You are on page 1of 14

Article

Cite This: Mol. Pharmaceutics XXXX, XXX, XXX−XXX pubs.acs.org/molecularpharmaceutics

Development of a Liposomal Formulation of Acetyltanshinone IIA


for Breast Cancer Therapy
Qi Wang,† Man Luo,† Na Wei,† Alex Chang,‡ and Kathy Qian Luo*,§

School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457

Department of Oncology, Johns Hopkins Singapore, Singapore 308433
§
Faculty of Health Sciences, University of Macau, Taipa, Macau, China
*
S Supporting Information
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via INDIAN INST OF TECH GUWAHATI on August 11, 2019 at 08:16:01 (UTC).

ABSTRACT: Acetyltanshinone IIA (ATA), synthesized in our group exhibiting


good anti-breast cancer effects, is expected to replace the commonly used anti-ER+
breast cancer (breast cancer cells overexpressing the estrogen receptor) drug
tamoxifen. To promote the clinical progress of ATA, polyethylene glycol (PEG)-
modified liposomes were used to encapsulate ATA along with improving its
bioavailability and in vivo anticancer efficiency. The resulting liposomal ATA
exhibited a spherical shape with an average size of 188.5 nm. In vitro evaluations
showed that liposomal ATA retained the anti-breast cancer efficacy of ATA while
exerting much less cytotoxicity toward noncancerous cells. Significantly,
pharmacokinetics analysis showed that the AUC0−24h of liposomal ATA was 59
times higher than that of free ATA, demonstrating increased bioavailability of ATA.
Preclinical experiments demonstrated that liposomal ATA reduced the growth of
ER-positive human breast tumor xenografts by 73% in nude mice, and the
liposomal ATA exhibited a much lower level of toxicity than that of free ATA with
respect to zebrafish larval mortality, body formation, and heart function during development. Moreover, 7-day and 21-day tissue
toxicity levels were determined in mice by intravenous administration of a maximum dosage of liposomal ATA (120 mg/kg).
The results showed no obvious tissue damage in major organs, including the heart, liver, spleen, kidney, and brain. In summary,
we have developed a clinical formulation of liposomal ATA with the high bioavailability and potent efficacy for the treatment of
ER-positive breast cancer.
KEYWORDS: anti-breast cancer drugs, acetyltanshinone IIA (ATA), mPEG-liposomes, bioavailability, pharmacokinetics,
toxicity study

1. INTRODUCTION Encapsulated by poly(ethylene glycol) methyl ether-block-


Breast cancer has become a major public health problem in the poly(lactide-co-glycolide) (mPEG-PLGA), the 24 h blood-
current society and suffered from the resistance of frequently circulation time of ATA was extended to 10-fold that of
used anti-breast cancer drugs including tamoxifen. Therefore, a unbound ATA.7 However, the majority of the ATA was
new chemotherapeutic agent for more effective breast cancer trapped in red blood cells rather than in plasma.
treatment has been urgently needed recently. Tanshinone IIA Herein, it is urgent to develop ATA into a clinical
(TIIA), one of the major hydrophobic components of formulation to promote its clinical process. In this report,
Danshen,1−3 has received major attention from the scientists Federal Drug Administration-approved liposomes have at-
for the high anticancer activity.4 Deriving from TIIA, tracted our attention that the liposome-based formulations
acetyltanshinone IIA (ATA) was synthesized in our group represented excellent biocompatibility and improved bioavail-
and was further approved to have high anti-breast cancer ability in vivo.8−12 Also, a conventional liposome is a mature
efficacy.5 Specifically, ATA displayed a strong growth- and widely used intravenous dosage form among all the
inhibitory effect on estrogen receptor-positive (ER+) breast contemporary formulations,8−12 which ensures the further
cancer cells through degrading the ERα protein.6 More clinical evaluation of liposomal ATA. In this report, the main
significantly, ATA produced a more potent growth inhibitory constituents of liposomal ATA are phospholipids and
activity toward ER+ breast cancer cells than that of tamoxifen, cholesterol, which are components of the mammalian cell
which is a commonly used anti-ER+ breast cancer drug.6
In light of improved therapeutic effects, it is of great Received: May 6, 2019
potential to develop ATA into a new anti-breast cancer drug. Revised: July 24, 2019
However, our initial pharmacokinetic study showed that ATA Accepted: July 26, 2019
was quickly removed from the plasma in both mouse and rats.

© XXXX American Chemical Society A DOI: 10.1021/acs.molpharmaceut.9b00493


Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

Figure 1. (A) Structural features of liposomal ATA. ATA molecules were encapsulated in the lipid bilayer of the liposome. The PEG chain is
located on the surface of the liposome. (B) SEM images. The liposomal ATA was uniformly distributed. (C) Size distributions. The average size of
liposomal ATA was 188.5 nm based on intensity. (D) Zeta potential. The zeta potential of liposomal ATA was −31.0 mV.

membrane, making them compatible with biological systems. guide the future manufacturing and development of liposomal
On the other hand, poly(ethylene glycol) methyl ether (PEG)- ATA into a new anti-breast cancer drug.
modified phospholipids were utilized to prevent the engulf-
ment of liposomes by blood cells (Figure 1A). Also, PEG 2. MATERIALS AND METHODS
chains are expected to increase the circulation time of 2.1. Materials. Pure phosphatidylcholine (PC3) and 1,2-
liposomal ATA in vivo, thus improving its bioavailability. distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy-
Furthermore, based on previous reports, we hypothesized that (polyethylene glycol)-2000] (ammonium salt) (DSPE-PEG)
the manufacture of liposomal ATA with a smaller particle size were obtained from LIPOID Pte. Ltd. (Singapore). Choles-
might facilitate its ability to reach tumor tissues through an terol, sodium dodecyl sulfonate (SDS), Tween 80, dialysis
enhanced permeability and retention (EPR) effect, resulting in sacks (Mw cutoff of 12,000 kDa), and 3-(4,5-dimethylthiazol-2-
an enhanced antitumor effect.13−16 yl)-2,5-diphenyltetrazolium bromide (MTT) were purchased
from Sigma-Aldrich (St. Louis, MO). Chloroform (CHCl3)
The liposomal ATA was prepared using a reverse-
and methanol (MeOH) were obtained from Merck Pte. Ltd.
evaporation method and was optimized through an orthogonal
(Singapore).
array test generating the drug loading efficiency of 12.35% with Dulbecco’s Minimum Essential Medium (DMEM), pen-
the size of 188.5 nm. The liposomal ATA formulation could be icillin/streptomycin (PS), and trypsin were purchased from
stably stored at 4 °C and diluted in 5% glucose in clinical GIBCO. Fetal bovine serum (FBS) was purchased from
storage and use. Specifically, important preclinical experiments HyClone. Human umbilical vein endothelial cells (HUVECs)
were conducted in this report including pharmacokinetic and MCF-7 breast cancer cells were obtained from the ATCC
profiles, anti-breast cancer efficacy, cytotoxicity, and small- (Manassas, VA).
animal toxicity to pave the way for clinical use of liposomal 2.2. Preparation and Characterization of ATA-Loaded
ATA. The information obtained from this study can be used to PEG-Liposomes (Liposomal ATA). A reverse-evaporation
B DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

method was used to prepare the liposomal ATA. First, PC3 buffered saline (PBS, pH 7.4) and incubated at 4 or 37 °C for
(120.8 mg), ATA (29.9 mg), DSPE-PEG (39.9 mg), and 1−7 days. Under all conditions, 1 mL of the suspension was
cholesterol (52.8 mg) were dissolved in chloroform (CHCl3, 5 collected at designated intervals; the size and zeta potential
mL). Distilled (DI) H2O (1.5 mL) was then added to the were detected using a laser electrophoretic light scatter
solution and treated with ultrasound (37% strength) for 3 min. analyzer (Malvern Nano-ZS Particle Sizer), and the EE% was
CHCl3 was then removed by rotary evaporation. Finally, DI measured using HPLC.
H2O (10 mL) was added to rehydrate it to obtain liposomal 2.6. Drug Release from Liposomes. The release of ATA
ATA, and the liposomal ATA was ultrasonicated for 8 min to from liposomes was investigated using dialysis and compared
produce a smaller particle size. Blank liposomes were prepared with the release of free ATA in the release medium (ultrapure
using the same method but without the addition of ATA to the water with 1% SDS). Six milliliters of the release medium
formulation. containing an equivalent amount of liposomal ATA or free
The size distribution and zeta potential of the liposomes ATA was placed in a dialysis sack (MWCO 12,000, Sigma).
were determined using a laser electrophoretic light scatter The sack was then immersed in 50 mL of the release medium
analyzer (Malvern Nano-ZS Particle Sizer). The morphology with gentle stirring at 37 °C. The samples (2 mL) were
was determined by scanning electron microscopy (SEM) removed from the sack at designated time intervals, and an
(JSM6700-FESEM, JEOL). Briefly, liposome samples were equal volume of the release medium was replenished at the
spread onto the surface of a carbon tape and then sputter- same time.22−25 The amount of released drug was determined
coated with gold under a vacuum. Afterward, the prepared by RP-HPLC as described in Section 2.3: “Determination of
samples were observed under a scanning electron microscope Encapsulation Efficiency and the Drug Loading Rate”. The
using magnification ranges of 5000−20000 with an electron similarity of the release profiles between the free ATA and
beam energy of 20 kV. liposomal ATA was evaluated by the similarity factor (f 2)
2.3. Determination of Encapsulation Efficiency and shown below
l ÄÅ ÉÑ−0.5 |
o
o Å ÑÑ o
o
oÅÅÅ o
the Drug Loading Rate. A solution of liposomal ATA (1

Ñ
f 2 = 50logm ÅÅ1 + ∑ t t ÑÑ Ñ }
mL) was centrifuged at a low speed of 500 rpm at 28 °C for 5
o
o Å o
o
n

oÅÇ Å Ñ
ÑÖ o
1
min to remove large particles. A fixed amount of the
n ~
( R − T ) × 100
supernatant containing liposomal ATA was diluted with n t=1
methanol, which will dissolve the liposome structure to release
ATA to the desired concentration, and the concentration of where n is the number of time points, Rt is the release value of
ATA in the supernatant was referred to as the concentration of liposomal ATA at time t, and Tt is the release value of free
the encapsulated drug. The concentration of ATA was ATA.26,27
measured simultaneously by RP-HPLC analysis (Shimadzu 2.7. Determination of the Pharmacokinetic Profile of
LC-20AT pump liquid chromatograph; chromatographic Liposomal ATA in Rats. Female SD rats (INVIVOS,
column: Kromasil 100-5C8, 250 mm × 4.6 mm, 5 μm). A Singapore) were randomly divided into two groups (n = 6):
mobile phase system consisting of methanol−H2O (80:20, v:v) free ATA and liposomal ATA. Free ATA was suspended in a
was pumped at a flow rate of 1 mL/min with a column solution of PEG300:ethanol:Tween 80 (60:25:15, v/v/v). The
temperature of 30 °C, and the column eluents were monitored free ATA suspension or liposomal ATA was administered at an
at a wavelength of 254 nm.7,17,18 The drug loading rate (DL%) equivalent dose of ATA at 20 mg/kg body weight via a single
and percentage of encapsulation efficiency (EE%) were intravenous injection in the tail vain. At different time points
calculated as follows: (15 min, 30 min, and 1, 2, 3, 4, 5, 6, 8, and 24 h), blood
samples (0.3 mL) were collected from the suborbital vein and
DL% = amount of drug in liposomes placed in heparinized tubes. The blood samples were
/amount of feeding materials centrifuged at 3000 rpm at 4 °C for 10 min to collect plasma.
The samples were then pretreated and injected into RP-HPLC
for detection of ATA as described in a previous work.6
EE% = amount of drug in liposomes Microsoft Excel PKsolver was used to analyze the pharmaco-
/amount of feeding drug kinetic data.
2.8. Cell Culture. HUVECs and MCF-7 cells were cultured
2.4. Thermal Analyses of Liposomal ATA. Thermal in a DMEM-based culture medium supplemented with 1%
analyses that include thermogravimetric (TG) investigation penicillin/streptomycin and 10% FBS. Cell cultures were
and differential thermal analysis (DTA) were performed using maintained at 37 °C in a humidified incubator supplemented
Diamond TG/DTA (PerkinElmer instrument).19−21 The with 5% CO2.
thermal decomposition was conducted on free ATA, PC3, 2.9. Determination of the Anticancer Efficacy and
cholesterol, DSPE-PEG, a physical mixture of the four Cytotoxicity of Liposomal ATA in Vitro. The MTT
components, and freeze-dried liposomal ATA. Approximately reduction ability was determined as an index of the metabolic
2 mg of a vacuum-dried coal sample with an average particle activity of the mitochondria, which can indicate cell viability.
diameter less than 200 μm was placed in the sample crucible. Briefly, MCF-7 cells or HUVECs were seeded in 96-well plates
The sample was heated in pure N2 from 30 to 400 °C at a and cultured overnight. Free ATA (dissolved in DMSO diluted
heating rate of 10 °C/min. The melting points of the samples in DMEM), liposomal ATA, and blank liposomes were added
were determined from thermal graphs. at different concentrations, and the cells were incubated for 24
2.5. Storage Stability of Liposomal ATA. The storage h. MTT solution (10 μL; 5 mg/mL) was then added into each
stability of liposomal ATA was investigated by measuring the well, and they were incubated for 4 h at 37 °C in the CO2
size, zeta potential, and EE% of liposomal ATA during the incubator. One hundred microliters of the solubilization
experiment. Liposomal ATA was diluted using phosphate- solution containing 1% SDS and 0.1% hydrochloric acid was
C DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

added to each well to dissolve the formazan crystals. After 8 h, zebrafish were used for each group in each experiment, and
the absorbance of the solubilized MTT formazan product was the zebrafish experiments were carried out at least four times.
measured using a spectrophotometer at an absorbance 2.11.3. Malformation and Mortality Rates of Zebrafish
wavelength of 595 nm, and the cell viability was calculated Embryos Exposed to Liposomal ATA. To examine the effects
from the optical density (OD) value of the tested sample and of free ATA and liposomal ATA on embryos during
the vehicle control (untreated cells) using the following development, several exposure durations were chosen. These
equation: cell viability (%) = OD tested sample/OD vehicle control × durations included 6−10 hpf (gastrula period), 14−24 h
100. Nine sets of data obtained in triplicate from three (important organ formation stage), 10−24 hpf (somite stage),
independent experiments were averaged to generate the final 24−48 hpf (pharyngeal phase), and 48−72 hpf (the incubation
data with the standard deviation, which are presented as the period). During each period, embryos were exposed to free
mean ± SD. ATA, liposomal ATA, or blank liposomes at the equivalent
2.10. Determination of the Antitumor Effect of concentration of 5 μM. At 96 hpf, larvae or embryos were
Liposomal ATA in Vivo. This mouse experiment was microscopically examined to determine the malformation and
conducted in accordance with the protocol approved by the mortality rates. The heart rate was also recorded at 24, 72, and
Institutional Animal Care and Use Committee, Nanyang 96 hpf.28−31 N = 20 fish/group.
Technological University, Singapore. To ensure that the 2.12. Toxicity Evaluation of Liposomal ATA on Mice.
estrogen receptor-positive breast cancer MCF-7 cells could BALB/C mice purchased from (INVIVOS, Singapore) were
grow into solid tumors in female mice, a tablet of 1.5 mg β- used to evaluate the toxicity of ATA and liposomal ATA in
estradiol 17-acetate was implanted subcutaneously seven days vivo. The maximum dosage of liposomal ATA was determined
prior to tumor inoculation into female nude mice (18−22 g, to be 120 mg/kg body weight as this is the maximum amount
6−8 weeks of age). MCF-7 cells (5 × 106) were mixed with of liposomal ATA that could be prepared in a 0.2 mL volume
Matrigel in a final volume of 0.2 mL and subcutaneously for intravenous injection. Twenty mice (10 female and 10
injected into the armpit of the mouse. male) were used in this study. A solution of 0.2 mL liposomal
After the tumor reached a minimum volume of 25 mm3, an ATA at a dose of 120 mg/kg body weight was intravenously
aliquot (200 μL) of PBS, blank liposomes, free ATA, or injected into the tail vain of each mouse. The behaviors of the
liposomal ATA (20 mg ATA/kg) was intravenously injected mice were observed, and the body weight was recorded after
into the tumor-bearing nude mice (n = 5 per group) every administration every other day. The mice were then sacrificed
other day. The tumor size and the weight of the nude mouse after 7 days (5 female and 5 male) or 21 days (5 female and 5
were measured and recorded during the treatment period. The male), and different organs such as the heart, liver, spleen,
tumor volume was determined by measuring its length and lung, kidney, and brain were collected, fixed in 4%
width and calculated using the following formula: V = (L × paraformaldehyde, and processed for H&E staining. The
W2)/2 where L is the longest dimension parallel to the skin damage in each organ was assessed and imaged to record any
surface, and W is the dimension perpendicular to L and parallel pathological changes.
to the surface. After the treatment period, the tumors and The maximal dosage of free ATA [dissolved in PEG300:e-
major organs were harvested. thanol:tween 80 = 60:25:15 (v/v/v) as a stock solution then
2.11. Toxicity Evaluation of Liposomal ATA in diluted using 0.9% NaCl at a volume ratio of 1:9 before
Zebrafish. 2.11.1. Zebrafish Maintenance and Fish Embryo injection] at 30 mg/kg body weight was also injected into
Generation. Male and female zebrafish were bred in separate BALB/C mice to evaluate the toxicity of free ATA as a
tanks. The temperature of the water in the tanks was control.32
maintained at 28.5 °C, and the pH value was within 6.8. 2.13. Statistical Analysis. Statistical analysis was
Adult zebrafish were placed in the upper level of a spawning performed using SPSS 10.0 software. Descriptive data were
tank the evening prior to mating at a ratio of female/male = expressed as the arithmetic mean value plus or minus the
2:1. The next morning after mating, the embryos were standard deviation. Statistics were performed for all compar-
collected from the lower level of the spawning tank, washed, isons. All quantitative results were obtained from at least
and kept in fresh E3 medium (13.7 mmol/L NaCl, 0.54 mmol/ triplicate samples. A t-test was applied to detect differences
L KCl, 0.025 mmol/L Na2HPO4, 0.044 mmol/L KH2PO4, between groups. In all evaluations, *p < 0.05 was considered
0.42 mmol/L NaHCO3, 1.3 mmol/L CaCl2, and 1.0 mmol/L statistically significant.
MgSO4, and the pH was adjusted to 7.2 before usage).
2.11.2. Toxicity Evaluation of Liposomal ATA in Zebrafish
3. RESULTS AND DISCUSSION
Larvae. The chorion surrounding the embryo was removed
enzymatically at 4 h post fertilization (hpf) following the 3.1. Preparation and Characterization of Liposomal
procedures described in the literature.28,29 At 6 hpf, the ATA. To increase the water solubility and bioavailability of
embryos were transferred to 96-well plates at one embryo per hydrophobic ATA, liposomes were utilized to encapsulate
well with 500 μL of E3 medium containing different ATA. Three components were utilized to make liposomes:
compositions: E3 medium alone (negative control), E3 PC3, DSPE-PEG, and cholesterol. PC3 and cholesterol possess
medium containing blank liposomes, various concentrations excellent biocompatibility in vivo. DSPE-PEG allows a longer
of liposomal ATA, or free ATA dissolved in DMSO. circulation in vivo for this drug delivery system, protects ATA
Unexposed embryos (embryos raised in E3 medium) were from degradation within the circulation system, and enhances
also incubated to monitor the natural quality of the embryos. the delivery of ATA to tumor tissues (Figure 1A).
Throughout the experiment, incubation solutions with To determine an optimal condition to formulate liposomal
freshly added ATA were replaced daily. Images of the zebrafish ATA, an orthogonal array test was designed with nine
were captured using a dissecting microscope. The heart rate experiments, [L9(34)]. This array test allowed us to examine
was also recorded as described previously.30,31 Twenty four critical experimental factors (A: ratio of PC3:cholesterol,
D DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

B: ratio of DSPE-PEG:ATA, C: ultrasonic time, and D:


homogeneous ultrasonic time) at three levels (Table S1).33,34
The reverse-phase evaporation method was employed to
prepare liposomal ATA. HPLC was used to measure the
concentration of ATA in each experiment from which the
percentage of encapsulation efficiency (EE%) and rate of drug
loading (DL%) were calculated (Table S2). The results of
array tests were further analyzed, and the optimal conditions to
formulate liposomal ATA were determined after comparing the
calculated parameters (Table S3): PC3:cholesterol = 9:4,
DSPE-PEG:ATA = 7, ultrasonic time = 3 min, and
homogeneous ultrasonic time w = 8 min. Under these
conditions, the EE% and DL% of liposomal ATA were found
to be 98.94% and 12.35%, respectively.
This high level of EE should prevent a burst release of
encapsulated ATA and, thus, is expected to produce a smooth
and steady drug release profile. In contrast, a moderate 12%
drug loading rate would satisfy the requirement for decreased
liposome usage, reduced costs, and improved compliance of
patients.
The functional performance of nanoparticle-based delivery
systems depends on the physicochemical properties of the
particles, such as size, morphology, charge, and physical state.
SEM analysis showed that the liposomal ATA was spherical
and not aggregated (Figure 1B). The average particle size of
liposomal ATA was found to be 188.5 nm with a relatively
narrow size distribution (Figure 1C). The nonaggregative
dispersion and relatively narrow size distribution of liposomal
ATA may ensure its smooth and steady drug release properties.
In addition, a small size is supposed to allow the easy
dissemination of liposomal ATA through the tumor neo-
vascularization. Consequently, the passive targeting ability of
liposomal ATA for tumors may be achieved. The zeta potential Figure 2. TG/DTA profiles of liposomal ATA and the materials used
to formulate ATA in an individual or mixed form. Four characteristic
was −31.0 mV, which suggests the stability of the liposomal absorption peaks appeared in the DTA curve when ATA, cholesterol,
ATA (Figure 1D). Likewise, due to the PEG chain on the DSPE-PEG, and PC3 were physically mixed together, while only one
surface of liposomes, an enhanced circulation period could be absorption peak appeared in the DTA curve in the formulated
obtained. Greater accumulation in the tumor tissue may be liposomal ATA.
anticipated for liposomal ATA, potentially further facilitating
its therapeutic effects and reducing the off-target effects on The mass loss of PC3 was complicated, occurring in the range
normal tissues. from 250 to 350 °C. The phase transition peak of PC3 was
3.2. Using Thermal Analysis (TG/DTA) to Study the 109.16 °C (Figure 2).
Properties of Liposomal ATA. The thermodynamic study of The individual phase transition peak of cholesterol, DSPE-
liposomal ATA was conducted to achieve three objectives: (1) PEG, PC3, and ATA can be found by analyzing the TG/DTA
to confirm the existence of ATA in liposomes, (2) to evaluate curves obtained from the four physically mixed components.
the stability of drug-loaded liposomes, and (3) to validate These four DTA peaks were found at 143.84 °C, 54.72 °C,
whether the drug-loaded liposomes were successfully prepared. 105.24 °C, and 165.23 °C, which corresponded to phase
The thermal analysis of liposomal ATA was investigated using transition temperatures of cholesterol (148.96 °C in the
TG/DTA. The results of pure and formulated components are cholesterol curve), DSPE-PEG (54.90 °C in the DSPE-PEG
shown in Figure 2. curve), PC3 (109.16 °C in the PC3 curve), and ATA (175.72
The TG/DTA curve of pure ATA showed that the mass loss °C in the ATA curve). In opposition to the physical mixture,
of ATA occurred in just one step in the temperature range of the curve for liposomal ATA showed only one phase transition
220−310 °C. This step consisted of an accelerated mass loss temperature (65.04 °C), which suggested that ATA was
reaching ∼100% that was associated with a small DTA molecularly dispersed within liposomes (Figure 2).
endothermic peak at 175.72 °C, which was presumably caused The blank liposomes and the physical mixture of liposomal
by the phase transition of ATA. The materials used to prepare materials of cholesterol, DSPE-PEG, and PC3 were also
liposomal ATA showed different TG/DTA profiles compared analyzed by TG/DTA. From the enlarged DTA curve, three
with ATA. For example, the mass loss of cholesterol occurred DTA peaks were detected from the components of liposome
in the temperature range from 220 to 330 °C that was materials. In contrast, only one DTA peak at 72.03 °C was
associated with a DTA endothermic peak after 300 °C. The detected from the blank liposomes (Figure S1), which is quite
phase transition peak of cholesterol was 148.96 °C. The mass different from the DTA peak at 65.04 °C from liposomal ATA
loss of DSPE-PEG occurred in the temperature range of 300− (Figure 2).
400 °C associated with the DTA endothermic peak after 350 The thermodynamic results indicated that the physical
°C. The phase transition peak of DSPE-PEG was 54.90 °C. mixture of four components of liposomal ATA would not affect
E DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

the state of their existence because each melting point of these stability features were observed at 37 °C. The size and zeta
components could be easily found in the thermodynamic curve potential did not significantly change in the first 5 days (Figure
of the physical mixture. In contrast, when these components 3A,B). However, smaller liposomes started to aggregate to
were molecularly integrated in the liposomal ATA, only one form larger liposomes after 5 days, resulting in a significant
melting point was detected, suggesting that the ATA molecules increase in average particle size from ∼190 to ∼380 nm and a
were dissolved in the lipid bilayer of liposomes. This result significant reduction of the average zeta potential from −42
confirmed that ATA was successfully and stably carried by the mV to nearly −68 mV. We do not expect that this aggregation
PEG-liposomes. problem will affect the in vivo efficacy of liposomal ATA as
3.3. Evaluating the Storage Stability of Liposomal they should be metabolized in the body in less than 3 days.
ATA. The size distribution, surface charge, and EE% of The EE% of liposomal ATA did not decrease at 37 °C in the
liposomal ATA were measured during a storage period of 7 first 4 days and was slightly reduced to ∼80% after 7 days
days to assess their storage stability. This information is (Figure 3C). In conclusion, liposomal ATA should be stored at
important for developing liposomal ATA into a clinical 4 °C to maintain its small particle size and high encapsulation
formulation. efficiency.
Figure 3A,B shows that the size and zeta potential of the The compatibility and stability of liposomal ATA with two
liposomal ATA remained constant at 4 °C during the 7 days of commonly used intravenous dosing media, saline (0.9% NaCl)
storage. In addition, the EE% remained at a very high level and isotonic glucose solution (5% glucose), were evaluated.
(>95%) in the first 4 days at 4 °C and was slightly reduced to After being incubated with 5% glucose at room temperature for
>80% from day 5 to 7 (Figure 3C). In contrast, different 24 h, the particle size of the liposomal ATA was reduced by 15
nm, and the zeta potential of liposomal ATA was reduced by 6
mV. In contrast, incubation with 0.9% NaCl resulted in a much
greater variation of the particle size and zeta potential of
liposomal ATA at 50 nm and 38 mV, respectively (Figure
S2A,B). More importantly, these changes occurred within the
first several hours of incubation. A significant reduction of EE%
was also detected when liposomal ATA was incubated with
0.9% NaCl for 12 h, while no reduction was observed when
liposomal ATA was incubated with 5% glucose for 12 h
(Figure S2C). These results indicated that liposomal ATA was
more stable in 5% glucose than in 0.9% NaCl, and therefore
liposomal ATA should be diluted with 5% glucose for
intravenous injection in future clinical applications.
The release profiles of ATA were compared between free
ATA and liposomal ATA in aqueous medium. The graph in
Figure 3D shows that most free ATA could not be released
from the dialysis bag into the surrounding medium, likely
because of its poor solubility, which could not produce a
sufficient concentration gradient between the two sides of the
dialysis membrane for the diffusion of ATA. However, ATA
exhibited a better release profile when it was encapsulated with
liposomes. For example, ∼78% of ATA was released from
liposomal ATA, while only ∼38% was released from free ATA
after 72 h. More interestingly, unlike the flat release profile of
free ATA, a steady increase in ATA release was observed over
the 72 h.
This result not only supported the conclusion that ATA
molecules were successfully encapsulated within liposomes, but
it also demonstrated that encapsulating ATA within liposomes
could prolong its release. Other important information
obtained from this study included the finding that the
liposomal ATA formulation could be more stably stored at 4
°C and diluted in 5% glucose for intravenous injection or
diffusion.
3.4. Determination of the Pharmacokinetic Profile of
Liposomal ATA in Rats. The most important objective of
Figure 3. Storage stability of liposomal ATA. Liposomal ATA was this study was to develop a clinical formulation of ATA with
stored in PBS at 4 or 37 °C for 7 days. (A) Size changes in liposomal improved bioavailability. To assess whether the formulated
ATA. The size of liposomal ATA was more stable at 4 °C. (B) Zeta
ATA could meet this goal, free and liposomal ATA at 20 mg/
potential of liposomal ATA. The zeta potential of liposomal ATA was
more stable at 4 °C. (C) EE% change in liposomal ATA. *p < 0.05, kg were administered to rats via a single intravenous injection.
***p < 0.001 compared with day 0. (D) Drug release profile. At different time points, blood samples were obtained from
Liposomal ATA displayed a gradual and prominent release profile rats, and the concentrations of ATA in plasma and whole
compared with free ATA. f 2 < 50 between free ATA and the blood were determined using a well-validated HPLC method.6
liposomal ATA group. The pharmacokinetic profiles shown in Figure 4 revealed that
F DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

Figure 4. Pharmacokinetic (PK) profiles of free and liposomal ATA from plasma and blood of rats after intravenous injection. N = 6 rats/group.

Table 1. Pharmacokinetic Parameters of Free ATA and Liposomal ATA in Rat Plasma and Whole Blooda
free ATA liposomal ATA ratio (lipo/free-ATA)
parameter plasma blood plasma blood plasma blood
CL{[(mg/kg)/μM]/min} 0.24 25 0.004 2672 0.02 106
AUC0−inf (μM·min) 85 882 5009 27,912 59 32
a
CL: clearance; AUC0−inf: area under the plasma concentration-time curve from time zero to infinity.

Figure 5. The cytotoxicity of free and liposomal ATA was evaluated by the MTT assay. (A) Liposomal ATA displayed a good growth inhibitory
effect in breast cancer MCF-7 cells. (B) Liposomal ATA was not toxic to normal endothelial HUVECs at concentrations ≤5 μM. *p < 0.05, **p <
0.01, ***p < 0.001 between tested samples and the control group. Data were collected from triplicate wells in each experiment repeated three
times.

the concentrations of ATA in plasma and whole blood from parameters. Compared with free ATA, the 24 h area under the
the free ATA-treated group were at a very low level at 15 min. curve (AUC0−24h) of liposomal ATA was increased 59-fold in
Compared with free ATA, liposomal ATA had significantly plasma and 32-fold in whole blood, whereas the clearance rate
higher concentrations of ATA in both plasma and whole blood. was reduced 50-fold in plasma and increased 106-fold in whole
We noticed that the concentration of ATA is higher in the blood. These results demonstrated that liposomal ATA
whole blood group than in the plasma group, which may be significantly extended the retention time and enhanced
due to the higher internalization of liposomal ATA by the bioavailability of ATA in vivo.
blood cells. This distribution of liposomal ATA in the blood 3.5. In Vitro Determination of the Anticancer Efficacy
cells may increase its storage ability and contribute to a longer and General Cytotoxicity of Liposomal ATA. As liposomal
circulation time of ATA in the blood stream. ATA was designed for intravenous injection, we wanted to
The pharmacokinetic parameters of ATA were then investigate whether liposomal ATA could retain its anticancer
calculated using Excel PKsolver. According to the requirements effect without damaging endothelial cells. To assess this
for selecting the pharmacokinetic calculation model, the values possibility, the viabilities of human breast cancer MCF-7 cells
of Akaike’s information criterion and residual sum of squares and human umbilical vein endothelial cells (HUVECs) were
(Re) were minimized, and the value of the determinate determined by the MTT assay after the cells were incubated
coefficient (r) was maximized. The free ATA and liposomal with various concentrations of free ATA, liposomal ATA, or
ATA belonged to the two-chamber model in whole blood. Free blank liposomes for 24 h.
ATA belonged to the one-compartment model in plasma, and The MTT results showed that, at 0.5 μM, liposomal ATA
liposomal ATA belonged to the two-chamber model in plasma. significantly reduced the viability of MCF-7 cells to ∼60%
The mean retention time was calculated using the statistical compared with the control group, while free ATA did not
moment. The main pharmacokinetic parameters of ATA in rat affect the viability of MCF-7 cells (Figure 5A), demonstrating
plasma and whole blood are listed in Table 1 and Tables S4 that formulating ATA with liposomes has increased its
and S5. cytotoxicity on breast cancer cells. Importantly, 0.5 μM
As shown in Table 1, the free ATA and liposomal ATA- liposomal ATA did not reduce the viability of endothelial
treated groups exhibited significantly different pharmacokinetic HUVECs (Figure 5B), suggesting that it has little toxicity
G DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

toward noncancerous cells. At elevated concentration ranges of After the MCF-7 cells had grown into solid tumors,
1−20 μM, both liposomal and free ATA produced a much liposomal ATA (20 mg/kg) was injected into the tumor-
stronger growth inhibitory effect on MCF-7 cells than that of bearing nude mice through the tail vein every other day for 3
the control group (Figure 5A). weeks. During the treatment period, both the tumor size and
We also calculated the concentration that could reduce 50% body weight were measured. As shown in Figure 6A, tumors
of the cell viability after a 24 h incubation. The 24 h IC50 grew rapidly in the control group, whereas in the free ATA-
values showed that liposomal ATA had a lower (0.4-fold) IC50 treated group, tumor growth was significantly reduced. More
value than that of free ATA in breast cancer MCF-7 cells and a importantly, liposomal ATA exerted an even better tumor
higher (1.5-fold) IC50 value in endothelial cells (Table 2). In growth inhibitory effect (73%) than that of free ATA (61%)
summary, these results suggested that liposomal ATA had a (Table 3).
higher anticancer efficacy than that of free ATA and was less
toxic to HUVECs in comparison to its free form. Table 3. Tumor Growth Inhibition Ratio
sample tumor size (mm3) inhibition ratio (%) p value
Table 2. IC50 Values of Liposomal ATA vs Free ATA
control 1,611 ± 612 0
ratio of IC50 ratio of IC50 blank liposomes 897 ± 360 44 0.023 (*)
24 h IC50 breast cancer (lipo/free endothelial (lipo/free
values MCF-7 cells ATA) HUVECs ATA) free ATA 625 ± 301 61 0.009 (**)
liposomal ATA 432 ± 428 73 0.007 (**)
liposomal 1.0 μM 0.4-fold 22 μM 1.5-fold
ATA
free ATA 2.3 μM 14 μM No significant body weight differences were found between
the treatment groups (free ATA, blank liposomes, and
liposomal ATA) and the control group injected with PBS
3.6. Evaluating the in Vivo Antitumor Effect of during the experiment, which suggested that liposomal ATA
Liposomal ATA. As ATA has been previously shown to did not cause serious side effects in nude mice (Figure 6B).
effectively inhibit the growth of ER+ breast cancer MCF-7 At the end of the animal experiment, tumor tissues were
cells, in this study, MCF-7 cells were utilized to establish removed and photographed. The images in Figure 6C revealed
human xenograft tumors in nude mice. A tablet of 1.5 mg β- that three out of four tumors from the liposomal ATA-treated
estradiol 17-acetate was first implanted dermatologically into mice had a much smaller size than the tumors from free ATA-
female mice, and seven days later, MCF-7 cells (5 × 106) were treated mice. We propose that the following reasons
mixed with Matrigel and subcutaneously injected into the contributed to a higher antitumor efficacy of liposomal ATA.
armpit of the mice. First, the encapsulation of ATA with liposomes significantly

Figure 6. Determination of the antitumor effect of liposomal ATA in a human breast tumor xenograft model. (A) Graphs of the tumor size during
the treatment period. Tumor growth was effectively suppressed after the treatment of liposomal ATA. (B) Body weight of nude mice during the
treatment. No apparent body weight loss was found in all four groups. (C) Pictures of harvested tumors at the end of the experiment. The
liposomal ATA treatment group had the smallest tumor size. N = 5 mice/group. *p < 0.05, **p < 0.01 between samples and the control group.

H DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

Figure 7. Toxicity evaluation of liposomal ATA in zebrafish embryos. (A) Types of induced malformations in zebrafish embryos at 96 hpf: (a)
normal development, (b) yolk sac absorption delay, (c) pericardial edema, and a (d, e) bent body axis. (B) Deformity and mortality rates of
zebrafish at 96 hpf. Less deformity and mortality were observed in zebrafish in the liposomal ATA-treated group compared with the free ATA-
treated group under the same treatment concentration. (C) Heartbeat of zebrafish. *p < 0.05, **p < 0.01 between samples and the control group.
N = 20 fish/group, and each experiment was performed at least four times.

increased their plasma concentration. Second, the average 3.7. Evaluation of the Potential Toxicity of Liposomal
particle size of liposomal ATA was 188.5 nm, which should ATA on Zebrafish. To date, our data have shown that the
facilitate its exit from the leaky tumor vascular system to reach newly formulated liposomal ATA displayed good antitumor
tumor cells and enhance its therapeutic effect. effects with little toxicity to human endothelial cells at ≤5 μM
On a separate note, we found that blank liposomes exhibited and did not reduce the body weight of tumor-bearing mice at a
a minor tumor growth inhibitory effect (Figure 6A). The dose of 20 mg/kg. As our ultimate goal for liposomal ATA is to
results of the MTT assay also showed that the blank liposome use it to treat cancer, it is necessary to evaluate its potential
reduced the viability of MCF-7 cells (Figure 5A). These two toxicity in vivo. We first utilized zebrafish because they are
cheap and easy to acquire in large quantities and because of the
observations suggested that internalized liposomes might affect
similarities of the zebrafish genome to the human genome and
the membrane integrity of tumor cells, which reduced cell statuses of both fish and humans as vertebrates.
viability and tumor growth. Further studies will be conducted In this experiment, zebrafish larvae at the age of 6 h post
to understand why blank liposomes produced a minor tumor fertilization (hpf) were exposed to different concentrations of
growth inhibitory effect, which can also help demonstrate that free or liposomal ATA, and the morphological changes were
the enhanced anticancer activity is due to the encapsulation of observed at 96 hpf. Moreover, the heart rates were measured at
ATA with liposomes but not due to the additive effect of blank 48, 72, and 96 hpf. During the treatment with free or liposomal
liposomes plus free ATA. ATA, various kinds and degrees of toxicity were observed over
I DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

Figure 8. Toxicity study of 5 μM free or liposomal ATA during zebrafish development. (A) Zebrafish embryos at different stages. (B) Deformity
and mortality rate of zebrafish at different periods during development. (C) Heartbeat of zebrafish at different developmental stages. *p < 0.05, **p
< 0.01 between samples and the control group.

the course of zebrafish development. At high concentrations, 90% at 5 μM. The combined rate of deformity and mortality
ATA resulted in death or malformations in the early was 40% at 2.5 μM. These results suggested that free ATA at 5
developmental period. Images of malformations observed in μM was toxic to zebrafish development.
this experiment are shown in Figure 7A, including pericardial In contrast, liposomal ATA showed much lower toxicity
edema, yolk sac absorption delay, and curvature of the spine. toward zebrafish than that of free ATA. The sum percentage of
The overall percentages of deformity and mortality are mortality and deformity caused by liposomal ATA was lower
shown in Figure 7B. A 5% mortality and no deformity were than that of free ATA. Importantly, no toxicity was observed at
observed in the control group. Blank liposomes did not cause a low concentration of 1 μM, and 10% mortality was detected
significant deformities or mortalities under the tested at 2.5 μM. In addition, the sum percentage of mortality and
concentrations. The data indicated that free ATA caused deformity did not exceed 50% at high concentrations of 10−20
extensive death or body deformation in zebrafish at high μM.
concentrations (5−20 μM). In particular, 100% mortality was Additionally, the heart rate (heartbeats per minute) of
recorded in zebrafish treated with 10 μM and 20 μM free ATA, zebrafish was measured to determine heart function (Figure
and the total amount of deformity and death rate exceeded 7C). In the control group, the heartbeat gradually increased
J DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

from 48 hpf to 96 hpf, which resulted from normal heart 8C, blank liposomes and liposomal ATA did not cause obvious
growth during zebrafish development. Similar to the control changes in the heartbeat compared with the control group in
group, blank liposomes also formed this normal pattern at all treatment periods. Free ATA did not cause notable changes
most liposome concentrations. Free ATA at 1−5 μM caused in the heartbeat in the 6−10 hpf and 14−24 hpf exposure
significant changes in the heartbeat compared with the control periods but significantly increased the heartbeat in the periods
group. No heartbeats could be detected in the zebrafish treated at 10−24, 24−48, and 48−72 hpf compared with the control
with high concentrations of free ATA (10−20 μM) as they had group. These results further confirmed that liposomal ATA was
all been killed. Compared with free ATA, lower toxicity was safer than free ATA.
observed in the liposomal-ATA treated group. No signs of Although encapsulating ATA with liposomes reduced its
cardiotoxicity were observed in fish larvae treated with toxicity, liposomal ATA still showed a negative impact during
liposomal ATA at 1−10 μM at 96 hpf. Although the heartbeat early embryonic development. This finding suggested that
of 20 μM at 48 hpf was significantly increased compared with liposomal ATA should not be administered to breast cancer
the control group, this elevation was not observed at 72 hpf patients who are pregnant.
and 96 hpf, which suggested that this side effect could be 3.9. Evaluating the Toxicity of Liposomal ATA in
overcome during fish development. Mice. The results of the toxicity study in zebrafish showed that
3.8. Determining the Effects of 5 μM Liposomal ATA liposomal ATA had much lower toxicity than that of free ATA.
on the Malformation and Mortality of Zebrafish In this study, the acute toxicity evaluation was conducted in
Embryos during Early Development. The above zebrafish mice, which is a required process for the development of
toxicity evaluation showed that 5 μM liposomal ATA did not liposomal ATA into a therapeutic agent. Acute toxicity refers
affect heart function and had much less toxicity than that of to the adverse effects of a substance caused by either a single
free ATA. In this experiment, we further compared the exposure or multiple exposures in a short period of time
differential toxicity between free and liposomal ATA (5 μM) (usually less than 24 h). Because liposomal ATA did not cause
during five major periods of zebrafish development (Figure any mice to die at the dose of 120 mg/kg, which is the highest
8A). dose that could be prepared, this maximum dose was used in
These five time periods are the (1) gastrula period (6−10 this study. Equal numbers of female and male mice (n = 10)
hpf, morphogenetic movements of involution, convergence, were administered with 120 mg/kg liposomal ATA via an
and extension form the epiblast, hypoblast, and embryonic intravenous injection. The saline group and blank liposomes
axis; through the end of epiboly), (2) somite stage (10−24 hpf, were used as controls.
somite, pharyngeal arch primordium, and neuromere develop- After injection, the mice were monitored for 21 days in
ment; primary organogenesis; earliest movement; tail appear- terms of behavioral changes related to the following:
ance), (3) important organ formation stage (14−24 h, primary respiratory system, motor system, convulsive reaction, reflex
organogenesis; earliest movement; the tail appears), (4) system, eyes, cardiovascular system, excretory system, and
pharyngeal phase (24−48 hpf, phylotypic-stage embryo; muscle tension (Table S6). The results revealed no death in
body axis straightens from its early curvature around the either female or male mice after dosing, and the following
yolk sac; circulation, pigmentation, and fins begin to develop), abnormal behaviors were observed. First, all control, liposomal
and (5) hatching period (48−72 hpf, completion of rapid ATA, and blank liposome-treated groups produced reflex
morphogenesis of primary organ systems; cartilage develop- irritation and were sensitive to noise and touch, suggesting
ment in the head and pectoral fin; hatching occurs neuromuscular toxicity. This response quickly recovered,
asynchronously) (Figure 8A). Mortality, abnormality, and implying that the injection process induced a transient stress
heart function were also assessed in each developmental period response. Second, liposomal ATA and blank liposomes
of zebrafish. increased spontaneous activities in mice, which could be
Figure 8B shows the overall deformity and mortality rates of quickly recovered, indicating the occurrence of a minor
zebrafish after the treatment of various samples in different recoverable motor malfunction. Third, liposomal ATA resulted
exposure periods. The sensitivity window was determined in transient salivation, which was related to disturbed
according to the statistical results. No deformity or mortality autonomic functions. In summary, liposomal ATA may have
was observed in the control group during all developmental toxic effects on motor function and autonomic function in
stages. The blank liposomes did not produce significant mice. Liposomal ATA did not cause strong acute toxicity in
toxicity in zebrafish, and less than 18% mortality or deformity male or female mice, and all the aforementioned symptoms
was observed in all tested periods. could be rapidly recovered (i.e., in 1 day), which suggested that
Free ATA caused the highest mortality (100%) during early liposomal ATA was quite safe in mice.
development at 6−10 hpf and resulted in lower mortality As the highest dose that could be prepared for free ATA is
(53.85%) in the later developmental periods at 10−24 hpf. 30 mg/kg, we used this dose to conduct the toxicity study of
Even lower mortality rates of 11.76% and 14.29% were free ATA. Mice appeared to have inspiratory difficulties and
detected in other periods of 14−24 hpf and 24−48 hpf, produced a wheezing sound when they breathed. These
respectively. These results suggested that free ATA mainly disorders involved malfunctions in pulmonary edema, respira-
affected the gastrula period and early somite stage of zebrafish tory secretion accumulation, and enhanced cholinergic
development. In contrast, liposomal ATA significantly function. However, at 120 mg/kg, which is 4 times higher
decreased the toxicity of ATA in zebrafish. For example, than free ATA, liposomal ATA did not cause such problems.
much lower mortality rates (29−31%) were recorded at These results further confirmed our previous findings in
exposure times of 6−10 and 10−24 hpf in comparison to free zebrafish: liposomal ATA is safer than free ATA.
ATA. In addition to observing behavioral changes, the body
In addition to evaluating larval mortality and abnormality, weights of the injected mice were measured at multiple time
zebrafish heart function was also examined. As shown in Figure points over 21 days. The graphs in Figure S3 show that the
K DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

weights of male and female mice increased slightly and that no


significant changes were observed between the liposomal ATA
and saline control group. These results showed that a single
intravenous dose of liposomal ATA at 120 mg/kg would not
affect the normal increment of mouse body weight.
Histopathology analysis was conducted for important
organs, including the heart, liver, spleen, lung, kidney, and
brain at 7 and 21 days post injection by H&E staining. The
images in Figures 9 and 10 showed no obvious necrosis or

Figure 10. Long effect of liposomal ATA on various organs in BALB/


C mice (female) 21 days after injection. Images of H&E staining show
that liposomal ATA caused light congestion in the lung (50×). N = 5
mice/group.

encapsulate a promising anticancer agent ATA into nano-


particles with a suitable size and further evaluate the preclinical
properties. Encapsulating ATA into liposomes significantly
increased its release capacity in vitro and increased the ATA
Figure 9. Short effect of liposomal ATA on various organs of BALB/ plasma concentration 59-fold in rats. The formulation of
C mice (female). Blank liposomes and liposomal ATA (at an liposomal ATA was stable for storage at 4 °C for at least 4 days
equivalent dosage of 120 mg/kg ATA) were injected into mice via a
and after being diluted with 5% glucose for intravenous
single intravenous injection. Images of H&E staining show no obvious
necrosis in the heart, liver, spleen, kidney, and brain 7 days after administration for 12 h. Liposomal ATA displayed strong
injection (50×). More red blood cells were found in lung tissues of potency toward ER+ breast cancer cells by inhibiting their
the liposomal ATA group. N = 5 mice/group. proliferation in vitro and by reducing tumor growth by 73% in
nude mice. At the maximum dose of 120 mg/kg, in 21 days,
liposomal ATA did not show obvious toxicity, such as death,
significant inflammation in all tissues excluding the lung, reduced body weight or damage to major organs. However,
indicating the relatively low toxicity of liposomal ATA. The liposomal ATA showed minor toxicity in zebrafish larvae
lung tissue of the liposomal ATA-treated group appeared during their early development and resulted in a slight
firmer and showed slight congestive phenomena compared abnormality in the lung tissue in mice. Thus, liposomal ATA
with the control group, indicating weak toxicity to the lung should not be given to cancer patients who are pregnant, and
tissue (Figures 9 and 10). A similar phenomenon was observed more clinical studies are needed to determine whether ATA
in the tissue sections of male mice (Figures S4 and S5). should be given to cancer patients with lung disease. The
successful development of ATA into a clinical formulation with
4. CONCLUSIONS improved bioavailability and better anticancer efficacy
How to construct a clinical formulation of anti-breast cancer provided an example for drug formulation. Furthermore, the
chemotherapeutic agent ATA with improved bioavailability is a information obtained from this study can be used to guide the
difficult challenge to meet during drug development. In this future manufacturing and development of liposomal ATA into
study, by using PEG-modified liposomes, we were able to a new anti-breast cancer drug.
L DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article


*
ASSOCIATED CONTENT
S Supporting Information
positive breast cancer cell growth by down-regulating the oestrogen
receptor. Cancer Lett. 2014, 346, 94−103.
(7) Wang, Q.; Wei, N.; Liu, X.; Chang, A.; Luo, K. Q. Enhancement
The Supporting Information is available free of charge on the of the bioavailability of a novel anticancer compound (acetyltan-
ACS Publications website at DOI: 10.1021/acs.molpharma- shinone IIA) by encapsulation within mPEG-PLGA nanoparticles: a
ceut.9b00493. study of formulation optimization, toxicity, and pharmacokinetics.
Orthogonal array test design with four critical factors at Oncotarget 2017, 8, 12013−12030.
three variable levels, results of the orthogonal array tests, (8) Duggan, S. T.; Keating, G. M. Pegylated liposomal Doxorubicin.
analysis of orthogonal array tests, pharmacokinetic Drugs 2011, 71, 2531−2558.
parameters of free and liposomal ATA in rat whole (9) Saw, P. E.; Park, J.; Lee, E.; Ahn, S.; Lee, J.; Kim, H.; Kim, J.;
Choi, M.; Farokhzad, O. C.; Jon, S. Effect of PEG pairing on the
blood after iv administration, pharmacokinetic parame-
efficiency of cancer-targeting liposomes. Theranostics 2015, 5, 746−
ters of free and liposomal ATA in rat plasma after iv 754.
administration, behavioral changes in mice after (10) Xing, H.; Hwang, K.; Lu, Y. Recent Developments of
injection of the maximum dose of liposomal ATA and Liposomes as Nanocarriers for Theranostic Applications. Theranostics
blank liposomes, TG/DTA profiles of the mixture of 2016, 6, 1336−1352.
ATA, cholesterol, and DSPE-PEG, blank liposomes, (11) Park, K. Enhanced immune responses by co-adsorption of
compatibility and stability of liposomal ATA, weight liposomal adjuvant formulations to the aluminum-antigen complex. J.
changes in female and male mice after injection of the Controlled Release 2018, 275, 269.
maximum dose of liposomal ATA and blank liposomes, (12) van der Maaden, K.; Heuts, J.; Camps, M.; Pontier, M.;
toxicity of liposomal ATA in male mice after injection of Terwisscha van Scheltinga, A.; Jiskoot, W.; Ossendorp, F.; Bouwstra,
the maximum dose of liposomal ATA and blank J. Hollow microneedle-mediated micro-injections of a liposomal HPV
liposomes, recovery of male mice after injection of the E743-63 synthetic long peptide vaccine for efficient induction of
maximum dose of liposomal ATA and blank liposomes cytotoxic and T-helper responses. J. Controlled Release 2018, 269,
(PDF) 347−354.


(13) Yang, Y.; Yang, Y.; Xie, X.; Cai, X.; Zhang, H.; Gong, W.;
Wang, Z.; Mei, X. PEGylated liposomes with NGR ligand and heat-
AUTHOR INFORMATION activable cell-penetrating peptide-doxorubicin conjugate for tumor-
Corresponding Author specific therapy. Biomaterials 2014, 35, 4368−4381.
*E-mail: kluo@um.edu.mo. Phone: 853-8822-4233. Fax: 853- (14) Li, Y.; Liu, R.; Yang, J.; Shi, Y.; Ma, G.; Zhang, Z.; Zhang, X.
8822-2314. Enhanced retention and anti-tumor efficacy of liposomes by changing
their cellular uptake and pharmacokinetics behavior. Biomaterials
ORCID 2015, 41, 1−14.
Qi Wang: 0000-0003-3694-5826 (15) Zhu, X.; Sun, Y.; Chen, D.; Li, J.; Dong, X.; Wang, J.; Chen, H.;
Notes Wang, Y.; Zhang, F.; Dai, J.; Pirraco, R. P.; Guo, S.; Marques, A. P.;
The authors declare no competing financial interest. Reis, R. L.; Li, W. Mastocarcinoma therapy synergistically promoted


by lysosome dependent apoptosis specifically evoked by 5-Fu@
ACKNOWLEDGMENTS nanogel system with passive targeting and pH activatable dual
function. J. Controlled Release 2017, 254, 107−118.
This study was supported by grants awarded to K.Q.L. from (16) Wang, Q.; Zhang, X.; Liao, H.; Sun, Y.; Ding, L.; Teng, Y.; Zhu,
the Johns Hopkins Singapore Research Fund, the Start-Up W.-H.; Zhang, Z.; Duan, Y. Multifunctional Shell-Core Nanoparticles
Fund from the Faculty of Health Sciences, and the Start-Up for Treatment of Multidrug Resistance Hepatocellular Carcinoma.
Research Grant (no. SRG2016-00068-FHS) from the Uni- Adv. Funct. Mater. 2018, 28, 1706124.
versity of Macau. We would like to thank Prof. Sierin Lim for (17) Zhang, W.; He, H.; Liu, J.; Wang, J.; Zhang, S.; Zhang, S.; Wu,
her kind help in managing the JHS Research Fund. Z. Pharmacokinetics and atherosclerotic lesions targeting effects of


tanshinone IIA discoidal and spherical biomimetic high density
REFERENCES lipoproteins. Biomaterials 2013, 34, 306−319.
(18) Meng, Z.; Meng, L.; Wang, K.; Li, J.; Cao, X.; Wu, J.; Hu, Y.
(1) Zhou, L.; Zuo, Z.; Chow, M. S. S. Danshen: an overview of its
Enhanced hepatic targeting, biodistribution and antifibrotic efficacy of
chemistry, pharmacology, pharmacokinetics, and clinical use. J. Clin.
Pharmacol. 2005, 45, 1345−1359. tanshinone IIA loaded globin nanoparticles. Eur. J. Pharm. Sci. 2015,
(2) Wang, X.; Morris-Natschke, S. L.; Lee, K. H. New developments 73, 35−43.
in the chemistry and biology of the bioactive constituents of Tanshen. (19) Deb, N. An investigation on solid-state pyrolytic decomposition
Med. Res. Rev. 2007, 27, 133−148. of barium trihemiaquatris(oxalato)lanthanate(III)decahydrate using
(3) Zhou, X.; Razmovski-Naumovski, V.; Chang, D.; Li, C.; Kam, A.; TG-DTA in air and DSC in nitrogen. J. Therm. Anal. Calorim. 2013,
Low, M.; Bensoussan, A.; Chan, K. Synergistic Effects of Danshen 112, 1415−1421.
(Salvia Miltiorrhiza Radix et Rhizoma) and Sanqi (Notoginseng Radix (20) Celiz, L. L.; Arii, T. Study on thermal decomposition of
et Rhizoma) Combination in Inhibiting Inflammation Mediators in polymers by simultaneous measurement of TG-DTA and miniature
RAW264.7 Cells. BioMed Res. Int. 2016, 2016, 5758195. ion trap mass spectrometry equipped with skimmer-type interface. J.
(4) Tian, H.; Ip, L.; Luo, H.; Chang, D. C.; Luo, K. Q. A high Therm. Anal. Calorim. 2014, 116, 1435.
throughput drug screen based on fluorescence resonance energy (21) Viera Dos Santos, B. R.; Montoya Urbina, M.; Souza, M. J. B.;
transfer (FRET) for anticancer activity of compounds from herbal Garrido Pedrosa, A. M.; Silva, A. O. S.; Sobrinho, E. V.; Castedo, R. V.
medicine. Br. J. Pharmacol. 2007, 150, 321−334. Preparation and characterization of Pt-dealuminated Y zeolite by TG/
(5) Tian, H.-L.; Yu, T.; Xu, N.-N.; Feng, C.; Zhou, L.-Y.; Luo, H.- DTA and TPR. J. Therm. Anal. Calorim. 2015, 119, 391−399.
W.; Chang, D. C.; Le, X.-F.; Luo, K. Q. A novel compound modified (22) Lin, J.; Wang, X.; Wu, Q.; Dai, J.; Guan, H.; Cao, W.; He, L.;
from tanshinone inhibits tumor growth in vivo via activation of the Wang, Y. Development of Salvianolic acid B-Tanshinone II A-
intrinsic apoptotic pathway. Cancer Lett. 2010, 297, 18−30. Glycyrrhetinic acid compound liposomes: formulation optimization
(6) Yu, T.; Zhou, Z.; Mu, Y.; de Lima Lopes, G.; Luo, K. Q. A novel and its effects on proliferation of hepatic stellate cells. Int. J. Pharm.
anti-cancer agent, acetyltanshinone IIA, inhibits oestrogen receptor 2014, 462, 11−18.

M DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX
Molecular Pharmaceutics Article

(23) Xu, J.; Luo, K. Q. Enhancing the solubility and bioavailability of


isoflavone by particle size reduction using a supercritical carbon
dioxide-based precipitation process. Chem. Eng. Res. Des. 2014, 92,
2542−2549.
(24) Zhang, J.; Li, Y.; Fang, X.; Zhou, D.; Wang, Y.; Chen, M.
TPGS-g-PLGA/Pluronic F68 mixed micelles for tanshinone IIA
delivery in cancer therapy. Int. J. Pharm. 2014, 476, 185−198.
(25) Soni, P.; Kaur, J.; Tikoo, K. Dual drug-loaded paclitaxel-
thymoquinone nanoparticles for effective breast cancer therapy. J.
Nanopart. Res. 2015, 17, 18.
(26) Rosenzweig, O.; Lavy, E.; Gati, I.; Kohen, R.; Friedman, M.
Development and in vitro characterization of floating sustained-
release drug delivery systems of polyphenols. Drug Delivery 2013, 20,
180−189.
(27) Nguyen, C.; Christensen, J. M.; Ayres, J. W. Compression of
coated drug beads for sustained release tablet of glipizide:
formulation, and dissolution. Pharm. Dev. Technol. 2012, 19, 10−20.
(28) Truong, L.; Moody, I. S.; Stankus, D. P.; Nason, J. A.;
Lonergan, M. C.; Tanguay, R. L. Differential stability of lead sulfide
nanoparticles influences biological responses in embryonic zebrafish.
Arch. Toxicol. 2011, 85, 787−798.
(29) Abdel-moneim, A.; Moreira-Santos, M.; Ribeiro, R. A short-
term sublethal toxicity assay with zebra fish based on preying rate and
its integration with mortality. Chemosphere 2015, 120, 568−574.
(30) Devi, G. P.; Ahmed, K. B. A.; Varsha, M. K. N. S.; Shrijha, B. S.;
Lal, K. K. S.; Anbazhagan, V.; Thiagarajan, R. Sulfidation of silver
nanoparticle reduces its toxicity in zebrafish. Aquat. Toxicol. 2015,
158, 149−156.
(31) Wang, Q.; Liu, P.; Sun, Y.; Gong, T.; Zhu, M.; Sun, X.; Zhang,
Z.; Duan, Y. Preparation and properties of biocompatible PS-PEG/
calcium phosphate nanospheres. Nanotoxicology 2014, 9, 190−200.
(32) Wang, Q.; Shen, M.; Zhao, T.; Xu, Y.; Lin, J.; Duan, Y.; Gu, H.
Low toxicity and long circulation time of polyampholyte-coated
magnetic nanoparticles for blood pool contrast agents. Sci. Rep. 2015,
5, 7774.
(33) Shahbazi, M.A.; Hamidi, M.; Mohammadi-Samani, S.
Preparation, optimization, and in-vitro/in-vivo/ex-vivo character-
ization of chitosan-heparin nanoparticles: drug-induced gelation. J.
Pharm. Pharmacol. 2013, 65, 1118−1133.
(34) He, G.; Zhang, L.; Zhou, D.; Zou, Y.; Wang, F. The optimal
condition for H2TiO3-lithium adsorbent preparation and Li+
adsorption confirmed by an orthogonal test design. Ionics 2015, 21,
2219−2226.

N DOI: 10.1021/acs.molpharmaceut.9b00493
Mol. Pharmaceutics XXXX, XXX, XXX−XXX

You might also like