ArticlePDF Available

Pharmacodynamic Analysis of Magnetic Resonance Imaging-Monitored Focused Ultrasound-Induced Blood-Brain Barrier Opening for Drug Delivery to Brain Tumors

Wiley
BioMed Research International
Authors:

Abstract and Figures

Microbubble-enhanced focused ultrasound (FUS) can enhance the delivery of therapeutic agents into the brain for brain tumor treatment. The purpose of this study was to investigate the influence of brain tumor conditions on the distribution and dynamics of small molecule leakage into targeted regions of the brain after FUS-BBB opening. A total of 34 animals were used, and the process was monitored by 7T-MRI. Evans blue (EB) dye as well as Gd-DTPA served as small molecule substitutes for evaluation of drug behavior. EB was quantified spectrophotometrically. Spin-spin (R1) relaxometry and area under curve (AUC) were measured by MRI to quantify Gd-DTPA. We found that FUS-BBB opening provided a more significant increase in permeability with small tumors. In contrast, accumulation was much higher in large tumors, independent of FUS. The AUC values of Gd-DTPA were well correlated with EB delivery, suggesting that Gd-DTPA was a good indicator of total small-molecule accumulation in the target region. The peripheral regions of large tumors exhibited similar dynamics of small-molecule leakage after FUS-BBB opening as small tumors, suggesting that FUS-BBB opening may have the most significant permeability-enhancing effect on tumor peripheral. This study provides useful information toward designing an optimized FUS-BBB opening strategy to deliver small-molecule therapeutic agents into brain tumors.
This content is subject to copyright. Terms and conditions apply.
Hindawi Publishing Corporation
BioMed Research International
Volume , Article ID ,  pages
http://dx.doi.org/.//
Research Article
Pharmacodynamic Analysis of Magnetic Resonance
Imaging-Monitored Focused Ultrasound-Induced Blood-Brain
Barrier Opening for Drug Delivery to Brain Tumors
Po-Chun Chu,1Wen-Yen Chai,1,2 Han-Yi Hsieh,1Jiun-Jie Wang,3Shiaw-Pyng Wey,3
Chiung-Yin Huang,4Kuo-Chen Wei,4and Hao-Li Liu1
1DepartmentofElectricalEngineering,Chang-GungUniversity,259Wen-Hwa1stRoad,Kwei-Shan,Tao-Yuan333,Taiwan
2Department of Diagnostic Radiology, Chang-Gung University and Memorial Hospital, 5 Fu-shin Street, Kwei-Shan,
Tao-Yuan 333, Taiwan
3Department of Medical Imaging and Radiological Sciences, Chang-Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan,
Tao-Yuan 333, Taiwan
4Department of Neurosurgery, Chang-Gung Memorial Hospital, 5 Fu-shin Street, Kwei-Shan, Tao-Yuan 333, Taiwan
Correspondence should be addressed to Hao-Li Liu; haoliliu@mail.cgu.edu.tw
Received  January ; Accepted  February 
Academic Editor: Fan-Lin Kong
Copyright ©  Po-Chun Chu et al. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Microbubble-enhanced focused ultrasound (FUS) can enhance the delivery of therapeutic agents into the brain for brain tumor
treatment. e purpose of this study was to investigate the inuence of brain tumor conditions on the distribution and dynamics
of small molecule leakage into targeted regions of the brain aer FUS-BBB opening. A total of  animals were used, and the
process was monitored by T-MRI. Evans blue (EB) dye as well as Gd-DTPA served as small molecule substitutes for evaluation of
drug behavior. EB was quantied spectrophotometrically. Spin-spin (R1) relaxometry and area under curve (AUC) were measured
by MRI to quantify Gd-DTPA. We found that FUS-BBB opening provided a more signicant increase in permeability with small
tumors. In contrast, accumulation was much higher in large tumors, independent of FUS. e AUC values of Gd-DTPA were well
correlated with EB delivery, suggesting that Gd-DTPA was a good indicator of total small-molecule accumulation in the target
region. e peripheral regions of large tumors exhibited similar dynamics of small-molecule leakage aer FUS-BBB opening as
small tumors, suggesting that FUS-BBB opening may have the most signicant permeability-enhancing eect on tumor peripheral.
is study provides useful information toward designing an optimized FUS-BBB opening strategy to deliver small-molecule
therapeutic agents into brain tumors.
1. Introduction
Focused ultrasound beams (FUS) in the presence of cir-
culating microbubbles can temporarily open the blood-
brain barrier (BBB opening) of capillaries in the central
nervous system (CNS) parenchyma []. Bursts of acoustic
ultrasound induce microbubble cavitation in the vasculature,
and the resultant shear stress temporarily disrupts tight
junctions to enhance blood-brain permeability. is BBB-
opening process can be carried out at moderate acoustic
pressures to minimize adverse eects on vascularture and
prevent damage to neurons [], while facilitating localized
delivery of chemotherapeutic agents from the vasculature to
the pathological brain parenchyma and CNS []. Since
more than % of the therapeutic agent normally cannot pen-
etrate CNS tight junctions [],thisnovelapproachprovides
a unique opportunity for local delivery of therapeutic agents
across the BBB and into the targeted site, thus opening a new
frontier of CNS drug delivery.
Brain tumors could potentially be treated by FUS-BBB
opening to enhance chemotherapeutic agent delivery. In
the United States, at least , patients are diagnosed
with glioblastoma multiforme (GBM) each year, comprising
more than half of the malignant primary brain tumors [],
BioMed Research International
Function
generator
Power
amplier
Power
meter
Targeted
position
Animal
FUS
transducer
(400 kHz)
Ultrasound
focal beam
Disgassed
water
F : Schematic showing the experimental setup of the focused ultrasound exposure system.
and chemotherapy is an important treatment modality [].
Recent preclinical studies showed that FUS-BBB opening
can eectively enhance local deposition and concentration of
chemotherapeutics including BCNU [], liposomal doxoru-
bicin [,], and chemodrugs carried by novel nanocarriers
[].
Currently, FUS-mediated CNS drug delivery is moni-
tored by magnetic resonance imaging (MRI) by intravenous
(IV) injection of gadolinium diethylenetriamine penta-acetic
acid (Gd-DTPA) contrast agent together with chemother-
apeutic drug. e MRI signal intensity increase caused by
the leakage of Gd-DTPA thus serves as an indicator to
estimate drug concentration [,,,]. Most studies have
focused on analyzing the eects of FUS exposure parameters
such as acoustic pressure amplitude, ultrasound frequency,
pulse length, pulse repetition frequency, exposure duration,
and microbubble dose on BBB opening [,]. How-
ever, pharmacodynamic analysis including the dynamics and
distribution of the specic molecular agent in the brain
is critical for evaluating specic drug delivery. MRI could
also be used for pharmacological endpoint evaluation using
concurrently administered MR contrast agents as surrogate
indicators of therapeutic drug concentrations. Although the
kinetics of contrast agent permeability of a defective blood-
brain barrier have been measured using MR compartment
modeling [], these studies were performed in normal
animal brains, and so far the detailed pharmacodynamic
behavior of contrast agents aer FUS-BBB opening remains
uncertain in brain tumors.
e purpose of this study was to conduct an MRI
pharmacodynamic analysis of FUS-BBB opening in brain
tumors in an animal model. Injected Gd-DTPA contrast
agent was used to characterize pharmacodynamic changes
as a function of time aer BBB opening, in both normal
andbrain-gliomaanimals.Wealsoattemptedtoestablish
the correlation between deposition of Gd-DTPA by in vivo
semiquantication and the quantitation of another surrogate,
Evans blue dye, by spectrophotometry aer sacrice. Finally,
we evaluated the pharmacodynamic changes aected by FUS-
BBB opening in various grades of gliomas.
2. Methods
2.1. FUS Setup. A focused ultrasound transducer was used
to generate ultrasound focal energy (IMASONIC, France;
diameter =  mm, radius of curvature =  mm, frequency =
 kHz, and electric-to-acoustic eciency = %) (Figure ).
An arbitrary function generator (A, Agilent, Palo Alto,
CA and DS, Stanford Research Systems, Sunnyvale, CA)
was used to generate the driving signal, which was then fed
into a radiofrequency power amplier (AB, Amplier
Research, Souderton, PA). e focal zone distribution of
the intensity of the ultrasound eld was measured in an
acrylic water tank lled with deionized, degassed water. e
measured diameter of the half-maximum pressure amplitude
wasmm,andthelengthoftheproducedfocalzonewas
 mm. Animals underwent isouorane anesthesia before
ultrasound treatment. e animal was laid prone and placed
directly under an acrylic water tank (with a window of
×cm
2at its bottom sealed with a thin lm to allow
entry of the ultrasound energy), using ultrasound gel to
ll the interspaces between the animal head and the thin-
lm window. SonoVue SF-coated ultrasound microbub-
bles (– 𝜇mmeandiameter,.𝜇L/kg; Bracco Diagnos-
tics Inc.) were IV administered by burst injection with
. mL of saline solution containing .mL heparin. Aer
injecting the microbubbles, burst-tone mode ultrasound at
a pressure of . MPa (peak negative value; measured in
the free-eld) was delivered to the brain with the center
of the focal zone positioned at a penetration depth of -
mm under the scalp (burst length =  ms, pulse repe-
tition frequency =  Hz, and total sonication duration =
 s).
BioMed Research International
I: before ∗∗ : group 2
animals only
Sacrice
Implant FUS
Intervention
Gd Gd Gd∗∗
IV inject
MRI
10 or 17 d 4 h
T
(a) (b)
(c)
1400
700
0
MBs
and EB
80 min
10 min 20 min 40 min 80 min
T1/R1/T2T1/R1/T2T1/R1/T∗∗
2
II: aer,
10 min
III: aer,
2 h∗∗
F : (a) Experimental protocol for st- and nd-group animal experiments. MRI images were acquired in time slots I/II for group
 animals and in time slots I/II/III for group  animals; double asterisks (∗∗) indicate group- experiments only. (b) FUS-exposed brain
of a normal animal with Evans blue extravasations to identify the location of BBB opening. (c) R1accumulating map showing Gd-DTPA
accumulation in the BBB-opening location of a normal animal over time.
2.2. Animal Experiment Design. All animal experiments
were approved by the Institutional Animal Care and Use
CommitteeofChangGungUniversityandadheredtothe
experimental animal care guidelines. A total of  animals
(male Sprague-Dawley rats (– g)) were used, including
normal (𝑛=18) and tumor animals (𝑛=16). Experi-
ments were divided into two groups. In group , the aim
was to conrm the correlation between Gd-DTPA leakage
(concentration measured by relaxometry) and Evans blue
(EB) dye (concentration measured spectrophotometrically)
aer FUS-BBB treatment. Subgroups included () normal rats
(𝑛=18) and () tumor rats (𝑛=4), and the rst subgroup
underwent FUS-BBB opening. Subgroups were conrmed
by dynamic contrast-enhanced (DCE) MRI with Gd-DTPA
(molecular weight =  Da). In addition, EB dye (molecular
weight =  Da) was IV injected into the animals, and
the amount of EB deposited in the brain was quantied
spectrophotometrically (procedure described below). In the
rst subgroup of group , contrast-enhanced T1-weighted
imaging was rst performed to estimate Gd-DTPA concen-
tration aer BBB opening, followed by T2-weighted imaging
to provide a reference of tumor morphology. e second
subgroup underwent the same scanning process without FUS
induced.
In experimental group , our aim was to monitor the
increase in Gd-DTPA accumulation in tumor-bearing ani-
mals aer conducting FUS-BBB opening. Animals were
divided into two subgroups: () animals receiving FUS
exposure  days aer tumor implantation (tumor volume
typically <. cm3) with FUS-BBB opening (𝑛=6)and
() animals receiving FUS exposure  days aer tumor
implantation (tumor volume typically >. cm3) with FUS-
BBB opening (𝑛=6). Tumor volume was measured by
T2-weighted MRI. In group , animals were subjected to
three -minute-long MR relaxometry-based imaging ses-
sions (before FUS exposure, and -min and -min aer
FUS exposure). Detailed experimental procedures are shown
in Figure .
2.3. Rat Brain Glioma Model. C glioma cells were harvested
by trypsinization and cultured at a concentration of  ×
5cells/mL for implantation. For intracranial injection into
the striatum of rat brains, cells were washed once with
phosphate buered saline (PBS). Male Sprague-Dawley rats
(– g) were anesthetized by intraperitoneal admin-
istration of ketamine (mg/kg) and immobilized on a
stereotactic frame. A sagittal incision was made through the
BioMed Research International
skin overlying the calvarium, and a small dental drill was used
to make a hole in the exposed cranium, . mm anterior and
mmlateraltothebregmaonthelesideofskull.Ccell
suspension ( mL) was injected at a depth of . mm from
the brain surface. e injection was performed over a -
minute period, and the needle was withdrawn over another
 minutes. Ten days aer implantation, tumor sizes were
measured by MRI.
2.4. Spectrophotometric Quantitation of Evans Blue Dye. EB
dye (% in saline) was IV injected ( mg/kg), and the
animalsweresacricedtwohourslater.Allanimalswererst
deeply anesthetized with % chloral hydrate and infused
with heparinized saline through the cardiac ventricle until
colorless infusion uid was obtained from the atrium. Aer
the rats had been sacriced by decapitation, the hemispheres
of the brain were separated along the transverse suture. en
both hemispheres were weighed and placed in formamide
( mL/ mg) at C for  h. e sample was centrifuged
for  mins at , rpm. e concentration of dye extracted
from each brain was determined spectrophotometrically at
 nm and was compared with a standard graph created
by recording optical densities from serial dilutions of EB in
.% sodium chloride solution. e EB tissue content was
quantied using a linear regression standard curve derived
from seven concentrations of the dye.
2.5. MRI. For in vitro measurements, Gd-DTPA (Omniscan,
. mL/kg, Magnevist) was diluted with physiological saline
to ., ., ., ., ., and . 𝜇M. Circular wells (inner
diameter =  mm) were lled with  𝜆of contrast agent
sample or physiological saline as control and were placed
in the MR scanner (Clinscan, Bruker, Germany;  Tesla).
Spin-lattice relaxivity maps were calculated from two T1-
weighted images with dierent ip angles (gradient recalled
echo sequence, TR/TE = . ms/. ms, slide thickness =
. mm, matrix =  ×, and ip angle = /). e
correlation between R1(= /T1)mappingandGd-DTPA
concentration was determined [].
In the animal experimental group, FUS-induced BBB
opening was monitored by MRI with a -Tesla magnetic reso-
nance scanner (Bruker ClinScan, Germany) and a -channel
surface coil. e mouse was placed in an acrylic holder,
positioned in the center of the magnet, and anesthetized with
isourane gas (-%) at – breaths/min during the entire
MRI procedure.
In the rst experimental group, the distribution and
dynamics of Gd-DTPA leakage were investigated imme-
diately aer conducting FUS-BBB opening. Aer FUS-
BBB opening, animals were immediately relocated into the
MR scanning room, and contrast-enhanced T1-weighted
images with dierent ip angles were acquired to calcu-
late spin-lattice relaxivity maps by transferring two images
with dierent ip angles (gradient recalled echo sequence,
TR/TE = . ms/. ms, slice thickness = . mm, slice
number=,matrix=×, and ip angle = /).
Images were sequentially acquired over  min with a time
interval of  seconds for area under the curve (AUC)
calculation. Upon completion of the th acquisition, a
diluted bolus of Gd-DTPA was IV injected through a
catheter at an infusion rate of  mL/s. In the second
experimental group, three sets of Gd-DTPA-leakage dis-
tribution/dynamics were investigated, including (I) before
FUS exposure, (II) immediately aer FUS exposure, and
(III) two hours aer FUS exposure. Immediately aer
conducting FUS-BBB opening, turbo spin echo (TSE) T2-
weighted images were obtained as a reference to identify
the tumor region (repetition time (TR)/echo time (TE) =
/ ms, FOV =  × mm2,in-planeresolution=
. ×. mm2, and slice thickness = . mm).
2.6. MR Analysis of Gd-DTPA Accumulation and Distribution
aer FUS-BBB Opening. In R1-map analysis, a region of
interest (ROI) was selected and compared with the non-
enhanced contralateral brain to determine the increase in
Gd-DTPA concentration caused by BBB opening. AUC maps
were then transferred from a series of time-dependent R1
maps (up to  min) to determine pharmacodynamic char-
acteristics of Gd-DTPA for comparison with the dynamics of
EB dye permeability. us, the total area (AUC) is given by
the following equation:
AUC80 min =∫𝐶𝑝𝑡⋅𝑑𝑡
𝑉,()
where 𝐶𝑝𝑡 are vertical segments under the Gd-DTPA con-
centration curve area and 𝑉is total ROI volume.
In experimental group , ROIs were selected in the tar-
geted tumor area which was based on the tumor dimensions
denedinT
2images (the same ROI as in the contralateral
brain was selected). e distribution and dynamics of Gd-
DTPA leakage were evaluated for dierent tumor sizes
including  days aer implantation (typically <. cm3)
and  days aer implantation (typically >. cm3)andwere
divided by the tumor dimension. ROI including the entire
tumor and the contralateral area were selected. Moreover,
in order to evaluate the homogeneity of Gd-DTPA leakage,
tumors with dimension >. cm3were further divided into
tumor core (inner half of the area) and tumor peripheral
(outer half of the area), based on T2images.
2.7. Histology. Albumin-bound EB dye was IV injected as
a bolus immediately aer sonication. BBB opening was
quantied as extravasation of EB. Tumor model animals
were sacriced about  h aer sonication and MR scanning.
Brain samples were serially sectioned ( 𝜇mthickness)using
the same slice direction as in MRI analysis. Representative
sections were stained with hematoxylin and eosin (HE).
Tumor morphology was histologically evaluated.
3. Results
BBB opening was clearly evidenced by staining with EB
dye. A typical image of a normal BBB-opened brain stained
with EB dye is shown in Figure (b). In addition, a series
BioMed Research International
100
80
60
40
20
0
1
2
3
4
5
6
7
8
(a)
20
15
10
5
0
−5 012345
Slope =3.2936
𝑦=3.2936𝑥 − 0.1446
𝑅2= 0.9991
Gd-DTPA (𝜇M)
R1(s1)
(b)
0.25
0.2
0.15
0.1
0.05
000.2 0.4 0.6 0.8 1
Evans blue (𝜇g)
𝑅2= 0.9992
OD (620 nm)
𝑦=0.1945𝑥 + 0.0353
(c)
F : (a) T1image and corresponding R1map for the in vitro Gd-DTPA phantom at increasing concentrations ( and : water; : . 𝜇M;
: . 𝜇M; : . 𝜇M; : . 𝜇M; : . 𝜇M; : . 𝜇M). (b) Dependence of R1on Gd-DTPA concentration; relaxivity was estimated as about
. s−1 mM−1. (c) Calibration curve of spectrophotometrically determined EB concentration. EB concentrations ranging from  to .𝜇g
were tested, resulting in O.D. readings of . to . at nm.
R1
Before FUS Aer FUS Before FUS Aer FUS Before FUS Aer FUS
NormalDay-10Day-17
1400
700
0
1400
700
0
1400
700
0
2
1
0
2
1
0
2
1
0
(a) (b) (c) (d)
AUC80 min
CE-T1T2
F : Typical MRI for normal animals (upper) as well as animals aer -day (middle) and -day (bottom) tumor implantations. (a)
Contrast-enhanced T1images before and aer FUS exposure. (b) T2images (aer FUS). (c) R1maps before and aer FUS exposure. (d) Area
under the R1curve over  minutes (denoted as AUC80 min) before and aer FUS exposure.
of R1maps obtained at dierent time points aer FUS-
BBB opening demonstrated the dynamic change in Gd-
DTPA accumulation in a normal brain, with particularly high
leakage at the sonication site (Figure (c)).
R1relaxivity of Gd-DTPA and ELISA measurements of
EB dye concentration were calibrated in vitro. e detected
R1-signal increased in a highly linear manner with Gd-DTPA
concentration (input concentrations of , ., ., , , and
𝜇M) as shown by the calibration curve (𝑟2= 0.9991)
(Figure (b)). e relaxivity of Gd-DTPA contrast agent was
found to be . at  Tesla. e detected ELISA signal also
increased in a highly linear manner with EB concentration
(Figure (c);𝑟2= 0.9992). ese calibration curves thus
allowed precise quantitation of Gd-DTPA and EB deposition
in the brain.
FUS-induced BBB opening was veried by CE-MRI.
Typ i cal CE-T1images, T2images, R1maps, and AUC maps in
normal, -day glioma, and -day glioma animals are shown
in Figure . In normal animals, the BBB-opened area was
clearly visible in T1-weighted images. T2-weighted images did
not show any evidence of FUS-induced damage at the target
location at pressure amplitudes of . MPa (Figure ). In the
BioMed Research International
Normal
2
1
0
010 20 40 60 80
0.8
0.6
0.4
0.2
0
Time (min)
Concentration (𝜇M)
R1(s1)
Target, w/o FUS
Target, with FUS
Contralateral
(a)
2
1
0
010 20 40 60 80
0.8
0.6
0.4
0.2
0
Time (min)
Concentration (𝜇M)
R1(s1)
Contralateral
Tumor, 10 d
Tumor, w/o FUS
Tumor, with FUS
(b)
2
1
0
010 20 40 60 80
0.8
0.6
0.4
0.2
0
Time (min)
Concentration (𝜇M)
R1(s1)
Contralateral
Tumor, 17 d
Tumor, w/o FUS
Tumor, with FUS
(c)
Target, w/o FUS
Target, with FUS
Contralateral
5500
3500
1500
0
1200
800
400
0
R1accumulation
Gd accumulation (𝜇M)
010 20 40 60 80
Time (min)
Normal
(d)
Contralateral
5500
3500
1500
0
1200
800
400
0
R1accumulation
Gd accumulation (𝜇M)
010 20 40 60 80
Time (min)
Tumor, 10 d
Tumor, w/o FUS
Tumor, with FUS
(e)
Contralateral
5500
3500
1500
0
1200
800
400
0
R1accumulation
Gd accumulation (𝜇M)
010 20 40 60 80
Time (min)
Tumor, 17 d
Tumor, w/o FUS
Tumor, with FUS
(f)
F : (a)–(c) R1change as a function of time before and aer FUS exposure for normal animals, -day and -day tumor animals. (d–f)
e corresponding R1accumulation as a function of time over  minutes for (a)–(c).
rst subgroup of experimental group , the R1-map signal of
the BBB-opening area was increased from . to about . by
FUS, and AUC maps showed an increase in accumulation of
Gd-DTPA deposition from  to about .
In tumor-bearing animals, the FUS-induced BBB area
clearly covered the tumor tissue (Figure ;small(-day)or
large (-day) tumors). Sonication resulted in increased Gd-
DTPA accumulation in the tumor and in the peripheral BBB-
opened area as evidenced by signal enhancement in the R1-
map images. AUC maps showed maintained high staining
intensities aer Gd-DTPA injection. In the small (-day)
tumors, FUS resulted in an increase of R1signal from  to
about . s−1 andanincreaseinGd-DTPAdepositionofabout
 (from  to ). However, in large (-day) tumors,
FUS did not lead to a signicant change in the R1-signal,
which increased from . to  s−1,ortheAUCvaluewhich
increased by only about  (from  to ).
e kinetics of Gd-DTPA accumulation were evaluated
aer a single sonication treatment in thirty animals (normal
rats: 𝑛=18; small-tumor model: 𝑛=6;large-tumor
model: 𝑛=6). An ROI from the BBB-opened area on T2-
weighted images (target) and the corresponding ROI from
the contralateral brain (contralateral) were used to infer Gd-
DTPA concentration from the R1signals and the AUC over
time in the -day tumors (volume <. cm3), -day tumors
(volume >. cm3), or normal controls aer sonication
(Figure ). Figures (a)(c) showed the comparison of
changes in R1as a function of time (from to  min)
for three typical animals. When considering the peak value
over the whole scanning process, FUS caused the highest
enhancement in R1signal of contrast agent in normal tissue
(from . to .; Figure (a)). Sonication also led to a large
increase in R1signal in -day tumor, from . to . s−1
(Figure (b)). However, the already high permeability of -
day tumor to Gd-DTPA was not signicantly increased by
FUS, from . to . s−1 (Figure (c)).
e corresponding AUC (accumulation of R1)asa
function of time (from  to  min) in these three animals is
showninFigures(d)(f). In the normal animal, total Gd-
DTPA accumulation in the BBB-opening area was increased
from . to . pmol by FUS (Figure (d)). Gd-DTPA
accumulation in the -day tumor increased from . to
. pmol, compared to only about  pmol over time in
the contralateral control hemisphere (Figure (e)). However,
BioMed Research International
2.5
2
1.5
1
0.5
00 80 0 80 0 80
0.8
0.6
0.4
0.2
0
Time aer FUS (min)
Contralateral
FUS site
Normal
Gd-DTPA concentration (𝜇M)
R1(s1)
Tumor, 10 d
Tumor, 17 d
(a)
Time aer FUS (min)
Normal
Ratio to control
8
6
4
2
0080
Tumor, 10 d
Tumor, 17 d
(b)
0
Contralateral
FUS site
Normal
5500
3500
1500
1200
800
400
0
Concentration by AUC (𝜇M)
R1AUC
Tumor, 10 d Tumor, 17 d
(c)
F : (a) Instantaneous R1measured immediately or  minutes aer FUS exposure in normal animals, -day and -day tumor animals.
(b) Ratio of instantaneous R1values, immediately and  min aer FUS. (c) Corresponding R1AUC of (a). Images were acquired in time slot
II.
Gd-DTPA accumulated to about the same high value with
and without FUS (. versus . pmol) in the -day
tumor (Figure (f )).
Gd-DTPA levels on the FUS-treated and contralateral
side were also evaluated at either min or  min aer a
single sonication treatment (Figure ). e Gd-DTPA signal
intensity increased in the BBB-opening area min aer
sonication as evidenced by an increase in the R1signal from
. to . s−1 for control and from . to . s−1 for
small-tumor animals. However, the contrast agent signals
in the FUS-enhanced large tumors were the same as for
the contralateral region (about . s−1). All R1signals in
these brain tissues returned to baseline (about . s−1)at
 min aer sonication. e ratio of R1between the FUS-
exposed and control areas went from . s−1 at  min to
. s−1 atmininnormaltissueandfromabout.to
s
−1 for the tumor-bearing animals (Figure (b)). e AUC
at  min and  min was compared between control and
BBB-opened brain regions. At  min aer sonication, the
accumulated Gd-DTPA concentration of the BBB-opened
area increased to . 𝜇M, compared to . 𝜇Mforthe
smalltumorandalimitedincreaseto𝜇Mforthelarge
tumor (Figure (c)).
Next we evaluated the correlation between EB leakage
(Figure (a)) and Gd-DTPA accumulation (estimated by
the AUC) in the same region of the brain. We found that
the accumulated distribution of Gd-DTPA (i.e., AUC) was
highly correlated with the distribution of EB (𝑟2= 0.8897)
(Figure (b)). us, R1-based pharmacodynamic analysis
providedareasonablemapofthepermeabilityoftheBBB-
disrupted region to EB dye over time. EB dye and Gd-
DTPA accumulation showed the same tendency of higher
overall permeability in tissues of large tumors and less depen-
dence on FUS treatment, as evidenced from the ratios of
accumulation between contralateral and FUS-treated regions
(Figure (c)).
Next, we analyzed Gd-DTPA deposition dynamics in
experimental group  animals by AUC analysis for three
individual time slots: (I) before FUS, (II) immediately aer
FUS, and (III)  hours aer FUS. Gd-DTPA concentration
BioMed Research International
Normal BBB group
Normal tumor group
Contralateral
FUS site
Tumo r s ite
Evans blue (𝜇M)
5
4
3
2
1
0
10 200 400 600 800 1000
Gd-DTPA (𝜇M)
𝑦 = 0.0051𝑥 − 0.3963
𝑟2=0.8897
(a)
Evans blue (𝜇M)
12
10
8
6
4
2
0
Normal, contra
Normal, FUS
Normal, FUS
Tumor, 10 d
Tumor, 17 d
Normal
Tumor, 10 d
Tumor, 17 d
(b)
Normal, FUS
8
6
4
2
0
102468
Ratio of Gd-DTPA
Ratio of Evans blue
Tumor, 10 d
Tumor, 17 d
(c)
F : (a) Correlation between quantied Evans blue concentration and R1-estimated Gd-DTPA concentration in normal (red circle) and
tumor (blue triangle) brains. Le arrow: contralateral side and right arrow: with FUS (red circle); normal tumor (blue triangle). (b) Evans
blue aer FUS exposure in the normal contralateral region, normal FUS targeting region, -day tumor region aer FUS, and -day tumor
region aer FUS. (c) Correlation of the ratios of FUS: contralateral concentrations of Evans blue and Gd-DTPA in normal and tumor-bearing
brains.
BioMed Research International
2.5
2
1.5
1
0.5
0
0.8
0.6
0.4
0.2
0
I/ before
FUS
II/ immediately
aer FUS
Normal
Gd-DTPA (𝜇M)
R1(s1)
Tumor, 10 d
Tumor, 17 d
III/ 2 hours
aer FUS
(a)
10
7
4
1
Ratio
I/ before
FUS
II/ immediately
aer FUS
Normal
Small tumor
Large tumor
III/ 2 hours
aer FUS
(b)
5500
3500
1500
0I/ before
FUS
II/ immediately
aer FUS
Normal
Gd-DTPA accumulation (𝜇M)
1200
800
400
0
R1AUC
Tumor, 10 d
Tumor, 17 d
III/ 2 hours
aer FUS
(c)
10
7
4
1
I/ before
FUS
II/ immediately
aer FUS
Ratio
Normal
Small tumor
Large tumor
III/ 2 hours
aer FUS
(d)
F : (a) Instantaneous R1values of the normal animals, -day and -day tumor animals at MRI acquisition time slots I, II, and III. (b)
Ratio of instantaneous R1to the value in time slot I. (c) Corresponding R1AUCof(a).(d)RatioofR
1AUCtothevalueintimeslotI.
and accumulation in the target area presented the same trend
at all three time points (Figure ). As before, we observed a
transient peak of R1andtheAUCintheBBB-openedbrain
just aer sonication (Figures (a) and (c)). However, the
increase in the ratio of R1or AUC again diered between
the normal and tumor-bearing animals (Figures (b) and
(d)). e ratio was highest in the normal BBB-opening
area (R1signal . times than before FUS; AUC: . times),
followed by the small-tumor model (R1signal: . times;
AUC: . times), with no signicant change in the large
tumor, conrming our previous observations that FUS did
not signicantly further aect permeability in large tumors.
ese ratios subsequently decreased at the time point  hours
aer FUS induction, returning to approximately the same
values of DCE-MRI as originally observed before sonication.
is result implied that at  hours, the BBB-opening area had
recovered to the same baseline permeability level to contrast
agent as prior to sonication.
HE staining of tumors  days aer implantation showed
even staining without scattered red blood cells in the absence
of FUS (Figures (a) and (b)). Tumor cells were charac-
terizedbydensenucleardistribution,andonlytinyareas
of gliosis inltrated with chronic inammatory cells and
some hemorrhaging were found (Figures (c) and (d)).
HE staining of tumors  days aer implantation revealed
a number of regions with extensive apoptosis and cavities
inthecoreofthetumor,andhemorrhagicstructureswith
scattered and spreading erythrocytes could be observed
 BioMed Research International
(a) (b) (c) (d)
(e) (f) (g)
100 𝜇m
(h)
F : HE staining. (a) and (b) Small (-day) tumor tissue without FUS exposure, x and x. (c) and (d) Small (-day) tumor tissue
with FUS exposure, x and x. (e) and (f) Large (-day) tumor tissue without FUS exposure, x and x. (c) and (d) Large (-day)
tumor tissue with FUS exposure, x and x.
5500
3500
1500
0
1200
800
400
0
Before
FUS
Immediately
aer FUS
Gd-DTPA concentration (𝜇M)
R1AUC
10-day tumor
17-day tumor
17-day tumor, peripheral
17-day tumor, core
2 hours
aer FUS
(a)
Immediately
aer FUS
R1concentration increase (%)
100
80
60
40
20
0
10-day tumor
17-day tumor
17-day tumor, peripheral
17-day tumor, core
2 hours
aer FUS
(b)
F : (a) R1AUC analysis of the core and peripheral subregions of -day tumor. (b) R1AUCincreaserelativetotimeslotI.
around discontinuous vasculature (Figures (e) and (f)),
supporting our ndings of high permeability of -day tumors
based on observation of Gd-DTPA deposition. is structure
did not change signicantly aer sonication (Figures (g) and
(h)) with hemorrhagic regions remaining similar to those in
the unexposed tumor.
Previous reports showed that the tumor core consists
of a bulky necrotic mass without functional vasculature,
whereas the tumor periphery maintains a high degree of
vasculature structure [,]. We therefore hypothesized that
microbubble-enhanced FUS exposure would have a bigger
eect on enhancing permeability in the peripheral tumor.
We further divided the -day tumor animals into core
and peripheral subregions and then repeated the MRI AUC
analysis (Figure ). We observed that, aer FUS exposure,
the -day peripheral tumor showed a similar trend to the -
day tumor, which showed a nearly .-fold of instantaneous
increase and % accumulation increase in Gd-DTPA, and
the permeability dropped signicantly two hours aer FUS
exposure to about half (%), which is similar to -day
tumor. In contrast, the -day tumor core mimicked the
behavior of the undivided -day tumor, accumulating high
BioMed Research International 
levels of Gd-DTPA both before and aer FUS exposure
(AUC increase of only % aer FUS exposure) (Figure ).
is relatively low increase in the AUC persisted when the
tumor core was re-evaluated at  hours aer FUS exposure
(%). ese observations suggest that the FUS-BBB opening
can provide the most pronounced drug delivery enhancing
eect on tumor peripheral or tumors with high-vascularity
stagetumors,yetonlyprovideslimitedeectsonbulkyand
necrotic tumors.
4. Discussion
is study demonstrated the pharmacodynamic characteris-
ticsofsmall-moleculeleakageatvariousstagesoftumorsaer
application of microbubble-enhanced FUS to open the BBB.
We analyzed two small molecules with similar molecular
weights to obtain complimentary data on pharmacodynamic
behavior. Gd-DTPA was used to provide contrast in MRI and
for semiquantitative verication of biodistribution in vivo,
andEBdyewasusedasameasureofdrugaccumulation
aer animal sacrice. ese two molecules, which normally
do not enter the brain parenchyma from the bloodstream,
could potentially be used as surrogate markers for drug
delivery. Although the dynamic distribution of Gd-DTPA
may dier from that of Evans blue, we demonstrated that the
AUC accumulation of Gd-DTPA analyzed by MRI was highly
correlated with EB accumulation in the brain (𝑟2= 0.8897),
implying that MRI AUC analysis of Gd-DTPA could predict
the concentration of EB accumulating in the brain, and may
thus have the potential to predict the pharmacodynamic
behavior and biodistribution of other therapeutic agents.
is study employed high-temporal-resolution dynamic
CE-MRI that could be utilized for small-molecule in vivo
distribution and semiquantication, as attempted in previous
studies [,]. e unique features provided by dynamic
CE-MRI include the capability of rapid evaluation and high
spatial resolution, as well as kinetic analysis to evaluate tumor
perfusion []. Positron emission tomography (PET) has
also been used for pharmacological studies in several tumor
types [,]. However, potential limitations of PET may
include its limited imaging resolution and complexity of
radiotracer synthesis. In small tumors, partial volume eects
may be signicant if the tumor size is less than twice the
resolution of the scanner []. MRI methods provide the
advantageofhavinggoodspatialresolutionequaltothat
of corresponding morphologic images. In addition, MRI
isminimallyinvasiveandposeslittlerisktopatients.We
used voxels of about . ×. ×mm
3to construct
images sucient for small-animal analysis. On the other
hand, PET relies on radiolabeled molecules that bind to
receptors to allow absolute quantication by detection of
isotopes. PET may also be limited by its high cost, limited
availability of radiotracer, and the need for a cyclotron as
well as onsite radiochemistry for radioisotope production
[]. PET involves comprehensive conjugation of radiotracers
and specic tailor-made molecules limiting its general use
for pharmacodynamic analysis. Although Gd-DTPA can-
not be directly conjugated to therapeutic molecules, the
detection of coadministered Gd-DTPA by CE-MRI is highly
correlated with targeted molecules, providing an excellent
tool for monitoring vasculature and evaluating tissue/tumor
permeability at high temporal/spatial resolution, suggesting
its continued usefulness for pharmacodynamic analysis of
brain drug delivery.
Tumor tissues are known to have high permeability due
to the presence of large endothelial cell gaps, incomplete
basement membrane, and the relative lack of pericyte or
smooth muscle association with endothelial cells [,].
In addition, the network of vasculature in solid tumors is
markedly dierent from the normal hierarchical branching
patterns and contains leaky vessel structures. Variations in
permeabilityarealsoassociatedwiththetumorgradeaswell
as various neoplastic eects that could disrupt the BBB [].
In this study, we observed that tumors with dierent levels of
progression showed dierent characteristics of blood-vessel
permeability and small-molecule accumulation. We found
that the AUC80 min in small tumors was 452 ± 122.5𝜇M,
whereas in large tumors it reached 754 ± 48.3𝜇M. More-
over, we conrmed that FUS-BBB opening provided a %
enhancement of accumulation of Gd-DTPA in small brain
tumors and a % enhancement at the large tumor periphery,
implying that FUS-BBB opening is an eective approach
to increase brain-tumor permeability and therefore enhance
delivery of therapeutic molecules.
Histological examination by HE staining showed that
smaller (-day) tumors had well-ordered vasculature with
fewer abnormal endothelial cell gaps (Figures (a) and (b)).
e blood vessel density in small tumor was lower, resulting
in less Gd-DTPA and EB accumulation. In contrast, -
day tumor tissues contained more large fenestrae (Figures
(e) and (f)), consistent with previous pathological ndings
that high-grade brain tumors contain neovasculature and
apoptotic tumor cells, leading to hyperpermeability [].
ese pathological changes are consistent with the increased
Gd-DTPA and EB accumulation that we observed in -day
tumor tissues before sonication.
Although AUC80 min correlated well with the pharmaco-
dynamic behavior of another small molecule (EB), Gd-DTPA
accumulation can be very dierent even under the same
FUS exposure conditions, for example, varying from .
to  𝜇M(Figure(b)).eselargevariationswerelikely
due to dierences in skull thickness and angle of incidence
between the FUS beam and the skull surface among the
animals [], and the presence of standing waves produced
in the skull cavity that alter the peak pressure at the target
position and thus the level of BBB opening []. Since FUS-
BBB opening may vary substantially, it is essential to perform
an AUC analysis during CE-MRI to monitor small-molecule
delivery into the brain for individual subjects and targets.
5. Conclusion
In this study, we characterized the dynamics of BBB opening
in normal and tumor tissues using DCE-MRI with Gd-
DTPA contrast agent, and related them to the concentrations
of Evans Blue determined from tissues aer sacrice. e
 BioMed Research International
concentrations of the surrogate tracer (Gd-DTPA) and EB
dye showed a strong linear correlation. With this dynamic
information of tumor permeability, the pharmacodynamic
model can be modied to eventually take into account
parameters that aect drug delivery over time. Tumor periph-
eral or high-vascular tumor may have the most signicant
benet on blood-brain or blood-tumor permeability increase,
which gives critical information when intending to apply
FUS for brain drug enhanced delivery. We hope to use such
pharmacodynamic predictions along with FUS-induced BBB
opening to develop a method for image-guided drug delivery
that can estimate the amount of drug that will be delivered to
tissues at each time point.
Conflict of Interests
e authors declare that there is no conict of interests.
Acknowledgments
e authors thank the Molecular Imaging Center, Chang
Gung Memorial Hospital. is work was supported by the
National Science Council, Taiwan (Grant nos. --
B--, --M-A-, and --E---
MY).
References
[] K. Hynynen, N. McDannold, N. Vykhodtseva, and F. A. Jolesz,
“Noninvasive MR imaging-guided focal opening of the blood-
brain barrier in rabbits,Radiology,vol.,no.,pp.,
.
[]K.Hynynen,N.McDannold,N.A.Sheikov,F.A.Jolesz,and
N. Vykhodtseva, “Local and reversible blood-brain barrier
disruption by noninvasive focused ultrasound at frequencies
suitable for trans-skull sonications,NeuroImage,vol.,no.,
pp. –, .
[] K.Hynynen,N.McDannold,N.Vykhodtsevaetal.,“Focaldis-
ruption of the blood-brain barrier due to -kHz ultrasound
bursts: a method for molecular imaging and targeted drug
delivery,Journal of Neurosurgery,vol.,no.,pp.,
.
[]M.Kinoshita,N.McDannold,F.A.Jolesz,andK.Hyny-
nen, “Targeted delivery of antibodies through the blood-brain
barrier by MRI-guided focused ultrasound,Biochemical and
Biophysical Research Communications,vol.,no.,pp.
, .
[] N. McDannold, N. Vykhodtseva, and K. Hynynen, “Eects of
acoustic parameters and ultrasound contrast agent dose on
focused-ultrasound induced blood-brain barrier disruption,
Ultrasound in Medicine and Biology,vol.,no.,pp.,
.
[]H.L.Liu,Y.Y.Wai,W.S.Chenetal.,“Hemorrhagedetec-
tion during focused-ultrasound induced blood-brain-barrier
opening by using susceptibility-weighted magnetic resonance
imaging,Ultrasound in Medicine and Biology,vol.,no.,pp.
–, .
[]H.L.Liu,P.H.Hsu,P.N.Chuetal.,“Magneticresonance
imaging enhanced by superparamagnetic iron oxide particles:
usefulness for distinguishing between focused ultrasound-
induced blood-brain barrier disruption and brain hemorrhage,
Journal of Magnetic Resonance Imaging,vol.,no.,pp.,
.
[]M.Kinoshita,N.McDannold,F.A.Jolesz,andK.Hynynen,
“Noninvasive localized delivery of Herceptin to the mouse
brain by MRI-guided focused ultrasound-induced blood-brain
barrier disruption,Proceedings of the National Academy of
Sciences of the United States of America,vol.,no.,pp.
, .
[]L.H.Treat,N.McDannold,N.Vykhodtseva,Y.Zhang,K.
Tam, and K. Hynynen, “Targeted delivery of doxorubicin to
the rat brain at therapeutic levels using MRI-guided focused
ultrasound,International Journal of Cancer,vol.,no.,pp.
–, .
[] J. Mei, Y. Cheng, Y. Song et al., “Experimental study on
targeted methotrexate delivery to the rabbit brain via mag-
netic resonance imaging-guided focused ultrasound,Journal of
Ultrasound in Medicine, vol. , no. , pp. –, .
[] H. L. Liu, M. Y. Hua, P. Y. Chen et al., “Blood-brain bar-
rier disruption with focused ultrasound enhances delivery of
chemotherapeutic drugs for glioblastoma treatment,Radiol-
ogy,vol.,no.,pp.,.
[] W. M. Pardridge, “e blood-brain barrier: bottleneck in brain
drug development,NeuroRx,vol.,no.,pp.,.
[] A. Jemal, R. Siegel, E. Ward et al., “Cancer statistics, ,CA:
A Cancer Journal for Clinicians,vol.,no.,pp.,.
[] S. A. Grossman and J. F. Batara, “Current management of
glioblastoma multiforme,Seminars in Oncology,vol.,no.,
pp.,.
[]L.H.Treat,N.McDannold,Y.Zhang,N.Vykhodtseva,
and K. Hynynen, “Improved anti-tumor eect of liposomal
doxorubicin aer targeted blood-brain barrier disruption by
MRI-guided focused ultrasound in rat glioma,Ultrasound in
Medicine and Biology,vol.,no.,pp.,.
[] H. L. Liu, M. Y. Hua, H. W. Yang et al., “Magnetic resonance
monitoring of focused ultrasound/magnetic nanoparticle tar-
geting deliver y of therapeutic agents to the brain,” Proceedings of
the National Academy of Sciences of the United States of America,
vol.,no.,pp.,.
[] K. Liu, G. Wen, X. F. Lv et al., “MR imaging of cerebral extraven-
tricular neurocytoma: a report of  cases,e American Journal
of Neuroradiology,.
[] N. McDannold, N. Vykhodtseva, and K. Hynynen, “Blood-
brain barrier disruption induced by focused ultrasound and cir-
culating preformed microbubbles appears to be characterized
by the mechanical index,Ultrasound in Medicine and Biology,
vol.,no.,pp.,.
[] N. Vykhodtseva, N. McDannold, and K. Hynynen, “Progress
and problems in the application of focused ultrasound for
blood-brain barrier disruption,Ultrasonics,vol.,no.,pp.
–, .
[]F.Y.Yang,W.M.Fu,W.S.Chen,W.L.Yeh,andW.L.Lin,
“Quantitative evaluation of the use of microbubbles with tran-
scranial focused ultrasound on blood-brain-barrier disruption,
Ultrasonics Sonochemistry,vol.,no.,pp.,.
[] F.Y.Yang,W.M.Fu,R.S.Yang,H.C.Liou,K.H.Kang,and
W. L. Lin, “Quantitative evaluation of focused ultrasound with
a contrast agent on blood-brain barrier disruption,Ultrasound
in Medicine and Biology,vol.,no.,pp.,.
[] J. J. Choi, J. A. Feshitan, B. Baseri et al., “Microbubble-size
dependence of focused ultrasound-induced bloodBrain barrier
BioMed Research International 
opening in mice in vivo,IEEE Transactions on Biomedical
Engineering,vol.,no.,pp.,.
[] R. Chopra, N. Vykhodtseva, and K. Hynynen, “Inuence of
exposure time and pressure amplitude on blood-brain-barrier
opening using transcranial ultrasound exposures,ACS Chemi-
cal Neuroscience,vol.,no.,pp.,.
[] J.Park,Y.Zhang,N.Vykhodtseva,F.A.Jolesz,andN.J.McDan-
nold, “e kinetics of blood brain barrier permeability and
targeted doxorubicin delivery into brain induced by focused
ultrasound,Journal of Controlled Release,vol.,no.,pp.–
, .
[] P. S. Tos and A. G. Kermode, “Measurement of the blood-
brain barrier permeability and leakage space using dynamic
MR imaging. . Fundamental concepts,Magnetic Resonance in
Medicine,vol.,no.,pp.,.
[] F. Vlachos, Y. S. Tung, and E. Konofagou, “Permeability
dependence study of the focused ultrasound-induced blood-
brain barrier opening at distinct pressures and microbubble
diameters using DCE-MRI,Magnetic Resonance in Medicine,
vol. , no. , pp. –, .
[] I. M. Noebauer-Huhmann, P. Szomolanyi, V. Juras, O. Kra,
M. E. Ladd, and S. Trattnig, “Gadolinium-based magnetic
resonance contrast agents at  tesla: in vitro T relaxivities in
human blood plasma,Investigative Radiology,vol.,no.,pp.
–, .
[] E. A. Neuwelt, P. A. Barnett, C. I. McCormick, L. G. Remsen, R.
A. Kroll, and G. Sexton, “Dierential permeability of a human
brain tumor xenogra in the nude rat: impact of tumor size and
method of administration on optimizing delivery of biologically
diverse agents,Clinical Cancer Research,vol.,no.,pp.
, .
[] H. Sarin, A. S. Kanevsky, H. Wu et al., “Eective transvascular
delivery of nanoparticles across the blood-brain tumor barrier
into malignant glioma cells,JournalofTranslationalMedicine,
vol. , article , .
[] A. R. Padhani and J. E. Husband, “Dynamic contrast-enhanced
MRI studies in oncology with an emphasis on quantication,
validation and human studies,Clinical Radiology,vol.,no.
, pp. –, .
[] P. S. Tos, G. Brix, D. L. Buckley et al., “Estimating kinetic
parameters from dynamic contrast-enhanced T()-weighted
MRI of a diusable tracer: standardized quantities and sym-
bols,Journal of Magnetic Resonance Imaging,vol.,no.,pp.
–, .
[] H. Olsson, C. Halldin, C. G. Swahn, and L. Farde, “Quan-
tication of [C]FLB  binding to extrastriatal dopamine
receptors in the human brain,JournalofCerebralBloodFlow
and Metabolism,vol.,no.,pp.,.
[] K.W.Kang,D.S.Lee,J.H.Choetal.,“QuanticationofF-
 FDG PET images in temporal lobe epilepsy patients using
probabilistic brain atlas, NeuroImage,vol.,no.,part,pp.
–,  .
[] S. L. Bacharach, S. K. Libutti, and J. A. Carrasquillo, “Measur-
ing tumor blood ow with H215O: practical considerations,
Nuclear Medicine and Biology, vol. , no. , pp. –, .
[] M. S. Alavijeh and A. M. Palmer, “Measurement of the pharma-
cokinetics and pharmacodynamics of neuroactive compounds,
Neurobiology of Disease,vol.,no.,pp.,.
[]H.F.Dvorak,L.F.Brown,M.Detmar,andA.M.Dvorak,
“Vascular permeability factor/vascular endothelial growth fac-
tor, microvascular hyperpermeability, and angiogenesis,e
American Journal of Pathology,vol.,no.,pp.,
.
[] L. E. Benjamin, D. Golijanin, A. Itin, D. Pode, and E. Keshet,
“Selective ablation of immature blood vessels in established
humantumorsfollowsvascularendothelialgrowthfactorwith-
drawal,Journal of Clinical Investigation,vol.,no.,pp.
, .
[] J. S. Taylor, P. S. Tos, R. Port et al., “MR imaging of tumor
microcirculation: promise for the new millennium,Journal of
Magnetic Resonance Imaging, vol. , no. , pp. –, .
[] L. F. Brown, K. T. Yeo, B. Berse et al., “Expression of vascular
permeability factor (vascular endothelial growth factor) by
epidermal keratinocytes during wound healing, Journal of
Experimental Medicine,vol.,no.,pp.,.
[] M. A. O’Reilly, A. Muller, and K. Hynynen, “Ultrasound
insertion loss of rat parietal bone appears to be proportional
to animal mass at submegahertz frequencies,Ultrasound in
Medicine and Biology, vol. , no. , pp. –, .
[] M. A. O’Reilly, Y. Huang, and K. Hynynen, “e impact
of standing wave eects on transcranial focused ultrasound
disruption of the blood-brain barrier in a rat model,Physics
in Medicine and Biology,vol.,no.,pp.,.
... To evaluate the performance of FSW-induced BBB opening with or without the presence of microbubbles, the following procedure was modified based on previous studies (Chu et al., 2013;Kung et al., 2018). The intensity levels for each parameter in this research are shown in Table 1 (calibration data obtained from Richard Wolf GmbH). ...
... To define the cut-off value, the positive EB-stained area of the histology sections was analyzed using a color histogram of Image J based on the differences in number of blue pixels between the FSW-applied side and the untreated side. UCA has been widely used with previous HIPU studies to induce BBB opening in the brain by enhancing cavitation (Yang et al., 2011;Chu et al., 2013), thus the effect of UCA on FSW was investigated by infusing various concentrations of SonoVue UCA from 2.5 × 10 2 to 2.5 × 10 8 microbubbles/kg (MBs/kg) body weight (the clinical concentration is 3 -15 × 10 6 MBs/kg) through the tail vein immediately before FSW treatment (N = 5). ...
... Moreover, the 1 h BBB opening duration at intensity level 0.1 (peak positive pressure 5.4 MPa; peak negative pressure −4.2 MPa; energy flux density 0.03 mJ/mm 2 ) with 1/5 CD (2 × 10 6 MBs/kg) of UCA is shorter than the 3 h in our previous research. A shorter BBB opening duration can reduce the risk of some bio-harmful substances crossing into the parenchyma and central nervous system during the opening period (Chu et al., 2013;Tominaga et al., 2016;Cho et al., 2017). ...
Article
Full-text available
Focused extracorporeal shockwave (FSW), one kind of focused high-intensity pulsed ultrasound, has been shown to induce blood-brain barrier (BBB) opening in targeted brain areas in rat animal models with minimal detrimental effects below threshold intensity levels or iterations. In the current study, we found that the thresholds could be further reduced by the addition of microbubbles (ultrasound contrast agents or UCA; SonoVue). FSW with 2 × 10⁶ MBs/kg of UCA (20% of clinical dosage) at an intensity level of 0.1 (peak positive pressure 5.4 MPa; peak negative pressure −4.2 MPa; energy flux density 0.03 mJ/mm²) resulting in a 100% BBB opening rate without detectable hemorrhage or apoptosis in the brain. Significantly reduced free radical production was found compared with 0.5 MHz focused ultrasound at a peak negative pressure of 0.44 MPa (1% duty cycle and 4 × 10⁷ MBs/kg of UCA). FSW devices offer advantages of commercial availability and high safety, and thus may facilitate future research and applications of focal BBB opening for oncological and pharmacological purposes.
... With microbubble administration at 0.15 ml/kg, FUS exposure levels below 0.8-MI have been previously shown to be a threshold for consistent BBB opening without significant erythrocyte extravasations, while acoustic pressure of 1.4-MI causes significant intracerebral hemorrhage 16,18,19,26,31 . We therefore regard the former as "adequate FUS exposure", while the later condition is regarded as "excessive FUS exposure". ...
... 17,18 . The effect was explained by the ability of FUS-BBB opening to enhance drug delivery to the peripheral normal brain tissue while improving drug retain in the tumor core without posting other biological hazard such as intracerebral hemorrhage 31,38 . In addition to improve drug delivering efficiency to brain tumor, FUS-BBB opening may offer physicians the chance to design protocols with less frequent injections or reduced dose to reduce systemic toxicity while enhance tumor control. ...
Article
Full-text available
Abstract Focused ultrasound (FUS) with the presence of microbubbles induces blood brain barrier (BBB) opening in targeted areas and facilitates drug delivery. However, recent studies have indicated that FUS-BBB opening with excessive exposure levels may be associated with inflammatory response and cellular/tissue damage. Multiple weekly FUS exposures have been shown to be safe for human subjects. However the effect of more frequent FUS exposures is still unknown. This study examines whether frequent focused ultrasound blood brain barrier opening is associated with aggravated behavioral, histopathologic change or brain tissue damage. Two protocols of focused ultrasound blood brain barrier opening were devised using different microbubble doses (0.15 µl/kg and 0.4 µl/kg). Focused ultrasound exposure at a threshold level of BBB-opening, below-threshold level, or above level for intracerebral hemorrhage were delivered every 2 days. Animal behavioral and physiological changes were examined and recorded. Brain tissue was examined for hemorrhage and apoptosis. Results indicate that frequent exposure of excessive focused ultrasound (1.4 mechanical index) produced minor and short-term behavioral changes despite significant tissue damage, while frequent BBB opening with threshold or below-threshold FUS exposure (0.33–0.8 mechanical index) did not cause behavioral or histological change. Immunofluorescent examination of rat brain tissue indicated that excessive doses of microbubble administration induce an apparent cellular apoptotic response, which may be exacerbated by intracerebral hemorrhage. Experimental results suggest that frequent focused ultrasound blood brain barrier opening with sufficient ultrasound exposure level and a microbubble dose can be safe and pose minimal risk to brain tissue.
... The radioactive counts were also correlated with the degree of BBBO, demonstrating the possibility of using SPECT to detect BBBO [39]. Figure 1 summarizes representative images for the various modalities used to detect and monitor BBBO. [40], (b) TI-weighted CE-MRI [33], (c) T2-weighted CE-MRI [41], (d) MRI T1 relaxometry to sequentially follow the distribution of penetrating Gd-DTPA [42], (e) fluorescent-tag dextran penetration using optical microscopy [43], (f) autoradiography [39], (g) dynamic ultrasound imaging [44], (h) DCE-MRI [37], (i) 99m Tc-DTPA SPECT [39], and (j) 68 Ga PET [45] (min. = minutes, ANT = anterior; Adapted from Chen et al. (2019)). ...
Article
Full-text available
The blood–brain barrier (BBB) protects the central nervous system (CNS) from invasive pathogens and maintains the homeostasis of the brain. Penetrating the BBB has been a major challenge in the delivery of therapeutic agents for treating CNS diseases. Through a physical acoustic cavitation effect, focused ultrasound (FUS) combined with microbubbles achieves the local detachment of tight junctions of capillary endothelial cells without inducing neuronal damage. The bioavailability of therapeutic agents is increased only in the area targeted by FUS energy. FUS with circulating microbubbles is currently the only method for inducing precise, transient, reversible, and noninvasive BBB opening (BBBO). Over the past decade, FUS-induced BBBO (FUS-BBBO) has been preclinically confirmed to not only enhance the penetration of therapeutic agents in the CNS, but also modulate focal immunity and neuronal activity. Several recent clinical human trials have demonstrated both the feasibility and potential advantages of using FUS-BBBO in diseased patients. The promising results support adding FUS-BBBO as a multimodal therapeutic strategy in modern CNS disease management. This review article explores this technology by describing its physical mechanisms and the preclinical findings, including biological effects, therapeutic concepts, and translational design of human medical devices, and summarizes completed and ongoing clinical trials.
... FUS-induced BBB disruption is usually assessed by MRI, fluorescence imaging, or post-FUS histological examination (Chu et al., 2013;Fan et al., 2014;Nhan et al., 2013;. ...
Chapter
The blood-brain barrier (BBB) is a unique interface between the cerebral circulation and the brain. It plays crucial roles in facilitating the brain nutrient uptake and waste removal, maintaining the stability of the micro-environment in the central nervous system (CNS), and protecting the CNS under physiological and pathological conditions. On the other hand, the BBB also hinders drug delivery to the brain. Various strategies have been explored for enhancing drug delivery across the BBB to the brain, and a variety of smart nano-drugs and biomaterials have been designed to pass the BBB to achieve targeted therapy of neurological and brain diseases. Meanwhile, to observe, assess, and quantify drug delivery across the BBB as well as the efficacy of drugs for the treatment of brain diseases, multiple optical imaging techniques and platforms have been developed. In this chapter, we systematically review the progresses and applications of optical imaging techniques for drug delivery to the brain.
... The correlation between MRI contrast and tissue accumulation of agents varies in the literature. Some studies have reported correlations between dyes 50,51 or therapeutics, such as reported by Kinoshita. Others have reported no correlation. ...
Article
Full-text available
Extensive studies on focused ultrasound (FUS)-mediated drug delivery through the blood–brain barrier have been published, yet little work has been published on FUS-mediated drug delivery through the blood-spinal cord barrier (BSCB). This work aims to quantify the delivery of the monoclonal antibody trastuzumab to rat spinal cord tissue and characterize its distribution within a model of leptomeningeal metastases. 10 healthy Sprague–Dawley rats were treated with FUS + trastuzumab and sacrificed at 2-h or 24-h post-FUS. A human IgG ELISA (Abcam) was used to measure trastuzumab concentration and a 12 ± fivefold increase was seen in treated tissue over control tissue at 2 h versus no increase at 24 h. Three athymic nude rats were inoculated with MDA-MB-231-H2N HER2 + breast cancer cells between the meninges in the thoracic region of the spinal cord and treated with FUS + trastuzumab. Immunohistochemistry was performed to visualize trastuzumab delivery, and semi-quantitative analysis revealed similar or more intense staining in tumor tissue compared to healthy tissue suggesting a comparable or greater concentration of trastuzumab was achieved. FUS can increase the permeability of the BSCB, improving drug delivery to specifically targeted regions of healthy and pathologic tissue in the spinal cord. The achieved concentrations within the healthy tissue are comparable to those reported in the brain.
... Gd accumulation in the focal spot during the first minutes after Gd administration has also been reported by others in rabbits but was not observed in rats. 36,37 The kinetics of Gd uptake, directly after FUS treatment and intravenous injection of Gd, are not fully understood. It is not straightforward to quantify the extent of BBB opening for pyruvate from contrastenhanced images, and therefore current results only give a rough indication of the transport dynamics of Gd-like large molecules in the focal spot. ...
Article
Full-text available
Imaging of hyperpolarized ¹³C-labeled substrates has emerged as an important MR technique to study metabolic pathways in real time in vivo. Even though this technique has found its way to clinical trials, in vivo dynamic nuclear polarization is still mostly applied in preclinical models. Its tremendous increase in SNR overcomes the intrinsically low MR-sensitivity of the ¹³C nucleus and allows real-time metabolic imaging in small structures like the mouse brain. However, applications in brain research are limited as delivery of hyperpolarized compounds is restrained by the blood-brain barrier (BBB). A local non-invasive disruption of the BBB could facilitate delivery of hyperpolarized substrates and creates opportunities to study metabolic pathways in the brain that are generally not within reach. In this work we designed a setup to apply BBB disruption in the mouse brain by MR-guided focused ultrasound (FUS) prior to MR imaging of ¹³C-enriched hyperpolarized [1-¹³C]-pyruvate and its conversion to [1-¹³C]-lactate. To overcome partial volume issues, we optimized a fast multi-gradient-echo imaging method (temporal resolution of 2.4 s) with an in-plane spatial resolution of 1.6x1.6 mm², without the need of processing large amounts of spectroscopic data. We demonstrated the feasibility to apply ¹³C imaging in less than one hour after FUS treatment and showed a locally disrupted BBB during the time window of the whole experiment. From detected hyperpolarized pyruvate and lactate signals in both FUS-treated and untreated mice, we conclude that even at high spatial resolution signals from the blood compartment dominate in the ¹³C-images, leaving the interpretation of hyperpolarized signals in the mouse brain challenging.
... At appropriate acoustic power density (or pressure), burst repetition rate, duty cycle, and sonication duration and in the presence of MBs with proper materials, sizes, and concentrations, FUS can achieve noninvasive, selective and localized disruption of BBB without visible damage to the brain tissue (Konofagou 2012;Deng 2010). FUS-induced BBB disruption has been widely assessed by using magnetic resonance imaging (MRI) or post-FUS histological examination (Yang et al. 2011: Chu et al. 2013Park et al. 2010). Indirect MRI collects the images of brain slices in the presence of a MRI contrast agent such as gadolinium to access the BBB disruption in different locations of the brain. ...
Chapter
Full-text available
The blood-brain barrier (BBB) is a dynamic barrier essential for maintaining the microenvironment of the brain. Although the special anatomical features of the BBB determine its protective role for the central nervous system (CNS) from blood-borne neurotoxins, however, the BBB extremely limits the therapeutic efficacy of drugs into the CNS, which greatly hinders the treatment of major brain diseases. This chapter summarized the unique structures of the BBB; described a variety of in vivo and in vitro experimental methods for determining the transport properties of the BBB and the permeability of the BBB to water, ions, and solutes including nutrients, therapeutic agents, and drug carriers; and presented recently developed mathematical models which quantitatively correlate the anatomical structures of the BBB with its barrier functions. Recent findings for modulation of the BBB permeability by chemical and physical stimuli were described. Finally, drug delivery strategies through specific trans-BBB routes were discussed.
Article
Natural and synthetic sugars have great potential for developing highly biocompatible and translatable chemical exchange saturation transfer (CEST) MRI contrast agents. In this study, we aimed to develop the smallest clinically available form of dextran, Dex1 (molecular weight, MW ~ 1 kDa), as a new CEST agent. We first characterized the CEST properties of Dex1 in vitro at 11.7 T and showed that the Dex1 had a detectable CEST signal at ~1.2 ppm, attributed to hydroxyl protons. In vivo CEST MRI studies were then carried out on C57BL6 mice bearing orthotopic GL261 brain tumors (n = 5) using a Bruker BioSpec 11.7 T MRI scanner. Both steady-state full Z-spectral images and single offset (1.2 ppm) dynamic dextran-enhanced (DDE) images were acquired before and after the intravenous injection of Dex1 (2 g/kg). The steady-state Z-spectral analysis showed a significantly higher CEST contrast enhancement in the tumor than in contralateral brain (∆MTRasym 1.2 ppm = 0.010 ± 0.006 versus 0.002 ± 0.008, P = 0.0069) at 20 min after the injection of Dex1. Pharmacokinetic analyses of DDE were performed using the area under the curve (AUC) in the first 10 min after Dex1 injection, revealing a significantly higher uptake of Dex1 in the tumor than in brain tissue for tumor-bearing mice (AUC[0-10 min] = 21.9 ± 4.2 versus 5.3 ± 6.4%·min, P = 0.0294). In contrast, no Dex1 uptake was foundling in the brains of non-tumor-bearing mice (AUC[0-10 min] = -1.59 ± 2.43%·min). Importantly, the CEST MRI findings were consistent with the measurements obtained using DCE MRI and fluorescence microscopy, demonstrating the potential of Dex1 as a highly translatable CEST MRI contrast agent for assessing tumor hemodynamics.
Preprint
Optogenetics, the genetic incorporation of light-sensitive proteins such as Channelrhodopsin-2 (ChR2) into target mammalian neurons, has enabled activation, silencing, and receptor subtype specific neuromodulation with high spatiotemporal resolution. However, the essential components of the ontogenetic system require invasive procedures with very few non-invasive alternatives preventing its use as a translational tool. The implantation of light emitting fibers deep within brain structures is both technically demanding and causes tissue scarring in target brain regions. To overcome these limitations, while maintaining the highly-tuned components of optogenetics we have developed a novel noninvasive alternative. Our approach replaces fibers with light-emitting radioluminescent particles (RLPs) that can be activated non-invasively with X-ray exposure. Here, we report successful noninvasive delivery of RLPs to target brain regions using MRI-guided focused ultrasound (FUS) blood brain barrier opening. In addition, FUS BBBO can be used to deliver viral vectors for light sensitive channel expression. Combined, these components can provide a completely non-invasive optogenetic system.
Article
Magnetic resonance image-guided focused ultrasound has emerged as a viable non-invasive technique for the treatment of central nervous system-related diseases/disorders. Application of mechanical and thermal effects associated with focused transcranial ultrasound has been studied extensively in pre-clinical models, which has paved the way for clinical trials. However, in vivo treatment evaluation techniques on drug delivery application via blood–brain barrier opening has not been fully explored. Current treatment evaluation techniques via magnetic resonance imaging are hindered by systemic toxicity resulting from free gadolinium delivery. Here we propose a novel treatment evaluation strategy to overcome limitations by (i) synthesizing liposomes that are dually labeled with gadolinium, a magnetic resonance imaging (MRI) contrast agent, and rhodamine, a fluorophore; (ii) applying a focused ultrasound (FUS)-mediated BBB opening technique to deliver the liposomes across vascular barriers, achieving local gadolinium enhancement while reducing systemic and unwanted regional toxic effects associated with free gadolinium; and (iii) utilizing the MRI modality to confirm the delivery as it is already included in the FUS treatment in clinic. Liposomes were secondarily labeled with a fluorescent marker to confirm results obtained by MRI quantification postmortem. Two different sizes, 77.5 nm (group A) and 140 nm (group B), of gadolinium- and fluorescence-labeled liposomes were fabricated using thin-film hydration followed by extrusion methods and determined their stability up to 6 h under physiologic conditions. Gadolinium signal was detected on contrast-enhanced T1-weighted MRI 5 h after the delivery of liposomes via the BBB opening approach with an ultrasound pulse of 0.42 MPa (estimate in water) combined with microbubbles. MRI contrast was enhanced significantly in sonicated regions compared with non-sonicated regions of the brain. This was due to the accumulation of labeled liposomes, which was confirmed by detection of rhodamine fluorescence in histologic sections. The relative increase in MRI signal intensity was greater for smaller liposomes (mean diameter = 77.5 nm) than larger liposomes (mean diameter = 140 nm), which suggested a greater accumulation of the smaller liposomes in the brain after ultrasound-mediated opening of the BBB. Our findings suggest that the dual-labeled nanocarrier platform can be established, the FUS-mediated BBB opening approach can be used to deliver it through vascular barriers and MRI can be used to evaluate the extent of nanocarrier delivery.
Article
Full-text available
The therapeutic efficacy of neurological agents is severely limited, because large compounds do not cross the blood-brain barrier (BBB). Focused ultrasound (FUS) sonication in the presence of microbubbles has been shown to temporarily open the BBB, allowing systemically administered agents into the brain. Until now, polydispersed microbubbles (1-10 ??m in diameter) were used, and, therefore, the bubble sizes better suited for inducing the opening remain unknown. Here, the FUS-induced BBB opening dependence on microbubble size is investigated. Bubbles at 1-2 and 4-5 ??m in diameter were separately size-isolated using differential centrifugation before being systemically administered in mice ( n = 28). The BBB opening pressure threshold was identified by varying the peak-rarefactional pressure amplitude. BBB opening was determined by fluorescence enhancement due to systemically administered, fluorescent-tagged, 3-kDa dextran. The identified threshold fell between 0.30 and 0.46 MPa in the case of 1-2 ??m bubbles and between 0.15 and 0.30 MPa in the 4-5 ??m case. At every pressure studied, the fluorescence was greater with the 4-5 ??m than with the 1-2 ??m bubbles. At 0.61 MPa, in the 1-2 ??m bubble case, the fluorescence amount and area were greater in the thalamus than in the hippocampus. In conclusion, it was determined that the FUS-induced BBB opening was dependent on both the size distribution in the injected microbubble volume and the brain region targeted.
Article
Persistent microvascular hyperpermeability to plasma proteins even after the cessation of injury is a characteristic but poorly understood feature of normal wound healing. It results in extravasation of fibrinogen that clots to form fibrin, which serves as a provisional matrix and promotes angiogenesis and scar formation. We present evidence indicating that vascular permeability factor (VPF; also known as vascular endothelial growth factor) may be responsible for the hyperpermeable state, as well as the angiogenesis, that are characteristic of healing wounds. Hyperpermeable blood vessels were identified in healing split-thickness guinea pig and rat punch biopsy skin wounds by their capacity to extravasate circulating macromolecular tracers (colloidal carbon, fluoresceinated dextran). Vascular permeability was maximal at 2-3 d, but persisted as late as 7 d after wounding. Leaky vessels were found initially at the wound edges and later in the subepidermal granulation tissue as keratinocytes migrated to cover the denuded wound surface. Angiogenesis was also prominent within this 7-d interval. In situ hybridization revealed that greatly increased amounts of VPF mRNA were expressed by keratinocytes, initially those at the wound edge, and, at later intervals, keratinocytes that migrated to cover the wound surface; occasional mononuclear cells also expressed VPF mRNA. Secreted VPF was detected by immunofluoroassay of medium from cultured human keratinocytes. These data identify keratinocytes as an important source of VPF gene transcript and protein, correlate VPF expression with persistent vascular hyperpermeability and angiogenesis, and suggest that VPF is an important cytokine in wound healing.
Article
Focused ultrasound has been recently found to capable of temporally and reversibly disrupt local blood-brain barrier (BBB) and opens new frontier in delivering varies type of drugs into brain for central nerve system (CNS) disorder treatment. In this study, we aim to investigate the feasibility of delivering 1, 3-bits (2-chloroethyl) -1-nitrosourea (BCNU) to treat glioblastoma in animal models and evaluate whether this approach would gain treatment efficacy. Under the presence of microbubbles administration, a 400-kHz focused ultrasound was employed to deliver burst-tone ultrasonic energy stimulation to disrupt BBB in animal brains transcranially, and in-vivo monitored by magnetic-resonance imaging (MRI). C6-glioma cells were cultured and implanted into Sprague-Dawley rats as the brain-tumor model. BCNU deposited in brain was quantified by using high-performance liquid chromatography (HPLC), and brain tissues were examined histologically. MRI was employed to longitudinal evaluate the brain tumor treatment including the analysis of tumor progression and animal survival. We confirmed that the focused ultrasound, under the secure ultrasonic energy level, can significantly enhance the BCNU penetration through BBB over 300% than control without cause hemorrhage. Apparent improvement of treatment efficacy achieved by combining focused ultrasound with BCNU delivery, including significant suppression of tumor growth and a prolonged animal survival. This study highly support that this treatment strategy could be clinically-relevant and may help to provide another potential strategy in increasing local chemotherapeutic drugs for brain-tumor treatment.
Article
Extraventricular neurocytoma is a rare entity, most frequently occurring in brain parenchyma outside the ventricular system. The purpose of this study was to characterize the MR imaging findings in a series of 9 patients with EVN verified by results of pathologic examination. All 9 EVNs were solitary and intracranially located. Eight lesions were well demarcated, and 3 showed intratumoral hemorrhage. The solid parts of 7 tumors were primarily isointense on T1-weighted images and heterogeneously enhanced on T1WI with contrast. Although cerebral EVNs can present a wide spectrum of appearances on MR, the imaging patterns appear to vary according to anatomic location and cellularity. Lesions in frontal or parietal lobes often present as well-demarcated large masses with cystic degeneration, hemorrhage, mild-to-moderate edema, and inhomogeneous enhancement. Moreover, the general isointensity of the solid parts of EVN on T1WI may be of some specificity.
Article
Dynamic contrast-enhanced magnetic resonance imaging (DCE MRI) is a method of imaging the physiology of the microcirculation. A series of recent clinical studies have shown that DCE MRI can measure and predict tumor response to therapy. Recent advances in MR technology provide the enhanced spatial and temporal resolution that allow the application of this methodology in the management of cancer patients. The September issue of this journal provided a microcirculation section to update readers on this exciting and challenging topic. Evidence is mounting that DCE MRI-based measures correlate well with tumor angiogenesis. DCE MRI has already been shown in several types of tumors to correlate well with traditional outcome measures, such as histopathologic studies, and with survival. These new measures are sensitive to tumor physiology and to the pharmacokinetics of the contrast agent in individual tumors. Moreover, they can present anatomical images of tumor microcirculation at excellent spatial resolution. Several issues have emerged from recent international workshops that must be addressed to move this methodology into routine clinical practice. First, is complex modeling of DCE MRI really necessary to answer clinical questions reliably? Clinical research has shown that, for tumors such as bone sarcomas, reliable outcome measures of tumor response to chemotherapy can be extracted from DCE MRI by methods ranging from simple measures of enhancement to pharmacokinetic models. However, the use of similar methods to answer a different question—the differentiation of malignant from benign breast tumors—has yielded contradictory results. Thus, no simple, one-size-fits-all-tumors solution has yet been identified. Second, what is the most rational and reliable data collection procedure for the DCE MRI evaluation? Several groups have addressed population variations in some key variables, such as tumor T10 (T1 prior to contrast administration) and the arterial input function Ca(t) for contrast agent, and how they influence the precision and accuracy of DCE MRI outcomes. However, despite these potential complications, clinical studies in this section show that some tumor types can be assessed by relatively simple dynamic measures and analyses. The clinical scenario and tumor type may well determine the required complexity of the DCE MRI exam procedure and its analysis. Finally, we suggest that a consensus on naming conventions (nomenclature) is needed to facilitate comparison and analysis of the results of studies conducted at different centers. J. Magn. Reson. Imaging 10:903–907, 1999. © 1999 Wiley-Liss, Inc.
Article
The blood-brain barrier (BBB) inhibits the entry of the majority of chemotherapeutic agents into the brain. Previous studies have illustrated the feasibility of drug delivery across the BBB using focused ultrasound (FUS) and microbubbles. Here, we investigated the effect of FUS-enhanced delivery of doxorubicin on survival in rats with and 9L gliosarcoma cells inoculated in the brain. Each rat received either: (1) no treatment (control; N = 11), (2) FUS only (N = 9), (3) IV liposomal doxorubicin (DOX only; N = 17), or (4) FUS with concurrent IV injections of liposomal doxorubicin (FUS+DOX; N = 20). Post-treatment by magnetic resonance imaging (MRI) showed that FUS+DOX reduced tumor growth compared with DOX only. Further, we observed a modest but significant increase in median survival time after a single treatment FUS+DOX treatment (p = 0.0007), whereas neither DOX nor FUS had any significant impact on survival on its own. These results suggest that combined ultrasound-mediated BBB disruption may significantly increase the antineoplastic efficacy of liposomal doxorubicin in the brain.
Article
Focused ultrasound (FUS) combined with a circulating microbubble agent is a promising strategy to non-invasively disrupt the blood-brain barrier (BBB) and could enable targeted delivery of therapeutics that normally do not leave the brain vasculature. This study investigated the kinetics of the BBB permeability using dynamic contrast-enhanced MRI (DCE-MRI) and the resulting payload of the chemotherapy agent, doxorubicin (DOX). We also investigated how the disruption and drug delivery were affected by a double sonication (DS) with two different time intervals (10 or 120 min). Two locations were sonicated transcranially in one hemisphere of the brain in 20 rats using a 690 kHz FUS transducer; the other hemisphere served as a control. For BBB disruption, 10 ms bursts were applied at 1 Hz for 60s and combined with IV injection of a microbubble ultrasound contrast agent (Definity; 10 μl/kg). DOX was injected immediately after the second location was sonicated. The transfer coefficient (K(trans)) for an MRI contrast agent (Gd-DTPA) was estimated serially at 4-5 time points ranging from 30 min to 7.5 hrs after sonication using DCE-MRI. After a single sonication (SS), the mean K(trans) was 0.0142 ± 0.006 min(-1) at 30 min and was two or more orders of magnitude higher than the non-sonicated targets. It decreased exponentially as a function of time with an estimated half-life of 2.22 hrs (95% confidence intervals (CI): 1.06-3.39 hrs). Adding a second sonication increased K(trans), and with a 120 min interval between sonications, prolonged the duration of the BBB disruption. Mean K(trans) estimates of 0.0205 (CI: 0.016-0.025) and 0.0216 (CI: 0.013-0.030) min(-1) were achieved after DS with 10 and 120 min delays, respectively. The half-life of the K(trans) decay that occurred as the barrier was restored was 1.8 hrs (CI: 1.20-2.41 hrs) for a 10 min interval between sonications and increased to 3.34 hrs (CI: 0.84-5.84 hrs) for a 120 min interval. DOX concentrations were significantly greater than in the non-sonicated brain for all experimental groups (p<0.0001), and 1.5-fold higher for DS with a 10 min interval between sonications. A linear correlation was found between the DOX concentration achieved and the K(trans) measured at 30 min after sonication (R: 0.7). These data suggest that one may be able to use Gd-DTPA as a surrogate tracer to estimate DOX delivery to the brain after FUS-induced BBB disruption. The results of this study provide information needed to take into account the dynamics BBB disruption over time after FUS.
Article
The need to discover and develop safe and effective new medicines is greatest for disorders of the CNS. A core requirement for an effective neurotherapeutic agent is an ability to cross the blood–brain barrier and remain in the brain interstitial fluid (ISF) for a sufficient duration and concentration to evoke the desired therapeutic effect. Measuring the free concentration of a neuroactive compound in brain ISF is therefore an essential step in the critical path towards the development of a CNS medicine. In vivo microdialysis provides a powerful method for the measurement of endogenous and exogenous substances in the ISF surrounding the probe and so it represents an important tool in CNS drug discovery. It can also be used to measure the pharmacodynamic response of neuroactive compounds by measuring neurotransmitters and second messengers. Another approach to measure both pharmacokinetics and the pharmacodynamics of neuroactive compounds is the measurement of receptor occupancy, which has the advantage of being applicable to the study of humans as well as experimental animals. Measurement of the pharmacokinetics and pharmacodynamics of neuroactive compounds clearly improve understanding of the efficacy and safety of drug candidates, which improves both the efficiency and the effectiveness of CNS medicines research.
Article
{Abstract 10.1002/mrm.1910170208.abs Leakage of Gd-DTPA through a defective blood-brain barrier is measured quantitatively using dynamic MRI scanning, in which repeated scans are made after a bolus injection. Image registration artifacts are minimized; a dose of 0.1 mM/kg and an IR sequence enable enhancement to be measured quantitatively. The triexponential enhancement curve is fitted to a theoretical model based on compartmental analysis. The transfer constant, or permeability surface area product per unit volume of tissue (k), and leakage space per unit volume of tissue (y) are measured. Estimates for a quickly enhancing multiple sclerosis lesion are k = 0.050 min−1