You are on page 1of 14

Journal of Controlled Release 272 (2018) 145–158

Contents lists available at ScienceDirect

Journal of Controlled Release


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

Light/magnetic hyperthermia triggered drug released from multi-functional T


thermo-sensitive magnetoliposomes for precise cancer synergetic
theranostics
Yuxin Guoa, Yang Zhangb, Jinyuan Maa, Qi Lia, Yang Lic, Xinyi Zhoua, Dan Zhaoa, Hua Songa,
⁎ ⁎
Qing Chena, , Xuan Zhua,
a
Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
b
State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health,
Xiamen University, Xiamen, China
c
College of Materials, Xiamen University, Xiamen, China

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

Keywords: Precise delivery of antineoplastic drugs to specific tumor region has drawn much attention in recent years.
Thermo-sensitive Herein, a light/magnetic hyperthermia triggered drug delivery with multiple functionality is designed based on
Magnetoliposomes methotrexate (MTX) modified thermo-sensitive magnetoliposomes (MTX-MagTSLs). In this system, MTX and
Light/magnetic hyperthermia oleic acid modified magnetic nanoparticles (MNPs) can be applied in biological and magnetic targeting.
Triggered release
Meanwhile, lipophilic fluorescent dye Cy5.5 and MNPs are encapsulated into the bilayer of liposomes, which can
Magnetic/folate receptor targeting
Fluorescence/magnetic resonance imaging
not only achieve dual-imaging effect to verify the MTX-MagTSLs accumulation in tumor region, but also provide
an appropriate laser irradiation region to release Doxorubicin (Dox) under alternating magnetic field (AMF).
Both in vitro and in vivo results revealed that MTX-MagTSLs possessed an excellent targeting ability towards HeLa
cells and HeLa tumor-bearing mice. Furthermore, the heating effect of MTX-MagTSLs was amplified 4.2-fold
upon combination with AMF and local precise near-infrared laser irradiation (808 nm) (DUAL-mode) to rapidly
reach the phase change temperature (Tm) of MTX-MagTSLs in 5 min compared with either AMF or laser sti-
mulation alone, resulting in a significantly enhanced release of Dox at tumor region and precise cancer syner-
getic theranostics.

1. Introduction Folate-mediated receptor targeting is found to be promising in li-


posomal drug delivery for various cancer cells, including ovarian
Following decades of research in drug delivery systems, nanoscale [9–11], breast [12,13], kidney, lung, brain, endometrial [14,15], and
liposomal formulations is considered as one of the most advanced de- colon cancer cells [16], all of which overexpress folate receptor on the
livery systems for cytotoxic drugs [1,2]. Long circulating Dox-loaded surface of cell membranes. Besides acting the role of anticancer drug,
liposomes (Caelyx®, Schering-Plough; DOXIL®, Ortho Biotech), which MTX also has a good targeting effect to folate receptor-overexpressed
have been used in clinical cancer therapy for many years [3], is proved cancer cells such as human cervical carcinoma (HeLa) cells, due to the
to reduce cardiotoxic side effects [4] and improve pharmacokinetic similar structure with folate [17–20]. MTX can be connected to phos-
profile compared with free Dox due to the enhanced permeability and pholipid molecules through amide reaction for functionalization of
retention effect (EPR) [5]. However, the therapeutic efficacy of lipo- TSLs, which introduce folate receptor-mediated active targeting
somal formulations is still undesired. Recent research reveal that it can strategy in the treatment of cancer [21]. Although several targeting
be further enhanced by either biological/physical targeting [6] or lo- strategies have been developed in recent decades, the folate receptor-
calized triggered drug release [7] from responsive liposomal formula- mediated active targeted delivery is still limited by the insufficient drug
tions to increase the drug concentration at the target site. Hence, tar- release at tumor site, which can be improved by local temperature-
geted and thermo-sensitive liposomes (TSLs) attract much attention due triggered drug release from the TSLs [22–24].
to the capacity of releasing encapsulated molecules when the tem- MNPs, in particular iron oxides, is approved by Food and Drug
perature reach near Tm caused by the decomposition of TSLs [8]. Administration for in vivo usage due to their low toxicity and good


Corresponding authors.
E-mail addresses: chenqing@xmu.edu.cn (Q. Chen), zhuxuan@xmu.edu.cn (X. Zhu).

https://doi.org/10.1016/j.jconrel.2017.04.028
Received 24 November 2016; Received in revised form 6 April 2017; Accepted 13 April 2017
Available online 23 April 2017
0168-3659/ © 2017 Published by Elsevier B.V.
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

biocompatibility. It is applied widely in biomedical fields [25,26]. For (Carlsbad, CA). Dimethyl sulfoxide (DMSO) was brought from Aladdin.
instance, MNPs can be used in magnetic targeting, thermal therapy, and HeLa cells and A549 cells were provided by American Type Culture
magnetic resonance imaging (MRI) [27,28], which allow to monitor the Collection (ATCC). Dulbecco's minimum essential medium (DMEM)
biodistribution in vivo non-invasively. Magnetic TSLs (MagTSLs, mag- was obtained from GE Healthcare Life Sciences HyClone Laboratories.
netic nanoparticles encapsulated within TSLs) appear to realize mag- Fetal bovine serum (FBS) was purchased from Zhejiang Tianhang
netocaloric mediated triggered drug release based on Brown and Neel Biological Technology Co., Ltd. (Hangzhou, China). Penicillin and
relaxations [29,30]. In order to achieve a high magnetocaloric con- streptomycin were from Beyotime Biotechnology (Shanghai, China). All
version efficiency, MNPs are embedded into the hydrophobic bilayer to other chemicals were analytical grade and were used without further
lead heat dissipating directly into the surrounding bilayer and bring the purification. The dialysis bags (MWCO = 3500) were from BIOSHARP
temperature across the Tm of the liposomes to achieve drug release (Hefei, China).
effect [31].
In addition, it is reported that MagTSLs can convert radiofrequency 2.2. Synthesis and characterization of DSPE-PEG2000-MTX
irradiation energy into heat energy under AMF to trigger drug release
when the temperature reach near the Tm of TSLs [32,33]. However, DSPE-PEG2000-MTX conjugate was synthesized by a previously de-
such a magnetocaloric way still suffers from several disadvantages, scribed method [21] with minor modification. Briefly, 16.4 mg of MTX,
including the injure of the health tissues and the low heating efficiency 4.2 mg of NHS, and 7.4 mg of DCC were dissolved in 2 mL of DMSO in a
which brings inefficient drug release, both of which could impede the round-bottomed flask, and reacted for 6 h under nitrogen atmosphere
clinical translation of the magnetocaloric therapy. In recent study, it is protection to activate the carboxyl group of MTX. Then, 20 mg of DSPE-
demonstrated that magnetocaloric effect of iron oxide-based nanocubes PEG2000-NH2 was added to the round-bottomed flask, which was dis-
can be significantly enhanced when in combination with laser treat- solved in 1.5 mL of DMSO containing 6 μL of TEA. The reaction mixture
ment [34]. Thus, we assume that the heating effect of MagTSLs can also was carried on stirring for an additional 72 h under nitrogen atmo-
be increased appreciably under the simultaneous stimulation of AMF sphere protection, and filtered to remove the precipitate after reaction
and laser, achieving a better enhanced drug release effect as well as mixture diluting into 2 times with water. The reaction progress was
reducing the side effects. monitored by thin layer chromatography (TLC) according to the re-
Incorporating both antineoplastic drugs and imaging agents within duced quantity of DSPE-PEG2000-NH2/MTX and increased amount of
a multi-functional carrier combined with active targeting, followed by DSPE-PEG2000-MTX. The prepared product was dialyzed against PBS
delivering and releasing the right drug to the right location at the right and DI water for 48 h respectively to completely remove excess MTX
time based on certain mechanism, which can achieve precise cancer and byproducts, and then lyophilized for 24 h to obtain solid DSPE–-
synergetic theranostics [35–37]. Herein, we design the MTX modified PEG2000–MTX. The impurities were further monitored by TLC to detect
TSLs encapsulating MNPs/Cy5.5 in lipid bilayer and Dox in hydrophilic whether been removed completely or not after dialysis. The final pro-
lumen, combining biological/physical targeting, light/magnetic medi- duct was analyzed by mass spectrometry (MS) and Fourier transform
ated triggered drug release, and fluorescence/MR imaging for the infrared spectrometry (FT-IR). MS was performed with a matrix assisted
cancer therapy. A conceptual scheme for MTX-MagTSLs targeting to laser desorption/ionization time-of-flight mass spectrum (MALDI-TOF-
tumor site and releasing Dox locally is shown in Fig. 1. Magnetic and MS), using DSPE-PEG2000-NH2 for comparison. FT-IR was determined
folate receptor targeting effect support and accelerate MTX-MagTSLs on a Nicolet Avatar370 Fourier Transform Infrared Spectrometer, using
targeting to tumor tissue from intravenous injection in nude mouse, DSPE-PEG2000-NH2 and MTX for comparison.
then proceed cell uptake and accelerate drug release at tumor site under
the simultaneous application of AMF and laser followed by working 2.3. Preparation of magnetic nanoparticles (MNPs)
finally in nuclear to cause tumor cell killing accurately.
To the best of our knowledge this is the first report committed to 1 mM of Iron (III) acetylacetonate was added into 9 mL of benzyl
precise medicine about using AMF and near-infrared laser irradiation alcohol, and then reacted under 200 °C for 10 h after adding 1.5 mL of
(808 nm) synchronously to stimulate the rapid release of drug from oil amine and 1.5 mL of oleic acid quickly, followed by centrifuging
multi-functional MagTSLs with dual-mode targeting and dual-mode 5 min with the speed of 10000 rpm. After removing of supernatant, the
imaging, which is expected to improve therapeutic efficacy, reduce the oleic acid-coated iron oxide NPs were dispersed in 10 mL of chloroform
side effects of Dox, and realize the precise cancer synergetic ther- for storage.
anostics.
2.4. Preparation of Dox loaded MTX-MagTSLs
2. Materials and methods
MTX-MagTSLs were prepared by thin film dispersion method. In this
2.1. Materials process, 15 mg of total lipids composed of DPPC:Chol:SA:DSPE-
MPEG2000:DSPE-PEG2000-MTX (mol ratio 67:17:13:1:2), containing
Dipalmitoylphosphatidylcholine (DPPC), 1,2-diacyl-SN-glycero-3- 5 mg of MNPs and 100 μg of Cy5.5, were dissolved in 10 mL of
phosphoethanolamine-N-[methoxy(poly(ethyleneglycol))-2000] (DSPE- chloroform in a round bottom flask. Then the chloroform was evapo-
MPEG2000), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[ami rated using a rotator evaporator (N-1100, TOKOY RIKAKIKAI CO.LTD)
no(polyethylene glycol)-2000] (DSPE-PEG2000-NH2), and N-hydro- at 50 °C to form a thin film on walls of eggplant type flask, starting at
xysuccinimide (NHS) were purchased from Shanghai Ponsure 0.05 MPa for 10 min, then increased to 0.9 MPa for 30 min, then the
Biotechnology (Shanghai, China). Cholesterol (Chol) was obtained from flask was frozen at − 20 °C for one night. For loading Dox with an
Shanghai Advanced Vehicle Technology Co. Ltd. (Shanghai, China). ammonium sulfate gradient loading method [38], 250 mM of ammo-
Octadecanamine (SA) was obtained from Sigma-Aldrich. Doxorubicin nium sulfate solution was added into the flask, followed by hydration at
(Dox) was from Beijing Huachunlianbo Technology Co., Ltd. (Beijing, 55 °C (above the Tm of MTX-MagTSLs) for 50 min while rotating at
China). Iron (III) acetylacetonate was purchased from Alfa Aesar. 110 rpm at atmospheric pressure using the rotary evaporator, followed
Methotrexate (MTX) was purchased from Bio Basic Inc. (Markham, by ultrasound for 30 min and being extrude through 450 nm, 200 nm,
Ontario, Canada). Dicyclohexylcarbodiimide (DCC) was purchased and 100 nm polycarbonate membranes using an extruder (Avanti Polar
from J&K Chemical Scientific Co. Ltd. (Beijing, China). Cy5.5 ester was Lipids, USA) for size control. The prepared MTX-MagTSLs were dia-
obtained from AAT Bioquest. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphe- lyzed against PBS for 2 h. 1 mg of Dox was then added into the sus-
nyltetrazo-lium bromide (MTT) was obtained from Life Technologies pension to incubate at 40 °C for 30 min. Unencapsulated Dox was

146
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Fig. 1. Schematic illustration of realizing theranostic functionalities using MTX-MagTSLs. The multi-functional MTX-MagTSLs can be targeted to tumor site under constant magnetic field
(CMF) and folate receptor targeting, followed by triggering Dox release under light/magnetic hyperthermia simulation synchronously to achieve the effect of drug treatment. The process
can be monitored by fluorescence and MR imaging.

removed by gel filtration using Sephadex G50 in a HR 10/30 column Weight Dox of encapsulation
Dox loading efficiency (%) =
(GE Healthcare) with phosphate buffered saline (PBS) as eluent at a Initial weight Total
flow rate of 1 mL/min.
The lipid concentration of liposomes was determined using phos-
phorus molybdenum blue spectrophotometry as described previously
2.5. Payload and encapsulation efficiency with minor modification [41] after the liposomes destruction with
chloroform and perchloric acid, then converted to phospho-molybdic
After addition of methyl alcohol for demulsification, the Dox con- acid by the addition of ammonium molybdate followed by being re-
centration of MTX-MagTSLs was determined by EnVision Multilabel duced to a blue colored complex by ascorbic acid during heating at
Reader (EnVision, USA) under 490/550 nm [39]. A calibration curve 65 °C for 20 min, which was determined colorimetrically at 819 nm
was used for quantification by calculating the Dox concentration of a using an ELISA reader (Multiskan™ Go, Thermo Fisher Scientific,
dilution series of free Dox in PBS (0.1–5 μg·mL− 1 of Dox concentra- America). A calibration curve was used for quantification by calculating
tion). The Dox encapsulation and loading efficiency was calculated the lipid concentration of a dilution series of monopotassium phosphate
using the following formula [40]: solution (0.4–2 μg·mL− 1 of phosphorus concentration). The con-
centration of iron of MTX-MagTSLs was measured by Inductively cou-
Weight Dox of encapsulation pled plasma mass spectrometry (7500CE, Agilent Technologies, USA)
Dox encapsulation efficiency (%) =
Initial weight Dox after being treated with aqua regia for dissolving iron and diluted with
2% dilute nitric acid. A calibration curve was used for quantification by

147
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

calculating the iron concentration of a dilution series of ferric chloride 2.8. In vitro release study
hexahydrate solution in 2% dilute nitric acid (5–120 ng·mL− 1 of iron
concentration). Then the encapsulation efficiency of MNPs was calcu- To determinate the release of Dox from MTX-MagTSLs with
lated by using the final quality of MNPs of MTX-MagTSLs (converted by 13.93 mM of iron, a cellulose membrane (molecular weight cut-off of
the final quality of iron) divided by the initial amount of MNPs (5 mg of 3500 kDa, Solarbio, China) was mounted between the donor and re-
the drying MNPs). The concentration of MTX of MTX-MagTSLs was ceptor compartments. The donor medium was consisted of 500 μL of
measured by UV-VIS spectrophotometer (UV-2550, SHIMADZU, Japan) free Dox solution or MTX-MagTSLs formulation with the same con-
due to its absorption to specific wavelengths of ultraviolet [21] after centration of Dox (0.182 mg·mL− 1). The receptor medium consisted of
demulsification and dilution with methanol. A calibration curve was 200 mL of PBS (10 mM, pH 7.4). During the dialysis, the temperature
used for quantification by calculating the MTX concentration of a di- was kept at 37 °C and 45 °C respectively. At pre-determined time in-
lution series of free MTX solution in methanol (1–25 μg·mL− 1 of MTX tervals, the amount of the released Dox was determined by EnVision
concentration). Multilabel Reader (EnVision, USA) under 490/550 nm from 0 to 24 h.

2.9. Hemolysis assay


2.6. Characterization
1 mL of fresh collected whole blood was centrifuged for 5 min at
X-ray diffraction (XRD, Rigaku Ultima IV) was used to determine 4 °C with 10000 rpm to isolate red blood cells, then it was washed three
the crystallinity and phase composition of MNPs. Magnetic properties of times with the isotonic icy PBS and suspended in 10 mL of icy PBS.
the MNPs and MTX-MagTSLs was measured with a vibrating sample 0.5 mL of the prepared solution above was added to 0.5 mL of MagTSLs
magnetometer (VSM). The T2-weighted MR images in vitro were con- at different phospholipid concentration (between 25 and 400 μg·mL− 1)
ducted on a 7-T MRI scanner (Varian, Palo Alto, CA, USA). To evaluate suspended in PBS, followed by an intensive mixing and retaining for 1 h
the stability of MTX-MagTSLs at 4 °C, the size distribution and zeta at 37 °C, then centrifuged 5 min with 3000 rpm. The absorbance value
potential of MTX-MagTSLs were determined using a Malvern zetasizer of supernatant liquid containing hemoglobin was determined at
(Malvern instrument, Worcestershire, UK) apparatus in a week, and the 560 nm. A similar volume of PBS was used as negative control and de-
changes of the encapsulation and loading efficiency of Dox of MTX- ionized (DI) water as the positive control. The percentage of hemolysis
MagTSLs were measured as well within 7 days. Each experiment was was calculated using the formula: hemolysis value (%) =
done in triplicate. The morphology of the MNPs and MTX-MagTSLs (Asample − Anegative control) / (Apositive control − Anegative control) × 100%.
were characterized by transmission electron microscopy (TEM, Tecnai
G2 Spirit, FEI, Hong Kong) and scanning electron microscopy (SEM, 2.10. Pharmacokinetic study
Zeiss SIGMA, Germany). The elemental analysis of MTX-MagTSLs was
detected by energy dispersive spectrometer (EDS, X-MaxN, Oxford, UK). 0.5 mL of blood was drawn from eyes of tumor-free healthy SD rats
The Tm of different lipids compositions was measured by differential after intravenous administration of free Dox and MTX-MagTSLs (Dox
scanning calorimetry (VP-Capillary, Malvern instrument, UK) between dose: 2 mg·kg− 1) at 5, 15, 30, 60, 120, 180, 240, 300, 420, 540, and
10 °C and 90 °C at a heating rate of 1 °C/min. 1440 min, respectively, followed by centrifuging 10 min at 10000 rpm.
Then 0.1 mL of supernatant was added into 0.3 mL of acid ethanol
solution (12.5 mL of concentrated hydrochloric acid mixed with
2.7. Specific absorption rate (SAR) measurement 250 mL of dehydrated alcohol then added into distilled water as to
500 mL). A calibration curve was used for quantification by calculating
After measuring the absorption spectra of MTX-MagTSLs with a the Dox concentration of a dilution series of free Dox in acidic ethanol
series of iron concentration (ranging from 2.5 to 15 mM) to investigate prepared above. Each group comprised six rats. The Dox concentration
its absorbing ability to 808 nm near-infrared light, the heating ability of of the solution was determined by EnVision Multilabel Reader
MNPs and MTX-MagTSLs with 13.93 mM of iron were measured by SAR (EnVision, USA) under 490/550 nm after mixing by vortexing. The
based time-dependent calorimetric measurements under AMF pharmacokinetic parameters were calculated by fitting the blood drug
(500 kHz, 20 kA·m− 1, the sample was positioned in the center of a 10- concentrations to a non-compartment model using WinNonlin
turn circular copper coil with a 8 cm outer diameter), 808 nm near- Professional Edition Version 2.1 (Pharsight Corporation, Mountain
infrared laser irradiation (0.8 W/cm2, the sample was placed just below View, California).
the laser source), and DUAL-mode (Laser 0.8 W/cm2, AMF 500 kHz,
20 kA·m− 1) for 5 min respectively. The PBS and TSLs were measured in 2.11. Cell culture
negative control mode under the same condition. The impacts of the
concentration of iron (ranging from 2.5 to 15 mM) and the laser power HeLa (human cervical carcinoma) and A549 (human lung adeno-
(0.3, 0.8, and 1.2 W/m2) to heat absorption capacity of MTX-MagTSLs carcinoma) cell lines were cultured in DMEM supplemented with 10%
were investigated with the same mode of treatment as well. An alter- fetal calf serum and 100 μg/mL of streptomycin/penicillin at 37 °C in a
nating magnetic field was produced by an AMF generator (SPG-10-II, humidified atmosphere of 5% CO2.
Shenzhen double-Power Technology Co. Ltd. China). Near-infrared
laser irradiation (808 nm) was produced by Stone Laser (STL 808CF- 2.12. Cellular uptake
10W, China). The temperature of the sample was monitored by a
thermocouple thermometer (TES-1310, Taiwan). The SAR value was The cells were seeded at the density of 1 × 105 cells per well in the
calculated using the following equation [42]: 6-well plates with complete DMEM. The cells were incubated at 37 °C in
5% CO2 for 24 h. The free Dox and MTX-MagTSLs (10 μg·mL− 1 at Dox-
△T m eq. dose) were added into HeLa cells and incubated further for 1 and
SAR = CP × V
△t mNP 4 h. The cells were washed with PBS, fixed with 4% paraformaldehyde
and stained with DAPI. The fluorescence intensity of cells was observed
where CP is the heat capacity of the medium, ΔT/Δt is the experimen- using a Leica TCS SP5 LCSM (Leica Microsystems, Mannheim,
tally observed heating rate, mV is the mass of the suspension, mNP is the Germany). Additionally, to investigate the targeting ability of MTX-
iron content of the suspension. MagTSLs against HeLa cells, MagTSLs and MTX-MagTSLs (10 μg·mL− 1
at Dox-eq. dose) were added into HeLa and A549 cells respectively, and

148
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

incubated further for 4 h. To investigate the impact of AMF and laser on 2.16. In vivo fluorescence imaging
the drug release, MTX-MagTSLs (10 μg·mL− 1 at Dox-eq. dose) were
added into HeLa cells and incubated further for 1 h after AMF (500 kHz, To evaluate the synergetic tumor targeting effect by using MTX and
20 kA·m− 1, 5 min) and DUAL-mode (0.8 W/cm2, 500 kHz, 20 kA·m− 1, CMF through the targeting capability of the TSLs, MagTSLs, MTX-TSLs,
5 min) respectively. MTX-MagTSLs, and MTX-MagTSLs/CMF, the HeLa tumor-bearing nude
mice were randomly divided into six groups (three mice per group),
2.13. Flow cytometry minimizing the difference of weights and tumor sizes in each group.
0.2 mL of PBS, TSLs, MagTSLs, MTX-TSLs, and MTX-MagTSLs at
To measure the internalization of free Dox, MagTSLs, and MTX- 2 mg·kg− 1 (Dox-eq. dose) were injected into the mice intravenously
MagTSLs in the absence of external stimulation of physical conditions, respectively to investigate the tumor targeting efficiency. Two groups
and MTX-MagTSLs under AMF (500 kHz, 20 kA·m− 1, 5 min), near-in- were injected with MTX-MagTSLs, and one of group was fixed with a
frared laser irradiation (808 nm, 0.8 W/cm2, 5 min), and DUAL-mode 10 mm diameter circular magnet on skin surface of tumor (MTX-
(0.8 W/cm2, 500 kHz, 20 kA·m− 1, 5 min) respectively, the HeLa and MagTSLs/CMF). All mice were used for fluorescence imaging of Cy5.5
A549 cells were incubated in 6-well plates at a density of 1 × 105 cells using a Maestro™ in vivo imaging system (Cambridge Research &
per well for 24 h at 37 °C in 5% CO2, followed by being cultured with Instrumentation, Woburn, MA, USA) at 4, 8, and 12 h after injection.
free Dox, MagTSLs, and MTX-MagTSLs (10 μg·mL− 1 at Dox-eq. dose) The tumor and major organs (heart, liver, spleen, lung, and kidney)
for 1 h or 4 h. The wells cultured with MTX-MagTSLs were treated with were excised at 12 h post-injection, followed by washing with 0.9%
no stimulation, AMF (500 kHz, 20 kA·m− 1, 5 min), 808 nm near-in- NaCl for the ex vivo imaging.
frared laser irradiation (0.8 W/cm2, 5 min), and DUAL-mode (Laser
0.8 W/cm2, AMF 500 kHz, 20 kA·m− 1, 5 min) respectively. After the 2.17. In vivo MRI
incubation for the prescribed period, the cells were washed with cold
PBS, harvested by 0.25% trypsin-EDTA, centrifuged with 1000 rpm for To examine the suitability of MTX-MagTSLs for in vivo MRI and its
5 min and resuspended in PBS. The fluorescence intensity of the cells dual-targeting (magnetic and folate receptor) ability, HeLa tumor-
was assessed using a Beckman Coulter ALTRA Epics flow cytometer bearing nude mice were intravenously injected with 0.2 mL of MTX-
(Beckman Coulter, CA, USA). MagTSLs at 2 mg·kg− 1 (Dox-eq. dose) for in vivo MRI. The mice in MTX-
MagTSLs/CMF group were fixed with a 10 mm diameter circular
magnet on skin surface of tumor (taken away before MRI), and the mice
2.14. Cytotoxicity study in control group were not fixed the magnet (without CMF). After in-
jection for 0, 4, 8, and 12 h, the pseudo-color processing T2-weighted
The MTT assay was used to evaluate the cytotoxicity of the Dox-free MR images were conducted on a 7-T MRI scanner (Varian, Palo Alto,
MagTSLs and Dox-free MTX-MagTSLs against HeLa cell, MTX-MagTSLs CA, USA).
against HeLa cell and A549 cells, and MTX-MagTSLs against HeLa cells
under AMF (500 kHz, 20 kA·m− 1), 808 nm near-infrared laser irradia- 2.18. In vivo anticancer effect
tion (0.8 W/cm2), and DUAL-mode (Laser 0.8 W/cm2, AMF 500 kHz,
20 kA·m− 1) respectively. The cells were seeded into 96-well plates at a HeLa tumor-bearing nude mice were randomly divided into seven
density of 5000 cells per well in 100 μL of complete DMEM, followed by groups and treated as follows: PBS, free Dox, MagTSLs, MTX-MagTSLs,
24 h of incubation, then added with Dox-free MagTSLs at different MTX-MagTSLs/CMF, MTX-MagTSLs/CMF/AMF, and MTX-MagTSLs/
phospholipid concentrations (25, 50, 100, 200, and 400 μg/mL) for CMF/AMF/Laser. Each group comprised six mice. The mice were in-
24 h to study the toxicity of Dox-free MagTSLs, free MTX and Dox-free jected intravenously with different formulation at 2 mg·kg− 1 (Dox-eq.
MTX-MagTSLs which contained the same MTX concentrations with dose) every two days. In addition, the MTX-MagTSLs/DUAL group was
MTX-MagTSLs (2.5–10 μg·mL− 1 at Dox-eq, 0.6–2.4 μg·mL− 1 at MTX- added to complementally verify dual-targeting ability for comparison
eq.) for 24 h to investigate the toxicity of MTX, free Dox/TSLs/ with the MTX-MagTSLs/CMF/DUAL group. The weight of every mice
MagTSLs/MTX-TSLs/MTX-MagTSLs at different Dox concentrations was recorded every two days. The tumor sizes were measured using a
(2.5, 5, and 10 μg·mL− 1) for 24 h respectively to investigate the toxicity caliper every two days and calculated as volume = (tumor
of MTX-MagTSLs, and MTX-MagTSLs at several concentrations (2.5, 5, length) × (tumor width)2 / 2, until the animals were given a eu-
and 10 μg·mL− 1 at Dox-eq.) for 12 h under no stimulation, AMF thanasia on day 15. Recording each group's survival situation as well.
(500 kHz, 20 kA·m− 1, 5 min), near-infrared laser irradiation (808 nm, For histological examination, the heart, liver, spleen, lung, kidney, and
0.8 W/cm2, 5 min), and DUAL-mode (0.8 W/cm2, 500 kHz, 20 kA·m− 1, tumor harvested from all groups were fixed in 10% buffered formalin,
5 min) respectively after 2 h incubation to inspect the toxicity of MTX- then embedded in paraffin, sectioned, stained with hematoxylin/eosin
MagTSLs under different conditions. The medium in each well was (H&E), and examined by digital microscopy (D-35578 Wetzlar, Leica
replaced by 100 μL of (3-(4,5)-dimethylthiazol-2-yl)-2,5-diphenylte- Microsystems CMS GmbH, German).
trazolium bromide dye (MTT) solution (500 μg/mL in DMEM), followed
by incubating for 4 h at 37 °C and 5% CO2. Then, the medium was re- 2.19. Data analysis
moved and added with 100 μL of dimethyl sulfoxide (DMSO) per well.
Absorbance was measured by using a spectrophotometer (Biotek, USA) Quantitative data were expressed as means ± standard deviation
at 490 nm, the relative cell viability (%) was calculated by (SD). The analysis of variance was performed to determine the sig-
(Abssample − Absbackground) / (Abscontrol − Absbackground) × 100%. nificance level among the tested groups and p values < 0.05 were
considered to be statistically significant.
2.15. Xenograft tumor mouse model
3. Results and discussion
All female BALB/c nude mice (16–18 g) were supplied by Xiamen
University Laboratory Animal Center. The HeLa tumor models were 3.1. Synthesis and characterization of DSPE-PEG2000-MTX
generated by subcutaneous injection of 2 × 106 cells in 0.1 mL of PBS
into the right shoulder, and the mice were used when the tumor volume Carboxyl group of MTX was activated and then reacted with amine
reached 60–100 mm3. All animal experiments were performed under a group of DSPE-PEG2000-NH2 to form stable amide linkage. After that,
protocol approved by Xiamen University Laboratory Animal Center. excessive amounts of MTX and other impurities were removed by

149
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

dialysis using PBS buffer/water. The reaction and purification process and hydrophilic Dox in hydrophilic lumen of MTX modified TSLs. As
were monitored by TLC. The peak of DSPE-PEG2000-MTX in the mass observed in TEM (Fig. 2B), the MNPs were uniformly scattered and
spectra (Fig. S1A) shifted a molecular weight of approximately 454 showed a diameter of about 4 nm, which was in agreement with the
(consistent with the molecular weight of MTX) to right. The infrared conclusion that a threshold maximum NPs diameter of 6.5 nm for in-
spectrum of DSPE-PEG2000-MTX (Fig. S1B) gave peaks at 1637 cm− 1 corporation of NPs into lipid bilayers [31]. The morphology of MTX-
for C]O stretching vibration absorption peak, 1486 cm− 1 for NeH MagTSLs was characterized by TEM (Fig. 2C) and SEM (Fig. S2). It
bending vibration absorption peak, and 1398 cm− 1 for CeN stretching could be seen that MTX-MagTSLs had a uniform size with oleic acid-
vibration absorption peak, derived from the amide bond in structure of coated iron oxide NPs scattered in the phospholipid bilayer, suggesting
DSPE-PEG2000-MTX. These results confirmed the successful synthesis of that magnetic nanoparticles were embedded in the lipid membrane.
DSPE-PEG2000-MTX which could be incorporated into TSLs later for The elemental analysis (Fig. S2) showed the content of major elements.
targeting to tumor cells via the folate receptor. Fig. 2D showed the UV–Vis spectra of the free MTX, Mag-TSLs, and
MTX-MagTSLs in methanol. The free MTX and MTX-MagTSLs gave
3.2. Synthesis and characterization of MNPs and MTX-MagTSLs peaks at around 300 nm deriving from the structure of MTX [43]. MTX
methanol solution revealed a good linear relationship between its
MNPs were synthesized using a high temperature decomposition concentration and absorption value at the maximum absorption wa-
method. Then MTX-MagTSLs were prepared by the thin film dispersion velength in the range of 1–25 μg·mL− 1 (Fig. 2E), which calculated the
method using MNPs and TSLs, followed by encapsulating Dox using final concentration of MTX of MTX-MagTSLs (42.7 μg·mL− 1) after de-
ammonium sulfate gradient loading method. The concentration of lipid, mulsification and dilution with methanol. It has been found that MTX-
iron, and Dox of MTX-MagTSLs were 3.12 mg·mL− 1, 13.93 mM, and MagTSLs tended to form monodisperse aggregates in the size of
0.182 mg·mL− 1 respectively. The encapsulation efficiency of MNPs in 107.5 ± 1.19 nm primitively (Fig. 2F) with almost no change after a
MTX-MagTSLs was 87.6 ± 1.83%. The encapsulation efficiency and week (112.3 ± 2.19 nm) as confirmed by DLS, and the zeta potential
loading efficiency of Dox were 72.74 ± 1.68% and 3.64 ± 0.08% also presented near 16.8 ± 1.2 mV primitively (Fig. 2G) with almost
respectively. The MTX-MagTSLs was schematically illustrated in no change after a week (Fig. 2H). Additionally, the encapsulation and
Fig. 2A. Hydrophobic MNPs/Cy5.5 was encapsulated in lipid bilayer loading efficiency of Dox in MTX-MagTSLs decreased lowly within a

Fig. 2. The characterization of MNPs and MTX-MagTSLs. (A) Schematic illustration of MTX-MagTSLs. (B) TEM image of MNPs. (C) TEM image of MTX-MagTSLs. (D) UV–Vis absorption
spectra of free MTX, Mag-TSLs, and MTX-MagTSLs in methanol. (E) Absorbance at 298 nm as a function of MTX concentration. (F) Particle size distribution of MTX-MagTSLs. (G) Zeta
potential distribution of MTX-MagTSLs. (H) The changes of size and zeta potential of MTX-MagTSLs within a week at 4 °C. (I) The changes of encapsulation efficiency and loading
efficiency of Dox in MTX-MagTSLs within a week at 4 °C.

150
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Fig. 3. Magnetic properties of MNPs and MTX-MagTSLs.


(A) XRD patterns of MNPs. (B) Magnetization loops of
MNPs and MTX-MagTSLs. (C) T2-weighted MR images of
MTX-MagTSLs at different iron concentrations. (D) T2 re-
laxation rates of MTX-MagTSLs at different iron con-
centrations.

week, the overall amplitude of which were < 5% and 0.3% respectively
(Fig. 2I). All results illustrated the good stability of MTX-MagTSLs.
As shown in the XRD spectrum of MNPs (Fig. 3A), the presence of
diffraction peaks at 30.2, 35.5, 43.2, 53.4, 57.0, and 62.7 coincided
well with the 〈220〉, 〈311〉, 〈400〉, 〈422〉, 〈511〉, and
〈440〉 facets of γ-Fe2O3 respectively [44], which indicated that the
formed γ-Fe2O3 nanocrystals loaded in MTX-TSLs are typically cubic
spinel structure. The MNPs and MTX-MagTSLs displayed good magnetic
properties. The magnetization hysteresis loop indicated the super-
paramagnetic nature of oleic acid-coated iron oxide NPs and MTX-
MagTSLs (Fig. 3B), in which the superparamagnetism of oleic acid-
coated iron oxide NPs was much higher than that of MTX-MagTSLs. The
reason was that oleic acid-coated iron oxide NPs embedded in the lipid
membrane would reduce the magnetic properties to a certain extent.
The MTX-MagTSLs could act as a T2 contrast agent for MRI, which
revealed the concentration dependent darkening effect (Fig. 3C). It
showed smaller concentration of MTX-MagTSLs which gave brighter
T2-weighted images. The transverse relaxivity (R2) of the MTX-
MagTSLs was measured to be 60.06 mM− 1 s− 1 (Fig. 3D).
The DSC thermogram (Fig. 4) of MTX-MagTSLs with a series of Fig. 4. Differential scanning calorimetric thermogram of liposomes with a series of
phospholipid components showed different main phase transition peaks phospholipid components.
and peak width respectively. It was observed that the liposomes com-
posed of DPPC showed a sharp phase transition at around 41 °C which
was correlated to its NIR photothermal effect. The decrease of gradual
was in agreement with previously published results [8,32], addition of
absorption was mainly due to the charge transfer and ligand transitions
cholesterol parts broadened the phase transition peak between 37 and
of iron [34]. The absorbance of MTX-MagTSLs at 808 nm increased
47 °C. Further addition of DSPE-MPEG2000 and DSPE-MPEG2000-MTX
linearly with the iron concentration (ranging from 2.5 to 15 mM)
caused sharper phase transition peak and shifted peak to higher tem-
(Fig. 5B). The heating temperature of PBS (Fig. 5C) and TSLs (Fig. 5D)
perature. The main phase transition peaks of MTX-MagTSLs shifted to
induced by different conditions have a minor increase in 5 min, in-
lower temperature when composed of more proportion of DPPC. To
dicated that there was almost no influence on the heating ability of
distinguish between Tm and normal body temperature, the proportion
MTX-MagTSLs under either AMF or laser. Fig. 5E and Fig. 5F showed
of phospholipid components was determined finally as DPPC:Chol:-
the time course of the temperature measured in MNPs and MTX-
SA:DSPE-MPEG:DSPE-PEG2000-MTX = 67:17:13:1:2 in mol ratio, with
MagTSLs suspension with the same iron concentration (13.93 mM)
Tm at around 45 °C, indicated that the prepared MTX-MagTSLs could
under different conditions respectively, from which the SAR values
improve drug release until 40 °C.
were calculated. Intriguingly, it could be calculated that MTX-MagTSLs
had an obviously higher SAR value under DUAL-mode (i.e. 621.5 W/g)
3.3. Measurement of heating ability of MTX-MagTSLs than that under both AMF (i.e. 171.6 W/g) and laser (i.e. 160.4 W/g)
condition, while MNPs had a relatively higher SAR value (i.e. 414.5 W/
Absorption spectra of MTX-MagTSLs were shown in Fig. 5A, which g) than MTX-MagTSLs under the laser, but lower value than MTX-

151
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Fig. 5. Heating capacity of PBS, TSLs, MNPs, and MTX-MagTSLs under different conditions. (A) Absorption spectra of MTX-MagTSLs at different iron concentrations [Fe] = 2.5–15 mM.
(B) Absorbance at 808 nm as a function of iron concentration. (C) Temperature increase of PBS in Laser (0.8 W/cm2), AMF (500 kHz, 20 kA·m− 1), and DUAL-mode (Laser 0.8 W/cm2,
AMF 500 kHz, 20 kA·m− 1) for 5 min respectively. (D) Temperature increase of TSLs in Laser (0.8 W/cm2), AMF (500 kHz, 20 kA·m− 1), and DUAL-mode (Laser 0.8 W/cm2, AMF 500 kHz,
20 kA·m− 1) for 5 min respectively. (E) Temperature increase of MNPs (CFe = 13.93 mM) under Laser (0.8 W/cm2), AMF (500 kHz, 20 kA·m− 1), and DUAL-mode (Laser 0.8 W/cm2, AMF
500 kHz, 20 kA·m− 1) for 5 min respectively. (F) Temperature increase of MTX-MagTSLs (CFe = 13.93 mM) under Laser (0.8 W/cm2), AMF (500 kHz, 20 kA·m− 1), and DUAL-mode (Laser
0.8 W/cm2, AMF 500 kHz, 20 kA·m− 1) for 5 min respectively. (G) Temperature increase of MTX-MagTSLs at different iron concentrations (2.5–15 mM) under DUAL-mode (Laser 0.8 W/
cm2, AMF 500 kHz, 20 kA·m− 1) for 5 min. (H) Temperature increase of MTX-MagTSLs with different NIR-laser powers under DUAL-mode (Laser 0.3–1.2 W/cm2, AMF 500 kHz,
20 kA·m− 1) for 5 min.

3.4. Doxorubicin release study in vitro

In in vitro release study, the comparative free Dox


(CDox = 0.182 mg·mL− 1) and Dox released from MTX-MagTSLs
(CDox = 0.182 mg·mL− 1, CFe = 13.93 mM) profiles following 24 h in-
cubation in PBS at 37 and 45 °C respectively could be seen in Fig. 6. Dox
released from MTX-MagTSLs at 37 °C in PBS was about 44% but around
83% at 45 °C in 24 h. In contrast, free Dox gave a burst release within
an hour, and was about 97% in PBS in 24 h. The above results showed
that the Dox released from MTX-MagTSLs could be controlled by ad-
justing the temperature, which indicated that a significant enhanced
Dox release could be realized with simultaneous application of AMF
and laser according to SAR results.

Fig. 6. Dox released profiles (CDox = 0.182 mg·mL− 1, CFe = 13.93 mM) in PBS (pH 7.4) 3.5. In vivo pharmacokinetics
at 37 °C and 45 °C (mean ± SD, n = 3).
No notable hemolytic analysis effects of Dox-free MagTSLs were
MagTSLs under DUAL-mode (i.e. 567.0 W/g). It is speculated that observed at phospholipid concentrations between 25 and 400 μg·mL− 1
photothermal effect of MNPs would reduce when encapsulated in li- (< 2% hemolysis) (Fig. S3), which confirmed good blood compat-
posomes. There had never been a coverage about an appreciable en- ibility. In the pharmacokinetics investigation of free Dox and MTX-
hancement of drug release under DUAL-mode stimulation for those MagTSLs, the results showed that Dox plasma concentration in both the
have either poor magnetocaloric effect or photothermal effect based free Dox and the MTX-MagTSLs group was highest at the completion of
multi-functional drug delivery so far, so the speculation is that general injection and then decreased, while the decreasing trend of the free Dox
magnetocaloric effect (due to small size of embedded MNPs) [45], in- group was faster noticeably than the MTX-MagTSLs group after in-
crease outstandingly under laser excitation for the reason that the travenous administration (Fig. S4). Compared to free Dox, the area
transition radiation of electron energy levels of MNPs and the change of under the curve (AUC) of MTX-MagTSLs (3.95 ± 0.46 h μg·mL− 1) was
spin states lead to the enhancement of Brownian and Néel relaxation, about four times greater than that of free Dox
which need further investigation on principle. SAR values were de- (1.01 ± 0.13 h μg·mL− 1). The elimination rate of MTX-MagTSLs and
termined as a function of laser power (ranging from 0.3 to 1.2 W/m2) free Dox were 60.49 ± 5.67 L h− 1 kg− 1 and 244.85 ±
−1 −1
and iron concentrations (ranging from 2.5 to 15 mM). The SAR values 19.18 L h kg , respectively. The elimination half-life (t1/2) of
increased with increasing laser power (Fig. 5G) and iron concentrations MTX-MagTSLs (32.36 ± 2.33 h) was approximately four times longer
(Fig. 5H). Tumor cells were generally found to be more heat-sensitive than that of free Dox (8.17 ± 1.03 h), indicating that MTX-MagTSLs
and vulnerable compared to normal cells between 42 and 47 °C [46,47]. increased the blood circulation time of Dox in vivo markedly. These
Therefore, 0.8 W/m2 was selected for the subsequent experiments. results could be explained by the great stability of the prepared long
circulating TSLs at normal body temperature, and regarded as a re-
ference in in vivo animals treatment cycle (2 days) later as well.

152
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

3.6. In vitro cellular uptake were used to assess the targeting efficacy of MTX and verify the toxicity
of MNPs. On the one hand, there was no significant difference of two
The folate receptor-mediated targeting ability for MTX-MagTSLs cell lines viability treated with TSLs and MagTSLs respectively, which
was studied against HeLa and A549 cells. Both Dox and Cy5.5 (as was in consistent with good safety features of the MNPs in Fig. 8A. On
markers of MTX-MagTSLs) were used as fluorescent dyes to investigate the other hand, the cytotoxicity of MTX-MagTSLs against HeLa cells
the uptake of MagTSLs and MTX-MagTSLs in cells by LCSM. Compared was higher than treated with free Dox, and MTX-MagTSLs induced
with free Dox and MagTSLs, the enhanced intracellular internalization more potent cytotoxicity than MagTSLs against HeLa cells, but smaller
of MTX-MagTSLs at Dox-eq. and Cy5.5-eq. dose in HeLa cells in 1 and trend against A549 cells. Such a cytotoxicity difference revealed that
4 h (Fig. 7A and B) indicated high selectivity of MTX-MagTSLs. In ad- MTX could be used to enhance cytotoxic effect selectively ascribed to
dition, the cellular uptake of MTX-MagTSLs by HeLa cells (folate re- folate receptor targeting ability and intrinsic toxicity, which was in
ceptor-positive) increased more than that of MTX-MagTSLs by A549 agreement with the previous reports [21]. The enhanced cytotoxicity of
cells (folate receptor-negative), and there was significant difference in free MTX and Dox-free MTX-MagTSLs compared with Dox-free
cellular uptake of MTX-MagTSLs towards these two cell models MagTSLs demonstrated certain toxicity of MTX at 1.2 and 2.4 μg·mL− 1
(Fig. 7B). All these results provided strong evidence that MTX could use of MTX concentration (Fig. 8C). In addition, the Dox-free MTX-MagTSLs
to be an effective selective targeted molecule, which would improve showed a little more cytotoxicity than free MTX due to the better
targeting efficiency and reduce side effects. Furthermore, the cellular membrane permeability [49,50]. Furthermore, compared with either
uptake of the MTX-MagTSLs treated by DUAL-mode increased signally single AMF or laser treating, the enhanced cytotoxicity of MTX-
more than being influenced by either no additional condition or single MagTSLs under DUAL-mode demonstrated that the simultaneous ap-
AMF treating condition (Fig. 7C), which demonstrated that the si- plication of AMF and laser could lead to enhance cytotoxicity of Dox
multaneous application of AMF and laser could enhance uptake of due to the acceleration of Dox release and uptake markedly (Fig. 8D).
MTX-MagTSLs in cancer cells and accelerate intracellular Dox release All these cell viability results were consistent with the results of early
markedly. The enhanced uptake and drug release using only AMF was
caused by two reasons. Firstly, the broad phase transition peak of MTX-
MagTSLs (Fig. 4) leaded to the phospholipid bilayer of MTX-MagTSLs
with increased permeability gradually before achieving the tempera-
ture of the phase transition peak value under AMF. Secondly, the
measured temperature in the experiment represented the macroscopical
temperature of the whole NPs suspension. MNPs embedded in MTX-
MagTSLs generated heat under AMF, followed by passing heat to sur-
roundings to cause solvent warming up, which means the microcosmic
local temperature of MTX-MagTSLs was higher than the whole NPs
suspension [48]. Therefore, although the temperature ascensional
range of MTX-MagTSLs under AMF was approximately 5.2 °C in 5 min
(Fig. 5F), it could still partly promote the release of Dox, and achieve
enhanced uptake and drug release in HeLa cells to a certain extent.

3.7. Flow cytometry

Flow cytometry was performed to confirm the targeting efficiency of


MTX-MagTSLs as a quantitative analysis of the cellular uptake. The
intracellular fluorescence intensity of Dox was enhanced as the in-
cubation time extended from 1 h (Fig. S5A) to 4 h (Fig. S5B) in HeLa
cells. The fluorescence intensity treated with free Dox was higher than
the MagTSLs group but lower than the MTX-MagTSLs group, indicated
that MTX had improved targeting efficacy. The intracellular fluores-
cence intensity of Dox was enhanced as the incubation time extended
from 1 h (Fig. S5C) to 4 h (Fig. S5D) in A549 cells, and there was no
obvious difference of fluorescence intensity between being treated with
MagTSLs and MTX-MagTSLs in A549 cells compared with in HeLa cells,
indicating the good folate receptor-mediated targeting efficacy of MTX-
MagTSLs. Furthermore, the MTX-MagTSLs under DUAL-mode exhibited
higher cellular uptake than either no additional condition or single
AMF treating for processing (Fig. S5E). The quantitative results were in
good agreement with the qualitative results (see Fig. 7), giving further
confirmation of improved cellular uptake efficiency of MTX-MagTSLs
and enhanced Dox release treated with AMF and laser simultaneously.

3.8. In vitro cell viability

HeLa and A549 cells were chosen to evaluate the cytotoxicity of Fig. 7. Qualitative cellular Dox uptake of the MTX-MagTSLs. (A) HeLa cells were treated
MTX-MagTSLs. No significant inhibiting effect against HeLa cells was with free Dox and MTX-MagTSLs respectively under 1 and 4 h. (B) HeLa and A549 cells
demonstrated by Dox-free MagTSLs at phospholipid concentrations were treated with MTX-MagTSLs respectively, and HeLa cells were treated with MagTSLs
between 25 and 400 μg·mL− 1 (Fig. 8A), which indicated Dox-free under 4 h. (C) HeLa cells were treated with MTX-MagTSLs under no stimulation, AMF
(500 kHz, 20 kA·m− 1, 5 min), and DUAL-mode (0.8 W/cm2, 500 kHz, 20 kA·m− 1, 5 min)
MagTSLs had almost no toxicity to HeLa cells within this concentration
respectively.
range. All the Dox loaded groups presented dose-dependent cytotoxicity
against both HeLa and A549 cells (Fig. 8B). Then these two cell lines

153
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

3.9. In vivo fluorescence and MR imaging

To evaluate the targeting capability of TSLs, MagTSLs, MTX-TSLs,


MTX-MagTSLs, and MTX-MagTSLs/CMF, the above materials were in-
jected intravenously into the HeLa tumor-bearing nude mice. In vivo
biodistribution was investigated by a non-invasive near-infrared (NIR)
optical imaging technique. The TSLs and MagTSLs group presented
weak fluorescence signal in tumor region compared with the MTX-TSLs
and MTX-MagTSLs group due to the lacking of folate receptor-mediated
targeting (Fig. 9A). The MTX-MagTSLs/CMF group presented a stronger
fluorescence signal in tumor region over 8 h compared with other
groups, which revealed that the amount of MTX-MagTSLs at tumor site
in the MTX-MagTSLs/CMF group was higher than groups with single
targeting (MTX-MagTSLs and MTX-TSLs) or without targeting
(MagTSLs and TSLs). Furthermore, the fluorescence signal of tumor
harvested from mice of all groups at 12 h post-injection from strong to
Fig. 7. (continued) weak was as follows, MTX-MagTSLs/CMF > MTX-MagTSLs ≈ MTX-
TSLs > MagTSLs ≈ TSLs (Fig. 9B), which verified the dual role of
magnetic and biological targeting ability of MTX-MagTSLs under CMF.
uptake and flow cytometry experiments, verified that MTX-MagTSLs The MTX-MagTSLs/CMF group demonstrated a much higher accumu-
could be used as effective targeted anti-tumor agents at the cellular lation at the tumor site and lower accumulation in other major organs
level, and should be further investigated in vivo. (Table 1), which clearly indicated the in vivo targeting ability of MTX-
MagTSLs/CMF and verified the dual role of magnetic and biological

Fig. 8. Relative cell viability of free Dox and different formulations at a series of either phospholipid or Dox concentrations. (A) HeLa cells were incubated with Dox-free MagTSLs at
different phospholipid concentrations (25, 50, 100, 200, and 400 μg/mL) for 24 h. (B) A549 cells (folate receptor-negative) and HeLa cells (folate receptor-positive) were incubated with
free Dox/TSLs/MagTSLs/MTX-TSLs/MTX-MagTSLs at different Dox concentrations (2.5, 5, and 10 μg/mL) for 24 h respectively. (C) HeLa cells were incubated with Dox-free MagTSLs,
free MTX, and Dox-free MTX-MagTSLs at different MTX concentrations (0.6, 1.2, and 2.4 μg/mL) for 24 h respectively. (D) HeLa cells were incubated with MTX-MagTSLs under no
stimulation, Laser (0.8 W/cm2, 5 min), AMF (500 kHz, 20 kA·m− 1, 5 min), and DUAL-mode (0.8 W/cm2, 500 kHz, 20 kA·m− 1, 5 min) (mean ± SD, n = 6). *p < 0.05, **p < 0.01,
***p < 0.001.

154
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Fig. 9. In vivo imaging of MTX-MagTSLs. (A) In vivo fluorescence images of the HeLa tumor-bearing nude mice at 4, 8, and 12 h after intravenous injection of PBS, MTX-MagTSLs, MTX-
MagTSLs/CMF, MagTSLs, TSLs, and MTX-TSLs respectively. (B) Ex vivo fluorescence images of the tumor excised from HeLa tumor-bearing nude mice treated with PBS, MTX-MagTSLs/
CMF, MagTSLs, MTX-MagTSL, TSLs, and MTX-TSLs at 12 h post-injection respectively. (C) In vivo pseudo-color processing T2-weighted MR images of MTX-MagTSLs with a magnet at 0, 4,
8, and 12 h post-injection. (D) In vivo pseudo-color processing T2-weighted MR images of MTX-MagTSLs without a magnet at 0, 4, 8, and 12 h post-injection. The arrow indicated the
tumor sites.

targeting of MTX. and 12 h after the intravenous injection of MTX-MagTSLs with a


To examine the suitability of MTX-MagTSLs for in vivo MRI, HeLa magnet at the tumor position (Fig. 9C). A sustained attenuation of the
tumor-bearing nude mice were further established for in vivo MRI T2 contrast signal at the tumor position was detected from 0 to 8 h after
evaluation using a 7-T MRI scanner. MR images were acquired for 4, 8, MTX-MagTSLs injection, indicating the effective accumulation of MTX-

155
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Table 1 was in consistent with in vitro anti-cancer effect of those. The result
In vivo biodistribution. demonstrated remarkable magnetic/MTX targeting function and in-
The fluorescence intensities of heart, liver, spleen, lung, kidney, and tumor excised
crease of Dox release under DUAL-mode. In addition, except for the free
from HeLa tumor-bearing nude mice treated with TSLs, MagTSLs, MTX-TSLs, MTX-
MagTSL, and MTX-MagTSLs/CMF at 12 h post-injection respectively. Dox group with relatively high toxicity, the body weights in other
groups had no significant changes in two weeks (Fig. 10B). The survival
Heart Liver Spleen Lung Kidney Tumor results of mice correlated to the therapy response were observed in
Fig. 10C. Compared to the free Dox and the MagTSLs group, the MTX-
TSLs 1.57E 6.57E 1.49E 1.51E 8.28E + 07 3.57E
+ 07 + 08 + 08 + 08 + 07 MagTSLs/CMF/AMF/Laser group showed improved survival sig-
MagTSLs 1.55E 6.66E 1.46E 1.49E 8.27E + 07 3.59E nificantly. Furthermore, the MTX-MagTSLs/DUAL group was added to
+ 07 + 08 + 08 + 08 + 07 verify dual-targeting ability compared with the MTX-MagTSLs/CMF/
MTX-TSLs 1.09E 5.35E 1.09E 9.01E 7.19E + 07 7.63E DUAL group. The tumor inhibition efficiency of MTX-MagTSLs/CMF/
+ 07 + 08 + 08 + 07 + 07
DUAL was obviously higher than MTX-MagTSLs/DUAL (Fig. S6A), the
MTX-MagTSLs 1.10E 5.28E 1.16E 8.97E 7.16E + 07 7.67E
+ 07 + 08 + 08 + 07 + 07 mice in both of two groups showed no significant body weight changes
MTX-MagTSLs/CMF 1.07E 4.62E 9.36E 4.47E 6.83E + 07 1.29E (Fig. S6B). The survival results of mice in MTX-MagTSLs/DUAL group
+ 07 + 08 + 07 + 07 + 08 was worse than the mice in MTX-MagTSLs/CMF/DUAL group due to
the insufficient targeting at tumor site and larger toxicity to other or-
gans under AMF/Laser triggering drug release without magnetic tar-
geting effect (Fig. S6C). All results suggested that MTX-MagTSLs/CMF/
MagTSLs at the tumor site via both magnetic targeting and folate re- AMF/Laser were highly effective for improving the therapeutic effects
ceptor-mediated targeting effect. The maximum decreased MRI signal of Dox while decreasing its associated toxicity.
intensities at tumor site was achieved at 8 h post-injection, varying Histological analysis by H&E staining of heart, liver, spleen, lung,
10.72% compared with the initiative value, followed by a gradual re- kidney, and tumor with different treatments at day 14 post-treatment
covery of MRI signal after 8 h post-injection, hinting the MTX-MagTSLs (Fig. 11) expressed that cell necrosis, lysis, and fragmentation occurred
could act as suitable negative (T2) contrast agents in MRI applications. in tumor tissue, while tight arrangement and intact shape in other tis-
As control, MR images of MTX-MagTSLs without a magnet at 0, 4, 8, sues when treated with MTX-MagTSLs under DUAL-mode. There was
and 12 h post-injection revealed a sustained attenuation of the T2 appearing an opposite result in the free Dox group and no obvious
contrast signal at the tumor position (Fig. 9D), and there was a rela- difference between other groups, which confirmed that combining AMF
tively lower MRI signal intensities attenuation (5.69%) compared with and laser could efficiently accelerate Dox release at tumor site, thereby
the MTX-MagTSLs/CMF group at tumor site, suggesting the less effec- achieved an optimal anti-tumor efficacy in vivo.
tive accumulation of MTX-MagTSLs in the tumor region without the
magnetic targeting effect. The metabolites of tumor including a large
amount of lactic acid from mainly anaerobic oxidation were different 4. Conclusions
from normal tissues (Warburg effect), which could cause the oppres-
sion, inflammation, the change of permeability, and the surrounding MTX modified thermo-sensitive magnetoliposomes, which consist of
edema under the development stage of tumor [51–53]. As a result of the DPPC:Chol:SA:DSPE-PEG2000:DSPE-PEG2000-MTX, has been developed
different composition and volume of distributive tissue fluids in the for precise cervical cancer therapy. The MTX-MagTSLs showed ex-
inflammatory mature tumor compared with other tissues, the mice cellent targeting ability, temperature sensitivity, and strong respon-
appeared a little strong signals in tumor before the administration due siveness to AMF and laser processing simultaneously, which improved
to more inherent hydrogen protons in the tumor region. Dox uptake and release into HeLa cells significantly, and also improved
tumor cell killing effects. Hence, both light/magnetic hyperthermia
3.10. In vivo anti-tumor efficacy triggered drug release and magnetic/MTX active targeting synergisti-
cally increased cytotoxicity to tumor cells and tissues while leading to a
The anti-tumor effects of MTX-MagTSLs in different groups were reduction of side effects compared with free Dox. Furthermore, it could
evaluated by measuring the tumor volume, weight, and survival si- be monitored in real time by fluorescence and MR imaging throughout
tuation of HeLa tumor-bearing nude mice. As shown in Fig. 10A, the the drug treatment. Therefore, this multi-functional liposomes can be
inhibition efficiency of different groups from high to low was shown as used as the potential carriers for precise cancer related diagnoses and
follows, MTX-MagTSLs/CMF/AMF/Laser > MTX-MagTSLs/CMF/ treatments.
AMF > MTX-MagTSLs/CMF > MTX-MagTSLs > MagTSLs, which

Fig. 10. In vivo anti-tumor activity. (A) Tumor growth curves, (B) changes of body weight, and (C) Kaplan-Meier survival curves of HeLa tumor-bearing nude mice after intravenous
injection of PBS, free Dox, MagTSLs, MTX-MagTSLs, MTX-MagTSLs/CMF, MTX-MagTSLs/CMF/AMF, and MTX-MagTSLs/CMF/DUAL at a Dox dose of 2 mg/kg every treatment (2 days)
within 14 days (mean ± SD, n = 6). *p < 0.05, ***p < 0.001.

156
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

Fig. 11. H&E stained tissues harvested from the mice with different treatments. (A) Control, (B) free Dox, (C) MagTSLs, (D) MTX-MagTSLs, (E) MTX-MagTSLs/CMF, (F) MTX-MagTSLs/
CMF/AMF, and (G) MTX-MagTSLs/CMF/DUAL.

Acknowledgements quantum dot system, J. Colloid Interface Sci. 480 (2016) 146–158.
[13] A. Sarkar, S. Ghosh, S. Chowdhury, B. Pandey, P.C. Sil, Targeted delivery of
quercetin loaded mesoporous silica nanoparticles to the breast cancer cells,
The research was financially supported by Social Development Biochim. Biophys. Acta 1860 (2016) 2065–2075.
Guidance Project of Fujian Province (No. 2014Y0074), Project of [14] P.S. Low, W.A. Henne, D.D. Doorneweerd, Discovery and development of folic-acid-
Xiamen Science and Technology Bureau (3502Z20163004), and based receptor targeting for imaging and therapy of cancer and inflammatory dis-
eases, Acc. Chem. Res. 41 (2008) 120–129.
Natural Science Foundation of Fujian Province (No. 2015J01348). [15] Z.Ü. Akal, L. Alpsoy, A. Baykal, Biomedical applications of SPION@APTES@PEG-
folic acid@carboxylated quercetin nanodrug on various cancer cells, Appl. Surf. Sci.
Appendix A. Supplementary data 378 (2016) 572–581.
[16] C.T. Kuo, W.S. Lee, Progesterone receptor activation is required for folic acid-in-
duced anti-proliferation in colorectal cancer cell lines, Cancer Lett. 378 (2016)
Supplementary data to this article can be found online at http://dx. 104–110.
doi.org/10.1016/j.jconrel.2017.04.028. [17] S. Lu, K.G. Neoh, C. Huang, Z. Shi, E.T. Kang, Polyacrylamide hybrid nanogels for
targeted cancer chemotherapy via co-delivery of gold nanoparticles and MTX, J.
Colloid Interface Sci. 412 (2013) 46–55.
References [18] M. Li, K.G. Neoh, R. Wang, B.Y. Zong, J.Y. Tan, E.T. Kang, Methotrexate-conjugated
and hyperbranched polyglycerol-grafted Fe(3)O(4) magnetic nanoparticles for tar-
[1] K. Loomis, K. McNeeley, R.V. Bellamkonda, Nanoparticles with targeting, triggered geted anticancer effects, Eur. J. Pharm. Sci. 48 (2013) 111–120.
release, and imaging functionality for cancer applications, Soft Matter 7 (2011) [19] P.T. Wong, S.K. Choi, Mechanisms and implications of dual-acting methotrexate in
839–856. folate-targeted nanotherapeutic delivery, Int. J. Mol. Sci. 16 (2015) 1772–1790.
[2] S. Mallick, J.S. Choi, Liposomes: versatile and biocompatible nanovesicles for effi- [20] C.F. Wang, E.M. Makila, M.H. Kaasalainen, M.V. Hagstrom, J.J. Salonen,
cient biomolecules delivery, J. Nanosci. Nanotechnol. 14 (2014) 755–765. J.T. Hirvonen, H.A. Santos, Dual-drug delivery by porous silicon nanoparticles for
[3] T.M. Allen, P.R. Cullis, Liposomal drug delivery systems: from concept to clinical improved cellular uptake, sustained release, and combination therapy, Acta
applications, Adv. Drug Deliv. Rev. 65 (2013) 36–48. Biomater. 16 (2015) 206–214.
[4] E. Tahover, Y.P. Patil, A.A. Gabizon, Emerging delivery systems to reduce doxor- [21] Y. Li, J. Lin, H. Wu, M. Jia, C. Yuan, Y. Chang, Z. Hou, L. Dai, Novel methotrexate
ubicin cardiotoxicity and improve therapeutic index: focus on liposomes, Anti- prodrug-targeted drug delivery system based on PEG–lipid–PLA hybrid nano-
Cancer Drugs 26 (2015) 241–258. particles for enhanced anticancer efficacy and reduced toxicity of mitomycin C, J.
[5] D.S. T., K.G. M., Pegylated liposomal doxorubicin, Drugs 71 (2011) 2531–2558. Mater. Chem. B 2 (2014) 6534–6548.
[6] B. Du, S. Han, H. Li, F. Zhao, X. Su, X. Cao, Z. Zhang, Multi-functional liposomes [22] L. Li, T.L. ten Hagen, M. Hossann, R. Suss, G.C. van Rhoon, A.M. Eggermont,
showing radiofrequency-triggered release and magnetic resonance imaging for D. Haemmerich, G.A. Koning, Mild hyperthermia triggered doxorubicin release
tumor multi-mechanism therapy, Nano 7 (2015) 5411–5426. from optimized stealth thermosensitive liposomes improves intratumoral drug de-
[7] J.P. May, S.D. Li, Hyperthermia-induced drug targeting, Expert Opin. Drug Deliv. livery and efficacy, J. Control. Release 168 (2013) 142–150.
10 (2013) 511–527. [23] B.E. Oude, E. Mastrobattista, R.M. Schiffelers, Strategies for triggered drug release
[8] A. Gharib, Z. Faezizadeh, S.A. Mesbah-Namin, R. Saravani, Preparation, char- from tumor targeted liposomes, Expert Opin. Drug Deliv. 10 (2013) 1399–1410.
acterization and in vitro efficacy of magnetic nanoliposomes containing the arte- [24] Z.S. Al-Ahmady, O. Chaloin, K. Kostarelos, Monoclonal antibody-targeted, tem-
misinin and transferrin, Daru 22 (2014) 44. perature-sensitive liposomes: in vivo tumor chemotherapeutics in combination with
[9] X. Jia, Y. Han, M. Pei, X. Zhao, K. Tian, T. Zhou, P. Liu, Multi-functionalized hya- mild hyperthermia, J. Control. Release 196 (2014) 332–343.
luronic acid nanogels crosslinked with carbon dots as dual receptor-mediated tar- [25] R. Das, J. Alonso, Z.N. Porshokouh, V. Kalappattil, D. Torres, M.H. Phan, E. Garaio,
geting tumor theranostics, Carbohydr. Polym. 152 (2016) 391–397. J.Á. García, J.L.S. Llamazares, H. Srikanth, Tunable high aspect ratio iron oxide
[10] L. Wang, K.G. Neoh, E.T. Kang, B. Shuter, Multifunctional polyglycerol-grafted Fe nanorods for enhanced hyperthermia, J. Phys. Chem. C 120 (2016) 10086–10093.
(3)O(4)@SiO(2) nanoparticles for targeting ovarian cancer cells, Biomaterials 32 [26] S.P. Mornet, S.B. Vasseur, F. Grasset, E. Duguet, Magnetic nanoparticle design for
(2011) 2166–2173. medical diagnosis and therapy, J. Mater. Chem. 14 (2004) 2161.
[11] J.Y. Lee, U. Termsarasab, J.H. Park, S.Y. Lee, S.H. Ko, J.S. Shim, S.J. Chung, [27] L. Liu, L. Tseng, Q. Ye, Y.L. Wu, D.J. Bain, C. Ho, A new method for preparing
H.J. Cho, D.D. Kim, Dual CD44 and folate receptor-targeted nanoparticles for mesenchymal stem cells and labeling with Ferumoxytol for cell tracking by MRI,
cancer diagnosis and anticancer drug delivery, J. Control. Release 236 (2016) Sci. Rep. 6 (2016) 26271.
38–46. [28] C. Ansari, G.A. Tikhomirov, S.H. Hong, R.A. Falconer, P.M. Loadman, J.H. Gill,
[12] I.B. Bwatanglang, F. Mohammad, N.A. Yusof, J. Abdullah, N.B. Alitheen, R. Castaneda, F.K. Hazard, L. Tong, O.D. Lenkov, D.W. Felsher, J. Rao,
M.Z. Hussein, N. Abu, N.E. Mohammed, N. Nordin, N.R. Zamberi, S.K. Yeap, In vivo H.E. Daldrup-Link, Development of novel tumor-targeted theranostic nanoparticles
tumor targeting and anti-tumor effects of 5-fluororacil loaded, folic acid targeted activated by membrane-type matrix metalloproteinases for combined cancer

157
Y. Guo et al. Journal of Controlled Release 272 (2018) 145–158

magnetic resonance imaging and therapy, Small 10 (2014) 566–575 (417). [41] N. Monteiro, M. Martins, A. Martins, N.A. Fonseca, J.N. Moreira, R.L. Reis,
[29] M. de Smet, S. Langereis, S. van den Bosch, H. Grull, Temperature-sensitive lipo- N.M. Neves, Antibacterial activity of chitosan nanofiber meshes with liposomes
somes for doxorubicin delivery under MRI guidance, J. Control. Release 143 (2010) immobilized releasing gentamicin, Acta Biomater. 18 (2015) 196–205.
120–127. [42] Y. Han, S.L. Lei, J.H. Lu, Y. He, Z.W. Chen, L. Ren, X. Zhou, Potential use of SERS-
[30] H. Grull, S. Langereis, Hyperthermia-triggered drug delivery from temperature- assisted theranostic strategy based on Fe3O4/Au cluster/shell nanocomposites for
sensitive liposomes using MRI-guided high intensity focused ultrasound, J. Control. bio-detection, MRI, and magnetic hyperthermia, Mater. Sci. Eng. C Mater. Biol.
Release 161 (2012) 317–327. Appl. 64 (2016) 199–207.
[31] E. Amstad, J. Kohlbrecher, E. Muller, T. Schweizer, M. Textor, E. Reimhult, [43] S. Lange, Conjugating Methotrexate to magnetite (Fe3O4) nanoparticles via tri-
Triggered release from liposomes through magnetic actuation of iron oxide nano- chloro-s-triazine, J. Mater. Chem. 2009 (2009) 6400–6406.
particle containing membranes, Nano Lett. 11 (2011) 1664–1670. [44] L.Y. Zhang, W. Zhang, Z. Zhou, C.M. Li, gamma-Fe2O3 nanocrystals-anchored
[32] P. Pradhan, J. Giri, F. Rieken, C. Koch, O. Mykhaylyk, M. Doblinger, R. Banerjee, macro/meso-porous graphene as a highly efficient adsorbent toward removal of
D. Bahadur, C. Plank, Targeted temperature sensitive magnetic liposomes for methylene blue, J. Colloid Interface Sci. 476 (2016) 200–205.
thermo-chemotherapy, J. Control. Release 142 (2010) 108–121. [45] M. Ma, Y. Wu, J. Zhou, Y. Sun, Y. Zhang, N. Gu, Size dependence of specific power
[33] Y. Chen, Y. Chen, D. Xiao, A. Bose, R. Deng, G.D. Bothun, Low-dose chemotherapy absorption of Fe3O4 particles in AC magnetic field, J. Magn. Magn. Mater. 268
of hepatocellular carcinoma through triggered-release from bilayer-decorated (2004) 33–39.
magnetoliposomes, Colloids Surf. B: Biointerfaces 116 (2014) 452–458. [46] A.B. Salunkhe, V.M. Khot, S.H. Pawar, Magnetic hyperthermia with magnetic na-
[34] A. Espinosa, R. Di Corato, J. Kolosnjaj-Tabi, P. Flaud, T. Pellegrino, C. Wilhelm, noparticles: a status review, Curr. Top. Med. Chem. 14 (2014) 572–594.
Duality of iron oxide nanoparticles in cancer therapy: amplification of heating ef- [47] B. Kneidl, M. Peller, G. Winter, L.H. Lindner, M. Hossann, Thermosensitive lipo-
ficiency by magnetic hyperthermia and photothermal bimodal treatment, ACS Nano somal drug delivery systems: state of the art review, Int. J. Nanomedicine 9 (2014)
10 (2016) 2436–2446. 4387–4398.
[35] T. Lammers, L.Y. Rizzo, G. Storm, F. Kiessling, Personalized nanomedicine, Clin. [48] D. Yoo, H. Jeong, S.H. Noh, J.H. Lee, J. Cheon, Magnetically triggered dual func-
Cancer Res. 18 (2012) 4889–4894. tional nanoparticles for resistance-free apoptotic hyperthermia, Angew. Chem. 52
[36] S. Kunjachan, J. Ehling, G. Storm, F. Kiessling, T. Lammers, Noninvasive imaging of (2013) 13047–13051.
nanomedicines and nanotheranostics: principles, progress, and prospects, Chem. [49] Y. Li, J. Lin, Y. Huang, Y. Li, X. Yang, H. Wu, S. Wu, L. Xie, L. Dai, Z. Hou, Self-
Rev. 115 (2015) 10907–10937. targeted, shape-assisted, and controlled-release self-delivery nanodrug for sy-
[37] C. von Roemeling, W. Jiang, C.K. Chan, I.L. Weissman, B.Y. Kim, Breaking down the nergistic targeting/anticancer effect of cytoplasm and nucleus of cancer cells, ACS
barriers to precision cancer nanomedicine, Trends Biotechnol. (2016). Appl. Mater. Interfaces 7 (2015) 25553–25559.
[38] W. Yuan, R. Kuai, Z. Dai, Y. Yuan, N. Zheng, W. Jiang, C. Noble, M. Hayes, [50] Y. Li, J. Lin, H. Wu, Y. Chang, C. Yuan, C. Liu, S. Wang, Z. Hou, L. Dai, Orthogonally
F.C. Szoka, A. Schwendeman, Development of a Flow-through USP-4 Apparatus functionalized nanoscale micelles for active targeted codelivery of methotrexate
Drug Release Assay to Evaluate Doxorubicin Liposomes, AAPS J. (2016). and mitomycin C with synergistic anticancer effect, Mol. Pharm. 12 (2015)
[39] Z. Liu, S. Tang, Z. Xu, Y. Wang, X. Zhu, L.-c. Li, W. Hong, X. Wang, Preparation and 769–782.
in vitro evaluation of a multifunctional iron silicate@liposome nanohybrid for pH- [51] W.B. Morrison, Inflammation and cancer: a comparative view, J. Vet. Intern. Med.
sensitive doxorubicin delivery and photoacoustic imaging, J. Nanomater. 2015 26 (2012) 18–31.
(2015) 1–13. [52] M.C. Moghadam, A. Deyranlou, A. Sharifi, H. Niazmand, Numerical simulation of
[40] Z. Peng, S. Li, X. Han, A.O. Al-Youbi, A.S. Bashammakh, M.S. El-Shahawi, the tumor interstitial fluid transport: consideration of drug delivery mechanism,
R.M. Leblanc, Determination of the composition, encapsulation efficiency and Microvasc. Res. 101 (2015) 62–71.
loading capacity in protein drug delivery systems using circular dichroism spec- [53] S.K. Lyons, Imaging mouse models of cancer, Cancer J. 21 (2015) 152–164.
troscopy, Anal. Chim. Acta 937 (2016) 113–118.

158

You might also like