You are on page 1of 10

ARTICLE IN PRESS

Ultrasound in Med. & Biol., Vol. 00, No. 00, pp. 110, 2019
Copyright © 2019 World Federation for Ultrasound in Medicine & Biology. All rights reserved.
Printed in the USA. All rights reserved.
0301-5629/$ - see front matter

https://doi.org/10.1016/j.ultrasmedbio.2019.02.024

 Original Contribution

MRI MONITORING AND QUANTIFICATION OF ULTRASOUND-MEDIATED


DELIVERY OF LIPOSOMES DUALLY LABELED WITH GADOLINIUM AND
FLUOROPHORE THROUGH THE BLOOD-BRAIN BARRIER

TAGEDPMUNA ARYAL,* IASON PAPADEMETRIOU,y YONG-ZHI ZHANG,* CHANIKARN POWER,*


NATHAN MCDANNOLD,* and TYRONE PORTERy,zTAGEDEN
* Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; y Department of
Mechanical Engineering, Boston University, Boston, Massachusetts, USA; and z Department of Biomedical Engineering, Boston
University, Boston, Massachusetts, USA
(Received 25 September 2018; revised 11 January 2019; in final from 28 February 2019)

Abstract—Magnetic resonance image-guided focused ultrasound has emerged as a viable non-invasive tech-
nique 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 bloodbrain barrier opening has not been fully explored. Current treatment evalua-
tion techniques via magnetic resonance imaging are hindered by systemic toxicity resulting from free gadolin-
ium 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 tech-
nique to deliver the liposomes across vascular barriers, achieving local gadolinium enhancement while reduc-
ing 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 sec-
ondarily 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 lipo-
somes 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. (E-mail: muna01@stanford.edu) © 2019
World Federation for Ultrasound in Medicine & Biology. All rights reserved.
Key Words: Nanocarrier, Focused Ultrasound, Cavitation, Brain drug delivery.

INTRODUCTION (Hynynen and Jones 2016). Mechanical and thermal effects


associated with FUS have been studied extensively in pre-
Transcranial image-guided focused ultrasound (FUS) has
clinical models, which has paved the way for their clinical
emerged as a viable non-invasive technique for the treat-
application (Carpentier et al. 2016; Coluccia et al. 2014;
ment of central nervous system-related diseases/disorders
Lipsman et al. 2018; Martin et al. 2009; McDannold et al.
2010). In one promising approach, researchers leveraged
Address correspondence to Muna Aryal, 1201 Welch Road, Stan- mechanical effects for the transient and focal opening of
ford, CA 94305, USA. E-mail: muna01@stanford.edu
Conflict of interest disclosure: The authors declare they have no the bloodbrain barrier (BBB) via microbubble cavitation
conflicts of interest. for brain drug delivery (Hynynen et al. 2001; Poon et al.

1
ARTICLE IN PRESS
2 Ultrasound in Medicine & Biology Volume 00, Number 00, 2019

2017). Furthermore, a dual-modality contrast agent, albu- well as improve subsequent penetration in brain tissue
min-shelled gadolinium-diethylenetriamine pentaacetic (Nance et al. 2014). Previously, we investigated the rela-
acid microbubbles, was designed for ultrasound and mag- tionship between MRI-derived tumor permeability esti-
netic resonance imaging (MRI) to assist BBB opening mates and the resulting tissue concentrations in rats that
(Liao et al. 2012). The use of imaging with this procedure received liposomal doxorubicin (DOX) (Aryal et al.
is critical to achieving both a tolerable and an efficacious 2015a). We found that the relationship between FUS-
outcome. Imaging is useful at each stage of the procedure, induced changes in tumor permeability and the MRI
from treatment planning to determine the target volume contrast agent gadolinium-diethylenetriamine pentaace-
and evaluate the skull in the ultrasound beam path for tic acid (Gd-DTPA) depended on the stage of the tumor.
phase aberration correction, to intra-procedural monitoring Importantly, at later stages of tumor growth, we did not
and feedback control over the acoustic parameters during observe an increase in permeability for the small mole-
ultrasound exposures (sonications). cule contrast agent, but we did find a significant increase
Post-sonication imaging is particularly important in liposomal drug delivery. This discrepancy was most
for evaluating whether unanticipated tissue damage was likely due to the difference in physicochemical proper-
produced, to confirm that BBB disruption (BBBD) ties between Gd-DTPA and liposomal DOX.
occurred throughout the targeted volume and to charac- Because of these issues, to map concentrations of
terize the level of permeabilization that was achieved. large therapeutic agents and nanocarriers, either new
BBBD has typically been evaluated using contrast- imaging agents with similar physical properties would
enhanced T1-weighted MRI (Hynynen et al. 2001). have to be developed or the drug itself would have to be
Quantitative estimates of contrast agent delivery and labeled directly. The latter approach has been used with
permeability changes can be performed using T1 map- FUS-induced BBBD, radiolabeled drugs, and single-pro-
ping and pharmacokinetic modeling (Park et al. 2012; ton emission computed tomography (SPECT) and positron
Vlachos et al. 2010). Several groups have investigated emission tomography (PET) imaging (Yang et al. 2012;
the use of these methods to estimate the tissue concentra- Ye et al. 2018). It is also possible to use gadolinium as a
tion of a drug or tracer agent after sonication (Chu et al. drug label for MRI (Frenzel et al. 2008). This approach
2013; Fite et al. 2017). In this approach, a water-soluble would be advantageous because of the higher spatial reso-
MRI contrast agent, such as chelated gadolinium, acts as lution that can be achieved with MRI and because stan-
a drug surrogate, and the relationship between the tissue dard MRI sequences can be used to confirm that no
concentrations of the MRI contrast agent and the thera- unexpected tissue damage occurred during sonication.
peutic agent is derived in experimental animal studies. Furthermore, recent clinical studies reported that the gado-
Presumably, these experimentally derived relationships linium accumulates in the brain, most notably in the den-
could be used in subsequent clinical studies. Being able tate nuclei and globus pallidus (Gulani et al. 2017; Kanda
to estimate local drug concentrations could be an impor- et al. 2015), which has raised concerns in the scientific
tant way to ensure that the treatment was effecti and uni- and clinical communities. If we succeed in designing a
formly applied. It is appropriate to use an MRI contrast liposome-based MR contrast agent, it will reduce the sys-
agent as a surrogate for a drug of comparable physico- temic toxicity, as well as unwanted regional toxicity, asso-
chemical properties for monitoring and estimating the ciated with free gadolinium. Therefore, we are
amount delivered to brain tissue through openings cre- envisioning the use of Gd-labeled liposomes primarily for
ated by transcranial ultrasound. However, many drugs fundamental, pre-clinical studies and perhaps in the clinic
have poor solubility in water or are substrates for circu- to investigate the pharmacokinetics and efficiency of
lating enzymes and thus must be packaged within nano- FUS-mediated brain drug delivery. So our long-term goal
carriers before systemic administration. Therefore, it is is to develop MR-detectable nanoparticles, particularly for
important to monitor and confirm delivery of the nano- finding an MR-based imaging method to non-invasively
carrier through ultrasound-induced openings in the BBB. predict how the pharmacodistribution of a given therapeu-
At present, FUS-mediated BBBD has been utilized to tic agent would change after ultrasound-mediated BBB
deliver several types of nanocarriers to the brain, includ- opening. The work described in this article is the first step
ing PEGylated liposomes (Kovacs et al. 2015), PEGy- toward that goal. Here, we designed a liposome-based
lated polymer nanoparticles (Nance et al. 2014), gold nanoparticle labeled with an MR contrast agent and fluoro-
nanoparticles (Diaz et al. 2014; Etame et al. 2012) and phore and investigated nanoparticle extravasation across
viral vectors (Hsu et al. 2013; Thevenot et al. 2012; the BBB after transcranial FUS in vivo via MRI and post-
Wang et al. 2015). These studies have established that mortem via spectrofluorometry in this study. We also
combination of FUS-mediated BBBD with nanocarriers incorporated a passive cavitation technique to monitor
can be used to enhance delivery of chemotherapy, cavitation activities during BBB opening. The study was
nucleic acids and contrast agents across the BBB, as designed to be a proof-of-concept, and the findings can be
ARTICLE IN PRESS
Liposome delivery through BBB  M. ARYAL et al. 3

used to guide the design of subsequent experiments in was measured in methanol with a spectrophotometer (Shi-
which changes in BBB penetration of drug-loaded nano- madzu, excitation/emission 560/595 nm, slit widths 1.5/5
particles as a function of acoustic parameters and/or cavi- nm). The fluorescence of methanol, PBS and PBS with
tation activity can be evaluated. 15% serum was measured as negative controls.

METHODS Animals
The Institutional Animal Care and Use Committees
Liposome preparation of Brigham and Women’s Hospital approved all animal
Fluorescence- and gadolinium-labeled liposomes experiments. Tests were performed in 16 male mice
were prepared by the thin-film hydration method. A rho- (Charles River Laboratories, Wilmington, MA, USA;
damine-labeled lipid (1,2-dipalmitoyl-sn-glycero-3- »35 g). Animals were divided into two groups based on
phosphoethanolamine (DPPE)-N-(lissamine rhodamine the size of PEGylated liposomal particles used for the
B sulfonyl) [ammonium salt], DPPE-rhodamine) and a study: group A, 7085 nm (n = 9), and group B,
gadolinium-labeled lipid (DTPA-bis(stearylamide) [gad- 130150 nm (n = 7). Two animals from group A and
olinium salt], Gd-DTPA-BSA) were used to incorporate one animal from group B were excluded because of
rhodamine and gadolinium on the head groups of the errors that occurred during fluorescence measurement or
lipid bilayer. The lipid composition was a molar ratio of during sonication. Before each procedure, the animals
58.459.5 mol% percent 1,2-dipalmitoyl-sn-glycero-3- were anesthetized via intraperitoneal injections of keta-
phosphocholine (DPPC), 35 mol% Gd-DTPA-BSA, 5 mine (80 mg/kg/h) and xylazine (10 mg/kg/h). A cathe-
mol% DSPE-PEG2k (1,2-distearoyl-sn-glycero-3-phos- ter was placed in the tail vein, and the hair on the scalp
phoethanolamine-N- [methoxy(polyethylene glycol)- was removed with clippers and depilatory cream. Body
2000] [ammonium salt]) and 0.51.6 mol% percent temperature was maintained throughout the experiment
DPPE-rhodamine. Lipids were dissolved in a 2:1 volume with a heated water pad.
ratio of chloroform: methanol, which was evaporated
over a stream of argon gas and left in a vacuum desicca- Sonication system
tor overnight to remove residual solvent. The lipid thin An air-backed, single-element, 690-kHz focused
film was rehydrated with phosphate-buffered saline piezoelectric transducer (diameter/radius of curvature:
(PBS) for 2 h at 66˚C with occasional stirring and pipet- 40 mm/30 mm) generated the ultrasound field. The trans-
ting. The lipid solution was sonicated at 20% power for ducer was assembled in-house. It was driven by an arbi-
2 min with a probe sonicator, and extruded sequentially trary waveform generator (33220 A, Agilent, Santa
through membrane filters with 1.0-mm pores (4 £), Clara, CA, USA) and radiofrequency (RF) amplifier
0.4-mm pores (6 £) and 0.2-mm pores (8 £) to prepare (240 L, E&I, Rochester, NY, USA); electric power was
liposomes with a mean diameter of 130150 nm (group measured with a power meter (E4419 B, Agilent) and
B). To prepare liposomes with a mean diameter of dual-directional RF coupler (C5948-10, Werlatone,
7085 nm (group A), the lipid solution was extruded in Cranberry Township, PA, USA). A radiation force bal-
the same manner with additional extrusion through ance (Mettler Toledo, Columbus, OH, USA) was used to
membrane filters with 0.1-mm pores (8 £) and 0.05-mm measure acoustic power, and a 0.2-mm-diameter needle
pores (12 £). The liposomes were measured by dynamic hydrophone (HNC-0200, Onda, Sunnyvale, CA, USA)
light scattering. The doses administered for group A was used to calibrate the transmitted pressure and map
(0.56 § 0.03 mmol total lipid/kg body weight) and group the acoustic field; its half-maximum diameter and length
B (0.51 § 0.08 mmol total lipid/kg body weight) were were 2.3 and 10 mm, respectively. These calibrations
similar. The calculated administered dose assumes no were used to estimate the peak negative pressure ampli-
loss of lipid because of extrusion. tude at the focus in water.
The experiments were performed under MRI guid-
Liposome stability ance (Fig. 1a). The transducer was mounted on a manually
To assess liposome stability under physiologic con- operated, MRI-compatible positioning system and cou-
ditions, liposomes were incubated at 37˚C with 15% pled to the mouse’s head with degassed, de-ionized water.
serum for 6 and 24 h. After incubation, liposomes were A transmit/receive surface coil (diameter/radius of curva-
measured via dynamic light scattering to assess aggrega- ture: 4 cm/3 cm) was placed under the mouse head, and
tion. Disassociation of rhodamine from liposomes was the system was placed in an animal 7-T MRI (Biospec,
assessed by filtration of liposomes through a 30-kDa cen- Bruker). T1-Weighted rapid acquisition with refocused
trifugal filter (Amicon Ultra, Sigma-Aldrich, St. Louis, echoes (RARE) images (parameters: TR/TE: 600/18 ms;
MO, USA) to collect solvent containing released rhoda- echo train length (ETL): 4; matrix size: 128 £ 128; slice
mine. The fluorescence of liposomes and liposome filtrate thickness/spacing: 1 mm/interleaved; field of view
ARTICLE IN PRESS
4 Ultrasound in Medicine & Biology Volume 00, Number 00, 2019

with 1.5- or 2-mm spacing. Each sonication was com-


bined with an intravenous injection of microbubbles
(Optison, Princeton, NJ, USA) administered at 20 mL/kg.
The interval between sonications was 12 min, which
allowed most of the residual microbubbles from the previ-
ous sonication to clear. To facilitate injections of such a
small volume, the agent was diluted in PBS to 0.1 times
its normal concentration. It was injected as a bolus
approximately 9 s before each sonication, followed by a
0.1-mL saline flush. MR images were obtained with a
transmit/receive MRI surface coil, which was placed
under the rat’s head. The MRI coil and positioning system
were assembled in-house. Before the mouse experiments,
we visualized heating in a silicone phantom using temper-
ature-sensitive MRI (T1-weighted RARE) to localize the
acoustic focal point in the MRI space.

Passive cavitation detection system


The acoustic emissions from cavitating microbub-
bles were recorded using a custom-built piezoelectric
transducer (center frequency: 1.5 MHz, bandwidth
» 30%, diameter: 8 mm) operated as a passive cavitation
detector (PCD). The procedure for measurement and
analysis of the emissions has been described previously
Fig. 1. (a) Schematic of the magnetic resonance imaging-
(Arvanitis et al. 2012; Aryal et al. 2017). Briefly, the
guided focused ultrasound (MRIgFUS) system used in this
work. The function generator, amplifier, and power meter were PCD was mounted adjacent to the focused transducer
located outside the MRI room. A PCD was used to monitor and aimed at the focal region to monitor the acoustic
acoustic emissions. (b) Sonication plan, monitoring and assess- emission produced during sonication. The detector was
ment of FUS-induced bloodbrain barrier disruption (BBBD) connected to a Notch filter (Allen Avionic, Mineola,
after microbubble-enhanced sonication. Axial MRI acquired
NY, USA) with a central frequency of 650 kHz. Filtered
during FUS-BBBD. Pre-FUS T2*-weighted images (T2*-WIs)
were used for treatment planning, and post-FUS *WIs were signals were routed through an amplifier (Standard
used to detect tissue damage. Post-FUS, contrast-enhanced T1- Research System, Sunnyvale, CA, USA) and recorded
weighted images (CE-T1-WIs) were acquired over 524 h to with a computer using a high-speed digitizing card
observe contrast enhancement after the delivery of liposomes (PXI-5124, National Instruments, Austin, TX, USA).
via ultrasound-mediated BBBD. Circles indicate the targeted
The time signal, frequency spectra and magnitude of the
spots; yellow curves outline the extent of gadolinium enhance-
ment resulting from BBB permeabilization. emission at different harmonics were displayed in real
time during each sonication using software developed
(FOV): 4 cm; number of excitations (NEX): 4, bandwidth: with MATLAB (The MathWorks, Natick, MA, USA)
64.1 kHz) were used to detect signal enhancement from and stored for later analysis. Emissions recorded during
the injected liposomes at different time points (t = 0.5, 5 sonication with microbubbles were normalized to data
and 24 h). T2*-Weighted imaging (parameters: TR/TE: obtained beforehand at the same locations and exposure
494.5/15 ms; matrix size: 128 £ 128; slice thickness: 0.8 levels without microbubbles. Frequency spectra were
mm; FOV: 3.5 cm; bandwidth: 50 kHz, NEX: 2) was used created from the time signals via fast Fourier transform.
to plan the sonication and to ensure petechiae, which can The strength of the relative integrated power spectra (in
result from excessive FUS exposures (Hynynen et al. dB-Hz) was calculated over 20-Hz bins for the harmon-
2001), did not occur (Fig. 2b). ics (FUS frequency £ 2 and 3), at the 1/2 subharmonic
(FUS frequency £ 1/2) and first and second ultrahar-
monics. For each animal, the mean signal for all bursts
Sonications and all sonications was calculated.
A peak negative pressure amplitude of 0.42 MPa
(estimate in water) was applied in burst mode (burst Fluorometric quantification of brain homogenates
length: 10 ms, pulse repetition frequency: 1 Hz, duration, To mark the targeted areas for tissue extraction, try-
60 s) for a single target. Sonication was applied at multi- pan blue (MP Biomedicals, Solon, OH, USA) was admin-
ple points, two or three minimally overlapping targets istered after the sonications. The agent was prepared for
ARTICLE IN PRESS
Liposome delivery through BBB  M. ARYAL et al. 5

Fig. 2. Contrast enhancement of brain tissue after focused ultrasound-induced bloodbrain barrier disruption (FUS-
BBBD) combined with microbubbles and its corresponding signal intensities. (a) Axial magnetic resonance images
acquired 24 h FUS-BBBD. Contrast-enhanced T1-weighted images (CE-T1-WIs) were used to verify contrast enhance-
ment at sonicated locations. Sonicated and control locations are indicated by red and blue squares, respectively. (b) Mea-
surement of gadolinium signal intensity on CE-T1-WIs for sonicated and non-sonicated (control) tissue. Gadolinium
signal intensity at sonicated locations increased by 26% compared with control, and the comparison was statistically sig-
nificant (p < 0.05). Bar = 5 mm.

each experiment. The agent was dissolved in 0.45% NaCl plane (perpendicular to the direction of ultrasound beam
and then boiled and filtered to avoid the formation of propagation). Every 20th section (20 mm apart) was
microcrystals. The solution was administered intrave- stained with hematoxylin and eosin (H&E). In each ani-
nously at a dose of 0.1 g trypan blue/kg body weight. mal, the sections with the greatest tissue effects were iden-
Twenty-four hours after the sonication, animals tified and examined in detail under high magnification; the
were deeply anesthetized with ketamine/xylazine and dimensions of any damaged area were measured.
sacrificed. A 30- to 90-mg volume of brain tissue was
harvested using trypan blue to differentiate sonicated Data analysis
and unsonicated brain regions. The brain tissue was Differences in signal intensity between sonicated
homogenized, resuspended in 100% methanol and cen- and non-sonicated brain were calculated in T1-weighted
trifuged to pellet debris, and fluorescence was measured MRI acquired 5 and 24 h after delivery of the liposomes.
using a spectrofluorophotometer (Shimadzu, 560 nm/ The means and standard deviations (SD) of signal inten-
585 nm excitation/emission wavelengths, 3 nm/5 nm slit sity were measured in regions of interest centered on the
widths, high sensitivity setting). Background fluores- sonicated target and in the same structure in the contra-
cence was assessed using brain homogenates from mice lateral hemisphere. Similarly, the means § SD of lipid
that did not receive liposome injections, and the back- concentrations on sonicated and non-sonicated tissue
ground was subtracted from the measurements. A cali- were determined based on the fluorescence measure-
bration curve was used to convert fluorescence signal ments. To compare gadolinium signal intensity and lipid
intensity to lipid mass (Supplementary Fig. S1). The concentrations between sonicated and non-sonicated tis-
mass of lipid was divided by the weight of brain tissue to sue in each group, two-tailed paired Student t-tests were
normalize for differences in mass of brain tissue har- used. Two-tailed unpaired Student t-tests were used to
vested, and the final data output was expressed as nano- compare gadolinium signal intensities, lipid concentra-
moles of lipid per gram of brain tissue. tions and mean harmonic strength between groups A and
B. Values of p < 0.05 were considered to indicate statis-
tical significance.
Histology
One mouse each from each group’s A and B was used
RESULTS
to evaluate the histologic effects. After transcardial perfu-
sion with normal saline (0.9% NaCl, 250 mL) to flush Liposome stability
unabsorbed Gd-labeled liposomal particles from the cere- To assess the stability of liposomes under physio-
bral vasculature, the brain was removed and flash-frozen. logic conditions, liposomes were incubated at 37˚C with
Frozen brains were sectioned into 8-mm slices in the axial 15% serum. No significant differences in the size of
ARTICLE IN PRESS
6 Ultrasound in Medicine & Biology Volume 00, Number 00, 2019

group A or B liposomes were observed after 6 h of incu- represented a statistically significant difference in group
bation (Supplementary Fig. S2). After 24 h of incuba- A (p = 0.017). The number of samples was not sufficient
tion, the mean diameter of the group A liposomes for statistical analysis in group B.
increased to »300 nm, whereas no significant change
was observed with the group B liposomes. After ultrafil- Fluorescence-labeled lipid concentrations
tration of the liposomes with a 30-kDa filter, no rhoda- In both experimental groups, the concentration of
mine was detectable in the filtrate of either group A or B rhodamine in the sonicated and non-sonicated (control) tis-
liposomes after incubation for 6 and 24 h (Supplemen- sues was measured 24 h after FUS-BBBD (Fig. 4). Mean
tary Fig. S2). The stability of the Gd-DTPA chelate fluorescence signal intensities in the sonicated hemisphere
bound to liposomes was not measured in the present were 0.83 § 0.22 nmol/g and 2.37 § 1.29 nmol/g in
study. In a previous report, less than 1% of Gd was groups A (n = 3) and B (n = 3), respectively. These values
released from Gd-DTPA chelate after 24 h of incubation were higher than those for the control groups, in which the
at 37˚C in the presence of serum (Frenzel et al. 2008). mean values were 0.51 § 0.3 nmol/g and 1.69 § 1.03
nmol/g, respectively. The increased signal intensity (62%
MRI findings and 40% in group A and B respectively) represented a sta-
The liposomes were labeled with Gd and Rho, thus tistically significant difference in group A (p = 0.02) but
enabling detection of extravasated liposomes in vivo via not in group B (p = 0.34). The mean ratio of fluorescence
MRI and postmortem via fluorometry. Contrast- signal intensities between the sonicated and control hemi-
enhanced T1-weighted images (CE-T1-WIs) revealed no spheres was 1.9 § 0.2 and 1.5 § 0.56 in groups A and B,
changes in signal intensity at 0.5 h after the sonication. respectively. The difference in this ratio for the two groups
At 5 and 24 h after FUS, CE-T1-WIs revealed a hyperin- was not statistically significant (p = 0.48).
tense area in sonicated tissue. In Figures 2a and 3a are
representative examples of CE-T1-WIs with a hyperin- Acoustic emission
tense area in sonicated tissue (red square in figures). A typical example of an acoustic emission spectrum
Measurement of signal enhancement on CE-T1-WIs is provided in Supplementary Figure S3. The data were
images of sonicated and control tissue is illustrated in obtained with a PCD with a center frequency of
Figures 2b and 3b. Mean gadolinium signal intensity in 1.5 MHz and were normalized to recordings acquired
the sonicated hemisphere was 7634.5 § 720 and 5216.9 without an injection of microbubbles. After normaliza-
§ 492 for groups A (n = 4) and B (n = 2), respectively. tion, only harmonic emissions (FUS frequency £ 2
These values were higher than the control volumes, and 3) and occasionally sub- or ultraharmonic emissions
where the mean values were 6053.8 § 210 and 4190.3 § (FUS frequency £ 1/2) were observed. A correlation
98 in these groups, respectively. The increased signal between liposome concentrations in brain tissue after
intensity (26% and 24% in groups A and B, respectively) FUS-mediated BBBD and strength of microbubble

Fig. 3. Contrast enhancement of brain tissue after focused ultrasound-induced bloodbrain barrier disruption (FUS-
BBBD) combined with microbubbles and its corresponding signal intensities. (a) Axial magnetic resonance image
(MRI) was acquired 24 h after FUS-BBBD. Contrast-enhanced T1-weighted images (CE-T1-WIs) were used to verify
contrast enhancement at sonicated locations. Sonicated and control locations are indicated by red and blue squares,
respectively. (b) Measurement of gadolinium signal intensity on CE-T1-WIs of sonicated and non-sonicated (control) tis-
sue. Gadolinium signal intensity at sonicated locations increased by 24% compared with control. Bar = 5 mm
ARTICLE IN PRESS
Liposome delivery through BBB  M. ARYAL et al. 7

Fig. 4. Lipid concentrations in brain tissue corresponding to group A and group B liposomes. Liposomes were labeled
with gadolinium (Gd) and the fluorophore rhodamine (Rho). Fluorescence was measured done 24 h after focused ultra-
sound-induced bloodbrain barrier disruption (FUS-BBBD). (a) For group A liposomes, 0.83 § 0.22 nmol/g (mean §
standard deviation) of lipid were measured in sonicated brain tissue (n = 3 mice), and had increased of 62% over unsoni-
cated brain tissue, which was a statistically significant (p < 0.05, two-tailed, paired t-test) difference. (b) For group B lip-
osomes, a concentration of 2.37 § 1.29 nmol/g (mean § standard deviation) of lipid was measured in sonicated brain
tissue (n = 3 mice), an increase of 40% over unsonicated brain tissue, which was not a statistically significant difference
(p = 0.34, two-tailed, paired t-test).

cavitation during FUS-mediated BBBD was noted in floor. Such emissions were produced in only 2 of 32
Supplementary Figure S3(d, e), particularly in group A. locations targeted in the group A animals.
Each data point represents an individual subject, and the
data points are generated by averaging acoustic strengths Histologic findings
from multiple sonication sites (2 or 3) of an individual Histologic analysis was performed on two mice,
subject. There was a good correlation between lipid con- one from each group, 24 h after Gd-Rho-labeled lipo-
centrations and harmonics (R2 = 0.67) and wideband some delivery via FUS-BBBD. In Figure 5 are example
strength (R2 = 0.535). There was no significant differ- H&E-stained sections from these animals. Both the soni-
ence in the strength harmonic (Supplementary Fig. S3b) cated and control locations appeared unaffected.
or wideband (Supplementary Fig. S3c) emissions for the
two experimental groups. However, 5 of 12 sonicated DISCUSSION
locations in the group B animals produced wideband
The results of this study indicate that delivery of
emissions that were clearly elevated above the noise
Gd-labeled liposomes to the brain after FUS-BBBD can

Fig. 5. Histologic appearance of the brain after sonication of group A or group B liposomes. The sonicated area is indi-
cated by the red square; the corresponding non-sonicated region in the contralateral hemisphere (black square) served as
a control. The sonicated and control locations both appeared unaffected. Hematoxylin and eosin. Bars = 5 mm.
ARTICLE IN PRESS
8 Ultrasound in Medicine & Biology Volume 00, Number 00, 2019

be monitored with MRI. Being able to directly image intact, particularly because accumulation was detected
drug-loaded nanoparticles with MRI can potentially in sonicated tissue at 5 h post- FUS BBBD.
ensure that an efficacious and reliable concentration of Direct comparison between groups A and B was
nanoparticles is obtained. We anticipate that this ability difficult, perhaps because of the relatively small sample
will be useful in pre-clinical studies, providing valuable numbers (n = 24 mice per group), but also because of
information that can guide the selection of acoustic other factors we observed during formulation of nano-
parameters chosen for clinical trials on ultrasound-medi- particles of two different sizes and fluorescence signal
ated liposomal drug delivery to the brain. Predicting the quantification in similar groups as described below. The
extent and “magnitude” of ultrasound-mediated BBBD signal intensity from Gd-labeled liposomes has been
can be challenging and depends on a number of factors reported to decrease with increasing liposome size (Fos-
including the amount of acoustic attenuation produced sheim et al. 1999; Ghaghada et al. 2008). This is implied
by the skull, the underlying vascular density and the in our current data as the MR signal in unsonicated brain
pharmacokinetics of the circulating microbubbles. These tissue was higher on average in group A than in group B.
factors are expected to be particularly challenging in the Further, although equivalent doses of liposomes were
heterogeneous vasculature in tumors. Having the means administered to groups A and B (based on mass of lipid
to visualize and potentially quantify the concentration of per kilogram body weight), the dose of group A lipo-
drug-loaded nanoparticles in vivo can provide important somes may have been lower than estimated because of
information that can be related to treatment outcomes in more extensive liposome extrusion. This is implied by
future studies. the higher fluorescence signal in control brain tissue for
The use of MRI for this purpose has advantages group B than group A. Nevertheless, group A liposomes
over other modalities. Others have found that radiola- and group B liposomes could be compared directly in
beled nanocarriers delivered across the BBB with FUS sonicated versus unsonicated brain tissue in a single
can be imaged with PET or SPECT imaging (Yang et al. mouse brain, as these factors are controlled in this cir-
2012; Ye et al. 2018). Although these methods excel in cumstance. Another strategy used to avoid this issue was
their sensitivity to small concentrations, MRI has a to compare the ratio of the signal in sonicated tissue
higher spatial resolution and can be used to ensure that with that in unsonicated brain tissue. The ratio of fluores-
tissue damage did not occur as a result of the sonications. cence signal intensity was slightly elevated for group A
Importantly, the use of Gd labeling offers the potential of (1.9) compared with group B (1.5), but the ratios were
serial imaging over extended periods. Even though the similar when detected via MRI (1.26 for group A vs.
distribution and elimination phases of free Gd-DTPA are 1.24 for group B). Thus, it is difficult to compare group
12 and 94 min, respectively (Weinmann et al. 1984), it is A and B liposomes directly.
still detectable up to 5 d after FUS-mediated vascular Others have investigated the time window for BBB
manipulation (Sun et al. 2015). The customized lipo- penetration of MR contrast agents ranging from <1 nm
somal-based Gd could be expected to have more exten- to 65 nm after FUS and reported that the time window
sive distribution, diffusion, and elimination phases, as it decreases with increasing agent size (Marty et al. 2012).
was detectable 5 h after BBBD in our study. We are A separate study found that there is a direct relation
excited about exploring in detail the characteristics of between cavitation activity and different applied pres-
the customized Gd in our future work. This study was sures during BBBD (Chen and Konofagou 2014). In the
designed to test a proof-of-concept on generating lipo- case of nanocarrier delivery after FUS BBBD, rhoda-
somal-based Gd-nanoparticles, delivering them via mine-labeled liposomes of 55, 120 and 200 nm were
FUS-BBBD and confirming their delivery via two differ- delivered across the BBB via FUS BBBD in a size-
ent imaging modalities, fluorescence, and MRI. We suc- dependent manner, with greater accumulation of 55-nm
ceeded in achieving our primary endpoints, and the liposomes than 120-nm liposomes (Shen et al. 2016).
findings generated several points for discussion. First, it Polymer nanoparticles have been delivered to the brain
is possible that the majority of injected liposomes were via FUS BBBD; it was reported that smaller nanopar-
intact after delivery to brain tissue via FUS and micro- ticles penetrated deeper into brain tissue than larger
bubbles. This is supported by the previous findings nanoparticles (Nance et al. 2014). Our results compare
where co-localization of fluorophores was evident after favorably with the findings of these studies, as collec-
liposome delivery, suggesting that the entire liposomal tively group A liposomes appeared to accumulate in
package crossed the BBB (Kovacs et al. 2015). The cir- brain tissue to a greater extent than group B liposomes
culation half-life of PEGylated liposomes has been after FUS BBBD. Group A liposomes resulted in a more
reported to range between 2 and 24 h in mice (Allen intense MR signal than group B. In addition, the accu-
1994; Moghimi and Szebeni 2003). Thus a significant mulation of group A liposomes in sonicated brain tissue
fraction of liposomes may have reached brain tissue was significantly greater than that in unsonicated brain
ARTICLE IN PRESS
Liposome delivery through BBB  M. ARYAL et al. 9

tissue, both via MRI and fluorescence quantification. subsequent penetration depth into brain tissue warrant
Although group B liposomes were observed visually to further investigation.
accumulate in sonicated brain tissue, accumulation lev-
els were not statistically significant compared with those CONCLUSIONS
in unsonicated brain tissue either via MRI- or fluores-
cence-based quantification. At present, not a single Overall, this work illustrates the proof-of-concept
report is available on making liposomes that are labeled of making liposomes that are labeled with an MR con-
with an MRI contrast agent to confirm BBB penetration trast agent and fluorophore, delivering them into the
and validate the MR signal with a different imaging brain parenchyma via ultrasound-mediated BBB open-
modality, such as fluorescence microscopy. Different ing and quantifying the delivered amount via MRI and
studies on “sonoporation” have indicated that different- fluorescence microscopy. Because the liposomes can be
sized agents can be delivered across the cell membrane visualized non-invasively with MRI and postmortem via
through the creation of pores that are present for a short fluorescence, they provide a useful tool to estimate the
time (Inoue et al. 2014; Lentacker et al. 2014; Rychak extent of nanocarrier extravasation through the blood
and Klibanov 2014; Sato et al. 2014; Tomizawa et al. brain barrier.
2013; Yamatomo et al. 2013). This implies that at a Acknowledgments—This work was supported by National Institutes of
given pressure, agents of different size can have different Health (NIH) Grants R25 CA153955 and P01 CA17464501.
drug penetration depths and different acoustic signatures
during BBBD with microbubbles. SUPPLEMENTARY MATERIALS
Nevertheless, we do not know whether the gadolin-
ium and fluorophore detected in the brain were still Supplementary material related to this article can
encapsulated in the liposomes or how far from the blood be found online at https://doi.org/10.1016/j.ultrasmed
vessels it had penetrated into the brain parenchyma over bio.2019.02.024.
time. Further work is needed to evaluate the dynamics of
liposome delivery into brain tissue. For example, it REFERENCES
would be interesting to investigate the kinetics and Allen TM. Long-circulating (sterically stabilized) liposomes for tar-
extent of penetration into brain tissue of Gd-labeled lipo- geted drug delivery. Trends Pharmacol Sci 1994;15:215–220.
somes compared with free Gd-DTPA after FUS BBBD. Arvanitis CD, Livingstone MS, Vykhodtseva N, McDannold N. Con-
trolled ultrasound-induced bloodbrain barrier disruption using
Given that in earlier work with doxorubicin, higher passive acoustic emissions monitoring. PloS One 2012;7:e45783.
concentrations were found in the brain when Lipo-DOX Aryal M, Park J, Vykhodtseva N, Zhang YZ, McDannold N. Enhance-
was injected before sonication (Aryal et al. 2015b), we ment in bloodtumor barrier permeability and delivery of lipo-
somal doxorubicin using focused ultrasound and microbubbles:
suspect that ultrasound could respond differently when it Evaluation during tumor progression in a rat glioma model. Phys
encounters larger molecules, 100 nm in this case, on their Med Biol 2015a;60:2511–2527.
way to BBBD disruption. Even though Lipo-Dox was not Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Multiple sessions
of liposomal doxorubicin delivery via focused ultrasound mediated
echogenic, the behaviors of microbubble oscillation, radi- bloodbrain barrier disruption: A safety study. J Control Release
ation force, microstreaming and so on could be expected 2015b;204:60–69.
to change if the larger molecules presented in the blood- Aryal M, Fischer K, Gentile C, Gitto S, Zhang YZ, McDannold N.
Effects on P-glycoprotein expression after bloodbrain barrier dis-
stream. Moreover, our finding of different acoustic signa- ruption using focused ultrasound and microbubbles. PloS One
tures with two different sizes of liposomes also suggests 2017;12:e0166061.
that this could be one of the mechanisms, but further Carpentier A, Canney M, Vignot A, Reina V, Beccaria K, Horodyckid
C, Karachi C, Leclercq D, Lafon C, Chapelon JY, Capelle L, Cornu
work is needed to support the given hypothesis. P, Sanson M, Hoang-Xuan K, Delattre JY, Idbaih A. Clinical trial
Lastly, nanocarrier features (e.g., size, shape, sur- of bloodbrain barrier disruption by pulsed ultrasound. Sci Transl
face functionality) may have a significant effect on nano- Med 2016;8:343.re2.
Chen H, Konofagou EE. The size of bloodbrain barrier opening
carrier delivery across the BBB via FUS BBBD. A induced by focused ultrasound is dictated by the acoustic pressure.
systematic characterization of these parameters and their J Cereb Blood Flow Metab 2014;34:1197–1204.
impact on nanoparticle penetration through openings Chu PC, Chai WY, Hsieh HY, Wang JJ, Wey SP, Huang CY, Wei KC,
Liu HL. Pharmacodynamic analysis of magnetic resonance imag-
created by FUS and microbubbles is in a nascent stage, ing-monitored focused ultrasound-induced bloodbrain barrier
as only a few studies have examined the effect of nano- opening for drug delivery to brain tumors. BioMed Res Int
carrier size, whereas none have explored shape, and two 2013;2013:627496.
Coluccia D, Fandino J, Schwyzer L, O’Gorman R, Remonda L, Anon J,
studies have examined nanocarriers functionalized with Martin E, Werner B. First noninvasive thermal ablation of a brain
ligands intended for markers present in the brain paren- tumor with MR-guided focused ultrasound. J Ther Ultrasound
chyma (Diaz et al. 2014; Etame et al. 2012). The impact 2014;2:17.
Diaz RJ, McVeigh PZ, O’Reilly MA, Burrell K, Bebenek M, Smith C,
of these parameters on nanoparticle diffusion through Etame AB, Zadeh G, Hynynen K, Wilson BC, Rutka JT. Focused
openings created by FUS and microbubbles and the ultrasound delivery of Raman nanoparticles across the bloodbrain
ARTICLE IN PRESS
10 Ultrasound in Medicine & Biology Volume 00, Number 00, 2019

barrier: Potential for targeting experimental brain tumors. Nanomed Dynamic study of bloodbrain barrier closure after its disruption
Nanotechnol Biol Med 2014;10:1075–1087. using ultrasound: A quantitative analysis. J Cereb Blood Flow
Etame AB, Diaz RJ, O’Reilly MA, Smith CA, Mainprize TG, Hynynen Metab 2012;32:1948–1958.
K, Rutka JT. Enhanced delivery of gold nanoparticles with thera- McDannold N, Clement GT, Black P, Jolesz F, Hynynen K. Transcra-
peutic potential into the brain using MRI-guided focused ultra- nial magnetic resonance imaging-guided focused ultrasound sur-
sound. Nanomed Nanotechnol Biol Med 2012;8:1133–1142. gery of brain tumors: Initial findings in 3 patients. Neurosurgery
Fite BZ, Kheirolomoom A, Foiret JL, Seo JW, Mahakian LM, Ingham 2010;66:323–332. discussion 332.
ES, Tam SM, Borowsky AD, Curry F-RE, Ferrara KW. Dynamic Moghimi SM, Szebeni J. Stealth liposomes and long circulating nano-
contrast enhanced MRI detects changes in vascular transport rate particles: Critical issues in pharmacokinetics, opsonization and pro-
constants following treatment with thermally-sensitive liposomal tein-binding properties. Prog Lipid Res 2003;42:463–478.
doxorubicin. J Control Release 2017;256:203–213. Nance E, Timbie K, Miller GW, Song J, Louttit C, Klibanov AL, Shih
Fossheim SL, Fahlvik AK, Klaveness J, Muller RN. Paramagnetic lipo- T-Y, Swaminathan G, Tamargo RJ, Woodworth GF, Hanes J, Price
somes as MRI contrast agents: Influence of liposomal physico- RJ. Non-invasive delivery of stealth, brain-penetrating nanopar-
chemical properties on the in vitro relaxivity. Magn Reson Imaging ticles across the bloodbrain barrier using MRI-guided focused
1999;17:83–89. ultrasound. J Control Release 2014;189:123–132.
Frenzel T, Lengsfeld P, Schirmer H, Hütter J, Weinmann HJ. Stability Park J, Zhang Y, Vykhodtseva N, Jolesz FA, McDannold NJ. The
of gadolinium-based magnetic resonance imaging contrast agents kinetics of blood brain barrier permeability and targeted doxorubi-
in human serum at 37 degrees C. Invest Radiol 2008;43:817–828. cin delivery into brain induced by focused ultrasound. J Control
Ghaghada K, Hawley C, Kawaji K, Annapragada A, Mukundan S. T1 Release 2012;162:134–142.
relaxivity of core-encapsulated gadolinium liposomal contrast Poon C, McMahon D, Hynynen K. Noninvasive and targeted delivery
agents—Effect of liposome size and internal gadolinium concentra- of therapeutics to the brain using focused ultrasound. Neurophar-
tion. Acad Radiol 2008;15:1259–1263. macology 2017;120:20–37.
Gulani V, Calamante F, Shellock FG, Kanal E, Reeder SB. Gadolinium Rychak JJ, Klibanov AL. Nucleic acid delivery with microbubbles and
deposition in the brain: Summary of evidence and recommenda- ultrasound. Adv Drug Deliv Rev 2014;72:82–93.
tions. Lancet Neurol 2017;16:564–570. Sato T, Mori S, Arai Y, Kodama T. The combination of intralymphatic
Hsu PH, Wei KC, Huang CY, Wen CJ, Yen TC, Liu CL, Lin YT, Chen chemotherapy with ultrasound and nano-/microbubbles is efficient
JC, Shen CR, Liu HL. Noninvasive and targeted gene delivery into in the treatment of experimental tumors in mouse lymph nodes.
the brain using microbubble-facilitated focused ultrasound. PloS Ultrasound Med Biol 2014;40:1237–1249.
One 2013;8:e57682. Shen Y, Guo J, Chen G, Chin CT, Chen X, Chen J, Wang F, Chen S,
Hynynen K, Jones RM. Image-guided ultrasound phased arrays are a Dan G. Delivery of liposomes with different sizes to mice brain
disruptive technology for non-invasive therapy. Phys Med Biol after sonication by focused ultrasound in the presence of microbub-
2016;61:R206–R248. bles. Ultrasound Med Biol 2016;42:1499–1511.
Hynynen K, McDannold N, Vykhodtseva N, Jolesz FA. Noninvasive Sun T, Samiotaki G, Wang S, Acosta C, Chen CC, Konofagou EE.
MR imaging-guided focal opening of the bloodbrain barrier in Acoustic cavitation-based monitoring of the reversibility and per-
rabbits. Radiology 2001;220:640–646. meability of ultrasound-induced bloodbrain barrier opening. Phys
Inoue H, Arai Y, Kishida T, Shin-Ya M, Terauchi R, Nakagawa S, Med Biol 2015;60:9079–9094.
Saito M, Tsuchida S, Inoue A, Shirai T, Fujiwara H, Mazda O, Thevenot E, Jord~ao JF, O’Reilly MA, Markham K, Weng YQ, Foust
Kubo T. Sonoporation-mediated transduction of siRNA amelio- KD, Kaspar BK, Hynynen K, Aubert I. Targeted delivery of self-
rated experimental arthritis using 3 MHz pulsed ultrasound. Ultra- complementary adeno-associated virus serotype 9 to the brain,
sonics 2014;54:874–881. using magnetic resonance imaging-guided focused ultrasound.
Kanda T, Osawa M, Oba H, Toyoda K, Kotoku J, Haruyama T, Take- Hum Gene Ther 2012;23:1144–1155.
shita K, Furui S. High signal intensity in dentate nucleus on unen- Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S,
hanced T1-weighted MR images: Association with linear versus Sueishi M. Sonoporation: Gene transfer using ultrasound. World J
macrocyclic gadolinium chelate administration. Radiology Methodol 2013;3:39–44.
2015;275:803–809. Vlachos F, Tung YS, Konofagou EE. Permeability assessment of the
Kovacs Z, Luciani P, Roth P, Leroux JC, Martin E, Werner B. Blood focused ultrasound-induced bloodbrain barrier opening using
brain barrier opening-based local delivery of 80 nm-sized liposomes in dynamic contrast-enhanced MRI. Phys Med Biol 2010;55:5451–5466.
mice using pulsed focused ultrasound. J Ther Ultrasound 2015;3:P24. Wang S, Olumolade OO, Sun T, Samiotaki G, Konofagou EE. Nonin-
Lentacker I, De Cock I, Deckers R, De Smedt SC, Moonen CTW. vasive, neuron-specific gene therapy can be facilitated by focused
Understanding ultrasound induced sonoporation: Definitions and ultrasound and recombinant adeno-associated virus. Gene Ther
underlying mechanisms. Adv Drug Deliv Rev 2014;72:49–64. 2015;22:104–110.
Liao AH, Liu HL, Su CH, Hua MY, Yang HW, Weng YT, Hsu PH, Weinmann HJ, Laniado M, Mützel W. Pharmacokinetics of GdDTPA/
Huang SM, Wu SY, Wang HE, Yen TC, Li PC. Paramagnetic per- dimeglumine after intravenous injection into healthy volunteers.
fluorocarbon-filled albumin(Gd-DTPA) microbubbles for the Physiol Chem Phys Med NMR 1984;16:167–172.
induction of focused-ultrasound-induced bloodbrain barrier open- Yamatomo N, Iwagami T, Kato I, Masunaga S-I, Sakurai Y, Iwai S,
ing and concurrent MR and ultrasound imaging. Phys Med Biol Nakazawa M, Ono K, Yura Y. Sonoporation as an enhancing
2012;57:2787–2802. method for boron neutron capture therapy for squamous cell carci-
Lipsman N, Meng Y, Bethune AJ, Huang Y, Lam B, Masellis M, Herr- nomas. Radiat Oncol 2013;8:280.
mann N, Heyn C, Aubert I, Boutet A, Smith GS, Hynynen K, Black Yang FY, Wang HE, Liu RS, Teng MC, Li JJ, Lu M, Wei MC, Wong
SE. Bloodbrain barrier opening in Alzheimer’s disease using TT. Pharmacokinetic analysis of 111In-labeled liposomal doxorubi-
MR-guided focused ultrasound. Nat Commun 2018;9:2336. cin in murine glioblastoma after bloodbrain barrier disruption by
Martin E, Jeanmonod D, Morel A, Zadicario E, Werner B. High-inten- focused ultrasound. PLoS ONE 2012;7:e45468.
sity focused ultrasound for noninvasive functional neurosurgery. Ye D, Sultan D, Zhang X, Yue Y, Heo GS, Kothapalli SVVN, Lueh-
Ann Neurol 2009;66:858–861. mann H, Tai Y, Rubin JB, Liu Y, Chen H. Focused ultrasound-
Marty B, Larrat B, Van Landeghem M, Robic C, Robert P, Port M, Le enabled delivery of radiolabeled nanoclusters to the pons. J Control
Bihan D, Pernot M, Tanter M, Lethimonnier F, Meriaux S. Release 2018;283:143–150.

You might also like