The presence of the BBB makes difficult the development of new treatments of brain diseases, or new radiopharmaceuticals for neuroimaging of brain. All of the products of biotechnology are large molecule drugs that do not cross the BBB. While it is assumed that small molecules are freely transported across the BBB, B98% of all small molecules are not transported across the BBB.1 The lack of small molecule transport across the BBB is illustrated by the whole body autoradiogram in Figure 1A. The mouse was euthanized 5 minutes after the intravenous (IV) injection of [14 C]-histamine, a small molecule of only 111 Da. Histamine rapidly enters the postvascular space of all organs of the body, with the exception of the brain and spinal cord. The anatomical site of the BBB is endothelial wall of the microvasculature in the brain.2 The density in the brain of the microvessels is shown at the light microscopic level for rat brain in Figure 1B, and at the electron microscopic level for the human brain in Figure 1C. The BBB is comprised of two membranes in series, the luminal and abluminal membranes of the brain capillary endothelium, which are separated by about 200 nm of endothelial cytoplasm. Owing to the presence of epithelial-like, high resistance tight junctions within the brain capillary endothelium, the intercellular pores that exist in the endothelial barriers in peripheral organs are absent in the endothelial barrier in the brain. In addition, there is minimal fluid-phase pinocytosis in brain capillary endothelium.2 The absence of paracellular or transcellular channels within the BBB means that molecules in the circulation gain access to brain ISF (interstitial fluid) via only one of the two mechanisms: (1) lipid-mediated free diffusion through the BBB or (2) carrier- or receptor-mediated transport (RMT) through the BBB. The BBB is the principal interface between blood and the ISF that bathes synaptic connections within the parenchyma of the brain. A separate barrier system in the brain is localized to the epithelial cells of the choroid plexus, which form the blood– cerebrospinal fluid (CSF) barrier (BCSFB). The choroid plexus is the principal interface between the blood and CSF that bathes the surface of the brain. The BBB is the fundamental problem blocking progress in the development of new therapeutics for brain disorders, or the development of new radiopharmaceuticals for imaging brain. This review will discuss both the principals of BBB drug transport, and strategies for the reengineering of drugs to enable BBB transport. The classical approaches to drug delivery to the brain, transcranial drug delivery or small molecules, are discussed, and the limita- tions of these strategies are reviewed. The endogenous carrier- mediated transport (CMT) and RMT systems within the BBB are discussed, and strategies are reviewed for the reengineering of pharmaceuticals that penetrate the brain from blood via access of the CMT and RMT systems within the BBB. HISTORICAL ORIGINS OF MODERN CONCEPTS ON BRAIN DRUG TRANSPORT Drug distribution into CSF is frequently used as an indicator of BBB transport. This is a misconception that stems from the lumping of the BBB and BCSFB systems as a single brain barrier. In fact, the BCSFB is leaky compared with the BBB, and all molecules in blood enter the CSF at a rate inversely related to molecular weight (MW).3 The finding of drug penetration into CSF is expected for any molecule, and does not provide information on rates of drug penetration across the BBB at the brain capillary endothelium. Thus, drug distribution in CSF is not an index of BBB transport, but rather is simply a measure of transport across the choroid plexus at the BCSFB. The idea that CSF composition reflects BBB transport is 100 years old, and finds its origins in Goldman’s vital dye experiments published in 1913.4 Before 1913, it was known that certain acidic vital dyes, such as trypan blue, are excluded from Department of Medicine, UCLA, Los Angeles, California, USA. Correspondence: Dr WM Pardridge, Department of Medicine, UCLA, Warren Hall 13-164, 900 Veteran Avenue, Los Angeles, CA 90024, USA. E-mail: wpardridge@mednet.ucla.edu Received 15 June 2012; revised 4 August 2012; accepted 9 August 2012; published online 29 August 2012 Journal of Cerebral Blood Flow & Metabolism (2012) 32, 1959–1972 & 2012 ISCBFM All rights reserved 0271-678X/12 $32.00 www.jcbfm.com
  • 2. entry into the brain following peripheral administration, and this observation was explained, not within the context of a barrier between blood and brain, but rather because the brain lacked the ‘receptors’ that bind the dye and sequester the molecule within the tissue. Goldman injected the trypan blue into the lumbar sac, or subdural space, of rabbits and observed vital dye staining of spinal cord and brain tissue, respectively.4 Thus, the brain did express vital dye binding sites. The failure to stain the brain following peripheral administration of the dye was hypothesized to be due to a ‘blut-gehirn schranke’ or BBB. However, in 1913, it was believed that the pathway of nutrient flux from blood to brain took place across the choroid plexus. The anatomical site of the BBB was erroneously placed at the choroid plexus.4 The idea that nutrient flux from blood to brain occurred across the capillaries within the parenchyma of brain was not accepted in 1913. By the 1930s, studies with brain-penetrating basic vital dyes showed that the dye entered the brain from blood across the walls of capillaries perfusing brain parenchyma.5 Broman6 and Friedemann5 clearly observed that the BBB was localized to the capillary wall in brain, and that drug entry into the CSF across the choroid plexus was an entirely separate problem from drug transport across the BBB. Indeed, in his 1942 review on the BBB, Friedemann5 stated that, ‘distribution between blood and CSF is an entirely different problem and remains outside the scope of this review.’ Nevertheless, the idea that the BBB and BCSFB are functionally equivalent, and that the CSF and ISF are functionally equivalent, is perpetuated in neuroscience text books.7 These ideas form the basis for bypassing the BBB with intrathecal drug delivery to the brain. In 2013, or 100 years after the publication of Goldman’s experiment defining the BBB, there are multiple ongoing clinical trials of intrathecal delivery to the human brain of drugs that do not cross the BBB. BRAIN DRUG TRANSPORT AND CEREBROSPINAL FLUID The brain has no lymphatic system. The CSF of the brain is produced at the choroid plexi of the ventricles, moves over the surface of the brain, and is absorbed into the general circulation across the arachnoid villi into the superior sagittal sinus of the venous bloodstream.8,9 In the human brain, there is about 100 to 140 mL of CSF, and this entire volume is turned over completely every 4 to 5 hours, or 4 to 5 times per day.10 In the mouse brain, there is about 40 mL of CSF, and the entire volume is turned over every 2 hours, or about 12 times per day.11 Cerebrospinal fluid is a fluid compartment in rapid equilibrium with the blood, which is due to the rapid rate of bulk flow (convection) of CSF from brain to blood. Conversely, drug movement into brain tissue from the CSF flow tracts occurs via diffusion. The differential rates of convection and diffusion create the paradox that drug injected into the CSF distributes easily to blood and poorly to brain beyond the ependymal surface. Poor Drug Transport from Cerebrospinal Fluid to Brain The poor distribution of drug into brain parenchyma is shown by the autoradiogram of rat brain prepared 20 hours after a single injection of [125 I]-BDNF (brain-derived neurotrophic factor) into a lateral ventricle.12 The drug distributes only to the ependymal surface of brain ipsilateral to the ventricular injection (Figure 2A). From the lateral ventricle, the drug moves via bulk flow to the third ventricle (3 V), then to the fourth ventricle, then over the surface of the brain, where the drug undergoes exodus from the CSF space to the venous blood. In contrast to the relative rapid rate of bulk flow of CSF out of brain, the transport of drug from the CSF to brain tissue is slow, and limited by diffusion. Diffusion decreases with the square of the distance. The limited distribution into brain from the CSF is also observed for small molecules. The concentration of water-soluble small molecules in brain tissue at increasing distances from the CSF surface was measured in the primate brain following the intracerebroventricular (ICV) injection of the drug.13 Small molecule drug concentration in the brain decreased logarithmically with each mm of distance from the CSF surface. There was an B10-fold decrease in drug concentration of small molecule with each mm of distance removed from the brain surface. In the case of a lipid soluble small molecule, which can diffuse into brain cells, there is a 10-fold decrease in drug concentration with each 500 mm of distance into the brain.14 The logarithmic decrease in drug concentration in the brain removed from the CSF surface is even steeper for a large molecule drug, which has a lower diffusion coefficient. To achieve a therapeutic drug level in the brain at 5 mm removed from the CSF surface, it is necessary to inject into the CSF a dose of drug that generates at least a 5-log increase in CSF drug concentration. This approach Figure 1. (A) Whole body autoradiogram of a mouse euthanized 5 minutes after the intravenous injection of [14 C]-histamine.124 (B) India ink visualization of complexity of the microvasculature of the cortex of rat brain.125 (C) Vascular cast of the microvasculature of the human cerebellar cortex. The distance between capillaries in brain is about 40 mm.126 Blood–brain barrier drug delivery WM Pardridge 1960 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 3. may cover drug distribution in the mouse brain, where all parts of the brain are within 3 to 5 mm of the CSF surface. However, in the human brain, the distance between brain and the CSF surface is up to 50 mm, which precludes significant drug distribution into brain parenchyma from the CSF space. A side effect of the ICV injection of very high doses of drug is the exposure of the ependymal surface of the brain to potentially toxic concentrations of drug. The ICV injection of nerve growth factor in rats or monkeys15 or basic fibroblast growth factor in rats16 causes leptomeningeal changes with axonal sprouting and astrogliosis at the ependymal surface of the brain. In addition to the macrocirculation of CSF through the ventricular system, there is a CSF microcirculation through the Virchow-Robin compartment, which is formed by the perivascular space around penetrating cortical precapillary arterioles, which emanate from the pial vessels on the surface of the brain.17 However, the fluid flow via the microcirculation is quantitatively small compared with the CSF macrocirculation. The rate of flow of the CSF macrocirculation is 2 mL/min in the rat,8 whereas the rate of flow of the Virchow-Robin microcirculation is only 5% of the CSF flow rate, or 0.1 mL/min in the rat.18 Rapid Drug Transport from Cerebrospinal Fluid to Blood Fishman and Christy19 observed nearly 50 years ago that an intrathecal injection of drug ‘is, in effect, equivalent to no more than a prolonged IV injection.’ The rapid movement of CSF to blood means that drug injected into the CSF compartment will also rapidly move to the general bloodstream. This creates the paradox that drugs injected into the CSF may have a pharma- cological effect within brain parenchyma, but only after transport of the drug from CSF to blood and then to brain across the BBB (Figure 2B). Working with barbiturates in dogs, Aird20 observed that the anesthesia was maintained at the same dose of drug, 0.2 mg per kg per minute, whether the barbiturate was infused by either the IV or the ICV route of administration. Drug injected into the CSF is rapidly distributed to blood, followed by entry into brain tissue from the blood via transport across the BBB. Drug concentration in the dialysate of an intracerebral dialysis fiber was measured following the ICV injection of drug; drug entry into the blood compartment was an obligatory intermediate step between the drug injection into the CSF and drug entry into the dialysis fiber placed within brain parenchyma.21 A secondary effect of the rapid movement of drug from CSF to blood is the finding of a pharmacological response following ICV injection, owing to the pharmacological activity of the drug in peripheral tissues. The effect of the neuropeptide, cholecystokinin, on feeding following ICV injection of the peptide is mediated via cholecystokinin transfer from CSF to blood, which enables activation of chole- cystokinin receptors in the stomach.22 Transnasal Drug Delivery to Brain The olfactory region of the submucous space of the nose is contiguous with the CSF flow tracts around the olfactory lobe. Therefore, intranasal drug administration could lead to direct delivery into the CSF providing (1) the drug is transportable across the nasal epithelium and (2) the drug is transportable across the arachnoid membrane separating the submucous space of the nose and the olfactory CSF compartment.23 The properties governing drug distribution from the nose to the olfactory CSF are similar to the properties determining drug transport across the BBB. A lipid soluble small molecule introduced into the nose would be expected to have some distribution into the olfactory CSF. However, a large molecule, or a water-soluble small molecule, would not enter olfactory CSF from the nose in the absence of local injury to the nasal epithelial barriers. Injury is induced in the nose following the intranasal instillation of volumes 4100 mL/ nares in humans.24 The volume that induces local injury in the mouse or rat nose is likely o10 mL/nares, and most studies of intranasal drug delivery to the brain in rodents use larger volumes, which may create nasal membrane disruption. There are species differences between humans and rodents with respect to the part of the nasal mucosa that corresponds to the olfactory region. Whereas the olfactory region of the nasal epithelium constitutes 50% of the nasal mucosa in the rat or mouse, the olfactory region is only 3% to 5% of the nasal mucosa in humans.25,26 Consequently, it is difficult to detect drug movement into CSF following intranasal drug delivery in humans. When vitamin B12 or melatonin was administered to humans with small volumes, for example, 70 mL/nares, no drug distribution to CSF was observed following intranasal drug administration.24 Convection-Enhanced Diffusion Drug delivery to the brain via the CSF is limited by the slow diffusion of drug from the ependymal surface of the brain (Figure 2A). So as to replace diffusion with bulk flow, drug delivery to the brain has been attempted with CED (convection-enhanced diffusion). In this approach, drug is placed in a peripheral reservoir connected to a pump, which is in line with a transcranial catheter. In theory, drug will penetrate the brain tissue by bulk flow, which should produce high drug levels in the brain at sites remote from the intracerebral catheter. In practice, drug concentrations in the brain decrease logarithmically with each mm of distance removed from the tip of the CED catheter in the brain. A logarithmic Figure 2. (A) Film autoradiogram of rat brain section 20 hours after the injection of [125 I]-BDNF (brain-derived neurotrophic factor) into the lateral ventricle (LV).12 (B) Solute distribution into brain parenchyma from the cerebrospinal fluid (CSF) compartment is limited by the slow rate of solute diffusion from the ependymal surface, relative to the rapid rate of bulk flow (convection) of CSF from the ventricles to the systemic circulation across the arachnoid villi. (C) Logarithmic decrease in the brain concentration of glial- derived neurotrophic factor (GDNF) in the brain of the Rhesus monkey relative to the distance from the intracerebral catheter used for brain drug delivery with convection-enhanced diffusion.127 Blood–brain barrier drug delivery WM Pardridge 1961 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 4. decrease in the brain concentration of GDNF (glial-derived neuro- trophic factor) is observed following the CED infusion of the neurotrophin in the monkey brain,27 and these data are plotted in Figure 2C. Instead of a broad distribution of the drug into brain tissue beyond the catheter, which would be expected for a convection or bulk flow process, there is a logarithmic decrease in drug distribution in the brain, which is typical of a diffusion process. It would appear that the resistance of brain tissue reduces convection within the brain beyond the catheter tip. Similar to intrathecal drug delivery to the brain, the CED approach requires exposure of parts of brain proximal to the catheter to very high drug levels, to achieve therapeutic levels of drug at sites of brain distal to the catheter (Figure 2C). Intrathecal or intracerebral drug delivery to the brain is limited by diffusion, which causes logarithmic gradients of drug concen- tration. In contrast, diffusion plays no role in the transvascular route of drug delivery to the brain. The distance between capil- laries in brain is about 40 mm (Figure 1C). Even a large molecule antibody drug diffuses 40 mm within a second. Therefore, once a drug is delivered across the BBB from blood, there is instanta- neous equilibration of drug throughout the entire brain volume. There are three mechanisms of trans-BBB transport of drug: (1) lipid-mediated transport of lipid soluble small molecules; (2) CMT transport of water-soluble small molecules that have an affinity for an endogenous BBB CMT system; and (3) RMT transport of large molecules that have a high affinity for an endogenous BBB RMT system. LIPID-MEDIATED TRANSPORT OF SMALL MOLECULES Almost all drugs for the brain presently in clinical practice are lipid soluble small molecules with an MW o400 Da. These drugs fit the dual criteria for lipid-mediated free diffusion across the BBB, which are (1) MW o400 Da threshold and (2) high lipid solubility, which is equivalent to low hydrogen bonding. In practice, very few small molecule drug candidates for the brain fit these dual criteria, and do not cross the BBB. Of 46,000 drugs in the Comprehensive Medicinal Chemistry database, only 6% are active in the brain, and these drugs treat only a select group of brain diseases, comprised of affective disorders and insomnia.28 In another study, only 12% of all drugs were CNS active, but if affective disorders are excluded, only 1% of all drugs are active in the CNS.29 CNS active drugs form r7 hydrogen bonds with solvent water.30 These considerations indicate that the BBB transport of a small molecule drug candidate can be predicted based on the chemical structure of the drug, which allows for computation of (1) MW and (2) hydrogen bonding. Molecular Weight Threshold Small molecule drug permeation through lipid membranes demonstrates an MW threshold, which is not predicted by the classical Overton rules for solute diffusion in water.31,32 Unlike diffusion through water, solute diffusion through lipid membranes is dependent on the molecular volume of the solute, which is generally proportional to solute MW or solute surface area. That is, when the size of the drug exceeds the volume threshold, then membrane permeation is less than predicted on the basis of the lipid solubility of the drug. The MW threshold of BBB drug transport of small molecules has been demonstrated previously in studies of drug penetration into the brain.33,34 Blood–brain barrier permeation decreases 100-fold when the size of the drug is increased from an MW of 300 Da, which corresponds to a surface area of 50 square angstroms, to an MW of 450 Da, which cor- responds to a surface area of 100 square angstroms.35 The molecular mechanism of the MW threshold phenomenon is consistent with the observation that transient water pores form in biological membranes, which are caused by the transient fluctuations in the conformation of the fatty acyl side chains of membrane lipids.36–38 Hydrogen Bonding The lipid solubility of a drug may be determined by measurement of the 1-octanol lipid partition coefficient. However, an important parameter, which can be determined just from an inspection of the drug structure, is the hydrogen bonding of the drug and solvent water, which is 55 molar. Diamond and Wright39 reviewed the rules for determination of the number (N) of hydrogen bonds a solute forms in water, for example, N ¼ 2 for each hydroxyl, N ¼ 3 for each primary amine, and N ¼ 4 for each amide group. Stein40 observed that there is a 10-fold decrease in membrane per- meation with each pair of hydrogen bonds on the solute, and this rule has been generally confirmed in studies of in vivo BBB transport of steroid hormones41 and oligopeptides.42 Consideration of the rules for hydrogen bonding show that even a simple dipeptide, glycylglycine, which forms eight hydrogen bonds with water, would have a very low BBB permeation. Conversely, if the glycylglycine was cyclized, the number of hydrogen bonds of the cyclic glycylglycine would be reduced to 4, and BBB transport would be predicted to increase by 100-fold for the cyclic form of the glycylglycine dipeptide, because glycine has no polar side chains. When confronted with the problem of a water-soluble drug that does not penetrate the BBB, the medicinal chemist is asked to fix the problem by lipidizing the drug. This can be done by blocking Table 1. BBB carrier-mediated transporters Carrier Substrate Genea product Kmb (uM) Vmax b,c (nmol/ min/g) Ccapd (pmol/ mgp) Vmax:Cape (molecules per second) Glucose D-glucose GLUT1 (SLC2A1) 11,000±1,400 1,420±140 139±46 600 Monocarboxylic acid L-lactic acid MCT1 (SLC16A1) 1,800±600 91±35 2.3±0.8 2,300 Neutral amino acid L-phenyl- alanine LAT1 (SLC7A5) 26±6 22±4 0.43±0.09 3,000 Basic amino acid L-arginine CAT1 (SLC7A1) 40±24 5±3 1.1±0.2 270 Purine nucleoside Adenosine CNT2 (SLC28A2) 25±3 0.75±0.08 o0.14 ND BBB, blood–brain barrier. a Solute carrier (SLC) group of transporter genes. b From Pardridge.47 c Per gram wet brain in the rat. d From Uchida et al.48 Data reported per mg isolated human brain capillary protein. e Vmax/Ccap ratio is the transporter turnover number, which is the number of substrate molecules transported per second by a single transporter molecule, and is based on 0.14 mg capillary protein per gram wet brain; it is assumed that 50% of the total capillary transporter is expressed on the endothelial luminal membrane; nd ¼ not determined. Blood–brain barrier drug delivery WM Pardridge 1962 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 5. hydrogen bond forming functional groups on the drug. For example, when the two hydroxyl groups of morphine are acetylated, to form heroin, the BBB penetration rate increases 100-fold.43 However, apart from the morphine/heroin example, there have been few successes in developing new drugs for human brain disorders by lipidization of a water-soluble drug. This is because the hydrogen blocking functional groups are rapidly hydrolyzed in vivo. If the hydrogen bond is blocked with a noncleavable group, then drug activity may be reduced. An alternative approach to the medicinal chemical modification of non-BBB-penetrating drugs is to focus on the endogenous CMT systems within the BBB. Instead of blocking hydrogen bonds, medicinal chemistry can be used to modify the structure of the drug to increase drug affinity for one of several endogenous carrier-mediated BBB transporters. CARRIER-MEDIATED TRANSPORT OF SMALL MOLECULES Discovery of Blood–Brain Barrier Carrier-Mediated Transport Systems Physiology of blood–brain barrier carrier-mediated transport. The first evidence of saturable or CMT of a solute across the BBB was demonstrated by Crone44 for D-glucose using the indicator dilution technique, which is a venous sampling-carotid artery injection method. Subsequently, the Brain Uptake Index technique, a tissue sampling-carotid artery injection method developed by Oldendorf45 , was used to characterize the saturable kinetics of BBB transport of multiple water-soluble metabolic substrates. The Michelis–Menten kinetics of BBB CMT for the major metabolic substrates were determined,46 and this analysis showed a wide range of affinity (Km) and maximal transport activity (Vmax) for the BBB nutrient transporters (Table 1). Molecular biology of blood–brain barrier carrier-mediated transport. The methods of molecular biology were employed to identify the principal gene product responsible for BBB transport of a given class of metabolic substrates. The BBB glucose carrier, which also transports other hexoses, including mannose, galactose, deox- yglucose, or 3-0-methyl glucose,49 is the glucose transporter type 1 or GLUT1.50 The BBB phenylalanine carrier, which also transports over 10 other large neutral amino acids,51 and to a lesser extent small neutral amino acids, was identified as the large neutral amino-acid transporter type 1 or LAT1.52 The BBB arginine carrier, which also transports other cationic amino acids (lysine, ornithine),45 was shown to be the cationic amino-acid trans- porter type 1 or CAT1.53 The BBB lactate carrier, which also transports other monocarboxylic acids (pyruvate, and the ketone bodies, acetoacetate, and b-hydroxybutryyrate),54 was identified as the monocarboxylic acid transporter type 1 or MCT1.55 The BBB adenosine transporter, which also transports other purine nucle- osides (guanosine, inosine), as well as the pyrimidine nucleoside, uridine,56 is the concentrative nucleoside transporter type 2 or CNT2 in the rat.57 Quantitation of blood–brain barrier transporter protein expression. Terasaki and colleagues58 determined the mass of transporters in isolated brain capillaries using the methods of LC–MS (liquid chromatography mass spectrometry). Capillaries were isolated from brain followed by digestion with trypsin. The tryptic peptides were separated and identified by LC–MS. Transporter-specific peptides were identified and quantitated by solid-phase synthesis of synthetic peptide standards. These peptide standards, which enable the quantification of the transporter expression, could be synthesized based on the known amino-acid sequences of the cloned transporters, as well as standard programs for predicting the sequence of tryptic peptides based on known amino-acid sequence of the target protein. The transporter concentrations in isolated human brain capillaries, Ccap, are listed in Table 2 for human GLUT1, MCT1, LAT1, and CAT1, and are compared with the transporter Vmax values determined in vivo in the rat. The comparison between rat Vmax values and human Ccap values was made, because the Ccap values are generally comparable for human and rodent brain capillaries.48 Transporter turnover number. The Vmax values are expressed per gram wet brain, whereas the Ccap values are expressed per mg of brain capillary protein. It is estimated that there is 0.17 mg capillary protein per gram brain, since there is 100 mg protein per gram brain, and the intracellular water volume of the brain capillary endothelial space, 1 mL/g,61 is 1/700 parts of the total water volume of brain. Based on this conversion, and assuming 50% of the capillary transporters are expressed on the luminal endothelial membrane, the Vmax/Ccap ratio can be computed, which is the transporter turnover number, or molecules of substrate transported by the carrier per second (Table 1). The estimated turnover numbers for BBB CMT systems range from 270 to 3,000 molecules/s. These calculations are approximations given the divergent in vitro and in vivo methodologies that were performed. Nevertheless, the estimated turnover rates approx- imate the turnover rate for other transporters, for example, the turnover number for the sucrose transporter (SUT1) is 500 mole- cules/s.62 To design drugs that access the CMT systems listed in Table 1, it is necessary to define the exact cellular location of the transporter at the brain microvasculature. A transporter identified in an isolated brain capillary preparation could be selectively expressed on the capillary endothelium, on the capillary pericyte, on smooth muscle cells of precapillary arterioles, or even on astrocyte endfeet, which remain adhered to the basement membrane of the isolated capillary. Following localization of the transporter to the capillary endothelium, it is necessary to determine whether the transporter is expressed on the luminal Table 2. Reengineering of protein drugs for BBB penetration as IgG Trojan horse fusion proteins Therapeutic class Protein therapeutic Target brain disease Animal models Lysosomal enzymes Iduronidase (IDUA) MPS-I (Hurler’s syndrome) Rhesus monkey, Hurler mouse Iduronate 2-sulfatase (IDS) MPS-II (Hunter’s syndrome) Rhesus monkey Decoy receptors Type II TNFa decoy receptor (TNFR) Stroke neurodegeneration Rhesus monkey, Mouse PD, Mouse stroke Neurotrophins Erythropoietin (EPO) Stroke Neurodegeneration Rhesus monkey, Mouse PD Glial-derived neurotrophic factor (GDNF) Neurodegeneration Stroke Rhesus monkey, Mouse PD Therapeutic antibodies Antiamyloid antibody (AAA) Alzheimer’s disease Rhesus monkey, Mouse AD BBB, blood–brain barrier. Primary references are given in Pardridge and Boado59 and Sumbria et al.60 Blood–brain barrier drug delivery WM Pardridge 1963 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 6. membrane, the abluminal membrane, or both. The cellular and subcellular expression of a BBB transporter is illustrated in the case of GLUT1 expression at the BBB. Expression of GLUT1 in Brain Is Localized to the Blood–Brain Barrier There are over 10 members of the sodium-independent GLUT glucose transporter gene family, and the first transporter identi- fied, GLUT1, was cloned from human HepG2 liver cells using an antibody to the human erythrocyte glucose transporter.63 Cloning of the cDNA allowed for prediction of the amino-acid sequence and a two-dimensional representation of the transporter struc- ture,63 which is shown in Figure 3A for the GLUT1 glucose transporter cloned from a bovine BBB cDNA library.64 Shortly after cloning from human liver cells, the same GLUT1 glucose trans- porter cDNA was cloned from a rat brain cDNA library, and the transporter was originally known as the brain-type glucose transporter.65 The GLUT1 glucose transporter was observed to be enriched at the brain microvasculature.66 However, studies of GLUT1 glucose transporter mRNA expression in the brain with the capillary depletion method showed that the GLUT1 glucose transporter was expressed in the brain only at the BBB, with little, if any, detection of GLUT1 transcript in mRNA devoid of capillary- derived mRNA.67 The GLUT1 Northern blot for capillary-depleted brain is illustrated in Figure 3B. The 2.9-kb GLUT1 glucose transporter mRNA was detected in Northern blotting of non- fractionated RNA from the brain (lane 1, left panel, Figure 3B). However, the GLUT1 glucose transporter mRNA was 495% removed with the capillary depletion method, which had no effect on the abundance of a common transcript, the 2.0-kb actin mRNA (Figure 3B). This finding indicated that GLUT1-mediated glucose transport at the BBB, but not at brain cells. The selective expression of the GLUT1 isoform at the BBB in the brain was subsequently confirmed with in situ hybridization, quantitative Western blotting,50 and quantitative immunogold electron micro- scopy.68 The GLUT1 glucose transporter is also expressed in other barrier structures in the brain, including the basolateral membrane of choroid plexus epithelium, and the apical membrane of certain regions of the ependymal epithelium.69 The localization of the immunoreactive GLUT1 glucose trans- porter at the human BBB was demonstrated with electron micro- scopy using an antibody directed against the human erythrocyte GLUT1 glucose transporter and a secondary antibody conjugated with 10 nm gold particles.70 GLUT1 glucose transporters are observed on the endothelial plasma membranes, but not within the neuropil of human brain (Figure 3C). The expression of the GLUT1 glucose transporter on both luminal and abluminal mem- branes at the human BBB illustrates how these two endothelial membranes operate in series to regulate BBB transport (Figure 3C). GLUT1 is also detected on the plasma membrane of the human erythrocyte (Figure 3C). The abundance of GLUT1 within the human erythrocyte plasma membrane is said to be highest of all transporters. Yet, the abundance of GLUT1 glucose transporter, expressed as transporter molecules per micron of membrane length, is 2.6-fold higher on the luminal membrane of the human brain capillary endothelium relative to the human red cell membrane.70 The abundance of the GLUT1 glucose transporter, Ccap, at the microvasculature of human brain is high, relative to other transporters, 139±46 pmol/mg protein, as determined by LC– MS (Table 1). The Ccap value determined with LC–MS conforms to a previous estimate of the Ccap of GLUT1, 102±25 pmol/mg protein, determined with a cytochalasin B radioreceptor assay in isolated brain capillaries.71 The Ccap of the GLUT1 glucose transporter is nearly 100-fold higher than the Ccap for other transporters (Table 1), which is attributed to the sole derivation of Figure 3. (A) Predicted secondary structure of the bovine blood–brain barrier (BBB) GLUT1 glucose transporter is shown, which is formed by 12 transmembrane domains, a glycosylated extracellular loop between transmembrane domains 1 and 2, and intracellular amino and carboxyl termini.128 (B) Northern blot of mRNA derived from either total rabbit brain (lane 1) or capillary-depleted rabbit brain (lane 2) for either the GLUT1 glucose transporter or actin.71 (C) Electron microscopic immunogold study of human brain with a primary antiserum against the purified human erythrocyte glucose transporter, and a secondary antibody conjugated with 10 nm gold particles, shows abundant expression of immunoreactive GLUT1 glucose transporter on the luminal (L) and abluminal (AL) membranes of the capillary endothelium in human brain.70 Blood–brain barrier drug delivery WM Pardridge 1964 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 7. energy in the brain from the combustion of glucose. Cerebral rates of glycolysis, 500 to 1,000 nmol per minute per gram, are over 100- fold greater than rates of amino-acid incorporation into brain proteins.46 Glucose supply in the brain is mediated solely via the activity of the BBB GLUT1 glucose transporter. Drug Transport into Brain via Blood–Brain Barrier Carrier-Mediated Transporters The characterization of the BBB GLUT1 glucose transporter exemplifies the extensive knowledge base on the molecular characteristics of a given BBB CMT system, and these systems have been recently cataloged with respect to the transporter abun- dance at the BBB.48 The cDNAs of almost all of the BBB CMT systems are available, and cell lines can be stably transfected with the transporter gene to enable high-throughput screening of drugs that have an acceptable affinity for a given BBB transporter. This knowledge base is not being exploited by the pharmaceutical industry for the discovery of drugs that cross the BBB on a given CMT system. However, there are historical examples of drugs that have an unexpected affinity for a BBB CMT system that may, or may not, be predicted on the basis of the drug structure. For example, L-DOPA is an effective treatment for PD (Parkinson’s disease), because this dopamine precursor is a large neutral amino acid that is transported across the BBB via the LAT1 neutral amino- acid carrier.51 L-DOPA is a straightforward example of a large neutral amino-acid drug that would be predicted to have an affinity for LAT1. However, there are examples of other drugs that have an affinity for LAT1 that are unexpected. Gabapentin is a water-soluble drug that is active in the CNS because the drug crosses the BBB on the large neutral amino-acid transporter,72 which is LAT1. The affinity of gabapentin for LAT1 is unexpected, because gabapentin is a g-amino acid, and LAT1 has an affinity only for a-amino acids. However, gabapentin is a cyclic form of a g-amino acid, which places the amino and carboxyl groups in a conformation that mimics a-amino acids. Paraquat is N,N0 -dimethyl-4,40 -bipyridinium, a widely used, highly water-soluble herbicide, which should not cross the BBB. However, paraquat is neurotoxic, which suggests that this double quaternary ammonium salt is BBB penetrating. Paraquat crosses the BBB via a process that is inhibited by valine, a large neutral amino acid,73 which suggests that paraquat is a substrate for LAT1. Melphalan is a polar alkylating agent used to treat brain cancer. Melphalan, or phenylalanine mustard, crosses the BBB via transport on the BBB large neutral amino-acid carrier.74 L-DOPA, gabapentin, paraquat, and melphalan are examples of BBB delivery via LAT1 of drugs that have structures that mimic the endogenous substrate, neutral amino acids. An alternative approach is to conjugate a drug, which normally does not cross the BBB, with a CMT substrate, for example, glucose, that does cross the BBB. This was attempted with oligopeptides, which were conjugated with glucose. However, the glucose moiety of the glycopeptide no longer was transported via GLUT1.75 Instead, the structure of the CMT substrate must be retained following conju- gation of the nontransportable drug. This is possible with ingenious uses of medicinal chemistry as applied to BBB structure–transport relationships. A novel example is the case where a nontranspor- table drug is coupled to the thiol group of L-cysteine.76 Cysteine is a small neutral amino acid, which has a low affinity for LAT1.45 However, when the drug is conjugated to the thiol group of cysteine, the small neutral amino acid is converted into a large neutral amino acid, and significant BBB transport via LAT1 was observed.76 This case shows that a CMT substrate can be used as a drug carrier, providing the original structural characteristic of the substrate class for the targeted CMT system is retained. Active Efflux Transporters and Enzymatic Blood–Brain Barrier The active efflux transporters (AET) serve to mediate the asymmetric efflux of metabolites and drugs from the brain to blood. The classical AET system within the BBB is p-glycoprotein, which is a product of the MDR (multidrug resistance) gene, MDR1, also named the ABC (ATP-binding cassette) subfamily B member 1 (ABCB1). Apart from p-glycoprotein, there are numerous other members of the ABC gene family of transporters that are expressed at the BBB, and these have been quantified by Terasaki and coworkers.48 The AET systems may work in concert with enzymes expressed within the BBB, such as P450 enzymes.77 Both the AET systems, and the enzymatic barriers are subject to modulation. Blood–brain barrier p-glycoprotein activity is sup- pressed by vascular endothelial growth factor,78 and the CYP1A1 and CYP1B1 P450 enzymes are upregulated by environmental toxins, such as dioxin derivatives, which activate the aryl hydrocarbon receptor at the BBB.79 In addition to environmental toxins, or growth factors, BBB permeability may be altered by disease states, including aging.80 RECEPTOR-MEDIATED TRANSPORT OF PROTEIN THERAPEUTICS Discovery of Blood–Brain Barrier Receptor-Mediated Transport Systems Insulin is present in the brain, but there is no insulin mRNA in the brain.81,82 Therefore, insulin in the brain is of peripheral origin and derived from the blood. Insulin in the blood is transported across the human BBB via RMT on the endogenous BBB insulin receptor.83 Similarly, transferrin (Tf) in blood is transported via RMT across the BBB on the endogenous human BBB Tf receptor (TfR).84 There are separate BBB receptors for the insulin-like growth factors85 and leptin.86 The BBB peptide receptors on either the animal or human BBB were identified using isolated brain capillaries and radioreceptor methodology. The brain capillaries are purified from the brain with a mechanical homogenization procedure and capillaries freshly isolated from bovine brain are shown in Figure 4A. The BBB peptide receptors may mediate functionally different processes, including (1) transcytosis of ligand from blood to brain; (2) reverse transcytosis from brain to blood; or (3) only endocytosis into the brain capillary endothelium without net transport across the endothelial cell (Figure 4B). Insulin RMT across the BBB from blood to brain was demonstrated with carotid artery infusion and thaw mount autoradiography of the brain.87 Transferrin transport from the blood to brain was similarly shown by internal carotid artery infusion and autoradiography.88 Transferrin efflux from the brain to blood was demonstrated with the Brain Efflux Index method,89 as applied to large molecules.90 IgG molecules are asymmetrically transcytosed only in the brain to blood direction via a Fc receptor (FcR) on the BBB.91 Confocal microscopy of brain shows the principal FcR on the brain capillary is the neonatal FcR or FcRn.92 The BBB FcR does not mediate the transport of IgG molecules in the blood to brain direction. Some BBB receptors do not mediate transcytosis across the endothelial barrier, but only mediate endocytosis into the endothelial cell. Acetylated LDL (low-density lipoprotein) is a ligand for the scavenger receptor (SR) on the BBB, and this system mediates the endocytosis of acetyl LDL from blood, but not the net transcytosis across the BBB into the brain.93 Scavenger receptor- related receptors, such as the LDL-related protein receptor (LRP) type 1, similarly mediate only endocytosis of ligands into cells, and not transcytosis.94 Proteins targeting LRP1 are taken up by brain at reduced rates compared with proteins that target the trans- cytosing receptor systems. The brain uptake of melanotransferrin (p97) or angiopep-2, ligands that target LRP1, is only 0.2% to 0.3% of injected dose (ID)/g in the mouse,95,96 which is 10-fold lower than the brain uptake of a monoclonal antibody (MAb) that targets the BBB TfR in the mouse.97 The BBB TfR mediates reverse transcytosis of apotransferrin in the brain to blood direction,90 in addition to influx of holo- transferrin from blood to brain.88 The reverse transcytosis is Blood–brain barrier drug delivery WM Pardridge 1965 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 8. enabled, because the TfR is also expressed on the abluminal membrane of the capillary endothelium in brain. In one study, the abluminal TfR was not detected with preembedding immune labeling methods.98 However, abluminal receptors on the endothelium in the brain cannot be detected with preem- bedding methods.99 Using confocal microscopy of isolated rat brain capillaries, and the OX26 rat MAb against the rat TfR, it was possible to identify the TfR on both luminal and abluminal membranes of the brain capillary endothelium (Figure 4C).100 The pathway of transcytosis through the BBB via the TfR was examined with electron microscopy. The OX26 TfRMAb was conjugated with 5 nm gold and infused in the carotid artery of rats for 10 minutes followed by saline clearance of the brain vasculature and perfusion fixation with glutaraldehyde.101 At the light microscopic level, the TfRMAb entrapped within the capillary endothelium was detected with a silver enhancement technique as shown in Figure 4D. There is no visible reaction product in the brain extravascular space (Figure 4D), owing to the differences in intraendothelial and postendothelial volumes in the brain. The intraendothelial volume is 1 mL/g brain and the postendothelial volume is nearly 3-log orders higher, 700 mL/g brain. Therefore, as the TfRMAb exits the intraendothelial volume, the antibody concentration is diluted 700-fold, and the postvascular antibody in the brain cannot be detected with immune labeling methods. Figure 4. (A) Light micrograph of freshly isolated bovine brain capillaries stained with trypan blue, which stains nuclei blue and luminal red blood cells yellow. The capillaries are isolated free of adjoining brain tissue. (B) Schematic diagram of different brain endothelial receptors, including the insulin receptor, the transferrin (Tf) receptor, the scavenger receptor (SR), which mediates only the endocytosis from blood into the endothelial compartment for ligands such as acetylated low density lipoprotein; and the neonatal Fc receptor (FcRn), which mediates the asymmetric transcytosis of IgG molecules selectively from brain to blood, but not blood to brain. (C) Confocal microscopy of isolated rat brain capillaries showing the expression of the Tf receptor on both luminal (L) and abluminal (AL) membranes of the endothelium.100 (D) Light micrograph of rat brain removed after a 10-minute carotid artery infusion of a conjugate of 5 nm gold and the mouse OX26 monoclonal antibody (MAb) against the rat TfR. The staining of the capillary compartment represents TfRMAb present in the intraendothelial compartment of the brain.101 (E) Electron microscopy of rat brain after perfusion with the conjugate of gold and TfRMAb shows the MAb concentrated in intraendothelial vesicles (intracellular arrow), as well as MAb molecules undergoing exocytosis from the endothelial compartment to the brain interstitial space (extracellular arrow).101 Blood–brain barrier drug delivery WM Pardridge 1966 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 9. The rapid postvascular distribution into the brain via transport on the BBB TfR was demonstrated with the capillary depletion method and autoradiography.88 At the electron microscopic level, the OX26 TfRMAb is detected in endosomes within the capillary endothelium (intracellular arrow, Figure 4E), and antibody is observed exocytosing into the ISF on the brain side of the BBB (extracellular arrow, Figure 4E). Blood–Brain Barrier Penetrating Genetically Engineered IgG Fusion Proteins The presence of endogenous RMT systems within the BBB, such as the insulin receptor or the TfR, enables the reengineering of recombinant proteins as BBB-penetrating pharmaceuticals with the use of molecular Trojan horse (MTH) technology.59 An MTH is an endogenous peptide, or a peptidomimetic MAb, which traverses the BBB on a specific RMT system. Insulin, per se, as an MTH is not preferred, since insulin causes hypoglycemia. Tf, per se, is not a preferred MTH, because the plasma concentration of endogenous Tf, 25 mmol/L, is so high that the Tf-binding site on the BBB TfR is fully saturated by endogenous Tf. Alternatively, RMT-specific MTHs may be peptidomimetic MAbs that bind exofacial epitopes on the BBB receptor, which are spatially removed from the endogenous ligand binding site. Once bound to the RMT system on the BBB, the MTH undergoes RMT across the BBB (Figure 4). An MAb against the human insulin receptor, designated the HIRMAb, undergoes rapid transport across the BBB of the Rhesus monkey in vivo, and the brain uptake is 2% to 3% of ID/brain.102 The HIRMAb crossreacts with the insulin receptor of Old World primates, but does not react with the insulin receptor of New World primates, such as the squirrel monkey, or lower animals such as rodents. The murine HIRMAb has been genetically engineered as either a chimeric or humanized antibody.103 There is no known MAb against the mouse insulin receptor that can be used as a BBB MTH. For BBB delivery in the mouse, a genetically engineered chimeric MAb against the mouse TfR, designated the cTfRMAb, has been produced.104 With the genes encoding the heavy chain and the light chain of either the engineered HIRMAb or the cTfRMAb, IgG fusion proteins may be genetically engineered as bifunctional molecules, which both cross the BBB via RMT and exert pharm- acological effects in brain, following transport into the brain. An IgG-type MTH is also preferred for genetic engineering of MTH fusion proteins, because the protein therapeutic can be fused to the carboxyl terminus of the heavy chain of the IgG, as shown in Figure 5A. It is essential that the bifunctionality of the IgG fusion protein be retained, and that the ‘head’ of the molecule retains high affinity (low nmol/L KD) binding to the BBB receptor, and that the ‘tail’ of the IgG fusion protein retains the pharmacological activity of the original protein therapeutic. If the protein therapeutic is a neurotrophin, there must be retention of high affinity binding of the IgG-neurotrophin fusion protein to the cognate neurotrophin receptor in brain. If an IgG-decoy receptor fusion protein is engineered, there must be retention of high affinity binding to the target cytokine in the brain. If an IgG- lysosomal enzyme fusion protein is produced, the enzyme activity of the IgG-enzyme fusion protein must be comparable to the native enzyme. If the therapeutic molecule is also an antibody, then the problem is the genetic engineering of a bispecific antibody, where there is retention of high affinity binding for both the BBB receptor and the target antigen. This was accomplished by fusion of an ScFv (single chain Fv) form of the therapeutic antibody to the carboxyl terminus of the heavy chain of the IgG.105 This molecular design places the monomeric ScFv antibody in a dimeric configuration, which restores high avidity binding to the target antigen of a bivalent therapeutic antibody. If the high affinity for the target BBB receptor is retained following engineering of the IgG fusion protein, then the rate of Figure 5. (A) Structure of an IgG fusion protein formed by fusion of a therapeutic protein to the carboxyl terminus of the heavy chain of a blood–brain barrier (BBB) targeting monoclonal antibody (MAb). (B) Brain uptake in the mouse, expressed as % injected dose (ID)/g brain, is shown for diazepam,110 a cTfRMAb–EPO fusion protein,59 a cTfRMAb–GDNF fusion protein,59 a cTfRMAb–TNFR fusion protein,59 and the cationic import peptide, tat.129 (C) Brain uptake in the Rhesus monkey is expressed as % ID/100 g brain, since the size of the primate brain is 100 g. Brain uptake is shown for fallypride,111 a HIRMAb–TNFR fusion protein,109 a HIRMAb–EPO fusion protein,107 the TNFR alone,109 EPO alone,107 and a human IgG1k isotype control antibody (hIgG1k), which is confined to the vascular compartment.108 (D) Film autoradiogram of Rhesus monkey brain removed 2 hours after the intravenous (IV) injection of [125 I]-HIRMAb–IDUA fusion protein, showing global distribution in brain with higher uptake in gray matter relative to white matter.130 Blood–brain barrier drug delivery WM Pardridge 1967 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 10. transport of the fusion protein across the BBB will be comparable to the rate of transport of the MTH without a fusion partner. For the mouse, several cTfRMAb fusion proteins were engineered, expressed, and tested for in vivo brain uptake activity. The brain uptake of the cTfRMAb fusion protein of EPO (erythropoietin), GDNF, or the type II TNF-a (tumor necrosis factor-a) decoy receptor (TNFR), is high, 2% to 3% ID/g brain at 60 minutes after an IV injection (Figure 5B). In contrast, the brain uptake of a cationic import peptide, tat, is low, 0.1% ID/g (Figure 5B), and this low level of brain uptake is comparable to the brain uptake of an IgG that does not cross the BBB in the mouse.106 Without fusion to the MTH, there is no BBB transport of the EPO alone,107 the GDNF alone,108 or the TNFR alone.109 The brain uptake of the cTfRMAb fusion proteins in the mouse is comparable to the brain uptake of a lipid soluble small molecule such as diazepam (Figure 5B), which is 1.8% ID/g at 10 minutes after an IV injection.110 Therefore, the brain uptake of the MTH fusion proteins in the mouse is comparable to the brain uptake of lipid soluble small molecules. Similar findings are made in the primate. In the Rhesus monkey, the brain uptake of the HIRMAb fusion proteins is 2% to 3% ID/ 100 g brain (Figure 5C), which is comparable to the peak brain uptake in the Rhesus monkey of fallypride, a lipid soluble small molecule.111 The brain uptake in the Rhesus monkey is expressed per 100 g brain, because the weight of the brain in this species is 100 g. The brain uptake in the Rhesus monkey of the TNFR alone, or EPO alone, is low, about 0.2% ID/100 g brain. This level of brain uptake does not represent a low level of BBB penetration, but rather sequestration of the protein within the vascular com- partment of brain. The primate brain uptake of an isotype IgG1 control antibody that does not cross the BBB is also 0.2% ID/100 g at 60 minutes after an IV injection (Figure 5C). The IgG fusion proteins undergo transcytosis through the BBB, and penetrate brain parenchyma. The global distribution within the brain of the fusion protein is shown by the brain scan of the Rhesus monkey at 2 hours after the IV injection of a fusion protein of the HIRMAb, and the human lysosomal enzyme, iduronidase (IDUA) (Figure 5D). The higher uptake in gray matter, as compared with white matter, is due to the higher vascular density in gray matter. The finding of transcytosis of the fusion proteins through the BBB and into brain parenchyma is corroborated by the in vivo CNS pharmacological effects of IV administration of the fusion proteins in mouse models of brain disease, including Hurler’s syndrome, PD, Alzheimer’s disease (AD), and stroke (Table 2). BLOOD–BRAIN BARRIER TRANSPORT OF PEPTIDE RADIOPHARMACEUTICALS FOR NEUROIMAGING The number of potential peptide radiopharmaceuticals that could be used to image brain disorders is large. However, peptide radiopharmaceuticals for the brain are not developed, because these large molecules do not cross the BBB. Peptide radio- pharmaceuticals can be reengineered for BBB transport with the combined use of MTH technology, avidin–biotin technology, and radiopharmaceutical technology. The peptide radiopharmaceuti- cal can be conjugated to the IgG MTH via a high affinity avidin– biotin linker, as shown in Figure 6A. An MTH–avidin fusion protein has been genetically engineered both for the HIRMAb, for primate or human applications, and for the cTfRMAb, for mouse studies.112,113 The IgG–avidin fusion protein is formulated in one vial. The monobiotinylated peptide radiopharmaceutical is for- mulated in a second vial. The two vials are mixed before IV administration. Owing to the very high affinity of avidin binding of biotin, there is instantaneous capture of the peptide radio- pharmaceutical by the MTH (Figure 6A). The efficacy of this approach has been demonstrated in imaging models in both primates and rodents, using either the Ab1–40 amyloid peptide of AD or epidermal growth factor (EGF) as the model peptide radiopharmaceutical. The Ab1–40 amyloid peptide is a potential peptide radio- pharmaceutical for the neuroimaging of brain amyloid in AD, as this peptide avidly binds amyloid plaque in sections of AD autopsy brain.114 However, the Ab1–40 amyloid peptide alone does not cross the BBB,113,115 and the absence of BBB transport prevents the development of this peptide as a radiopharmaceutical for brain imaging in AD. The [N-biotinyl]-Ab1–40 was radioiodinated and injected alone IV in the Rhesus monkey.115 No measureable brain uptake of the peptide was observed by brain scanning (Figure 6B). However, in a second Rhesus monkey, the [N-biotinyl]- Ab1–40 was first conjugated to the HIRMAb via a streptavidin linker before IV injection of the HIRMAb/Ab complex. Now, the peptide radiopharmaceutical rapidly penetrated the BBB in the primate (Figure 6C). Following reengineering with the MTH technology, the brain uptake of the Ab1–40 amyloid peptide is comparable to the brain uptake of lipid soluble small molecule amyloid imaging agents. The brain uptake of the [N-biotinyl]-Ab1–40 alone is very low in the mouse, 0.1% ID/g, but the brain uptake of the peptide increases to 2.1% ID/g following conjugation to the Figure 6. (A) Conjugate of an IgG–avidin fusion protein and a monobiotinylated peptide radiopharmaceutical. The biotin group on the peptide binds to the avidin domain of the fusion protein to form a high affinity linkage between the IgG molecular Trojan horse and the peptide radiopharmaceutical. (B, C) Scans of Rhesus monkey brain at 3 hours after the intravenous (IV) injection of either [125 I, N-biotinyl]-Ab1–40 alone (B), or [125 I, N-biotinyl]-Ab1–40 conjugated to the HIRMAb (monoclonal antibody) molecular Trojan horse via a streptavidin linker (C).115 (D) A rat brain tumor model was produced by the intracerebral injection of human U87 glioma cells in nude rats. Immunocytochemistry (ICC) of brain removed 16 days after tumor implantation with an antibody to the human EGFR shows abundant expression of the EGFR in the brain tumor.119 (E) Scan of rat brain removed 2 hours after the IV injection of [111 In-DTPA, Lys-biotinyl]–EGF conjugated to the TfRMAb mole- cular Trojan horse via a streptavidin linker. The section used for the film autoradiography was parallel to the section used for the ICC study in (D).119 Blood–brain barrier drug delivery WM Pardridge 1968 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 11. cTfRMAb–avidin fusion protein.113 This level of brain uptake of a peptide radiopharmaceutical is comparable to the brain uptake of a lipid soluble small molecule brain amyloid imaging agent, which is 2% to 3% ID/g in the mouse.116 The complex of the HIRMAb– avidin fusion protein and [N-biotinyl]-Ab1–40 retains high affinity binding to the amyloid plaques in autopsy sections of AD brain.117 The [N-biotinyl]-Ab1–40 can be radiolabeled with 124-iodine for positron emission tomography imaging of the brain. Alternatively, the [N-biotinyl]-Ab1–40 can be conjugated with a chelator moiety and labeled with 111-indium118 for single photon emission computed tomography. Epidermal growth factor is a potential peptide radiopharma- ceutical for imaging brain cancers that overexpress the EGF receptor (EGFR). Epidermal growth factor was monobiotinylated and conjugated with diethylenediaminepentaacetic acid (DTPA) and radiolabeled with 111-indium. A rat model of human brain cancer was developed with the intracranial injection of human U87 glioma cells in nude rats.119 At autopsy, immunocytoche- mistry with an antibody against the human EGFR showed selective expression of the EGFR in the brain tumor (Figure 6D). IV injection of the [111 In-DTPA, biotinyl]-EGF alone enabled no imaging of the brain tumor in vivo, because EGF does not cross the BBB.120 However, when the [111 In-DTPA, biotinyl]-EGF was conjugated to the TfRMAb with a streptavidin linker, there was robust imaging of the EGFR-positive brain cancer following IV injection of the BBB-targeted peptide radiopharmaceutical (Figure 6E). BLOOD–BRAIN BARRIER TRANSPORT OF ANTISENSE RADIOPHARMACEUTICALS FOR NEUROIMAGING Antisense radiopharmaceuticals are potential large molecule agents for imaging in brain the gene expression of sequence specific mRNA. However, antisense agents do not cross the BBB.121 An ideal antisense imaging agent is the peptide nucleic acid (PNA). Peptide nucleic acids are resistant to nucleases, as compared with phosphodiester-oligodeoxynucleotides (ODN), and have less nonspecific binding than phosphorothioate-ODNs. Peptide nucleic acid binding to target mRNAs does not trigger RNase H activity as is observed with ODNs.122 To evaluate BBB- targeted PNAs as antisense imaging agents for the brain, an intracranial RG-2 brain cancer model in rats was developed.123 Gene expression within the tumor was assessed for glial fibrillary acidic protein (GFAP) and caveolin-1a (CAV). The sequence of the GFAP-targeted and CAV-targeted PNAs is shown in Figure 7A. The amino terminus of the PNA was conjugated with DTPA for radiolabelling with111 In (Figure 7A). The amide backbone of the PNA contained a carboxyl terminal lysine (Lys) residue and the e-amino group of the Lys residue was monobiotinylated (Figure 7A). Confocal microscopy showed the intracranial brain tumor underexpressed the GFAP protein and overexpressed the CAV protein (Figure 7B). Northern blot analysis showed the level of the GFAP and CAV mRNAs in the tumor paralleled the protein, as the GFAP mRNA was underexpressed, and the CAV mRNA was overexpressed.123 The biotin-GFAP-PNA or the biotin-CAV-PNA was conjugated to the TfRMAb via a streptavidin linker. Conjugation of the PNA to the MTH did not affect binding of the PNA to the target transcript. This was demonstrated with Northern blotting using the labeled PNA as the probe, and cloned GFAP or CAV RNA as the target.123 Following IV injection of either the [111 In-DTPA, biotinyl]-GFAP PNA alone, or the [111 In-DTPA, biotinyl]-CAV PNA alone, there was no imaging of brain, as these molecules do not cross the BBB. Following the IV injection of the [111 In-DTPA, biotinyl]-GFAP PNA/TfRMAb conjugate, the nontumor brain was imaged, but there was no signal over the tumor Figure 7. (A) Structures are shown for model peptide nucleic acids (PNAs) with sequences that target either the rat glial fibrillary acidic protein (GFAP) mRNA or the rat caveolin-1a (CAV) mRNA.123 The methionine initiation codon sequence is underlined. (B) Confocal microscopy of brain from a rat with an intracranial RG-2 glioma is shown after double labeling with an antibody against GFAP (green channel) and an antibody against CAV (red channel).123 (C) Scan of brain removed from a rat with an intracranial RG-2 glioma at 1 hour after the intravenous (IV) injection of the [111 In-DTPA, Lys-biotinyl]-GFAP PNA conjugated to the TfRMAb (monoclonal antibody) via a streptavidin linkage. The image over the tumor is of low intensity, as the RG-2 tumor does not express the GFAP mRNA.123 (D) Scan of brain removed from a rat with an intracranial RG-2 glioma at 6 hours after the IV injection of the [111 In-DTPA, Lys-biotinyl]-CAV PNA conjugated to the TfRMAb via a streptavidin linkage. The image over the tumor is of high intensity, as the RG-2 tumor expresses abundant CAV mRNA.123 Blood–brain barrier drug delivery WM Pardridge 1969 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 12. (Figure 7C). Therefore, the in vivo brain scan with the targeted GFAP-PNA antisense radiopharmaceutical confirmed the in vitro finding of minimal expression of the GFAP mRNA in the glial tumor. Following the IV injection of the [111 In-DTPA, biotinyl]–CAV PNA/ TfRMAb conjugate, the tumor showed an intense signal, owing to overexpression of the CAV mRNA in the tumor (Figure 7D). The in vivo brain scans (Figures 7C and 7D) of the GFAP or CAV mRNAs with BBB-targeted antisense radiopharmaceuticals paralleled the immune detection of GFAP or CAV proteins in autopsy sections (Figure 7B), and the levels of the GFAP or CAV mRNAs in the tumor.123 Peptide and antisense radiopharmaceuticals have the potential to greatly expand the diagnostic capabilities of modern neuroimaging. However, these molecules need to be reformulated for BBB transport, and this is possible with the combined use of MTH and avidin–biotin technologies. In summary, small molecule drugs may be reengineered to enable transport across the BBB via the CMT systems expressed in the brain capillary endothelium. Large molecule drugs, including recombinant proteins, and peptide and antisense radiopharma- ceuticals may be delivered across the BBB with MTHs that target the endogenous RMT systems expressed within the brain capillary endothelium. The reengineering of pharmaceuticals for BBB transport is enabled with knowledge on the endogenous CMT and RMT systems within the BBB. DISCLOSURE/CONFLICT OF INTEREST The author is a consultant to ArmaGen Technologies. REFERENCES 1 Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx 2005; 2: 3–14. 2 Brightman MW, Reese TS. Junctions between intimately apposed cell mem- branes in the vertebrate brain. J Cell Biol 1969; 40: 648–677. 3 Reiber H, Felgenhauer K. Protein transfer at the blood cerebrospinal fluid barrier and the quantitation of the humoral immune response within the central ner- vous system. Clin Chim Acta 1987; 163: 319–328. 4 Mott FW. The late Professor Edwin Goldmann’s investigations on the central nervous system by vital staining. Br Med J 1913; 2: 871–873. 5 Friedemann U. Blood-brain barrier. Physiol Rev 1942; 22: 125–145. 6 Broman T. The possibilities of the passage of substances from the blood to the central nervous system. Acta Psych Neurol 1941; 16: 1–16. 7 Rowland LP. Blood-brain barrier, cerebrospinal fluid, brain edema, and hydro- cephalus. In: Kandel ER, Schwartz JH (eds) Principles of Neural Science. 2nd edn (Elsevier Science Publishing: New York, NY, 1985, pp 837–844. 8 Davson H. The cerebrospinal fluid. Handbook of Neurochemistry 1969; 2: 23–48. 9 Oldendorf WH. Cerebrospinal fluid formation and circulation. Prog Nucl Med 1972; 1: 336–358. 10 Cutler RW, Page L, Galicich J, Watters GV. Formation and absorption of cerebrospinal fluid in man. Brain 1968; 91: 707–720. 11 Rudick RA, Zirretta DK, Herndon RM. Clearance of albumin from mouse sub- arachnoid space: a measure of CSF bulk flow. J Neurosci Methods 1982; 6: 253–259. 12 Yan Q, Matheson C, Sun J, Radeke MJ, Feinstein SC, Miller JA. Distribution of intracerebral ventricularly administered neurotrophins in rat brain and its correlation with trk receptor expression. Exp Neurol 1994; 127: 23–36. 13 Blasberg RG, Patlak C, Fenstermacher JD. Intrathecal chemotherapy: brain tissue profiles after ventriculocisternal perfusion. J Pharmacol Exp Ther 1975; 195: 73–83. 14 Fung LK, Shin M, Tyler B, Brem H, Saltzman WM. Chemotherapeutic drugs released from polymers: distribution of 1,3-bis(2-chloroethyl)-1-nitrosourea in the rat brain. Pharm Res 1996; 13: 671–682. 15 Day-Lollini PA, Stewart GR, Taylor MJ, Johnson RM, Chellman GJ. Hyperplastic changes within the leptomeninges of the rat and monkey in response to chronic intracerebroventricular infusion of nerve growth factor. Exp Neurol 1997; 145: 24–37. 16 Yamada K, Kinoshita A, Kohmura E, Sakaguchi T, Taguchi J, Kataoka K et al. Basic fibroblast growth factor prevents thalamic degeneration after cortical infarction. J Cereb Blood Flow Metab 1991; 11: 472–478. 17 Marin-Padilla M, Knopman DS. Developmental aspects of the intracerebral microvasculature and perivascular spaces: insights into brain response to late- life diseases. J Neuropathol Exp Neurol 2011; 70: 1060–1069. 18 Szentistvanyi I, Patlak CS, Ellis RA, Cserr HF. Drainage of interstitial fluid from different regions of rat brain. Am J Physiol 1984; 246: F835–F844. 19 Fishman RA, Christy NP. Fate of adrenal cortical steroids following intrathecal injection. Neurology 1965; 15: 1–6. 20 Aird RB. A study of intrathecal, cerebrospinal fluid-to-brain exchange. Exp Neurol 1984; 86: 342–358. 21 de Lange EC, Danhof M, de Boer AG, Breimer DD. Critical factors of intracerebral microdialysis as a technique to determine the pharmacokinetics of drugs in rat brain. Brain Res 1994; 666: 1–8. 22 Crawley JN, Fiske SM, Durieux C, Derrien M, Roques BP. Centrally administered cholecystokinin suppresses feeding through a peripheral-type receptor mechanism. J Pharmacol Exp Ther 1991; 257: 1076–1080. 23 Kristensson K, Olsson Y. Uptake of exogenous proteins in mouse olfactory cells. Acta Neuropathol 1971; 19: 145–154. 24 Merkus P, Guchelaar HJ, Bosch DA, Merkus FW. Direct access of drugs to the human brain after intranasal drug administration? Neurology 2003; 60: 1669–1671. 25 Westin U, Piras E, Jansson B, Bergstrom U, Dahlin M, Brittebo E et al. Transfer of morphine along the olfactory pathway to the central nervous system after nasal administration to rodents. Eur J Pharm Sci 2005; 24: 565–573. 26 Graff CL, Pollack GM. Nasal drug administration: potential for targeted central nervous system delivery. J Pharm Sci 2005; 94: 1187–1195. 27 Salvatore MF, Ai Y, Fischer B, Zhang AM, Grondin RC, Zhang Z et al. Point source concentration of GDNF may explain failure of phase II clinical trial. Exp Neurol 2006; 202: 497–505. 28 Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem 1999; 1: 55–68. 29 Lipinski CA. Drug-like properties and the causes of poor solubility and poor permeability. J Pharmacol Toxicol Methods 2000; 44: 235–249. 30 Ajay Bemis GW, Murcko MA. Designing libraries with CNS activity. J Med Chem 1999; 42: 4942–4951. 31 Cohen BE, Bangham AD. Diffusion of small non-electrolytes across liposome membranes. Nature 1972; 236: 173–174. 32 Lieb WR, Stein WD. Non-Stokesian nature of transverse diffusion within human red cell membranes. J Membr Biol 1986; 92: 111–119. 33 Levin VA. Relationship of octanol/water partition coefficient and molecular weight to rat brain capillary permeability. J Med Chem 1980; 23: 682–684. 34 van de Waterbeemd H, Camenisch G, Folkers G, Chretien JR, Raevsky OA. Esti- mation of blood-brain barrier crossing of drugs using molecular size and shape, and H-bonding descriptors. J Drug Target 1998; 6: 151–165. 35 Fischer H, Gottschlich R, Seelig A. Blood-brain barrier permeation: molecular parameters governing passive diffusion. J Membr Biol 1998; 165: 201–211. 36 Trauble H. The movement of molecules across lipid membranes: a molecular theory. J Membrane Biol 1971; 4: 193–208. 37 Hingson DJ, Diamond JM. Comparison of nonelectrolyte permeability patterns in several epithelia. J Membr Biol 1972; 10: 93–135. 38 Marrink SJ, Jahnig F, Berendsen HJ. Proton transport across transient single-file water pores in a lipid membrane studied by molecular dynamics simulations. Biophys J 1996; 71: 632–647. 39 Diamond JM, Wright EM. Molecular forces governing non-electrolyte permeation through cell membranes. Proc R Soc Lond B Biol Sci 1969; 171: 273–316. 40 Stein WD. The Molecular Basis of Diffusion Across Cell Membranes. Academic Press: New York, NY, 1967, pp 66–125. 41 Pardridge WM, Mietus LJ. Transport of steroid hormones through the rat blood- brain barrier. J Clin Invest 1979; 64: 145–154. 42 Chikhale EG, Ng KY, Burton PS, Borchardt RT. Hydrogen bonding potential as a determinant of the in vitro and in situ blood-brain barrier permeability of peptides. Pharm Res 1994; 11: 412–419. 43 Oldendorf WH, Hyman S, Braun L, Oldendorf SZ. Blood-brain barrier: penetration of morphine, codeine, heroin, and methadone after carotid injection. Science 1972; 178: 984–986. 44 Crone C. Facilitated transfer of glucose from blood into brain tissue. J Physiol 1965; 181: 103–113. 45 Oldendorf WH. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am J Physiol 1971; 221: 1629–1639. 46 Pardridge WM, Oldendorf WH. Transport of metabolic substrates through the blood-brain barrier. J Neurochem 1977; 28: 5–12. 47 Pardridge WM. Recent advances in blood-brain barrier transport. Annu Rev Pharmacol Toxicol 1988; 28: 25–39. 48 Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie J et al. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J Neurochem 2011; 117: 333–345. 49 Pardridge WM, Oldendorf WH. Kinetics of blood-brain transport of hexoses. Biochim Biophys Acta 1975; 382: 377–392. Blood–brain barrier drug delivery WM Pardridge 1970 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM
  • 13. 50 Pardridge WM, Boado RJ, Farrell CR. Brain-type glucose transporter (GLUT-1) is selectively localized to the blood-brain barrier. Studies with quantitative western blotting and in situ hybridization. J Biol Chem 1990; 265: 18035–18040. 51 Pardridge WM, Oldendorf WH. Kinetic analysis of blood-brain barrier transport of amino acids. Biochim Biophys Acta 1975; 401: 128–136. 52 Boado RJ, Li JY, Nagaya M, Zhang C, Pardridge WM. Selective expression of the large neutral amino acid transporter at the blood-brain barrier. Proc Natl Acad Sci USA 1999; 96: 12079–12084. 53 Stoll J, Wadhwani KC, Smith QR. Identification of the cationic amino acid transporter (System y þ ) of the rat blood-brain barrier. J Neurochem 1993; 60: 1956–1959. 54 Cremer JE, Cunningham VJ, Pardridge WM, Braun LD, Oldendorf WH. Kinetics of blood-brain barrier transport of pyruvate, lactate and glucose in suckling, weanling and adult rats. J Neurochem 1979; 33: 439–445. 55 Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR. Expression of monocarboxylate transporter MCT1 by brain endothelium and glia in adult and suckling rats. Am J Physiol 1997; 273: E207–E213. 56 Cornford EM, Oldendorf WH. Independent blood-brain barrier transport systems for nucleic acid precursors. Biochim Biophys Acta 1975; 394: 211–219. 57 Li JY, Boado RJ, Pardridge WM. Cloned blood-brain barrier adenosine transporter is identical to the rat concentrative Na þ nucleoside cotransporter CNT2. J Cereb Blood Flow Metab 2001; 21: 929–936. 58 Ohtsuki S, Uchida Y, Kubo Y, Terasaki T. Quantitative targeted absolute pro- teomics-based ADME research as a new path to drug discovery and develop- ment: methodology, advantages, strategy, and prospects. J Pharm Sci 2011; 100: 3547–3559. 59 Pardridge WM, Boado RJ. Reengineering biopharmaceuticals for targeted delivery across the blood-brain barrier. Methods Enzymol 2012; 503: 269–292. 60 Sumbria RK, Boado RJ, Pardridge WM. Brain protection from stroke with intra- venous TNFa decoy receptor-Trojan horse fusion protein. J Cereb Blood Flow Metab 2012, in press. 61 Gjedde A, Christensen O. Estimates of Michaelis-Menten constants for the two membranes of the brain endothelium. J Cereb Blood Flow Metab 1984; 4: 241–249. 62 Schulz A, Beyhl D, Marten I, Wormit A, Neuhaus E, Poschet G et al. Proton-driven sucrose symport and antiport are provided by the vacuolar transporters SUC4 and TMT1/2. Plant J 2011; 68: 129–136. 63 Mueckler M, Caruso C, Baldwin SA, Panico M, Blench I, Morris HR et al. Sequence and structure of a human glucose transporter. Science 1985; 229: 941–945. 64 Boado RJ, Pardridge WM. Molecular cloning of the bovine blood-brain barrier glucose transporter cDNA and demonstration of phylogenetic conservation of the 50 -untranslated region. Mol Cell Neurosci 1990; 1: 224–232. 65 Birnbaum MJ, Haspel HC, Rosen OM. Cloning and characterization of a cDNA encoding the rat brain glucose-transporter protein. Proc Natl Acad Sci USA 1986; 83: 5784–5788. 66 Flier JS, Mueckler M, McCall AL, Lodish HF. Distribution of glucose transporter messenger RNA transcripts in tissues of rat and man. J Clin Invest 1987; 79: 657–661. 67 Boado RJ, Pardridge WM. The brain-type glucose transporter mRNA is specifically expressed at the blood-brain barrier. Biochem Biophys Res Commun 1990; 166: 174–179. 68 Farrell CL, Pardridge WM. Blood-brain barrier glucose transporter is asymme- trically distributed on brain capillary endothelial lumenal and ablumenal mem- branes: an electron microscopic immunogold study. Proc Natl Acad Sci USA 1991; 88: 5779–5783. 69 Farrell CL, Yang J, Pardridge WM. GLUT1 glucose transporter is present within apical and basolateral membranes of brain epithelial interfaces and in micro- vascular endothelia with and without tight junctions. J Histochem Cytochem 1992; 40: 193–199. 70 Cornford EM, Hyman S, Swartz BE. The human brain GLUT1 glucose transporter: ultrastructural localization to the blood-brain barrier endothelia. J Cereb Blood Flow Metab 1994; 14: 106–112. 71 Dwyer KJ, Pardridge WM. Developmental modulation of blood-brain barrier and choroid plexus GLUT1 glucose transporter messenger ribonucleic acid and immunoreactive protein in rabbits. Endocrinology 1993; 132: 558–565. 72 Wang Y, Welty DF. The simultaneous estimation of the influx and efflux blood- brain barrier permeabilities of gabapentin using a microdialysis-pharmacokinetic approach. Pharm Res 1996; 13: 398–403. 73 Shimizu K, Ohtaki K, Matsubara K, Aoyama K, Uezono T, Saito O et al. Carrier- mediated processes in blood--brain barrier penetration and neural uptake of paraquat. Brain Res 2001; 906: 135–142. 74 Cornford EM, Young D, Paxton JW, Finlay GJ, Wilson WR, Pardridge WM. Mel- phalan penetration of the blood-brain barrier via the neutral amino acid trans- porter in tumor-bearing brain. Cancer Res 1992; 52: 138–143. 75 Witt KA, Gillespie TJ, Huber JD, Egleton RD, Davis TP. Peptide drug modifications to enhance bioavailability and blood-brain barrier permeability. Peptides 2001; 22: 2329–2343. 76 Killian DM, Hermeling S, Chikhale PJ. Targeting the cerebrovascular large neutral amino acid transporter (LAT1) isoform using a novel disulfide-based brain drug delivery system. Drug Deliv 2007; 14: 25–31. 77 Ghosh C, Gonzalez-Martinez J, Hossain M, Cucullo L, Fazio V, Janigro D et al. Pattern of P450 expression at the human blood-brain barrier: roles of epileptic condition and laminar flow. Epilepsia 2010; 51: 1408–1417. 78 Hawkins BT, Sykes DB, Rapid Miller DS. reversible modulation of blood-brain barrier P-glycoprotein transport activity by vascular endothelial growth factor. J Neurosci 2010; 30: 1417–1425. 79 Dauchy S, Miller F, Couraud PO, Weaver RJ, Weksler B, Romero IA et al. Expression and transcriptional regulation of ABC transporters and cytochromes P450 in hCMEC/D3 human cerebral microvascular endothelial cells. Biochem Pharmacol 2009; 77: 897–909. 80 Zeevi N, Pachter J, McCullough LD, Wolfson L, Kuchel GA. The blood-brain barrier: geriatric relevance of a critical brain-body interface. J Am Geriatr Soc 2010; 58: 1749–1757. 81 Coker III GT , Studelska D, Harmon S, Burke W, O’Malley KL. Analysis of tyrosine hydroxylase and insulin transcripts in human neuroendocrine tissues. Brain Res Mol Brain Res 1990; 8: 93–98. 82 Kojima H, Fujimiya M, Matsumura K, Nakahara T, Hara M, Chan L. Extrapancreatic insulin-producing cells in multiple organs in diabetes. Proc Natl Acad Sci USA 2004; 101: 2458–2463. 83 Pardridge WM, Eisenberg J, Yang J. Human blood-brain barrier insulin receptor. J Neurochem 1985; 44: 1771–1778. 84 Pardridge WM, Eisenberg J, Yang J. Human blood-brain barrier transferrin receptor. Metabolism 1987; 36: 892–895. 85 Duffy KR, Pardridge WM, Rosenfeld RG. Human blood-brain barrier insulin-like growth factor receptor. Metabolism 1988; 37: 136–140. 86 Golden PL, Maccagnan TJ, Pardridge WM. Human blood-brain barrier leptin receptor. Binding and endocytosis in isolated human brain microvessels. J Clin Invest 1997; 99: 14–18. 87 Duffy KR, Pardridge WM. Blood-brain barrier transcytosis of insulin in developing rabbits. Brain Res 1987; 420: 32–38. 88 Skarlatos S, Yoshikawa T, Pardridge WM. Transport of [125I]transferrin through the rat blood-brain barrier. Brain Res 1995; 683: 164–171. 89 Kakee A, Terasaki T, Sugiyama Y. Brain efflux index as a novel method of ana- lyzing efflux transport at the blood-brain barrier. J Pharmacol Exp Ther 1996; 277: 1550–1559. 90 Zhang Y, Pardridge WM. Rapid transferrin efflux from brain to blood across the blood-brain barrier. J Neurochem 2001; 76: 1597–1600. 91 Zhang Y, Pardridge WM. Mediated efflux of IgG molecules from brain to blood across the blood-brain barrier. J Neuroimmunol 2001; 114: 168–172. 92 Schlachetzki F, Zhu C, Pardridge WM. Expression of the neonatal Fc receptor (FcRn) at the blood-brain barrier. J Neurochem 2002; 81: 203–206. 93 Triguero D, Buciak J, Pardridge WM. Capillary depletion method for quantifica- tion of blood-brain barrier transport of circulating peptides and plasma proteins. J Neurochem 1990; 54: 1882–1888. 94 Nazer B, Hong S, Selkoe DJ. LRP promotes endocytosis and degradation, but not transcytosis, of the amyloid-beta peptide in a blood-brain barrier in vitro model. Neurobiol Dis 2008; 30: 94–102. 95 Karkan D, Pfeifer C, Vitalis TZ, Arthur G, Ujiie M, Chen Q et al. A unique carrier for delivery of therapeutic compounds beyond the blood-brain barrier. PLoS One 2008; 3: e2469. 96 Che C, Yang G, Thiot C, Lacoste MC, Currie JC, Demeule M et al. New Angiopep- modified doxorubicin (ANG1007) and etoposide (ANG1009) chemotherapeutics with increased brain penetration. J Med Chem 2010; 53: 2814–2824. 97 Zhou QH, Boado RJ, Lu JZ, Hui EK-W, Pardridge WM. Monoclonal antibody-glial- derived neurotrophic factor fusion protein penetrates the blood-brain barrier in the mouse. Drug Metab Dispos 2010; 38: 566–572. 98 Roberts RL, Fine RE, Sandra A. Receptor-mediated endocytosis of transferrin at the blood-brain barrier. J Cell Sci 1993; 104(Pt 2): 521–532. 99 Vorbrodt AW. Ultracytochemical characterization of anionic sites in the wall of brain capillaries. J Neurocytol 1989; 18: 359–368. 100 Huwyler J, Pardridge WM. Examination of blood-brain barrier transferrin receptor by confocal fluorescent microscopy of unfixed isolated rat brain capillaries. J Neurochem 1998; 70: 883–886. 101 Bickel U, Kang YS, Yoshikawa T, Pardridge WM. In vivo demonstration of sub- cellular localization of anti-transferrin receptor monoclonal antibody-colloidal gold conjugate in brain capillary endothelium. J Histochem Cytochem 1994; 42: 1493–1497. 102 Pardridge WM, Kang YS, Buciak JL, Yang J. Human insulin receptor monoclonal antibody undergoes high affinity binding to human brain capillaries in vitro and Blood–brain barrier drug delivery WM Pardridge 1971 & 2012 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972
  • 14. rapid transcytosis through the blood-brain barrier in vivo in the primate. Pharm Res 1995; 12: 807–816. 103 Boado RJ, Zhang YF, Zhang Y, Pardridge WM. Humanization of anti-human insulin receptor antibody for drug targeting across the human blood-brain barrier. Biotechnol Bioeng 2007; 96: 381–391. 104 Boado RJ, Zhang Y, Wang Y, Pardridge WM. Engineering and expression of a chimeric transferrin receptor monoclonal antibody for blood-brain barrier delivery in the mouse. Biotechnol Bioeng 2009; 102: 1251–1258. 105 Boado RJ, Zhang YF, Zhang Y, Xia CF, Pardridge WM. Fusion antibody for Alz- heimer’s disease with bidirectional transport across the blood-brain barrier and abeta fibril disaggregation. Bioconjug Chem 2007; 18: 447–455. 106 Lee HJ, Engelhardt B, Lesley J, Bickel U, Pardridge WM. Targeting rat anti-mouse transferrin receptor monoclonal antibodies through blood-brain barrier in mouse. J Pharmacol Exp Ther 2000; 292: 1048–1052. 107 Boado RJ, Hui EK, Lu JZ, Pardridge WM. Drug targeting of erythropoietin across the primate blood-brain barrier with an IgG molecular Trojan horse. J Pharmacol Exp Ther 2010; 333: 961–969. 108 Boado RJ, Pardridge WM. Comparison of blood-brain barrier transport of glial- derived neurotrophic factor (GDNF) and an IgG-GDNF fusion protein in the rhesus monkey. Drug Metab Dispos 2009; 37: 2299–2304. 109 Boado RJ, Hui EK, Lu JZ, Zhou QH, Pardridge WM. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J Biotechnol 2010; 146: 84–91. 110 Greenblatt DJ, Sethy VH. Benzodiazepine concentrations in brain directly reflect receptor occupancy: studies of diazepam, lorezepam, and oxazepam. Psycho- pharmacology (Berl) 1990; 102: 373–378. 111 Christian BT, Vandehey NT, Fox AS, Murali D, Oakes TR. Converse AK, et al. The distribution of D2/D3 receptor binding in the adolescent rhesus monkey using small animal PET imaging. Neuroimage 2009; 44: 1334–1344. 112 Boado RJ, Zhang Y, Xia CF, Wang Y, Pardridge WM. Genetic engineering, expression, and activity of a chimeric monoclonal antibody-avidin fusion protein for receptor-mediated delivery of biotinylated drugs in humans. Bioconjug Chem 2008; 19: 731–739. 113 Zhou QH, Lu JZ, Hui EK, Boado RJ, Pardridge WM. Delivery of a peptide radio- pharmaceutical to brain with an IgG-avidin fusion protein. Bioconjug Chem 2011; 22: 1611–1618. 114 Maggio JE, Stimson ER, Ghilardi JR, Allen CJ, Dahl CE, Whitcomb DC et al. Reversible in vitro growth of Alzheimer disease beta-amyloid plaques by deposition of labeled amyloid peptide. Proc Natl Acad Sci USA 1992; 89: 5462–5466. 115 Wu D, Yang J, Pardridge WM. Drug targeting of a peptide radiopharmaceutical through the primate blood-brain barrier in vivo with a monoclonal antibody to the human insulin receptor. J Clin Invest 1997; 100: 1804–1812. 116 Ono M, Cheng Y, Kimura H, Cui M, Kagawa S, Nishi R et al. Novel 18F-labeled benzofuran derivatives with improved properties for positron emission tomo- graphy (PET) imaging of beta-amyloid plaques in Alzheimer’s brains. J Med Chem 2011; 54: 2971–2979. 117 Sumbria RK, Boado RJ, Pardridge WM. Imaging amyloid plaque in Alzheimer’s disease brain with a biotinylated Abeta peptide radiopharmaceutical conjugated to an IgG-avidin fusion protein. Bioconjug Chem 2012; 23: 1318–1321. 118 Kurihara A, Pardridge WM. Abeta(1-40) peptide radiopharmaceuticals for brain amyloid imaging: (111)In chelation, conjugation to poly(ethylene glycol)-biotin linkers, and autoradiography with Alzheimer’s disease brain sections. Bioconjug Chem 2000; 11: 380–386. 119 Kurihara A, Pardridge WM. Imaging brain tumors by targeting peptide radio- pharmaceuticals through the blood-brain barrier. Canc Res 1999; 54: 6159–6163. 120 Kurihara A, Deguchi Y, Pardridge WM. Epidermal growth factor radio- pharmaceuticals: 111In chelation, conjugation to a blood-brain barrier delivery vector via a biotin-polyethylene linker, pharmacokinetics, and in vivo imaging of experimental brain tumors. Bioconjug Chem 1999; 10: 502–511. 121 Pardridge WM, Boado RJ, Kang YS. Vector-mediated delivery of a polyamide (‘peptide’) nucleic acid analogue through the blood-brain barrier in vivo. Proc Natl Acad Sci USA 1995; 92: 5592–5596. 122 Knudsen H, Nielsen PE. Antisense properties of duplex- and triplex-forming PNAs. Nucleic Acids Res 1996; 24: 494–500. 123 Suzuki T, Wu D, Schlachetzki F, Li JY, Boado RJ, Pardridge WM. Imaging endo- genous gene expression in brain cancer in vivo with 111In-peptide nucleic acid antisense radiopharmaceuticals and brain drug-targeting technology. J Nucl Med 2004; 45: 1766–1775. 124 Pardridge WM. Misconceptions about the blood-brain barrier. In: Pardridge WM moderator. Blood-brain barrier: interface between internal medicine and brain Ann Int Med, 1986; 105: 82–95. 125 Bar T. The vascular system of the cerebral cortex. Adv Anat Embryol Cell Biol 1980; 59: I-VI, 1–62. 126 Duvernoy H, Delon S, Vannson JL. The vascularization of the human cerebellar cortex. Brain Res Bull 1983; 11: 419–480. 127 Pardridge WM. Biopharmaceutical drug targeting to the brain. J Drug Target 2010; 18: 157–167. 128 Pardridge WM, Boado RJ. Molecular cloning and regulation of gene expression of blood-brain barrier glucose transporter. In: Pardridge WM (ed The Blood-Brain Barrier: Cellular and Molecular Biology. Raven Press: New York, 1993, pp 347–362. 129 Sarko D, Beijer B, Garcia Boy R, Nothelfer EM, Leotta K, Eisenhut M et al. The pharmacokinetics of cell-penetrating peptides. Mol Pharm 2010; 7: 2224–2231. 130 Boado RJ, Zhang Y, Xia CF, Wang Y, Pardridge WM. Genetic engineering of a lysosomal enzyme fusion protein for targeted delivery across the human blood- brain barrier. Biotechnol Bioeng 2008; 99: 475–484. Blood–brain barrier drug delivery WM Pardridge 1972 Journal of Cerebral Blood Flow & Metabolism (2012), 1959 – 1972 & 2012 ISCBFM