Sunteți pe pagina 1din 14

ISSN (print): 1743 5889 | ISSN (online): 1748 6963

I.F.: 5.005 as per Thomson Reuters report July 2018 PreliminaryRCesearch Article
ommunication
For reprint orders, please contact: reprints@futuremedicine.com

Biphasic magnetic nanoparticles–nanovesicle


hybrids for chemotherapy and self-controlled
hyperthermia
Aim: The aim was to develop magnetic nanovesicles for chemotherapy and self-controlled hyperthermia
that prevent overheating of tissues. Materials & methods: Magnetic nanovesicles containing paclitaxel and
a dextran-coated biphasic suspension of La0.75Sr0.25MnO3 and Fe3O4 nanoparticles (magnetic nanoparticles)
were developed. Results: Encapsulation efficiencies of magnetic nanoparticles and paclitaxel were 67 ± 5
and 83 ± 3%, respectively. Sequential release performed at 37°C for 1 h followed by 44°C for another 1 h
(as expected for intratumoral injection), showed a cumulative release of 6.6% (109.6 µg), which was above
the IC50 of the drug. In an alternating current magnetic field, the temperature remained controlled at 44°C
and a synergistic cytotoxicity of paclitaxel and hyperthermia was observed in MCF-7 cells. Conclusion: Magnetic
nanovesicles containing biphasic suspensions La0.75Sr 0.25MnO3 and Fe3O 4 nanoparticles encapsulating
paclitaxel have potential for combined self-controlled hyperthermia and chemotherapy.
Original submitted 15 May 2012; Revised submitted 7 March 2013

KEYWORDS: biphasic suspension n cancer n chemotherapy n magnetic nanovesicle Manashjit Gogoi1,


n self-controlled hyperthermia Haladhar D Sarma2,
Dhirendra Bahadur3
Magnetic nanoparticle (MNP)-mediated hyper- for combined hyperthermia and chemotherapy
& Rinti Banerjee*1
1
Wadhwani Research Centre in
thermia is considered to be a potential candidate using doxorubicin [13]. However, therapeutic Biosciences & Bioengineering,
for treating cancer [1]. In complex tumor masses, efficacy was limited due to low encapsulation of Department of Biosciences &
oxygen-deprived hypoxic cells hardly respond to Fe3O4 nanoparticles (24 ± 6%) in the magnetic Bioengineering, Indian Institute of
Technology, Bombay,
radiation therapy [2–4], and chemotherapeutic vesicles. Drug-conjugated MNPs, magnetic Mumbai 400076, India
agents cannot reach the tumor sites in sufficient nano­vesicles and nano­hydrogels were reported 2
Radiation Biology & Health Sciences
Division, Bhabha Atomic Research
amounts due to a lack of proper blood circulation for combined hyperthermia and chemotherapy Centre, Mumbai 400085, India
and high intratumoral pressure [5,6]. Under these application [14–16]. 3
Department of Metallurgical
Engineering & Materials Science,
circumstances, hyperthermia can be effective in Homogenous intratumoral distribution of Indian Institute of Technology,
treating cancer because it can kill tumor cells magnetic nanomaterials is a challenging task and Bombay, Mumbai 400076, India
by damaging the cellular structures [7,8]. Hyper­ it determines the therapeutic efficacy of the hyper- *Author for correspondence:
Tel.: +91 222 576 7868
thermia not only makes the cancer cells vulner- thermia treatment [17,18]. Fe3O4 nano­particles Fax: +91 222 572 3480
able to radiation therapy [9], but also enhances are extensively used in magnetic nano­vesicles. rinti@iitb.ac.in
the bioavailability of drugs at tumor sites [10] by One disadvantage of these Fe3O4 nano­particles
increasing the blood supply to the tumor region. is their high Curie temperature (Tc; 580°C for
Magnetic nano­vesicle-based hyperthermia has Fe3O4 and 477°C for Fe2O3 nano­particles, respec-
been found to be effective in treating different tively) [18], which causes generation of hot spots
types of cancers in animal models [11,12]. Hyper- during hyperthermia that leads to overheating of
thermia in combination with chemotherapy or tissues adjacent to tumor cells [19,20]. Application
radiotherapy was reported to have improved the of MNPs with a Tc of approximately 44°C can
treatment efficacy without additional systemic be used to avoid overheating of tissues [20–23].
toxicity [13]. Drug-loaded magnetic nano­vesicles At the Tc, nanoparticles lose their magnetism
preferentially extravasate in tumor sites and and become paramagnetic and, hence, further
remain there via enhanced permeability and heating is not possible. This process is called
retention effect, which reduces their side effects on self-­controlled hyperthermia. Kuznetsov et al.
normal cells. Repeated doses of magnetic hyper- reported the potential of Cu–Ni alloy nano­
thermia with cationic magnetic nano­vesicles particles in hyperthermia application [20]. MNPs
were successfully demonstrated to be effective in from the perovskite family with a Tc in the range
treating tumor-bearing animals [11,12]. Pradhan of 42–46°C have been explored for self-controlled
et al. reported the application of folate and hyperthermia [21–23]. However, their studies were
magnetic-targeted thermosensitive nanovesicles limited to synthesis and a preliminary evaluation part of

doi:10.2217/NNM.13.90 © 2013 Future Medicine Ltd Nanomedicine (Epub ahead of print) ISSN 1743-5889
PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

of the heating ability of nanoparticles and did (FITC), a model fluorescent dye that enables the
not coencapsulate the MNPs with drugs within magnetic nanovesicles formulation for in vitro
nanovesicles. cellular imaging. Similarly, near infrared (NIR)
In this article, we report the use of pacli- dyes may be conjugated for in vivo imaging.
taxel-loaded thermosensitive magnetic nano­ The nanovesicle formulation was optimized for
vesicles containing a dextran-coated biphasic temperature sensitivity and their potential for
suspension of nanoparticles for combined chemotherapy and self-controlled hyperthermia
self-­
c ontrolled cancer hyperthermia and was evaluated.
chemotherapy. Paclitaxel is an effective anti­
neoplastic agent, which basically polymerizes Materials & methods
the cellular microtubules, and thereby inhibits „„ Materials
the normal tubule dynamics required for cell Analytical grade chemicals were used through-
division and replication [24]. The lipid bilayer of out the study without any further purification.
these nanovesicles consists of 1,2-distearol-sn-­ FeCl 2 .4H 2O, FeCl 3.6H 2O and sulforhoda-
phosphatidylcholine (DSPC) and cholesterol. mine B were procured from Sigma-Aldrich (MO,
DSPC is a thermosensitive lipid with a phase USA) and La 2O3 was procured from Indian
transition temperature of 58°C, and cholesterol Rare Earths Ltd (Cochin, India). DSPC was
is an important part of plasma membranes of purchased from Lipoid GmbH (Ludwigshafen,
almost all eukaryotic cells. The addition of Germany). Cholesterol was purchased from Loba
cholesterol reduces the transition temperature Chemicals (Mumbai, India). Anticancer drug
of saturated phospholipids such as DSPC and paclitaxel (purity: >99%) was supplied by Dabur
broadens transition peak. At a high concentra- India Ltd (Ghaziabad, India). For cell culture,
tion of cholesterol (50 mol%), the gel-to-liquid human breast cancer cell line (MCF-7) and
transition peak completely disappears [9,25]. In mouse fibroblast cell line (L929) were purchased
our formulation, we used 34 mol% of chol­ from the National Centre for Cell Science (Pune,
esterol. The dextran-coated biphasic nano­ India). Cells were maintained as a monolayer
particle suspension contained La0.75Sr 0.25MnO3 culture in modified Eagle medium and DMEM
(LSMO) and Fe3O4 nanoparticles in a ratio of supple­mented with 10% fetal bovine serum and
10:1. LSMO is a rare earth manganate, and 1% antibiotic antimycotic solution, respectively,
its Tc can be tuned to approximately 44°C by at 37°C in a humidified incubator containing
controlling the amount of divalent Sr2+ ion 5% CO2. Experiments were performed when
(x) (≤0.2  x  ≤0.4). It is expected that LSMO cells became 70–80% confluent.
nano­p articles will control the temper­ature
while Fe3O4 nanoparticles (possessing higher Synthesis of Fe3O4 & LSMO nanoparticles
heating ability) will provide sufficient heating Fe3O4 nanoparticles were synthesized by the
as well as make the biphasic suspension more coprecipitation method [15]. The black-colored
biocompatible. LSMO nanoparticles have a final solution was cooled down to room tem-
low specific absorption rate or heating abil- perature and particles were allowed to settle
ity, whereas Fe3O4 nanoparticles have a higher down with the help of a magnet. Magnetically
specific absorption rate, hence mixtures can be separated particles were washed successively with
used for temperature-tuned behavior. 5% NH4OH solution, acetone and ultrapure
One of the earlier studies from our group water (resistivity: 18.2 MWcm) three times with
reported that biphasic gels of LSMO and Al- each solvent, followed by an intermediate soni-
doped maghemite nanoparticles are suitable for cation (at 20 kHz, 250 W; Vibronics Pvt Ltd,
self-controlled hyperthermia [26]. To the best Mumbai, India) of 5 min between two wash-
of our knowledge, this is the first study report- ing cycles. LSMO was synthesized using the
ing the application of a biphasic suspension of method reported in the literature [18,19] with
nanoparticle-encapsulated magnetic nanovesicles little modification. Briefly, La 2O3, SrCO3 and
for cancer therapy. In fact, to date, there is no MnCO3 were separately dissolved in nitric acid
report of LSMO nanoparticle-loaded magnetic and mixed with citric acid and ethylene glycol
nanovesicles. The present study is focused on the in a ratio of 0.75[La 3+]:0.25[Sr2+]:[Mn 2+]:1.5[ci
preparation of magnetic nanovesicle formulation, tric acid]:2.25[ethylene glycol] and the pH was
characterization and its application for combined adjusted to 9 by adding NH4OH. The mixture
chemotherapy and self-controlled hyper­thermia. was stirred and heated to 80–90°C to remove
Dextran present in the biphasic suspension was water, and a pink-colored gel was formed. The
conjugated with fluorescein isothiocyanate resulting gel was heated to 250°C, which led to

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
the formation of black dry powder after auto- field ranging from -20 to 20 kOe. The M-H
combustion and the dry powder was annealed curves were recorded at room temperature. Mag-
at 700 and 800°C for 1 h. The annealed powder netization values, measured in millielectromag-
was then ground to a fine powder with a mor- netic units, were later normalized and expressed
tar and pestle. The ground powder was initially in electromagnetic units per gram. In order to
washed with ethanol followed by ultrapure water determine the Tc, magnetization versus tempera-
to remove the impurities. The magnetic particles ture measurements were carried out at 100 Oe.
were then dried in an oven. The Tc value was calculated from a derivative
of the magnetization versus temperature curve.
Preparation of dextran-coated MNPs & In order to confirm the coating of dextran
FITC conjugation on the LSMO and the biphasic suspension of
Dextran was dissolved in 0.5 M NaOH solu- LSMO and Fe3O4 nanoparticles, Fourier trans-
tion. LSMO nanoparticles (annealed at 700 form infrared (FTIR) analysis was performed
and 800°C) were dispersed in 0.5 M NaOH using the Magna 550 FTIR spectrophotometer
solution by sonication. The dextran-to-MNP (Nicolet Instruments Corp., WI, USA). For this,
ratio was always kept at 5:1 (w/w). MNPs were magnetic fluid was first frozen at -20°C and then
kept for sonication and the dextran solution was lyophilized. Lyophilized samples were then used
added dropwise. After another 1 h of sonication, to analyze the characteristic FTIR peaks.
the nanoparticles were centrifuged at 500 and Upon application of an alternating magnetic
750 rpm to remove larger particles, and finally field, the temperature of the magnetic fluid
centrifuged at 1000 rpm. Unbound dextran was increases due to Néel and Brownian relaxations.
removed. Dextran-coated MNPs were resus- The specific absorption rate (SAR) of the MNP
pended in water and sonicated for 1 h to obtain suspensions were calculated from the tempera-
a stable magnetic fluid suspension. Similarly, ture versus time curve. For this, 1 ml of mag-
mixtures of LSMO and Fe3O4 nanoparticles in netic nanovesicles was taken in a 15-ml falcon
1:0, 10:0.33 and 10:1 (w/w) ratios of the two tube and the tube was placed at the center of the
components were coated with dextran. copper coil of the radiofrequency (RF) generator
For the cellular internalization study, the (CLF-5000, Comdel, MA, USA) at a 423-kHz
dextran­-coated biphasic suspension was conjug­ frequency and field strength of 10 kA/m. Tem-
ated with FITC by slightly modifying the pro- peratures of the magnetic material suspension
tocol described by Shiigi and Slomich [27]. For were measured with an alcohol thermometer.
this, the biphasic suspension was dialyzed against The SAR was then calculated using Equation 1 [15]:
0.15 M sodium chloride for 48 h, followed by
dialysis against 0.05 M bicarbonate-buffered SAR = C^1/mM h (E quation 1)
saline (pH 8.5) for 7 h. The biphasic suspension
was then dialyzed against a 100 µg/ml of FITC where C is the specific heat capacity of the
in 0.05 M bicarbonate-buffered saline (pH 9.2) solution in joules/gram-1/Kelvin-1. In all experi-
for a further 14–16 h, and finally the reaction was ments, specific heat capacity was taken as
stopped by dialyzing against 0.02 M phosphate- 4.18 J/g-K (value for water), and mM is the mass
buffered saline (pH 7.4) for 2–3 h. It is note­ fraction of magnetic material in the suspension.
worthy to mention that all dialysis experiments In order to evaluate the biocompatibility of
were performed at room temperature. dextran-coated LSMO and biphasic suspension
of nanoparticles, both the materials were incu-
Characterization of MNPs bated in DMEM at 37°C for 48 h. The concen-
Crystallographic structures of Fe3O4 and LSMO tration of materials was 10 mg/ml. After 48 h,
nanoparticles were analyzed by the X’Pert the materials were centrifuged at 8000 rpm for
Diffract­ometer (Philips/PANalytical, Almelo, 15 min so that all of the MNPs were settled. The
The Netherlands) with a Cu Ka radiation wave- supernatants containing leached-out extracts of
length of 1.54 Å. The mean crystal size of the the nanoparticles were further diluted with com-
particles was calculated by using the Scherrer plete media and used for the bio­compatibility
equation [28]. Magnetic properties of Fe3O4 as study. For this, cells were seeded in a 96-well
well as LSMO nanoparticles were measured microtiter plate (1 × 104 cells/200 µl per well
using a vibrating sample magnetometer (model in one plate) in DMEM, supplemented with
7410, Lake Shore, OH, USA). For this, the dry 10% fetal bovine serum and 1% antibiotic anti­
powder of the nano­particles was placed inside mycotic solution. After 24 h, old media was
the sample holder and exposed to a magnetic discarded and the extracts of dextran-coated

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

MNPs at different concentrations of MNPs total amount of drug added during the process of
ranging from 0.0156–5 mg/ml were mixed nanovesicle formation. Encapsulation efficiency
with fresh media and added to the cells. The was then calculated using (Equation 3):
cells were then incubated again for 48 h. Cell
viability was determined by a sulforhodamine B % drug encapsulation = 61 - ^ DS/DT h@ # 100
assay [29]. (E quation 3)

Preparation of thermoresponsive where, DS is the drug present in the super­


magnetic nanovesicles natant and DT is the total amount of drug used
Magnetic nanovesicles were prepared by the during thin-film formation.
thin-film hydration method [15]. For this,
DSPC:cholesterol (8:2 w/w) were taken in a Drug-release studies
round-bottom flask and dissolved in a 2:1 chloro- Initially taking calcein as a model drug, release
form and methanol mixture. The solvent was then studies were performed according to the method
removed at 40°C by a rotary vacuum evaporator described by Anand et al., with nanovesicles hav-
(Superfit Continental Pvt Ltd, Mumbai, India) ing different lipid ratios of DSPC:cholesterol
to form a thin film. For drug-loaded magnetic (i.e., 9:1, 8:2 and 7:3) [30]. Results showed that
nanovesicles, the lipid-to-drug ratio was main- DSPC to cholesterol in the ratio of 8:2 is the
tained at 10:1 (w/w). Hydration of the film was most thermosensitive composition (data not
performed with MNPs or FITC-tagged MNPs shown). Drug-release studies of paclitaxel-
for 1 h at 60°C. The lipid-to-MNP ratio was 4:1 loaded DSPC:cholesterol (8:2) nanovesicles
(w/w). The nanovesicles thus formed were then were carried out at 37 and 44°C in a magnetic
successively extruded through 400 and 100 nm stirrer. In total, 1 ml of drug-loaded nano­vesicles
membranes using an extruder (Avanti Polar was put in a dialysis bag and placed in a beaker
Lipids, AL, USA) to make small unilamellar containing 50 ml of release media containing
nano­vesicles. Free MNPs and drugs were removed methanol:water in a 1:3 (volume:volume) ratio.
by centrifuging the magnetic nanovesicle suspen- Media was stirred using a mechanical stirrer and
sion at 1000 rpm for 10 min and 17000 rpm for the temperature of the media was maintained
25 min, respectively. either at 37 or 44°C for all experiments. A total
of 2 ml of the sample was drawn at different
Characterization of magnetic predetermined time points and substituted with
nanovesicles equal amounts of media to maintain the sink
The size and zeta-potential of the magnetic conditions.
nanovesicles were determined by dynamic light In another set of experiments, a release study
scattering (Brookhaven Instruments Corp., NY, was carried out for 24 h at 37°C followed by a
USA) and a zeta-potential analyzer (ZetaPALS, release study at 44°C for another 1 h. The objec-
Brookhaven Instruments Corp.). In order to tive of this set of experiments was to mimic the
determine the encapsulation of MNPs in nano­ temperature changes expected on intravenous
vesicles (Equation 2), magnetic nanovesicles were injection in vivo. It was considered that nano­
dissolved in concentrated HCl and then fil- vesicles take 24 h to reach the tumor site follow-
tered through Whatman filter paper of pore ing intravenous injection and then an alternating
size 0.2 µm. MNP concentration in the filtered magnetic field would be applied for another 1 h.
solution was determined by inductively coupled Similarly, one more set of sequential drug-release
plasma atomic emission spectroscopy (ICP-AES). studies was performed to mimic the temperature
changes expected in vivo, considering that mag-
% MNPs encapsulation = netic nanovesicle formulation was administered
Amount of MNP in formulation intratumorally where a drug-release study was
100
Amount of MNP used during hydration # performed at 37°C for 1 h followed by a release
(E quation 2) study at 44°C for another 1 h.

Paclitaxel-encapsulation efficiency was deter- Cellular internalization of magnetic


mined by estimating the amount of unencap- nanovesicles
sulated drug present in the supernatant after The cellular internalization study of FITC-tagged,
centrifugation of the nanovesicle sample at nanoparticle-loaded, DSPC:cholesterol (8:2) mag-
17,000 rpm for 25 min. The amount of drug in netic nanovesicles was performed with a MCF-7
the supernatant was then subtracted from the cell line. Cells were seeded (1 × 105 cells/ml per

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
well) in 24-well micro­titer plates with or without solution. After a 24-h incubation, old media was
12-mm glass cover slips in DMEM supplemented discarded and magnetic nanovesicles were diluted
with 10% fetal bovine serum and 1% antibiotic in complete media at different concentrations of
antimycotic solution. After 24 h of incubation, drug ranging from 50 to 600 nM. After adding the
old media was removed and 50 µl (50 µg MNPs) magnetic nanovesicles, cells were incubated again
of FITC-tagged, nanoparticle-loaded magnetic for 48 and 72 h. Cell viability was then determined
nano­vesicles were added with fresh media. The according to the aforementioned method.
cells were then incubated for 3 h in a CO2 incu- In vitro hyperthermia with magnetic nano­
bator. After 3 h, the cells were washed with phos- vesicles was performed in the MCF-7 cell line
phate-buffered saline and fixed on the glass cover under an alternating magnetic field. For this,
slips with 10% formaldehyde solution. The glass 12 × 105 cells were divided equally and put
cover slips were mounted on glass slides and fixed into two sterile polypropelyne centrifuge tubes
with glutaraldehyde. Confocal imaging of the cells (15 ml; Tarsons Ltd, Kolkata, India) with
was performed with the IX81 con­focal microscope complete media, and centrifuged at 2000 rpm
(Olympus, Tokyo, Japan) and using FV-500 soft- for 5 min to form pellets. The media was dis-
ware (Olympus). The cells were excited at 490 nm carded and magnetic nanovesicles with/without
and emission was captured at 530 nm. 112 nM of the drug in complete medium were
The cells seeded without glass cover slips were added to the pellets and cells were resuspended.
trypsinized and retrieved after washing out the A total of 6 × 105 cells suspended in complete
incubated FITC-tagged, nanoparticle-loaded media were taken in another 15-ml sterile poly-
magnetic nanovesicles. Trypsinized cells were prolyne tube. All three tubes were placed in
centrifuged for 5 min at 2000 rpm to form pellets. the copper coil of a RF generator (CLF-5000)
These cellular pellets were dissolved in concen- at a fixed frequency of 423 kHz and a field of
trated HCl and MNP concentration in the filtrate 10 kA/m one by one. During the experiment,
solutions were determined by ICP-AES (ARCOS the temperature was 44 ± 0.5°C for 15 min. The
from M/s, SPECTRO, Kleve, Germany). cells were then seeded in a 24-well plate with
six replicates and incubated for 48 h. For the
In vivo stability evaluation of magnetic control group, 6 × 105 cells were also seeded in
nanovesicles a 24-well plate with six replicates and incubated
In order to evaluate in vivo stability of mag- for 48 h. The viable cells were then counted by
netic nanovesicles, experiments were conducted the trypan blue dye exclusion method using a
on female Swiss mice (bodyweight: 20–23 g; hemocytometer.
6–8 weeks old) that were bred and reared in Valeriote’s method was used to evaluate the
the animal house facilities of Bhabha Atomic combined effect of magnetic hyperthermia and
Research Centre (Mumbai, India). The animal chemotherapy [13]. If A, B and (A + B) represent
experiment was conducted according to the rel- the percentage cell viability after treatment A,
evant national laws relating to animal experi- B and the combination of treatment A and B,
mentations. For this study, animals were ran- respectively, the combined effects were defined
domly divided into three groups (n = 4). Mag- as: synergistic: (A + B) < (A × B)/100; additive:
netic nanovesicles with 2 and 1 mg MNPs were (A + B) = (A × B)/100; subadditive: (A × B)/100
administered into the animals of groups 1 and < (A + B) < A, if A < B; interference: A < (A + B)
2, respectively, through the tail vein, whereas no < B if A < B; and antagonistic: B < (A + B), if
injection was given to group 3 animals (control). A < B.
After 24 h, all of the animals were observed for
any toxicity and sacrificed, blood samples were „„ Statistical analysis
collected in glass vials containing 150 µl of 4% All experiments were repeated at least three times
EDTA solution and evaluated for packed cell and results were expressed as mean ± stand-
volume (PCV) and agglomeration of MNPs. ard deviation. Statistical analysis was carried
out using t-tests (paired t-test with unequal
Cytotoxicity study of MNPs variances). An event is considered as significant
& hyperthermia when p < 0.05 (with 95% CI).
For in vitro cytotoxicity study, MCF-7 cells
were seeded in two 96-well microtiter plates Results
(1 × 104 cells/200 µl per well in one plate) in „„ Characterization of MNPs
modified Eagle medium supplemented with 10% X-ray diffraction results (Supplementary Figure  S1;
fetal bovine serum and 1% antibiotic antimycotic see online at www.futuremedicine.com/doi/

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

suppl/10.2217/NNM.13.90) confirmed the nano­particles are uniformly dispersed (Figure 2A).


formation of Fe3O4 and LSMO nanoparticles. The size of nanoparticles is in the range of
The size of Fe3O4 and LSMO nanoparticles were 10–30 nm. It was confirmed from the x-ray dif-
10 and 25 nm, respectively. Vibrating sample fraction results that the smaller particles were
magnetometer studies showed that saturation Fe3O4 nano­particles, while larger-sized parti-
magnetization of LSMO nanoparticles annealed cles were LSMO nano­particles. Annealing at a
at 700°C was 15.34 emu/g and its Tc was 48.6°C high temperature led to a formation of bridges
(Figure 1). As we increased the annealing tempera- among the LSMO particles and, therefore, the
ture, the magnetization values as well as the Tcs actual size of particles became more than the
of magnetic materials increased. The saturation size determined by x-ray diffraction [18]. A bio-
magnetization and Tc of LSMO nanoparticles compatibility study of extracts of LSMO and
annealed at 800°C were 21 emu/g and 59.7°C, biphasic nanoparticle suspension was performed
respectively (Supplementary Figure S2). in a L929 cell line (Figure 2B). Extracts of both
Dextran-coated MNPs were lyophilized and the materials were incubated with cells for
FTIR was studied. Dextran had peaks at 3400, 48 h. Cell viability for the extract of dextran-
2927 and 1649 cm-1 due to the presence of O-H, coated LSMO was 81% after 48 h of incuba-
C-H and C-C bonds, respectively, as shown in tion, whereas for the extract of dextran-coated
Supplementary Figure  S3. In dextran-coated MNPs, biphasic nanoparticle suspension it was 82%.
the corresponding peaks were at 3404, 2921 and Results of a SAR experiment showed that the
1632 cm-1. From the FTIR results, it is clear that the suspension of LSMO nanoparticles annealed
peaks in dextran due to O-H, C-H and C-C bonds at 700°C caused an increase in temperature of
are shifted. This indicates that there is interaction up to 33.8°C in 15 min, whereas during the
between dextran and the MNPs as the magnetic same time period, temperature due to sus-
particles become coated with dextran. pension of LSMO nanoparticles annealed at
A transmission electron microscope 800°C rose to 37.6°C, which might result in
image of biphasic suspension confirmed that prolonged exposure of the animal model dur-
ing in vivo experiments (Supplementary Figure S4).
The addition of Fe3O4 nanoparticles increased
A 25 the temperature within a short span of time.
Magnetization

20
Hence, Fe3O4 nanoparticles were added in dif-
15
ferent amounts and the SAR of these biphasic
(emu/g)

10
suspensions were evaluated. Finally, a biphasic
5
suspension of LSMO to Fe3O 4 nanoparticles
0 Field (kOe) 10 (10:1) was selected for further experiments.
-20 -10 -5 20
-10 This biphasic suspension reached 42.4°C in
-15 700°C 4.5 min. The SAR value of magnetic nano­
-20 800°C vesicles was 8.9 W/g and it took 13 min to raise
-25 the temperature to 44°C.

„„ Characterization of magnetic
B 2.5 1.00 × 10-4 nanovesicles
The mean hydrodynamic diameter of
Magnetization (emu/g)

2 0.00 × 10-4 DSPC:cholesterol magnetic nanovesicles was


dm/dT (emu/°C)

1.5 -1.00 × 10-4 225 ± 45 nm and its zeta-potential was -41 ± 5 mV.


A transmission electron microscope image of
1 emu/g -2.00 × 10-4 the magnetic nanovesicles shows that the size of
dm/dT (emu/°C) the nanovesicles is in the range of 180–200 nm
0.5 -3.00 × 10-4
(Figure 3A). Figure 3B shows the diffraction pattern
0 -4.00 × 10-4 of magnetic nanovesicles. The rings and bright
25 35 45 55 65 75 85 95 105 115 spots indicate the presence of MNPs within the
Temperature (°C) nano­vesicles. Figure 3C shows the transmission elec-
tron microscope image of a blank nanovesicle and
Figure 1. Magnetic properties of La0.75Sr0.25MnO3 nanoparticles. (A) M-H loop Figure 3D is its corresponding diffraction pattern.
recorded at room temperature for La0.75Sr0.25MnO3 nanoparticles calcined at 700 and The paclitaxel and MNP encapsulation effi-
800°C. (B) Curie temperature of La0.75Sr0.25MnO3 nanoparticles calcined at 700°C. ciency of magnetic nanovesicles were 83 ± 3 and
dm/dT: First order derivative of magnetization with respect to temperature. 67 ± 5%, respectively.

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
Drug release from DSPC:cholesterol (8:2) A
nanovesicles
A calcein-release study showed that
DSPC:cholesterol (8:2) nanovesicle formulation
was more thermosensitive than the other two for-
mulations (DSPC:cholesterol 9:1 and 7:3 w/w).
Therefore, the paclitaxel release was carried out
with DSPC:cholesterol (8:2) nanovesicle formula-
tion at 37 and 44°C. The paclitaxel release from
this formulation was 3.9 (33.54 µg) and 5.2%
(44.7 µg; p < 0.05), at 37 and 44°C, respec-
tively, as shown in Figure 4A . In order to mimic the
expected in vivo temperature change on the intra-
venous injection, a sequential paclitaxel release
with DSPC:cholesterol (8:2) nanovesicles was
performed at 37°C for 24 h, followed by release at
200 nm
44°C for another 1 h. The cumulative drug release
at the end of the study was 8.5% (120.7 µg) and,
thus, higher than those carried out only at 44°C. B 120
The drug-release profile is shown in Figure 4C. The BS
drug-release profile during the transition of tem- LSMO
100
perature is shown in Figure 4D. At the end of the sec-
ond sequential release study of sequential release at
80
Cell viability (%)

37°C for 1 h followed by 44°C for 1 h (as expected


for intratumoral injection), the cumulative drug
release was 6.6% (109.6 µg; shown in Figure 4B). 60

Cellular internalization of magnetic


40
nanovesicles
Therapeutic effects of drug-loaded nano­particles
are demonstrated to be dependent on the cellu- 20
lar internalization and sustained retention by the
malignant cells [31,32]. Although there are a num- 0
ber of differences between in vitro and in vivo pro- Control 0.16 0.31 0.63 1.25 2.50 5.00
cesses, in vitro experiments can provide some basic Dose of MNPs (mg/ml)
ideas regarding the intracellular movement and
accumulation of nanoparticles within the cells. Figure 2. Characterization of biphasic suspension. (A) Transmission electron
In vitro cellular internalization studies were car- microscopy image of BS; and (B) cell viability of dextran-coated BS and LSMO
ried out with FITC-tagged biphasic nanoparticle nanoparticles in the L929 cell line.
BS: Biphasic suspension; LSMO: La0.75Sr0.25MnO3; MNP: Magnetic nanoparticle.
suspension-loaded magnetic nanovesicles. Cellular
uptake of magnetic nanovesicles was qualitatively levels of hemoglobin, mean corpuscular volume
and quantitatively determined by using con­focal and PCV were evaluated (shown in Figur e  6).
microscopy of FITC-tagged biphasic nanoparticle The hemoglobin level in the control group was
suspension-loaded magnetic nanovesicles in 15.5 ± 0.4 and 14.8 ± 0.2 g/dl for group 1 and
MCF-7 cell lines and ICP-AES (Figure 5). ICP-AES 15.4 ± 0.6 g/dl for group 2 animals. No sig-
results suggested that 49% of the total MNPs were nificant change was observed in hemoglobin
internalized after 3 h of incubation (Figure  5E). levels, mean corpuscular volume or PCV on
Moreover, these FITC-tagged biphasic nano­ administration.
particle suspension-loaded magnetic nanovesicles
could also be used for in vitro cellular imaging. In vitro cytotoxicity of magnetic
nanovesicles
In vivo stability evaluation of magnetic A cytotoxicity study of paclitaxel-loaded mag-
nanovesicles netic nanovesicles was carried out by incubat-
In order to evaluate the stability of the mag- ing for 48 and 72 h. Results of the cytotoxicity
netic nanovesicles in plasma and to evaluate the studies are shown in Figure 7. It was observed that
feasibility of intravenous administration, the for 48-h treatment groups, the percentage cell

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

A AC magnetic fields were shown in Figure 8B. The


results showed that there was no effect of an
AC magnetic field on the cells. It was observed
that the cell viability for cells treated with mag-
netic nanovesicles mediating hyperthermia was
74.2%. Interestingly, this value (74.2%) reduced
to 24.3%, when the cells were subjected to a
B combination of hyperthermia and chemo­therapy
(drug: 112 nM). The result suggested that a
combination of hyperthermia and chemotherapy
act synergistically on the MCF-7 cell line. Con-
sequently, the efficacy of treatment improved
2 nm-1 due to the combined effect of these treatments.

C Discussion
We developed novel biphasic MNP–nanovesicle
200 nm
hybrids for self-controlled hyperthermia and
D chemotherapy. The thermosensitive nanovesicles
coencapsulated a biphasic suspension consisting
of Fe3O4 and LSMO nanoparticles and pacli-
taxel. The biphasic suspension was optimized
for self-controlled hyperthermia. LSMO nano-
1000 nm particles with a low SAR value regulated the
2 nm-1
overall temperature gained by the suspension,
Figure 3. Transmission electron micrograph and diffraction patterns of whereas Fe3O4 nanoparticles were responsible for
nanovesicles. (A) Transmission electron microscopy images of magnetic liposomes speeding up the heating process. A fine balance
containing biphasic suspension of La0.75Sr0.25MnO3 and iron oxide nanoparticles in between both the nanoparticles is necessary to
10:1 ratio. The size of magnetic liposomes is in the range of 180 to 250 nm. achieve a hyperthermia temperature within 1 h.
(B) The diffraction pattern of magnetic liposomes; (C) a transmission electron
The possibility of using a biphasic gel of
microscopy image of a blank liposome; and (D) a diffraction pattern of a blank
liposome. LSMO and Al-substituted maghemite nano-
particles for self-controlled hyperthermia has
viability sharply reduced for magnetic nano­ been successfully demonstrated in our earlier
vesicles until 200 nM and did not change sig- work [26]; however, the work was limited to the
nificantly even after increasing the dose. The evaluation of heating rates and did not evaluate
IC50 value of paclitaxel-loaded magnetic nan- the encapsulation within a biocompatible lipid
ovesicles was determined using the method men- nanovesicle. Concerns regarding the toxicity of
tioned in our earlier paper [15], and the value was LSMO nanoparticles have largely limited their
112 nM (95.64 µg). Therefore, a hyper­thermia biomedical applications. The cellular responses
experiment was performed using magnetic nano­ to LSMO nanoparticles have been largely unex-
vesicles containing 112 nM paclitaxel with a plored. In the present study, the encapsulation
48-h incubation period. After a 72-h incubation of biphasic suspensions of Fe3O4 nanoparticles
period, cell viability was less than 20% for all and LSMO within thermosensitive nano­vesicles
the concentrations of paclitaxel-loaded magnetic has been achieved. An in vitro biocompatibility
nanovesicles. experiment with L929 cell lines showed that the
extract of the biphasic suspension did not show
In vitro hyperthermia under an any toxicity up to a 5 mg/ml concentration. Fur-
alternating current magnetic field thermore, no toxic effects were observed on intra-
MCF-7 cells were subjected to hyperthermia venous in vivo administration. The study paves
under an alternating current (AC) magnetic the way for exploration of further biomedical
field with bare and drug-loaded magnetic nano­ applications of hybrid MNPs for self-controlled
vesicles containing 112 nM paclitaxel. On the hyperthermia.
other hand, control cells were not subjected to Phospholipids, such as DSPC, with high tran-
any treatment. Hyperthermia experiments were sition temperature are required for maintaining
conducted at 44°C temperature for 15 min and a gel state in the body. The inclusion of choles-
the result was presented in Figure 8. Temperature terol in DSPC liposomes at various molar ratios
profiles during hyperthermia experiments and has been reported to affect the clearance rates

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
A 6 B 7 37°C 44°C

Transition period
5 6
Cumulative drug release (%)

Cumulative drug release (%)


5
4
4
3
3
2
2

1 Release at 44°C 1
Release at 37°C

0 0
0 10 20 30 40 50 60 70 0 30 60 90 120 150
Time (min) Time (min)

C 10 37°C D 10
37°C 44°C

Transition period
9
Cumulative drug release (%)
Cumulative drug release (%)

8
9
7

5 8

3
7
2

0 6
0 5 10 15 20 25 30 23 23.5 24 24.5 25 25.5
Time (min) Time (min)

Figure 4. Drug release studies from nanovesicles under different physiological conditions.
(A) Drug release from magnetic liposomes at 37 and 44°C. (B) Sequential drug release (i.e., in order
to mimic the expected temperature change in vivo following intratumoral injection) was performed at
37°C for 1 h followed by a release study at 44°C for another 1 h. (C) Sequential drug release (i.e., in
order to mimic the expected temperature change in vivo following intravenous injection) was
performed at 37°C for 24 h followed by a release study at 44°C for another 1 h. (D) Enlarged image
of drug-release profile showing the transition period between 37 and 44°C.

of pure DSPC liposomes in vivo. At 20 mol% cells and reduce the entry of nanovesicles into
cholesterol, the circulation half-life was found the tumor tissue [34–36]. For example, Hong et al.
to be approximately 30 min, compared with showed that the plasma area under the curve for
seconds for pure DSPC vesicles. The inclusion doxorubicin-loaded PEGylated nanovesicles was
of 30 mol% cholesterol further extended the approximately twofold more than doxorubicin
half-life of the vesicles to 5 h. The cholesterol entrapped in non-PEGylated DSPC:cholesterol
effect on circulation time plateaued at 30 mol%, nanovesicles [37]. However, at the end of 72 h,
without further benefits of higher amounts of the concentration of doxorubicin entrapped
cholesterol addition [33]. in non-PEGylated DSPC:cholesterol nano­
Sterically stabilized PEGylated nanovesicles vesicle in tumor tissue was 1.44-times more
can escape from opsonization by plasma proteins than doxorubicin in PEGylated nanovesicles.
and are characterized by a long blood circulation These results suggest that non-PEGylated nan-
time. However, the presence of large molecules, ovesicles composed of DSPC and cholesterol
such as polyethylene glycol, on the surface of are suitable for accumulation at cancerous sites.
nanovesicles may minimize the interaction with Such nano­vesicles have advantages of being

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

A B

50 µm 50 µm

C D

E 60
internalization (%)

50
40
30
20
MNP

10
0 50 µm 50 µm
Control Treated

Figure 5. Confocal laser scanning microscopy images showing the cellular internalization of
magnetic liposomes in the MCF-7 cell line. (A) Fluorescence images of treated cells and
(B) merged image treated cells. (C) Fluorescence image of control cells and a (D) differential
interference contrast image of control cells. (E) The quantitative estimation of MNPs in cells.
MNP: Magnetic nanoparticle.

thermo­sensitive, lower in cost and easier to stability of the magnetic nanovesicles. The mag-
manufacture than PEGylated nanovesicles. netic nanovesicles neither formed any aggregate
In our study, during in vivo evaluation, mag- in blood, nor caused any acute toxic effects.
netic nanovesicles containing biphasic suspen- During the hyperthermia experiment the
sion up to 2 mg of MNPs was administered maximum temperature achieved was 44°C.
intravenously and the level of hemoglobin, mean This showed that the temperature of the mag-
corpuscular volume and PCV percentage did netic nanovesicles was stabilized at 44°C without
not change significantly, suggesting the colloidal any overheating. The temperature of magnetic

A 18 B 60 C 50

15 50 40
Hemoglobin (mg/dl)

12 40
30
PCV (%)
MCV (fl)

9 30
20
6 20
10
3 10

0 0 0
Control 24 h Control 24 h Control 24 h
Time point Time point Time point

Control Group 1 Group 2

Figure 6. The in vivo stability evaluation of magnetic liposomes. (A) Hemoglobin level, (B) MCV level and (C) PCV percentage.
MCV: Mean corpuscular volume; PCV: Packed cell volume.

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
nano­vesicles did not increase beyond this tempera- A 120
ture, even after applying an alternating magnetic
field due to the sharp decrease in saturation mag- 100
netization value in the vicinity of the Tc. The tem-

Cell viability (%)


80
perature is well below their actual Tc (i.e., 48.6°C),
this is due to the dead mass of dextran and lipids. 60
The magnetic nanovesicles have high MNP
and drug encapsulation efficiencies. High encap- 40
sulation of the drug and MNPs are necessary 20
for achieving a better therapeutic index with a
lesser amount of delivery systems. In most lit- 0
erature, the encapsulation efficiency of MNPs 0 50 100 200 250 400 500 600
Concentration of PTX (nM)
has been low when coencapsulated with an
anticancer drug. The other approach used to
B 120
increase this efficiency has been to functional-
ize the surfaces of MNPs and then conjugate the 100
drug [38]. Generally, the conjugation chemistry is Cell viability (%)

quite complex and the method of cleavage of the 80


drug to allow release of the drug at the target site 60
needs to be carefully controlled. Further asso-
ciation of the drug on the surface, rather than 40
within the nanocarrier, may influence the bio­
20
distribution of MNPs [39] by altering the physical
and/or surface characteristics of the bare MNPs 0
(e.g., hydrodynamic size, stability and magneti- 0 50 100 200 250 400 500 800
zation) [40–43]. Magnetic nanovesicles can play an Concentration of PTX (nM)
important role in this regard, and we formulated
PTX Magnetic liposomes
DSPC:cholesterol magnetic nanovesicles with-
out compromising the encapsulation efficiencies
Figure 7. Comparative cytotoxicity studies of magnetic liposomes and
of MNPs and the drug, as well as the original
marketed taxol formulation in MCF-7 cells. (A) 48-h incubation period and
physiological properties of MNPs. (B) 72-h incubation period.
In vitro cellular internalization showed that PTX: Paclitaxel.
magnetic nanovesicles were internalized by the
cells. The panel for control cells was dark, sug- changes expected in vivo following systemic
gesting that there was negligible autofluorescence administration of magnetic nano­vesicles, a
from the cells during the in vitro experiment, sequential drug-release experiment was per-
suggesting the feasibility of using these mag- formed where a release study was carried out
netic nanovesicles for in vitro optical imaging. at 37°C for 24 h, followed by a release study
However, interference due to auto­fluorescence at 44°C for another 1 h. The cumulative drug
and photobleaching cannot be ruled out during release during sequential release increased to
in vivo applications. In that case, NIR-sensitive 8.5% (i.e., 120.7 µg). However, the drug that
dyes may be used instead of FITC, which have is released at 37°C can lead to systemic toxicity.
very low interference from the tissue auto­ In case of a second sequential release study,
fluorescence. Moreover, problems such as pho- where it was considered that the AC magnetic
tostability, photobleaching and low quantum field would be applied after 1 h of intratumoral
yield can be overcome by using the NIR dyes or injection of magnetic nanovesicles, the cumula-
NIR dye-encapsulated nano­particles for tumor tive drug release was 6.6% (109.6 µg), and the
detection and imaging [44]. entire amount was released within the tumor
Considering the fact that hyperthermia mass. Systemic toxicity due to the drug would be
treatment can be applied continuously for a avoided. Hence, intratumoral administration of
maximum of 1 h, drug release studies were lim- magnetic nanovesicles was preferred to systemic
ited to 1 h. The percentage of cumulative drug administration for self-controlled hyperthermia
release was low due to the hydrophobic nature and drug delivery. It was expected that upon
of paclitaxel, but was sufficient for a therapeutic intratumoral injection that the drug would act as
effect as it was found to be higher than the IC50 a depot within the nanovesicles and continue to
of paclitaxel. In order to mimic temperature release slowly within the tumor mass. Achieving

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


PRreliminary Communication
eview Authors Gogoi, Sarma, Bahadur & Banerjee

A self-controlled magnetic vesicles on intravenous


120
administration would depend on the percentage
100 of the drug-loaded magnetic nanovesicles reaching
the tumor site.
Cell viability (%)

80
In the hyperthermia experiments, we chose
60 the dose of drug in magnetic nanovesicles
equal to its IC50 value. The combination of
40
paclitaxel and heat killed 75.7% cells within
20 15 min. Hyperthermia can kill the cells by
irreversibly damaging the cellular complexes
0 required for DNA synthesis and repair. The
Control AMF Heat 112 nM PTX Heat +
112 nM PTX extent of cell killing may vary depending
Treatment upon the temperature of treatment and the
duration of experiment. In an in vitro study,
B 50 Prasad et al. demonstrated that the cytotoxic
effect of magnetic hyperthermia is time and
40 temperature dependent [47]. They showed
that magnetic hyperthermia killed the cells
Temperature (°C)

30 by irreversibly damaging the cytoskeleton.


In our study, upon validating the results of
20 hyperthermia and chemotherapy with Veleri-
ote’s method, it was found that the combined
10 effect of chemotherapy and hyperthermia was
Magnetic liposomes
Only AMF
synergistic in nature. In cancer therapy, dif-
0 ferent modalities are used in combination to
0 10 20 30 40 achieve a synergistic effect and improve the
Time (min) therapeutic index. This helps in reducing the
dose of individual therapeutic modalities and,
hence, the side effects of individual modal-
Figure 8. Cellular toxicity of MCF-7 cell line during hyperthermia experiments.
ity’s can be minimized. Pradhan et al. [13] and
(A) A hyperthermia experiment was performed under an AMF with bare and drug-
loaded magnetic liposomes (112 nM paclitaxel). During the experiment, the field Kulshrestha et al. [15] showed the synergistic
strength was constantly maintained. The AMF group of cells were exposed to only effect of hyperthermia and chemotherapy in
the AMF. (B) The temperature profile during hyperthermia experiment as well as different cancer cell lines but did not have
only for the AMF. any self-control in the hyperthermia as only
AMF: Alternating current magnetic field; PTX: Paclitaxel.
Fe3O4 nanoparticles were entrapped in those
nanovesicles. Ito et al. demonstrated that the
sustained drug release is very important in can- combined effect of 4-S -cysteaminylphenol
cer therapy as it prevents cancer from relapsing treatment and hyperthermia on B16 melanoma
and developing resistance against the drug [45,46]. cells was additive in nature [48]. This may be
While there are advantages of a regional therapy, due to the nature of the action of the drug and
intratumoral injection also has the disadvantage duration of hyperthermia. In the present study
of being invasive, depending on the location of the effects of hyperthermia and drug delivery
the tumor, and may cause an uneven distribution. were found to be synergistic. This suggests the
Moreover, it is worth mentioning that the cumula- potential application of DSPC:cholesterol-
tive drug releases for both the sequential release based thermosensitive magnetic nanovesicles
studies were more than the IC50 of magnetic nano­ encapsulating biphasic suspensions of LSMO
vesicles formulation (95.64 µg). Our preliminary and Fe3O4 nanoparticles for chemotherapy and
study suggests that there was no aggregation of self-controlled hyperthermia.
the magnetic vesicles on intravenous administra-
tion. However, the half-life of the magnetic vesi- Conclusion & future perspective
cles in plasma on systemic administration and A novel magnetic nanovesicle containing pacli-
the biodistribution needs to be determined. It taxel and a biphasic suspension of Fe3O4 and
is expected that even a 10% localization of the LSMO nanoparticles were prepared by a thin-
magnetic vesicles at the tumor site would allow a film hydration method. To the best of our
sufficient temperature rise within 1 h and a drug knowledge, this is the first magnetic nanovesicle
release above the IC50. However, the success of the prepared from a biphasic magnetic nanohybrid

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group


Biphasic magnetic nanoparticles–nanovesicle hybrids Preliminary Communication
for combined chemotherapy and self-controlled Financial & competing interests disclosure
hyperthermia. Dextran-coated biphasic nano- The financial support provided by the Department of
particle suspension was conjugated with FITC, Information Technology and the Nanomission, Department
and coencapsulated with paclitaxel within the of Science and Technology are gratefully acknowledged. The
thermosensitive magnetic nano­vesicles. The IC50 authors have no other relevant affiliations or financial
of this magnetic nanovesicle was 112 nM. The involvement with any organization or entity with a finan­
combined effect of hyperthermia and chemo- cial interest in or financial conflict with the subject matter
therapy was synergistic in nature while con- or materials discussed in the manuscript apart from those
trolling the temperature at 44°C. The biphasic disclosed.
nanoparticle nanovesicle hybrid has the potential No writing assistance was utilized in the production of
for combined self-­controlled hyper­thermia and this manuscript.
chemotherapy.
Ethical conduct of research
Acknowledgements The authors state that they have obtained appropriate insti­
The authors thank the Centre for Research in Nanotech­ tutional review board approval or have followed the princi­
nology and Nanoscience for using their instrumentation ples outlined in the Declaration of Helsinki for all animal
facilities. experimental investigations.

Executive summary
Need for self-controlled hyperthermia and chemotherapy
ƒƒ Magnetic materials that can prevent overheating of tissues are required for combined hyperthermia and temperature-triggered drug
delivery.
ƒƒ Magnetic biphasic suspension of La0.75Sr0.25MnO3 and iron oxide nanoparticles were developed for self-controlled hyperthermia and
chemotherapy.
Results of encapsulation and proof of principle
ƒƒ Encapsulation efficiencies of paclitaxel and magnetic nanoparticles were 83 ± 3 and 67 ± 5%, respectively.
ƒƒ Magnetic nanovesicles coencapsulating La0.75Sr0.25MnO3, iron oxide nanoparticles and paclitaxel were biocompatible.
ƒƒ The IC50 of magnetic nanovesicles in the MCF-7 cell line was 112 nM.
ƒƒ Combined self-controlled hyperthermia and chemotherapy in an AC magnetic field showed synergistic effects in cytotoxicity in MCF-7
cell lines and maintained the temperature at 44°C.
Conclusion
ƒƒ Magnetic nanovesicles containing La0.75Sr0.25MnO3 and iron oxide nanoparticles and coencapsulating paclitaxel have potential for
self-controlled hyperthermia and trigger responsive drug delivery.

nn Synthesis and application of magnetic hyperthermia. Crit. Rev. Oncol. Hematol.


References nanovesicles and nanohydrogels for cancer 43(1), 33–56 (2002).
Papers of special note have been highlighted as:
n of interest
hyperthermia and chemotherapy. 9 Kong G, Dewhirst MW. Hyperthermia and
nn of considerable interest 4 Kizaka-Kondoh S, Inoue M, Harada H, nanovesicles. Int. J. Hyperthermia 15(5),
Hiraoka M. Tumor hypoxia: a target for 345–370 (1999).
1 Jordan A, Scholz R, Maier-Hauff K et al.
Presentation of a new magnetic field therapy selective cancer therapy. Cancer. Sci. 94(12), 10 Kong G, Braun RD, Dewhirst MW.
system for the treatment of human solid 1021–1028 (2003). Characterization of the effect of hyperthermia
tumors with magnetic fluid hyperthermia. 5 Jain RK. Normalization of tumor vasculature: on nanoparticle extravasation from tumor
J. Magn. Magn. Mater. 225(1–2), 118–126 an emerging concept in antiangiogenic vasculature. Cancer Res. 61, 3027–3032
(2001). therapy. Science 307, 58–62 (2005). (2001).
n Reports the application of ferrofluid for 6 Bouzin C, Feron O. Targeting tumor stroma 11 Yanase M, Shinkai M, Honda H,
hyperthermia application. and exploiting mature tumor vasculature to Wakabayashi T, Yoshida J, Kobayashi T.
improve anti-cancer drug delivery. Drug Resist. Intracellular hyperthermia for cancer using
2 Schneiderman MH, Hofer KG,
Updat. 10(3), 109–120 (2007). magnetite cationic nanovesicles: an in vivo
Schneiderman GS. Targets for radiation-
study. Jpn J. Cancer Res. 89, 463–469 (1998).
induced cell death: when DNA damage 7 Streffer C. Molecular and cellular mechanisms
doesn’t kill. Radiat. Res. 155(4), 529–535 of hyperthermia. In: Thermoradiotherapy and 12 Ito A, Tanaka K, Honda H, Abe S,
(2001). Thermo­chemotherapy. Seegenschimicdt MH, Yamaguchi H, Kobayashi T. Complete
Fessenden P, Vernon CC (Eds). Springer regression of mouse mammary carcinoma
3 Gogoi M, Jaiswal MK, Banerjee R,
Verlag, Berlin, Germany, 47–74 (1995). with a size greater than 15 mm by frequent
Bahadur D. Magnetic nanovesicles and
repeated hyperthermia using magnetite
hydrogels towards cancer therapy. nn Mechanism of cell death due to
nanoparticles. J. Biosci. Bioeng. 96(4),
In: Magnetic Nanoparticles from Fabrication to hyperthermia.
364–369 (2003).
Clinical Applications. Thanh NTK (Ed.). 8 Hildebrandt B, Wust P, Ahlers O et al.
CRC Press, London, UK, 479–498 (2011). nn Application of cationic magnetic
The cellular and molecular basis of
nanovesicles for in vivo hyperthermia.

future science group www.futuremedicine.com doi:10.2217/NNM.13.90


Preliminary Communication Gogoi, Sarma, Bahadur & Banerjee

13 Pradhan P, Giri J, Rieken F et al. Targeted 25 McMullen TPW, McElhaney RN. Differential without polyethylene glycol coating in C-26
temperature sensitive magnetic nanovesicles for scanning calorimetric studies of the interaction tumor-bearing mice is surface coating with
thermo-chemotherapy. J. Control. Release of cholesterol with distearoyl and dielaidoyl polyethylene glycol beneficial? Clin. Cancer
142(1), 108–121 (2010). molecular species of phosphatidylcholine, Res. 5, 3645–3652 (1999).
n Application of folate and magnetic-targeted phosphatidyl­ethanolamine, and 38 Kohler N, Sun C, Fichtenholtz A, Gunn J,
phosphatidylserine. Biochemistry 36, Fang C, Zhang M. Methotrexate-
magnetic nanovesicles for cancer therapy.
4979–4986 (1997) immobilized poly(ethylene glycol) magnetic
14 Dilnawaz F,Singh A, Mohanty C, Sahoo SK.
26 Prasad NK, Hardel L, Duguet E, Bahadur D. nanoparticles for MR imaging and drug
Dual drug loaded superparamagnetic iron
Magnetic hyperthermia with biphasic gel of delivery. Small 2, 785–792 (2006).
oxide nanoparticles for targeted cancer therapy.
La1-xSrxMnO3 and maghemite. J. Magn. Magn. 39 Chouly C, Pouliquen D, Lucet I, Jeune JJ,
Biomaterials 31(13), 3694–3706 (2010).
Mater. 321, 1490–1492 (2009). Jallet P. Development of superparamagnetic
15 Kulshrestha P, Gogoi M, Bahadur D, Banerjee
27 Shiigi SM, Slomich M. Heterologous antisera nanoparticles for MRI: effect of particle size,
R. In vitro application of paclitaxel loaded
to mouse brain. In: Selected Methods in Cellular charge and surface nature on biodistribution.
magnetonanovesicles for combined
Immunology. Mishell BB, Shiigi MS (Eds). WH J. Microencapsul. 13, 245–255 (1996).
chemotherapy and hyperthermia. Colloid.
Freeman Co., CA, USA, 247–257 (1980). 40 Arias JL, Ruiz MA, Gallardo V, Delgado AV.
Surf. B Biointerfaces 96, 1–7 (2012).
28 Cullity BD, Graham CD. Introduction to Tegafur loading and release properties of
16 Zhang J, Misra RDK. Magnetic drug-targeting
Magnetic Materials (2nd Edition). John Wiley magnetite/poly(alkylcyanoacrylate) (core/
carrier encapsulated with thermosensitive smart
& Sons Inc., NJ, USA (2008). shell) nanoparticles. J. Control. Release
polymer: core-shell nanoparticle carrier and
29 Vichai V, Kirtikara K. Sulforhodamine B 125, 50–58 (2008).
drug release response. Acta Biomater. 3,
838–850 (2007). colorimetric assay for cytotoxicity screening. 41 Kim SY, Lee YM. Taxol-loaded block
Nat. Protoc. 1, 1112–1126 (2006). copolymer nanospheres composed of
17 Shido Y, Nishida Y, Suzuki Y, Kobayashi T,
30 Anand P, Huilgol N, Banerjee R. Effect of methoxy poly(ethylene glycol) and
Ishiguro N. Targeted hyperthermia using
fluidizing agents on paclitaxel penetration in poly(epsilon-caprolactone) as novel
magnetite cationic nanovesicles and an
cervical cancerous monolayer membranes. anticancer drug carriers. Biomaterials 22,
alternating magnetic field in a mouse
J. Membr. Biol. 219, 83–91 (2007). 1697–1704 (2001).
osteosarcoma model. J. Bone Joint Surg. Br.
92(4), 580–585 (2010). 31 Zhang Y, Tang L, Sun L et al. A novel 42 Chattopadhyay P, Gupta RB. Supercritical
paclitaxel-loaded poly(epsilon-caprolactone)/ CO2 based production of magnetically
18 Vasseur S, Duguet E, Portier J et al.
Poloxamer 188 blend nanoparticle overcoming responsive micro- and nanoparticles for drug
Lanthanum manganese perovskite
multidrug resistance for cancer treatment. Acta targeting. Ind. Eng. Chem. Res.
nanoparticles as possible in vivo mediators for
Biomater. 6, 2045–2052 (2009). 41, 6049–6058 (2002).
magnetic hyperthermia. J. Magn. Magn. Mater.
302(2), 315–320 (2006). 32 Jin C, Bai L, Wu H, Tian F, Guo G. 43 Zhu L, Ma J, Jia N, Zhao Y, Shen H.
Radiosensitization of paclitaxel, etanidazole and Chitosan-coated magnetic nanoparticles as
nn Synthesis of Curie temperature-tuned
paclitaxel+etanidazole nanoparticles on hypoxic carriers of 5-fluorouracil: preparation,
magnetic nanoparticles.
human tumor cells in vitro. Biomaterials 28, characterization and cytotoxicity studies.
19 Liangruksa M, Ganguly R, Puri IK. Parametric Colloid. Surf. B Biointerface 68, 1–6 (2009).
3724–3730 (2007).
investigation of heating due to magnetic fluid
33 Semple SC, Chonn A, Cullis PR. Influence of 44 Luo S, Zhang E, Su Y, Cheng T, Shi C.
hyperthermia in a tumor with blood perfusion.
cholesterol on the association of plasma proteins A review of NIR dyes in cancer targeting and
J. Magn. Magn. Mater. 323(6), 708–716
wit liposomes. Biochemistry 35, 2521–2525 imaging. Biomaterials 32, 7127–7138 (2011).
(2011).
(1996). 45 Links M, Brown R. Clinical relevance of the
20 Kuznetsov AA, Leontiev VG, Brukvin VA et al.
34 Gabizon A, Chemla M, Tzemach D, Horowitz molecular mechanisms of resistance to anti-
Local radiofrequency-induced hyperthermia
AT, Goren D. Nanovesicle longevity and cancer drugs. Expert Rev. Mol. Med. 1999,
using CuNi nanoparticles with therapeutically
stability in circulation: effects on the in vivo 1–21 (1999).
suitable Curie temperature. J. Magn. Magn.
Mater. 311(1), 197–203 (2007). delivery to tumors and therapeutic efficacy of 46 Krishna R, Mayer L. Applications of
encapsulated anthracyclines. J. Drug Targeting nanovesicle technology to overcome
21 Pollert E, Knízek K, Marysko M, Kaspar P,
3, 391–398 (1996). multidrug resistance in solid tumors.
Vasseur S, Duguet E. New Tc-tuned magnetic
35 Parr MJ, Masin D, Cullis PR, Bally MB. In: Targeting of Drugs 6: Strategies for Stealth
nanoparticles for self-controlled hyperthermia.
Accumulation of liposomal lipid and Therapeutic Systems (NATO ASI Series).
J. Magn. Magn. Mater. 316 (2), 122–125
encapsulated doxorubicin in murine Lewis lung Gregoriadis G, McCormack B (Eds). Plenum
(2007).
carcinoma: the lack of beneficial effects by Press, NY, USA, 95–108 (1998).
22 Shlyakhtin OA, Leontiev VG, Oh Y, Kuznetsov
coating nanovesicles with poly(ethylene glycol). 47 Prasad NK, Rathinasamy K, Panda D,
AA. New manganite-based mediators for self-
J. Pharmacol. Exp. Ther. 280, 1319–1327 Bahadur D. Mechanism of cell death induced
controlled magnetic heating. Smart Mater.
(1997). by magnetic hyperthermia with nanoparticles
Struct. 16(5), N35–N39 (2007).
36 Mayer LD, Cullis PR, Bally MB. Designing of g-MnxFe2-xO3 synthesized by a single step
23 Prasad N, Rathinasamy K, Panda D, process. J. Mater. Chem. 17, 5042–5051
therapeutically optimized liposomal anticancer
Bahadur D. Tc-tuned biocompatible suspension (2007).
delivery systems: lessons from conventional
of La0.73Sr0.27MnO3 for magnetic hyperthermia.
nanovesicles. In: Medical Applications of 48 Ito A, Fujioka M, Yoshida T et al.
J. Biomed. Mater. Res. B Appl. Biomater. 85,
Nanovesicles. Lasic DD, Papahadjopoulos D 4-S-cysteaminylphenol-loaded magnetite
409–416 (2008).
(Eds). Elsevier, Amsterdam, The Netherlands, cationic nanovesicles for combination therapy
24 Rowinsky EK, Donehower RC. Paclitaxel 231–257 (1998). of hyperthermia with chemotherapy against
(Taxol). N. Engl. J. Med. 332, 1004–1014 malignant melanoma. Cancer Sci.
37 Hong RL, Huang CJ, Tseng YL et al. Direct
(1995). 98, 424–430 (2007).
comparison of liposomal doxorubicin with or

doi:10.2217/NNM.13.90 Nanomedicine (Epub ahead of print) future science group

S-ar putea să vă placă și