You are on page 1of 8

Journal of Drug Delivery Science and Technology 77 (2022) 103882

Contents lists available at ScienceDirect

Journal of Drug Delivery Science and Technology


journal homepage: www.elsevier.com/locate/jddst

Effect of ultrasonic parameters on gene transfection efficiency and cell


viability of the multifunctional microbubble in vitro
Tho Anh Thi Tran a, Toan Phi Nguyen a, Nhung Hong Thi Duong a, Duy Hieu Truong b,
Bac Xuan Nguyen a, Cuong Khac Bui c, Lap Thi Nguyen a, *
a
Hanoi University of Pharmacy, Hanoi, 10000, Viet Nam
b
Quang Binh Pharmaceutical Joint-stock Company, Quang Binh, 510000, Viet Nam
c
Vietnam Military Medical University, Hanoi, 10000, Viet Nam

A R T I C L E I N F O A B S T R A C T

Keywords: Studies have reported that microbubbles combined with focused ultrasound (FUS) can increase the membrane
Acoustic parameter permeability of tumor cells by triggering membrane perforation (sonoporation) to improve brain tumor-targeted
Brain tumor gene delivery. However, few studies focused on how to increase the efficiency of gene delivery to brain tumor
Focused ultrasound
cells with minor cell damage. The aim of this in vitro study was to find optimal parameters for efficient gene
Gene delivery
transfer into glioma cells using the VEGFR2-targeted cationic microbubbles gene vector (MB) for future brain
Microbubble
tumor-targeted gene delivery. Transfection efficiency and cell viability were used to determine the optimal
transfection parameters. Various MB concentrations (2 × 107 to 16 × 107/mL), post-transfection incubation
times (1–3 days), acoustic pressures (0–1400 kPa), duty cycles (0.5–5%), pulse repetition frequencies (PRF)
(0.5–2000 Hz) and cycle numbers (25–10000) were investigated. The results showed that the MB concentration
of 4 × 107 MB/ml and the incubation time of 2 days were optimal conditions for maximum gene transfection
efficiency and minimum cell damage. In addition, gene transfection efficiency and cell viability depended
strongly on the acoustic exposure conditions. The optimal FUS parameters include an acoustic pressure of 700
kPa, a duty cycle of 5%, a PRF of 5Hz, and a cycle number of 10000. These findings support those MB con­
centrations and FUS parameters of the MB-mediated gene delivery system influence gene transfer efficiency and
cell viability. They also describe our novel development of multifunctional MB for FUS-triggered gene delivery in
brain tumor cells.

1. Introduction responses they have caused raise safety concerns, prompting the
development of non-viral delivery methods [3]. The development of
Brain cancer is one of the causes that make patients most likely die non-viral vectors, typically the gene-carrying microbubble system, has
from [1]. Conventional brain tumor treatments include surgery, radia­ improved gene transfer efficiency, specificity, and biosafety [4].
tion, and chemotherapy. Adversities in brain tumor treatments are due Microbubbles are gas-encapsulating particles that can be surrounded by
to numerous anatomical and physiological characteristics of the brain, an electrically charged membrane capable of trapping nucleic acid
making it difficult for drugs to invade, such as endothelial cell imper­ molecules.
meability, local enzymes, receptors, and, especially, the blood-brain Gene delivery by microbubbles, on the other hand, still has relatively
barrier [2]. It is urgent to develop a suitable drug delivery system to low efficiency compared to viral or other delivery methods such as lip­
effectively deliver anti-cancer drugs to tumor tissue to meet these ofection and electroporation [5]. This phenomenon likely results from
challenges. insufficient numbers of microbubbles within the focused ultrasound
Gene therapy is a promising approach to brain cancer treatment. (FUS) field or in direct contact with the cell membrane. Also, glioma
Successful gene therapy requires the safe and effective transport of cancer cells are typically surrounded by normal neural cells, so a tumor
therapeutic genes across the cell membrane. Although viral vectors are cell-targeted gene delivery platform needs to be developed to scale back
effective for gene transfection, the inflammatory and immunogenic off-target effects. A previous study indicated that the structure of outer

* Corresponding author.
E-mail address: lapnt@hup.edu.vn (L.T. Nguyen).

https://doi.org/10.1016/j.jddst.2022.103882
Received 19 June 2022; Received in revised form 2 October 2022; Accepted 7 October 2022
Available online 13 October 2022
1773-2247/© 2022 Elsevier B.V. All rights reserved.
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

microbubbles’ shells allows them to construct covalent bonds with Biotec (Germany). Alarma blue assay was purchased from AbDSerotec,
various ligands, which establishs a novel targeted microbubble gene Oxford, UK. The red firefly luciferase gene (pLUC, 6.6 kb) was purchased
delivery system [6]. Therefore, a more refined design that involves from Origene Technologies, USA. Plasmid Maxi Kit was obtained from
modifying particular antibodies or ligands that bind to Macherey-Nagel, Germany. The U87 glioma cell line was obtained from
disease-associated molecular markers expressed on vascular endothelial American Type Culture Collection. All other reagents were of analytical
cells of the brain tumor is essential to promote local aggregations of grade.
microbubbles to increase gene transfection efficiency.
The vascular endothelial growth factor receptor 2 (VEGFR2) among 2.2. Plasmid preparation
angiogenesis molecular markers is a key player. VEGFR2 is a high-
affinity receptor for vascular endothelial growth factor-A (VEGF-A) An expression vector for the pLUC was all driven by a cytomegalo­
[7]. VEGFR2 expression has been associated with tumor growth and virus (CMV) promoter. Plasmid DNA was purified from the culture of
aggressiveness in various cancers [8]. VEGFR2 is also highly expressed Escherichia coli DH5a with the Plasmid Maxi Kit, following the protocol
in tumor cells and endothelial cells in blood vessels that nurture brain provided by the manufacturer. The plasmid DNA purity was assessed by
tumors [9]. Thus, attaching exogenous antibodies against VEGFR2 to the measuring absorption at 260 nm wavelength (A260) by NanoDrop
microbubble’s outer shell is a promising strategy to target specific sites (NanoDrop 2000, Thermo Fisher Scientific, IL, USA). The A260:A280
in the vicinity of tumors to unload therapeutic genes. ratio of plasmid DNA was between 1.8 and 1.9. The quality of the pu­
In addition, the combination of microbubbles and FUS increases gene rified plasmid was further analyzed by enzyme restriction and subse­
transfer efficiency. The principle is that microbubbles are vehicles to quent electrophoresis on a 0.8% agarose gel.
deliver therapeutic genes, and ultrasound forms “openings” between
cells by ultrasonic-activated oscillations, which also break the micro­ 2.3. Preparation of DNA-loaded VEGFR2 targeted cationic microbubbles
bubbles [10]. The cavitation effects from circulating microbubbles can
produce local intracellular gap junctions of the cerebral vesicular The cationic microbubbles with anti-VEGFR2 antibodies were pre­
endothelial cells, endocytotic cellular uptake, and vessel disruption. pared using the thin-film hydration technique described in a previous
Through sonoporation, microbubble cavitation may enhance and study [14]. Firstly, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC),
develop drug delivery routes, allowing pharmaceutical substances to be 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyeth­
taken up. In particular, microbubbles oscillate steadily or violently ylene glycol)-2000] (DSPE-PEG 2000), 1,2-distearoyl-sn-glycero-3-
grow/collapse in response to ultrasound depending on the applied phosphoethanolamine-N-[biotinyl(polyethylene glycol)-2000] (DSPE–
acoustic pressure. Dynamic microbubble motion combined with fluid PEG2000–Biotin) and 1,2-dipalmitoyl-3-trimethylammonium-propane
motion can exert mechanical stresses on the surrounding tissues, (DPTAP) were dissolved in chloroform at a weight ratio of 9:1:1:1, and
resulting in reversible cell membrane penetration that can help with drained via rotary evaporator to form a dried film. Secondly, the film was
medication administration. Additionally, intense microbubble collapse hydrated with a 5 μl/ml glycerol-PBS buffer in an airtight vial. The gas in
(inertial cavitation) causes secondary effects such as micro-jet and the vial was removed and refilled with perfluoropropane (C3F8) gas.
shockwaves, which enhance delivery capacity. Cavitation-induced ef­ Thirdly, the vial was shaken by an agitator for 45 s to produce micro­
fects can result in cell morphology changes and long-term bio-effects (e. bubbles with a hydrophobic gas core and lipid exterior. To eliminate free
g., temporal cell permeability changes, cell lysis), which can help lipids, biotinylated microbubbles were centrifuged for 2 min at 6000 rpm
improve therapeutic agent delivery and uptake [11]. (Mini-micro centrifuge, Bertec Enterprise Co., Ltd., Taiwan) and resus­
Moreover, the results obtained from a previous study indicated that pended in glycerol-PBS buffer.
each tumor cell line has its optimum parameter combination and opti­ Fourthly, the biotinylated microbubbles were then incubated for 10
mum sonoporation efficiency [12]. Another study also revealed that min at room temperature with 1 mg of avidin (in an aqueous solution of
transfection efficiency and loss of cell viability could be attributed to the 10 mg/ml). Next, the microbubbles were centrifuged again to eliminate
chosen ultrasound parameters [13]. Therefore, proper optimization of excess avidin and resuspended in glycerol-PBS buffer. Fifthly, 75 μg of
ultrasound parameters must be done to achieve therapeutic results and CD309 (VEGFR-2) biotinylated antibody was added to the avidin-
minimize unwanted damage to healthy tissues. biotinylated microbubble suspension and incubated for 10 min. The
In a previous study, a multifunctional microbubble system that microbubbles were centrifuged again and resuspended to remove excess
carries both a gene encoding luciferase and an anti-VEGFR2 antibody to biotinylated antibodies. Finally, an appropriate amount of pLUC in
target brain tumors was fabricated [14]. However, the quantitative deionized water was incubated with the microbubbles by slowly mixing
dependence of sonoporation on FUS exposure parameters was not well for 30 min to speed up DNA adsorption onto the microbubble surface.
documented and, therefore, is the subject of this study. The gene Hence, DNA-loaded VEGFR2 targeted cationic microbubbles (MB) were
encoding luciferase, an enzyme that produces bioluminescence, has formed.
been used for detection and biological effect evaluation. In combination
with FUS, the influence of microbubbles’ concentrations and acoustic 2.4. Evaluating pLUC gene transfection and cell viability in vitro
exposure parameters on gene transfection and cell viability using the
microbubble system mentioned above was determined in vitro. Gene transfection and cell viability were assessed using U87 glioma
cells with a modified protocol based on previous studies [15,16]. More
2. Materials and methods specifically, U87 glioma cells were cultured in Dulbecco’s Modified
Eagle’s medium/Ham’s Nutrient Mixture F-12 (DMEM/F12), supple­
2.1. Materials mented with 10% fetal bovine serum, penicillin/streptomycin at 37 ◦ C.
The day before the experiments, 4 × 104 cells per well were seeded in a
The lipids, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 24-well plate. Then, the VEGFR2-targeted cationic microbubbles were
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyeth­ incubated with 2 μg pLUC in 4 ml DMEM for 30 min at room tempera­
ylene glycol)-2000] (DSPE-PEG 2000), 1,2-distearoyl-sn-glycero-3- ture. After the medium in a 24-well plate was removed, DNA-loaded MB
phosphoethanolamine-N-[biotinyl(polyethylene glycol)-2000] (DSPE– were added to each well. An inverted setup promoted cell targeting ef­
PEG2000–Biotin) and 1,2-dipalmitoyl-3-trimethylammonium-propane ficiency by providing close contact between floated MB and cells for 10
(DPTAP), were obtained from Avanti Polar Lipids, AL, USA. Avidin min and then turning upright for FUS treatment. Next, serial acoustic
was purchased from Sigma Aldrich, MO, USA. CD309 (VEGFR-2) biotin, parameters of FUS (acoustic pressure, cycle number, and pulse repeti­
a human monoclonal antibody (0.5 mg/ml), was obtained from Miltenyi tion frequencies (PRF)) and concentration of MB (2-16 × 107 MB/mL)

2
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

were applied to optimize the transfection efficiency. FUS exposure 3. Results


(acoustic pressures of 250–1400 kPa, cycle numbers of 25–10000, PRF
of 0.5–2000 Hz, duty cycles of 0.0125–5%, and exposure duration of 1 3.1. Effect of MB concentration and post-transfection incubation time
min) was applied with a 6 mm diameter FUS probe (model V302,
Olympus NDT Inc., MA, USA). An RF power amplifier (150A100B, AR, The cells were exposed to FUS at a center frequency of 1 MHz and a
PA, USA) was used to generate the sonication. Four hours following FUS constant acoustic pressure of 700 kPa, a cycle number of 10000, a PRF of
sonication, the cells were washed with fresh PBS, and a fresh culture 5Hz, a duty cycle of 5%, exposure time of 1 min, and 1.5 μg pLUC per
medium was then added. A minimum of 15 replicate data points were well. A range of MB concentrations ((2-16) × 107 MB/ml) and post-
collected at each tested condition. The pLUC expression and cell transfection incubation times (1, 2, and 3 days) were evaluated for
viability were individually measured by bioluminescence imaging and their effects on gene transfection efficiency and cell viability. As shown
Alarma blue assay. in Fig. 1A, the transfection efficiency increased first and then decreased
For bioluminescence assays, pLUC transfected cells were measured with the increase of microbubble concentrations. On day one and day
1–3 days following ultrasound exposure by bioluminescence imaging two post-transfection, the luciferase expression reached a peak at an MB
(IVIS-200, Xenogen Corporation, Ca, USA). Thirty seconds before im­ concentration of 4 × 107 MB/ml. However, after three days post-
aging, D-luciferin (150 μg/mL) was added to each well (Biosynth AG, transfection, the luciferase expression reached a peak at an MB con­
Staad, Switzerland). Photon flux for each well was measured at 1.5 min centration of 8 × 107 MB/ml. On the other hand, the results from Fig. 1B
after adding D-luciferin by an IVIS charge-coupled device camera. Serial showed that the cell viability tended to decrease with increasing con­
1-min image frames were collected until the bioluminescent signal peak centration of microbubbles: the highest was at the MB concentration of
was reached. Then, image analysis of all frames was performed by 2 × 107 MB/ml, then gradually decreased to the lowest at 16 × 107 MB/
quantifying the average radiance (photon/s/cm2/steradian, p/s/cm2/ ml (Tukey HSD, p < 0.001).
sr) of each wall via Living Image 3.0 software (Caliper Life Sciences, MA, The highest average radiances on day one and day two post-
USA). transfection were (14.46 ± 2.35) × 103 and (18.52 ± 1.55) × 103 (p/
The cell viability was converted by the percentage of formazan s/cm2/sr), respectively. At the MB concentration of 4 × 107 MB/ml, the
production assaying the Alarma blue assay with a plate reader (Tecan cell viability remained above 70%. However, the highest average radi­
Infinite M200, Tecan Trading AG, Switzerland) at an absorbance ance on day three post-transfection was (18.04 ± 0.39) × 103 (p/s/cm2/
wavelength of 600 nm. Alarma blue (5 mg/mL in serum-free medium) sr) at the MB concentration of 8 × 107 MB/ml, while the cell viability
was added to each well and incubated at 37 ◦ C for 2 h. The absorbance of was quite low (45.77 ± 0.93%). Considering the transfection efficiency
each plate was measured with a fluorescence plate reader system at a and cell viability, the MB concentration of 4 × 107 MB/ml and the time
wavelength of 570 nm and a reference wavelength of 600 nm. The cell of two days post-transfection were determined as optimal for subsequent
viability was calculated according to the formula provided below. experiments.

(εOX )600nm × A570nm − (εOX )570nm × A600nm


cell viability (%) = 3.2. Effect of acoustic pressure
(εOX )600nm × A0,570nm − (εOX )570nm × A0,600nm
The inertial cavitation dose (ICD) and subharmonic intensity were
Where εox = molar extinction coefficient of Alamar Blue™ oxidized form
measured in this experiment. Investigating the acoustic properties of
(blue), A = absorbance of experimental wells, and A0 = absorbance of
MB, the thresholds for both stable and inertial cavitation of MB occurred
positive growth control wells (cells with Alamar Blue without test
at 250 kPa and 350 kPa, respectively (Fig. 2).
agent).
The influence of acoustic pressure on gene transfection efficiency
and cell viability was further investigated with varied acoustic pressures
2.5. Statistical analysis
(250–1400 kPa). Cells were insonated with FUS at a center frequency of
1 MHz and a constant cycle number of 10000, PRF of 5 Hz, the duty
Data are presented for each condition as mean ± standard deviation
cycle of 5%, exposure time of 1 min, the MB concentration of 4 × 107
(SD) from three separate experiments. All statistical tests were per­
MB/ml, and 1.5 μg of pLUC per well. The luciferase expression of cells
formed using R Statistical Software (version 2.14.0). Statistical signifi­
was observed on day two post-transfection. The results are presented in
cance of observed effects was determined by analysis of variance
Fig. 3.
(ANOVA) and Post hoc Tukey tests. A p-value < 0.05 was considered
As shown in Fig. 3A, increasing acoustic pressure from 250 to 400
statistically significant.
kPa provided no significant increase in luciferase expression (group a,
Tukey HSD, p > 0.05). After that, increasing the acoustic pressure from

Fig. 1. (A) Gene transfection efficiency and (B) cell viability under various microbubble concentrations and post-transfection incubation time. The two-
way ANOVA test showed that each group of microbubble concentration and time after ultrasound exposure demonstrated luciferase expression statistically sig­
nificant (ANOVA, p < 0.001) and cell viability statistically significant (ANOVA, p < 0.001).

3
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

acoustic pressure from 700 kPa to 900 kPa (group b and c, Tukey HSD, p
< 0.001). Taken together, Fig. 3A and B suggested that we should keep
the optimal acoustic pressure at 700 kPa for all subsequent tests.

3.3. Effect of PRF, duty cycle, and cycle number

3.3.1. Effect of PRF and duty cycle


The PRF is an important parameter of burst waveforms. The effect of
PRF (0.5–5Hz) and duty cycle (0.5–5%) on the gene transfection effi­
ciency and cell viability are shown in Fig. 4. Cells were insonated with
FUS at a center frequency of 1 MHz and a constant acoustic pressure of
700 kPa, cycle number of 10000, exposure time of 1 min, with the MB
concentration of 4 × 107 MB/ml, and 1.5 μg pLUC per well.
The result clarified that increasing the PRF and duty cycle while
maintaining a constant cycle number of 10000 resulted in a significant
increase in luciferase expression (groups a, b, and c, Tukey HSD, p <
Fig. 2. Inertial cavitation dose and subharmonic intensity of gene-loaded
0.001) (Fig. 4A) and a decrease in cell viability (group d, a, b, and c,
microbubbles under various acoustic pressure.
Tukey HSD, p < 0.001) (Fig. 4B). As shown in Fig. 4A and B, the
transfection efficiency increased with the increase of PRF and duty cycle
400 kPa to 700 kPa resulted in a significant increase in luciferase until reaching its highest level, as PRF and duty cycle were 5 Hz and 5%,
expression (group a, b, and c, Tukey HSD, p < 0.001). However, respectively, while the cell viability remained close to 70%.
increasing the acoustic pressure from 700 kPa to 1400 kPa resulted in a
significant decrease in luciferase expression (group c, d, e, and f, Tukey 3.3.2. Effect of PRF and cycle number
HSD, p < 0.001). Cells were insonated with FUS at the center frequency of 1 MHz and
Meanwhile, in Fig. 3B, the cell viability showed a significant constant acoustic pressure of 700 kPa, the duty cycle of 5%, exposure
decrease with the increase in acoustic pressure. When the acoustic time of 1 min, the MB concentration of 4 × 107 MB/ml, and 1.5 μg pLUC
pressure increased from 300 kPa to 700 kPa, the cell viability decreased per well. The effect of PRF and cycle number is shown in Fig. 5.
but remained above 70% (group a and b, Tukey HSD, p < 0.001). Increasing the cycle number from 25 to 10000 while reducing the
However, there is a sudden decrease in cell viability when increasing the PRF from 2000 Hz to 5 Hz resulted in a significant increase in luciferase

Fig. 3. (A) Gene transfection efficiency and (B) cell viability under various acoustic pressure. A. Gene transfection efficiency (a, b, c, d, e, f: different subgroups
that are statistically significant (Tukey HSD, p < 0.001)); B. Cell viability (d, a, b, c: different subgroups that are statistically significant (Tukey HSD, p < 0.001)).

Fig. 4. (A) Gene transfection efficiency and (B) cell viability under various duty cycles and pulse repetition frequencies (PRF). A. Gene transfection effi­
ciency (a, b, c: different subgroups that are statistically significant (Tukey HSD, p < 0.001)); B. cell viability (d, a, b, c: different subgroups that are statistically
significant (Tukey HSD, p < 0.001)).

4
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

Fig. 5. (A) Gene transfection efficiency and (B) cell viability under various pulse repetition frequencies (PRF) and cycle numbers. A. Gene transfection
efficiency (a, b, c, d: different subgroups that are statistically significant (Tukey HSD, p < 0.001)); B. cell viability (a, b, c: different subgroups that are statistically
significant (Tukey HSD, p < 0.001)).

expression, as shown in Fig. 5A (group a, b, c, and d, Tukey HSD, p < number of 10000, a PRF of 5 Hz, a duty cycle of 5%, exposure time of 1
0.001). On the other hand, this caused a significant decrease in cell min, the MB concentration of 4 × 107 MB/ml, 1.5 μg pLUC per well, and
viability (group a, b, and c, Tukey HSD, p < 0.001) (Fig. 5B). The gene a luciferin concentration of 150 μg/ml. We examined the groups that
transfection efficiency reached a maximum level when the PRF and lacked one or more than one of the treatments, including FUS, MB,
cycle numbers were 5 Hz and 10000, respectively, while the cell pLUC, and luciferin.
viability remained close to 70%. Luciferin, the luciferase substrate, was responsible for the charac­
teristic light emission. The luciferase expression and cell viability are
3.3.3. Effect of cycle number and duty cycle shown in Fig. 7. There was no luciferase expression in groups where cells
Cells were insonated with FUS at the center frequency of 1 MHz and lacked one or more treatments, including FUS, MB, pLUC, and luciferin.
constant acoustic pressure of 700 kPa, PRF of 5 Hz, exposure time of 1 Group 7 showed that no gene delivery was observed without MB, indi­
min, and the MB concentration of 4 × 107 MB/ml and 1.5 μg pLUC per cating that FUS alone does not affect gene delivery. In addition, in the
well. The effect of cycle number and duty cycle is shown in Fig. 6. absence of FUS (group 8), no gene delivery occurred under any condi­
The results showed that increasing the cycle number and duty cycle tions, confirming that MB alone cannot deliver the gene. Group 8 still
resulted in a significant increase in luciferase expression (group a, b, c, showed no luciferase expression because MB size (≈1 μm) was too large
and d, Tukey HSD, p < 0.001 (Fig. 6A) and a decrease in cell viability and too solid for cell endocytosis and gene transfection. This indicated
(group c, a, and b, Tukey HSD, p < 0.001) (Fig. 6B). In addition, the that cavitation and sonoporation triggered by FUS were required for
highest transfection efficiency achieved at cycle number and duty cycle gene transfection in cells. When these components were sufficient, the
were 10000 and 5%, respectively, while the cell viability remained close gene transfection efficiency increased sharply in group 9, demonstrating
to 70%. that these factors were unlikely to be independent, and the process was
Therefore, with the results from Figs. 4–6, we selected a cycle strongly related to cavitation and cavitation-induced activities. Cell
number of 10000, a duty cycle of 5%, and a PRF of 5Hz as the optimum viability varied between groups but remained around 70%. Group 5 and
parameters for the follow-up experiments. 9 showed lower cell viability due to MB cavitation.

3.4. Effect of other exposure factors 4. Discussion

After choosing optimal data from the six parameters discussed above Microbubble-mediated ultrasound has great potential for clinical
(MB concentration, time after ultrasound exposure, acoustic pressure, translation in oncology because it is safe and non-invasive. This
PRF, duty cycle, and cycle number), cells were insonated with FUS at a approach can create temporary and reversible openings in vessel walls
center frequency of 1 MHz and a constant pressure of 700 kPa, a cycle and cellular membranes through sonoporation, allowing enhanced

Fig. 6. (A) Gene transfection efficiency and (B) cell viability under various cycle numbers and duty cycles. A. Gene transfection efficiency (a, b, c, d: different
subgroups that are statistically significant (Tukey HSD, p < 0.001)); B. Cell viability (c, a, b: different subgroups that are statistically significant (Tukey HSD, p
< 0.001)).

5
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

Fig. 7. Gene transfection efficiency and cell viability under exposure factors.
**: cell viability and gene transfection efficiency are statistically significant (Tukey HSD, p<0.01).

transport of therapeutic agents across these biological barriers in the luciferase gene. This trend is similar to the results shown in some pre­
insonated region. Most studies have shown successful delivery of vious studies [12,18–22]. For instance, Shapiro et al. found that the
ultrasound-guided microbubble systems. However, the reported de­ concentration of microbubbles must be kept in a very tight range to
livery efficiency is inconsistent. Thus, parameter optimization may need achieve enhanced sonoporation. Their results indicated that delivery of
to be assessed to enable maximum gene transfection efficiency and 5 × 105 MB/ml, which is lower than our result of 4 × 107 MB/ml, led to
minimum cell death. This study tested the hypothesis that varying significantly enhanced transfection compared to lower and higher dos­
acoustic parameters of FUS-mediated gene delivery by MB influence ages of microbubbles. Below this concentration, there may be too little
efficiency and cell viability to find the most suitable parameters for impact from microbubble oscillation, and the microbubbles may be
efficient gene transfection on glioma cells for future brain tumor- more rapidly destroyed [18]. However, if the microbubble concentra­
targeted gene therapy. The effects of microbubble concentration, time tion is too high, the microbubbles may shield each other from the
after ultrasound exposure, and FUS parameters (acoustic pressure, duty acoustic field, and the effect would be attenuated. Also, too many
cycle, PRF, and cycle number) on gene transfection efficiency and cell microbubbles could cause increased tissue damage [18].
viability were investigated in a glioma cell line. We chose the U87 gli­ Results from Fig. 1 also initially evaluated the effect of time after
oma cell line because human glioblastomas are thought to be initiated gene transfer on the efficiency of MB combined with FUS. The cell
by glioma stem-like cells, and these cells express higher levels of VEGF viability tended to decrease with post-transfection incubation time with
receptor 2 (VEGFR-2) [17]. Therefore, this VEGFR2-targeted MB was increasing microbubble concentration, probably due to increased
fabricated to increase gene delivery efficiency by taking advantage of membrane damage with larger microbubble-to-cell ratios. However,
the active targeting capability. within the limitations of the study, it cannot be conducted at the next
Prior to evaluation of cell viability and gene transfection, physical time point after gene transfection (after three days), so the study has not
characteristics of the tested microbubble have been assessed, which proven the specific mechanism of changes in the gene-delivery system
include the binding efficiency of antibodies onto the microbubble’s over time, as well as where is the most appropriate timeline for gene
surface (data not shown). The amount of antibody added to create the transfection and cell viability. In the subsequent study, we will conduct a
targeted microbubbles was excessive enough compared to the total more detailed analysis of the effect of time (at more time points) to find
estimated area of microbubbles. Thus, we indirectly assumed that the out the mechanism of the microbubble system combined with
number of antibodies on the surface of each microbubble was abundant ultrasound.
enough to target the cells. Next, we examined the acoustic properties of MB. Stable cavitation
The experimental conditions used appear critical for efficient gene and inertial cavitation of MB started at 250 kPa and 350 kPa, respec­
expression in cells. First, we looked at the effect of MB concentration on tively (Fig. 2). Results also indicated that subharmonic intensity and ICD
gene delivery efficiency. Fig. 1 indicated that the transfection capacity were in a strong positive correlation with acoustic pressure. An increase
tended to gradually increase to a threshold and then decrease over time in acoustic pressure under the threshold of inertial cavitation caused
at all concentrations. Meanwhile, cell viability tended to decrease over non-linear expansion and compression of the MB and resulted in the
time when increased microbubble concentration. This result can be emission of non-linear harmonic signals at multiples of the transmitted
explained by lower cell viability, so these damaged cells were unable to frequency, which was known as stable cavitation. Also, the oscillating
increase gene expression. Too high a concentration of microbubbles can MB generated steady microstreaming and large shear forces [23].
cause cytotoxicity and mass cell death and reduce the expression of the Higher acoustic pressure over the threshold caused forced expansion

6
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

and compression of MB, resulting in bubble disruption. saturate or eventually decrease as extremely large membrane pores are
Furthermore, increasing the acoustic pressure from 400 kPa to 700 created, which is considered irreparable damage to most cell mem­
kPa resulted in a significant increase in luciferase expression (groups b branes. These pores allow macromolecules to pass and subsequently
and c, Tukey HSD, p < 0.001). This result indicated that an acoustic cause cell lysis [33]. Therefore, optimizing the parameters for
pressure threshold greater than 400 kPa was required for gene trans­ microbubble-mediated ultrasound transfection is inevitable to have a
fection because inertial cavitation of MB started from 400 kPa (Fig. 3A). safe and efficient transport of therapeutic materials.
However, increasing the acoustic pressure from 700 kPa to 1400 kPa There is a limitation associated with our study. The study was con­
resulted in a noticeable decrease in luciferase expression (groups d, e, ducted only on one glioma cell line, namely U87 glioma cell line. It
and f, Tukey HSD, p < 0.001). Such a reduction in gene transfection is would be interesting to determine whether our results of optimal ul­
that the cells have reached the cytotoxic threshold (Fig. 3B). Increasing trasonic parameters are the case for other glioma cell lines or not. Hence,
sonoporation also resulted in a larger proportion of cells that are unable further studies need to be done on additional human glioma cell lines.
to repair their damaged cell membranes [12,24,25]. The reason may be However, we contend that a similar trend of cell viability and gene
that high acoustic pressure will enhance the cytotoxic process, as pre­ transfection efficiency in other glioma cell lines would also be observed
viously proven in some studies [26–31]. Cell membrane permeability due to sonoporation. In addition, the reason why we chose U87 cell line
increases with decreasing acoustic pressure, and viability falls. The is because it is the most widely used human glioblastoma cell line in
number of permeabilized cells, on the other hand, decreases as the brain cancer research. Glioblastoma multiforme is also the most com­
pressure amplitude increases. Cells do not appear to be permeabilized or mon malignant brain cancer. Therefore, in this study, we want to focus
destroyed below a certain level. Higher than a threshold of 2–3 MPa on U87 cell line only.
acoustic pressures (higher than our result) may be less favorable for
producing reversible permeabilization, according to R. Karshafian et al. 5. Conclusion
[27]. Therefore, gene expression and cell viability should both be
evaluated when optimizing acoustic pressure. To sum up, an acoustic This study assessed the effects of microbubble’s concentration, post-
pressure threshold of 400 kPa is required for the microbubble transport transfection time, and ultrasound parameters (acoustic pressure, duty
system to start gene-transferring and gene transfection efficiency rea­ cycle, PRF, cycle number) on gene transfection and cell viability. The
ches its peak at a negative pressure of 700 kPa. effect of each microbubble’s concentration and time after ultrasound
Furthermore, a similar increasing trend as acoustic pressure is also exposure on gene transfection and cell viability are all statistically sig­
observed with PRF, duty cycle, and cycle number increasing (Figs. 4–6). nificant (two-way ANOVA, p < 0.001). In vitro models also show that the
When investigating the effects of the duty cycle (a gradual increase from higher the duty cycle, PRF, and cycle number are, the more efficiently
0.5% to 5%) and the PRF (a gradual increase from 0.5 Hz to 5 Hz) on the gene transfection is conveyed (Tukey HSD, p < 0.001). Simulta­
luciferase gene expression and cell viability, we found that increasing neously, cell viability decreases gradually (Tukey HSD, p < 0.001).
the duty cycle from 0.5% to 5% and the PRF from 0.5 Hz to 5 Hz showed Together, on day two post-transfection, microbubble concentration of 4
the average bioluminescence level tended to increase rapidly. Mean­ × 107 MB/mL, acoustic pressure of 700 kPa, the duty cycle of 5%, PRF of
while, the ability to transfer genes tends to decrease. At a duty cycle 5 Hz, and cycle number of 10000, the gene transfection and cell viability
value of 5% and a PRF of 5 Hz, the mean bioluminescence was highest, of microbubbles transport system combined with ultrasound are the
and cell viability was lowest. However, at these values, the cell viability most optimal in our experiment setting. This is the basis for conducting
was still higher than 70% (Fig. 4). The higher the duty cycle was, the further studies to evaluate the gene transfection of the microbubbles
greater inertial cavitation was created, where a higher percentage of transport system combined with ultrasound in targeting tumors in vivo.
microbubbles lasted longer between pulses.
Similarly, in Fig. 5, the higher cycle number generated greater in­ Funding
ertial cavitation, where a higher level of microbubbles endured longer
between pulses. This result can be explained by the increased number of This work was supported by the Ministry of Science and Technology
focused ultrasound cycles, leading to the formation of a larger mem­ of Vietnam (Code: NDT.65.TW/19).
brane hole, creating an effect that helps the microbubble to be more
stable in the interval between pulses. This is an essential factor deter­ Author statement
mining the success of using multifunctional transport systems in future
in vivo model studies. The importance of cycle numbers might be a Tho Anh Thi Tran: Conceptualization, Methodology, Investigation,
valuable characteristic of microbubbles. More control over the degree of Formal analysis, Writing - Original Draft. Toan Phi Nguyen: Writing -
sonoporation in vivo may be achieved by increasing cycle number to Original Draft, Writing - Review and Editing, Visualization. Nhung Hong
improve sonoporation or reducing cycle number to ameliorate cell death Thi Duong: Investigation, Data Curation, Visualization. Duy Hieu
in more sensitive tissues. Truong: Writing - Review and Editing. Bac Xuan Nguyen: Supervision.
Results from Fig. 6 demonstrated that luciferase expression was Cuong Khac Bui: Resources. Lap Thi Nguyen: Funding acquisition,
heavily dependent on cycle number, with more transfection efficiency at Project administration.
a higher cycle number of 10000. It can be concluded that the influence of
ultrasound parameters such as cycle numbers, PRF, and duty cycles on Declaration of competing interest
the gene transfection of microbubbles carrying genes encoding lucif­
erase in vitro. None.
In general, acoustic pressure, cycle number, duty cycles, and PRF are
directly proportional to gene transfection and inversely proportional to Data availability
cell viability (Figs. 3–6), which is also the case for previous conclusions
[25,32]. The overall trend for the data is that increases in transfection The data that has been used is confidential.
efficiency are associated with decreased cell viability. These experi­
mental observations strongly suggest that optimization is needed since Acknowledgments
the maximum transfection rate and cell viability cannot be obtained
together. The justification may be that these increased ultrasound pa­ The authors wish to convey their gratitude for the financial support
rameters change membrane pore size, thereby facilitating gene transfer. from the Ministry of Science and Technology of Vietnam and helpful
Nonetheless, increasing these parameters to too high may tend to expert support from Prof. Chih-Kuang Yeh and his lab members (En-Ling

7
T.A.T. Tran et al. Journal of Drug Delivery Science and Technology 77 (2022) 103882

Chang and Ching-Hsiang Fan), National Tsinghua University, Taiwan. [18] G. Shapiro, et al., Multiparameter evaluation of in vivo gene delivery using
ultrasound-guided, microbubble-enhanced sonoporation, J. Contr. Release 223
(Feb 10 2016) 157–164, https://doi.org/10.1016/j.jconrel.2015.12.001.
References [19] K. Okada, N. Kudo, K. Niwa, K. Yamamoto, A basic study on sonoporation with
microbubbles exposed to pulsed ultrasound, J. Med. Ultrason. 32 (1) (2005) 3–11,
[1] N.G. Zaorsky, et al., Causes of death among cancer patients, Ann. Oncol. 28 (2) https://doi.org/10.1007/s10396-005-0031-5, 2005/03/01.
(Feb 1 2017) 400–407, https://doi.org/10.1093/annonc/mdw604. [20] M. Ward, J. Wu, J.F. Chiu, Experimental study of the effects of Optison
[2] K. Aldape, et al., Challenges to curing primary brain tumours, Nat. Rev. Clin. concentration on sonoporation in vitro, Ultrasound Med. Biol. 26 (7) (Sep 2000)
Oncol. 16 (8) (Aug 2019) 509–520, https://doi.org/10.1038/s41571-019-0177-5. 1169–1175, https://doi.org/10.1016/s0301-5629(00)00260-x.
[3] S. Nayak, R.W. Herzog, Progress and prospects: immune responses to viral vectors, [21] D.L. Miller, J. Quddus, Sonoporation of monolayer cells by diagnostic ultrasound
Gene Ther. 17 (3) (Mar 2010) 295–304, https://doi.org/10.1038/gt.2009.148. activation of contrast-agent gas bodies, Ultrasound Med. Biol. 26 (4) (May 2000)
[4] S. Patil, et al., The development of functional non-viral vectors for gene delivery, 661–667, https://doi.org/10.1016/s0301-5629(99)00170-2, oi:.
Int. J. Mol. Sci. 20 (21) (Nov 4 2019), https://doi.org/10.3390/ijms20215491. [22] N. Qu, et al., Breast cancer cell line phenotype Affects sonoporation efficiency
[5] R. Bekeredjian, P.A. Grayburn, R.V. Shohet, Use of ultrasound contrast agents for under optimal ultrasound microbubble conditions, Med. Sci. Mon. Int. Med. J. Exp.
gene or drug delivery in cardiovascular medicine, J. Am. Coll. Cardiol. 45 (3) (Feb Clin. Res. 24 (Dec 14 2018) 9054–9062, https://doi.org/10.12659/MSM.910790.
1 2005) 329–335, https://doi.org/10.1016/j.jacc.2004.08.067. [23] H. Yu, S. Chen, A model to calculate microstreaming-shear stress generated by
[6] S. Ibsen, C.E. Schutt, S. Esener, Microbubble-mediated ultrasound therapy: a oscillating microbubbles on the cell membrane in sonoporation, Bio Med. Mater.
review of its potential in cancer treatment, Drug Des. Dev. Ther. 7 (2013) 375–388, Eng. 24 (1) (2014) 861–868, https://doi.org/10.3233/BME-130878.
https://doi.org/10.2147/DDDT.S31564. [24] Y. Qiu, et al., The correlation between acoustic cavitation and sonoporation
[7] M. Shibuya, Differential roles of vascular endothelial growth factor receptor-1 and involved in ultrasound-mediated DNA transfection with polyethylenimine (PEI) in
receptor-2 in angiogenesis, J. Biochem. Mol. Biol. 39 (5) (Sep 30 2006) 469–478, vitro, J. Contr. Release 145 (1) (Jul 1 2010) 40–48, https://doi.org/10.1016/j.
https://doi.org/10.5483/bmbrep.2006.39.5.469. jconrel.2010.04.010.
[8] D.J. Hicklin, L.M. Ellis, Role of the vascular endothelial growth factor pathway in [25] M.M. Forbes, R.L. Steinberg, W.D. O’Brien Jr., Examination of inertial cavitation of
tumor growth and angiogenesis, J. Clin. Oncol. 23 (5) (Feb 10 2005) 1011–1027, Optison in producing sonoporation of Chinese hamster ovary cells, Ultrasound
https://doi.org/10.1200/JCO.2005.06.081. Med. Biol. 34 (12) (Dec 2008) 2009–2018, https://doi.org/10.1016/j.
[9] B.L. Falcon, S. Chintharlapalli, M.T. Uhlik, B. Pytowski, Antagonist antibodies to ultrasmedbio.2008.05.003.
vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents, [26] G. Guo, et al., Mechanical and dynamic characteristics of encapsulated
Pharmacol. Ther. 164 (Aug 2016) 204–225, https://doi.org/10.1016/j. microbubbles coupled by magnetic nanoparticles as multifunctional imaging and
pharmthera.2016.06.001. drug delivery agents, Phys. Med. Biol. 59 (22) (Nov 21 2014) 6729–6747, https://
[10] V. Sboros, Response of contrast agents to ultrasound, Adv. Drug Deliv. Rev. 60 (10) doi.org/10.1088/0031-9155/59/22/6729.
(Jun 30 2008) 1117–1136, https://doi.org/10.1016/j.addr.2008.03.011. [27] R. Karshafian, P.D. Bevan, R. Williams, S. Samac, P.N. Burns, Sonoporation by
[11] S.M. Chowdhury, L. Abou-Elkacem, T. Lee, J. Dahl, A.M. Lutz, Ultrasound and ultrasound-activated microbubble contrast agents: effect of acoustic exposure
microbubble mediated therapeutic delivery: underlying mechanisms and future parameters on cell membrane permeability and cell viability, Ultrasound Med.
outlook, J. Contr. Release 326 (Oct 10 2020) 75–90, https://doi.org/10.1016/j. Biol. 35 (5) (May 2009) 847–860, https://doi.org/10.1016/j.
jconrel.2020.06.008. ultrasmedbio.2008.10.013.
[12] D. Shi, et al., Influence of tumor cell lines derived from different tissue on [28] M. Wang, Y. Zhang, C. Cai, J. Tu, X. Guo, D. Zhang, Sonoporation-induced cell
sonoporation efficiency under ultrasound microbubble treatment, Ultrason. membrane permeabilization and cytoskeleton disassembly at varied acoustic and
Sonochem. 38 (2017) 598–603, https://doi.org/10.1016/j.ultsonch.2016.08.022, microbubble-cell parameters, Sci. Rep. 8 (1) (2018) 3885, https://doi.org/
2017/09/01/. 10.1038/s41598-018-22056-8, 2018/03/01.
[13] A. Rahim, S.L. Taylor, N.L. Bush, G.R. ter Haar, J.C. Bamber, C.D. Porter, Physical [29] Y. Qiu, C. Zhang, J. Tu, D. Zhang, Microbubble-induced sonoporation involved in
parameters affecting ultrasound/microbubble-mediated gene delivery efficiency in ultrasound-mediated DNA transfection in vitro at low acoustic pressures,
vitro, Ultrasound Med. Biol. 32 (8) (Aug 2006) 1269–1279, https://doi.org/ J. Biomech. 45 (8) (May 11 2012) 1339–1345. https://doi: 10.1016/j.jbiomech
10.1016/j.ultrasmedbio.2006.04.014. .2012.03.011.
[14] E.L. Chang, et al., Angiogenesis-targeting microbubbles combined with ultrasound- [30] Z. Fan, H. Liu, M. Mayer, C.X. Deng, Spatiotemporally controlled single cell
mediated gene therapy in brain tumors, J. Contr. Release 255 (Jun 10 2017) sonoporation, Proc. Natl. Acad. Sci. U. S. A. 109 (41) (Oct 9 2012) 16486–16491,
164–175, https://doi.org/10.1016/j.jconrel.2017.04.010. https://doi.org/10.1073/pnas.1208198109.
[15] M. Cochran, M.A. Wheatley, In vitro gene delivery with ultrasound-triggered [31] H.R. Guzman, D.X. Nguyen, S. Khan, M.R. Prausnitz, Ultrasound-mediated
polymer microbubbles, Ultrasound Med. Biol. 39 (6) (Jun 2013) 1102–1119, disruption of cell membranes. I. Quantification of molecular uptake and cell
https://doi.org/10.1016/j.ultrasmedbio.2013.01.013. viability, J. Acoust. Soc. Am. 110 (1) (Jul 2001) 588–596, https://doi.org/
[16] Y. Zhang, et al., Ultrasound-mediated gene transfection in vitro: effect of ultrasonic 10.1121/1.1376131.
parameters on efficiency and cell viability, Int. J. Hyperther. 28 (4) (2012) [32] C.X. Deng, F. Sieling, H. Pan, J. Cui, Ultrasound-induced cell membrane porosity,
290–299, https://doi.org/10.3109/02656736.2012.665568. Ultrasound Med. Biol. 30 (4) (Apr 2004) 519–526, https://doi.org/10.1016/j.
[17] X. Yao, et al., Vascular endothelial growth factor receptor 2 (VEGFR-2) plays a key ultrasmedbio.2004.01.005.
role in vasculogenic mimicry formation, neovascularization and tumor initiation [33] Y. Taniyama, et al., Local delivery of plasmid DNA into rat carotid artery using
by Glioma stem-like cells, PLoS One 8 (3) (2013), e57188, https://doi.org/ ultrasound, Circulation 105 (10) (Mar 12 2002) 1233–1239, https://doi.org/
10.1371/journal.pone.0057188. 10.1161/hc1002.105228.

You might also like