ArticlePDF Available

Abstract and Figures

Introduction To date, numerous iron-based nanostructures have been designed for cancer therapy applications. Although some of them were promising for clinical applications, few efforts have been made to maximize the therapeutic index of these carriers. Herein, PEGylated silica-coated iron oxide nanoparticles (PS-IONs) were introduced as multipurpose stimuli-responsive co-delivery nanocarriers for a combination of dual-drug chemotherapy and photothermal therapy. Methods Superparamagnetic iron oxide nanoparticles were synthesized via the sonochemical method and coated by a thin layer of silica. The nanostructures were then further modified with a layer of di-carboxylate polyethylene glycol (6 kDa) and carboxylate-methoxy polyethylene glycol (6 kDa) to improve their stability, biocompatibility, and drug loading capability. Doxorubicin (DOX) and cisplatin (CDDP) were loaded on the PS-IONs through the interactions between the drug molecules and polyethylene glycol. Results The PS-IONs demonstrated excellent cellular uptake, cytocompatibility, and hemocompatibility at the practical dosage. Furthermore, in addition to being an appropriate MRI agent, PS-IONs demonstrated superb photothermal property in 0.5 W/cm² of 808 nm laser irradiation. The release of both drugs was effectively triggered by pH and NIR irradiation. As a result of the intracellular combination chemotherapy and 10 min of safe power laser irradiation, the highest cytotoxicity for iron-based nanocarriers (97.3±0.8%) was achieved. Conclusion The results of this study indicate the great potential of PS-IONs as a multifunctional targeted co-delivery system for cancer theranostic application and the advantage of employing proper combination therapy for cancer eradication.
This content is subject to copyright. Terms and conditions apply.
ORIGINAL RESEARCH
Doxorubicin/Cisplatin-Loaded Superparamagnetic
Nanoparticles As A Stimuli-Responsive Co-Delivery
System For Chemo-Photothermal Therapy
This article was published in the following Dove Press journal:
International Journal of Nanomedicine
Mona Khafaji
1
Masoud Zamani
2
Manouchehr Vossoughi
2,3
Azam Iraji zad
1,4
1
Institute for Nanoscience and
Nanotechnology, Sharif University of
Technology, Tehran 14588-89694, Iran;
2
Institute for Biotechnology and
Environment (IBE), Sharif University of
Technology, Tehran, Iran;
3
Department of
Chemical and Petroleum Engineering, Sharif
University of Technology, Tehran, Iran;
4
Department of Physics, Sharif University of
Technology, Tehran 14588, Iran
Introduction: To date, numerous iron-based nanostructures have been designed for cancer
therapy applications. Although some of them were promising for clinical applications, few
efforts have been made to maximize the therapeutic index of these carriers. Herein,
PEGylated silica-coated iron oxide nanoparticles (PS-IONs) were introduced as multipurpose
stimuli-responsive co-delivery nanocarriers for a combination of dual-drug chemotherapy
and photothermal therapy.
Methods: Superparamagnetic iron oxide nanoparticles were synthesized via the sonochem-
ical method and coated by a thin layer of silica. The nanostructures were then further
modied with a layer of di-carboxylate polyethylene glycol (6 kDa) and carboxylate-meth-
oxy polyethylene glycol (6 kDa) to improve their stability, biocompatibility, and drug loading
capability. Doxorubicin (DOX) and cisplatin (CDDP) were loaded on the PS-IONs through
the interactions between the drug molecules and polyethylene glycol.
Results: The PS-IONs demonstrated excellent cellular uptake, cytocompatibility, and hemo-
compatibility at the practical dosage. Furthermore, in addition to being an appropriate MRI
agent, PS-IONs demonstrated superb photothermal property in 0.5 W/cm
2
of 808 nm laser
irradiation. The release of both drugs was effectively triggered by pH and NIR irradiation. As
a result of the intracellular combination chemotherapy and 10 min of safe power laser
irradiation, the highest cytotoxicity for iron-based nanocarriers (97.3±0.8%) was achieved.
Conclusion: The results of this study indicate the great potential of PS-IONs as a multi-
functional targeted co-delivery system for cancer theranostic application and the advantage
of employing proper combination therapy for cancer eradication.
Keywords: iron oxide nanoparticles, chemo/photothermal therapy, dual-drug delivery,
control release
Introduction
Cancer is the second leading global cause of death that inicts more people every
year as lifestyles change and breast cancer is the most prevalent cancer type among
females, with one in every eight women suffering from it in their lifetime.
1,2
The
current clinical cancer therapies including chemotherapy, radiotherapy, and surgery
lack the desirable effectiveness and are associated with severe side effects.
35
For
these reasons, nding an improved cancer therapy method has been the subject of
extensive research efforts worldwide.
It has been proven that a mixed use of anticancer drugs and/or treatment methods
enhances the treatment efciency by synergistic therapeutic effect and by overcoming
Correspondence: Mona Khafaji
Institute for Nanoscience and
Nanotechnology, Sharif University of
Technology, PO Box 11365-11155, Tehran
14588-89694, Iran
Tel +98-21-66164123
Fax +98-21-66164117
Email mona.khafaji@gmail.com
Manouchehr Vossoughi
Department of Chemical and Petroleum
Engineering, Sharif University of
Technology, PO Box 11365-8639, Tehran,
Iran
Tel +98-21-66164104
Fax +98-21-66005417
Email vosoughi@sharif.edu
International Journal of Nanomedicine Dovepress
open access to scientic and medical research
Open Access Full Text Article
submit your manuscript | www.dovepress.com International Journal of Nanomedicine 2019:14 87698786 8769
http://doi.org/10.2147/IJN.S226254
DovePress © 2019 Khafaji et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
php and incorporate the Creative Commons Attribution Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the
work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
drug resistance.
6,7
Today, clinics practically benetfromthe
enhanced therapeutic effect of radiotherapy and/or dual
drug chemotherapy by two drugs of different action
mechanisms after the surgery has been performed.
Nevertheless, due to the nonspecicity and high dosage of
drugs in these methods, a combined use of them intensies
the side effects and deteriorates the quality of life for
patients.
8,9
Over the last decade, with advancement in
science and technology, scientists have sought a combina-
tion therapy for enhancing the therapeutic index while
reducing side effects. In this regard nanotechnology has
been the most sought after science to which the major
share of research in this eld is dedicated. The unique
properties that some materials represent at the nanoscale
enable them to be used for innovative applications; due to
their very small size and surface properties, some types of
nanoparticles can accumulate in specic tissues or cells and
demonstrate a great potential for application in cancer diag-
nosis and therapy.
4,6,10
The application of nanostructures as
drug carriers
11
and bioimaging
12
and thermal treatment
agents
13
has been studied for more than a decade. Some
of these nanostructures, eg gold
13-15
and iron,
16
can produce
heat as a response to external stimuli such as a magnetic
eld or laser irradiation. By specic surface modication of
these nanoparticles, anticancer drugs can be loaded on them
making them good candidates for a combination chemo and
thermal therapy.
12,17
However, sometimes the high degree
of complexity of the synthesis and modication process
practically limits the applicability of nanostructures.
Among all the investigated nanostructures superpara-
magnetic iron oxide nanoparticles (SPIONs) have attracted
extensive attention in cancer therapy due to their signi-
cant biocompatibility, biodegradability, ease of synthesis
and surface modication, magnetic targeting
18
excellent
contrast for magnetic resonance imaging (MRI)
19
and
application in thermal treatments.
19,20
SPION-based
nanostructures with various modications have been stu-
died as simultaneous drug carriers and hyperthermia
agents.
21,22
However, since their application as photother-
mal agents in comparison to magnetic hyperthermia pro-
vides a better control over the temperature increase it
increases the temperature in the exact target site most
modications on SPIONs in the recent years have been
performed with the goal of combination chemo- and
photothermal-therapy.
17,2325
The primary goal of all the research efforts in this area is
to achieve a more effective therapy with minimized side
effects. Diverse novel nanostructures have been studied but
they could not always improve the therapeutic index.
Indeed, although these new structures have their own mer-
its, the employed design failed to improve the efcacy of
both drug delivery and photothermal therapy. Therefore, the
maximum cytotoxicity was achieved when the amount of
drug was rather signicant or an unsafe laser power was
applied.
2628
For example, incorporation of graphene into
SPION-based nanostructure signicantly enhances the drug
loading content, yet a desirable photothermal cell killing
cannot be achieved unless an intense and unsafe laser power
is employed.
29,30
Not to mention, the biocompatibility of
graphene is still a challenge for the scientic community as
there are numerous contradictory reports on it.
31,32
In order
to take advantage of the surface plasmon resonance proper-
ties of gold in the photothermal therapy, gold nanoshell was
incorporated to SPION-based nanocarriers. However, due to
the weak NIR absorption of spherical gold nanostructures, it
was inevitable that a high-intensity laser power would be
used to achieve a suitable photothermal cell killing.
14,15
Furthermore, preparation of these nanostructures usually
follows a complicated and multi-step synthesis procedure
which practically limits their application. Therefore, the
need for a safe method with an easily synthesizable struc-
ture that could enhance the efciency of combined chemo-
photothermal therapy is well felt.
Herein, for the rst time, we present PEGylated silica-
coated SPIONs (PS-IONs) as dual drug carriers for
Doxorubicin (DOX) and Cisplatin (CDDP), and as photo-
thermal and MRI-contrast agents. By loading two drugs
with different action mechanisms on PS-IONs, it would be
possible to invade the cancer cells by a three-branched
shotgun design, ie targeted drug delivery, dual drug che-
motherapy, and photothermal therapy. The physicochem-
ical properties of the synthesized nanoparticles were
examined by transmission electron microscopy (TEM),
vibrating sample magnetometer (VSM), X-ray diffraction
(XRD), Fourier transform infrared spectroscopy (FT-IR),
dynamic light scattering (DLS), magnetic resonance ima-
ging (MRI), and by exposure to a 808 nm laser beam. The
cytocompatibility and cellular uptake of PS-IONs were
investigated both quantitatively and qualitatively using
broblast and breast cancer cells, respectively. The inu-
ence of PS-IONs on blood coagulation cascade and red
blood cells was also studied. Furthermore, the perfor-
mance of PS-IONs as a controlled release system was
measured by in vitro simulating the photothermal therapy
and acidic cancerous condition. Finally, the effectiveness
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8770
of combined therapy on the viability of breast cancer cells
was investigated.
Materials And Methods
Materials
Iron (III) chloride hexahydrate (FeCl
3
.6H
2
O), iron (II) chlor-
ide tetrahydrate (FeCl
2
.4H
2
O), chromium trioxide (CrO
3
),
polyethylene glycol (PEG, Mw 6 kDa), methoxypolyethylene
glycol (m-PEG, Mw 6 kDa), ammonia (NH
3
, 25%), tetraethy-
lorthosilicate (TEOS), potassium chloride (KCl), disodium
hydrogen phosphate (Na
2
HPO
4
), potassium dihydrogen phos-
phate (KH
2
PO
4
), sodium chloride (NaCl), ethylenediaminete-
traacetic acid (EDTA), acetone, and isopropanol were
purchased from Merck. Cis-diammineplatinum (II) dichloride
(CDDP), (3-aminopropyl) trimethoxysilane (APTMS), N-(3-
dimethylaminopropyl)-Hydroxysuccinimide (NHS), trypsin,
penicillin-streptomycin, and 3-(4, 5-dimethylthiazol-2-yl)-2,
5-diphenyltetrazolium bromide (MTT) were obtained from
Sigma-Aldrich.
The dicarboxylate-PEG and carboxylated m-PEG were
achieved from oxidation of PEG and m-PEG using Jones
reagent as described by Lele.
13,33
Hydrochloric acid (HCl, 37%) and methanol were pur-
chased from Chemlab (Belgium). Doxorubicin hydrochlor-
ide (DOX) was obtained from Pzer (Perth, Australia).
Fetal bovine serum (FBS), RPMI 1640 and high glucose
DMEM cell culture mediums were purchased from
Thermo Fisher Scientic (Gibco, USA).
All chemicals were of analytical grade and used as
received without further purications. Deionized (DI)
water with a resistivity of 18.2 MΩand double distilled
water were used for the preparation of all chemical and
biological samples, respectively.
Synthesis Of PS-IONs
The sonochemical method was used to synthesize iron
oxide nanoparticles (IONs) in order to achieve smaller
particles with a narrow size distribution.
34
Briey,
FeCl
2
.4H
2
O (0.002 moles) and FeCl
3
.6H
2
O (0.004
moles) were dissolved in 2 M HCl and titrated with 4%
ammonia solution at 3032°C in the ultrasonic bath for
1 hr. The produced particles were immediately rinsed
several times with methanol to neutralize the pH and
were dispersed in 50 mL of absolute ethanol.
In the next step, IONs were coated by silica according
to Stöbers method.
34
30 mL of the just-prepared IONs
suspension was alkalinized to pH 810 using diluted
ammonia solution and was put in an ultrasonic bath at
3032°C. After 15 min, 1.8 mL of TEOS was added to
the suspension. After 1 hr, 25 µL of APTMS was added to
the reaction mixture to cease the silica coating process and
also to functionalize the surface with amine groups.
Finally, the as-prepared core-shell nanostructures were
separated from the solution by the use of an external magnetic
eld and rinsed three times with ultrapure water and was
diluted to 30 mL. The structures were then further modied
with a layer of dicarboxylate PEG (6 kDa) and carboxylated-
methoxypolyethylene glycol (6 kDa) to improve their biocom-
patibility and drug loading capability. To this end, 1 mL of the
silica-coated SPIONs suspension was diluted 5 times and was
sonicated for 5 min. Then a diluted solution of an extra amount
of dicarboxylate PEG (18 mg) and m-PEG (36 mg) was added
to the silica-coated iron oxide nanoparticles and stirred with an
equivalent amount of EDC (1.7 mg)/NHS (1 mg) for 2 hrs.
The nal product was rinsed three times and kept as a suspen-
sion in ethanol for future use.
Characterization Of The PS-IONs
Furier transform infrared spectra were recorded on an ABB
Bomem MB-100 spectrometer in the range of 4004000 cm
1
.
Magnetic properties of Fe
3
O
4
nanoparticles (IONs) and PS-
IONs were evaluated by MDK Kawir Magnetic vibrating
sample magnetometery. The applied magnetic eld was in
the range of 0 to 9 kOe. The morphology of PS-IONs was
observed by transmission electron microscopy (Zeiss EM10C,
80 kV). Crystallographic information and mean particle size of
samples were obtained from X-ray diffraction. A P Analytical
XPert PRO MPD (The Netherlands) X-ray diffractometer
with CuKα-irradiation (λ= 1.5406 A, 10<2θ<80)was
used to measure the X-ray diffraction patterns.
The platinum concentration was quantied using
inductively coupled plasma optical emission spectroscopy
(ICP-OES, SPECTRO ARCOS). The uorescence spectra
of doxorubicin were collected on a uorescence spectro-
photometer (Varian, Cary Eclipse) at room temperature.
Magnetic resonance images were recorded using a 3 T
clinical MRI scanner (Magnetotrio, Siemens). The relax-
ivity was determined using spin-echo acquisition utilizing
a repetition time (TR) of 3000 ms and 32 echo-times (TE)
ranging from 12 to 384 ms where the eld of view was
7 cm, slice thickness was 3 mm and acquisition matrix was
256×128. The transverse relaxation time (T
2
) of water
protons was obtained by tting a logarithmic curve to the
mean of the measured MR signals. By linear least square
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8771
tting of 1/T
2
(s
1
) versus iron concentration (mM) the
transverse relaxivity, r
2,
was calculated.
All the samples were irradiated by a Hi-Tech
Optoelectronic (China) optic ber coupled continuous wave
diode laser at 808±5 nm (0.5 W/cm
2
).
In Vitro Cellular Assay
Cell Culture
Human breast cancer cells (MCF7; National Cell Bank of
Iran) and a mouse broblast cell line (L929; National Cell
Bank of Iran) were used as a model of cancer and normal
cells to evaluate the cytotoxicity of PS-IONs. The MCF7
and L929 cells were cultured in high glucose DMEM and
RPMI mediums contained 10% FBS and 1% penicillin/
streptomycin, respectively. The cultures were incubated at
37°C in a humidied atmosphere of 5% CO
2
.The mediums
were replaced every other day and upon 70% conuency,
the cultures were subcultured.
Cell Viability Assay
MTT assay was carried out to determine the viability of
normal broblast cells after one, two, and three days of
cell culture in the presence of different amounts of PS-
IONs. Briey, 1 mL of cell suspension (50,000 cells/mL)
were plated in plasma treated 24-well plates and cultured
for 24 hrs to allow them to attach, and then exposed to a
serial concentration of PS-IONs in nine different groups
(10 to 500 µg/mL) and further incubated at 37°C in a
humidied atmosphere of 5% CO
2
. The cell viability was
measured using MTT assay standard protocol.
13
The per-
cent of cell viability was calculated as follows:
% cell viability ¼
absorbance of samples
absorbance of blank
absorbance of control
absorbance of blank
100 (1)
MTT assay was also performed in order to compare the
cytotoxicity effect of CDDP and DOX with that of PS-
IONs and drug-loaded PS-IONs on MCF7 human breast
cancer cells. The laser effect was also evaluated on each
group and results were presented as the average records of
three replications. Briey, the one-day cultured cells (33
wells, 50,000 cells/well) were divided into eleven groups
and exposed to a dened concentration of the drugs and/or
PS-IONs, as presented in Table 1. After overnight incuba-
tion, in order to study the photothermal therapy effective-
ness of PS-IONs, samples of ve groups were exposed to a
laser beam (0.5 W/cm
2
, 10 min) and further incubated
overnight. The viability of the cells in each group was
measured using MTT standard protocol at the end of the
third day.
Cellular Uptake
An elemental analysis method was used to quantitatively
examine the cellular uptake of PS-IONs. To do so,
MCF7 cells (50,000 cell/well) were seeded in 24-well
plates and cultured overnight. Then, the culture medium
was replaced with a new medium containing a series of
PS-IONs concentrations followed by incubation at 37C
for 24 hrs. After rinsing with phosphate buffer saline
(PBS) solution (1 mL×3), the cells were detached using
trypsin solution and collected in separate vials. The cells
were then disintegrated using a high concentration nitric
acid at 70°C to release the PS-IONs in the solution for
elemental measurement by ICP-OES. Non-treated cells
were used as blank and results were averaged from three
replications.
In Vitro Hemocompatibility Assay
APTT and PT assay: Prothrombin time (PT) and activated
partial thromboplastin time (APTT) were applied to evaluate
the thrombogenic activities of PS-IONs. Briey, a speci c
amount of 2 mg/mL PS-IONs suspension in PBS was mixed
with the human blood plasma which was drawn from healthy
adult volunteer and mixed with 3.2% sodium citrate as an
anticoagulation agent to obtain the nal concentration of
200 μg/mL of PS-IONs and incubated for 30 min at 37°C.
Afterward, to 0.1 mL plasma solution, the specicreagentsof
PT and APTT test were added and left in 37°C for 3 min.
Finally, using a photo-optical clot detection instrument
(Coatron M1, TECO, Germany), the clotting times were
recorded.
Peripheral blood smear test: To obtain a suspension of
200 μg/mL of PS-IONs, a specic amount of 2 mg/mL PS-
IONs suspension in PBS was mixed with fresh blood
(EDTA-anticoagulated) and was shacked continuously for
10 mins. Then in 40 min time intervals a drop of the blood
sample was spread on a glass coverslip followed by xa-
tion with methanol at room temperature then the blood
cells were dyed using Giemsa stain.
It should be mentioned that the use of relevant human
material was approved by Pasteur Institute of Iran ethics
committee. For all the investigations involving human
subjects (cellular and blood experiments), a written
informed consent has been obtained from the involved
participants.
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8772
Drug Loading And Release
In order to load the drugs on the nanoparticles, 30 mg of
PS-IONs was dispersed in 30 mL of deionized water.
Different amounts of DOX and CDDP were added to the
suspension to reach the nal concentration of 1540 ppm
for each of them and the mixture was shaken for 72 h at
room temperature. Then, the drug-loaded nanoparticles
were separated using an external magnetic eld and rinsed
several times with deionized water to remove the unat-
tached drugs. All supernatants containing the unloaded
DOX and/or CDDP were collected. The amount of
unloaded DOX was determined by measuring uorescence
emission at 592 nm (excitation at 485 nm). The amount of
unloaded CDDP was calculated by measuring the platinum
content using an elemental analysis (ICPOES) relative to
a calibration curve obtained under identical conditions.
The drug loading content (DLC) and drug loading ef-
ciency (DLE) were calculated according to equations 2
and 3, respectively.
DLC ¼WD=Wn
ðÞ100 (2)
DLE ¼WtWu
Wt

100 (3)
where the W
D
,W
n
,W
u
, and W
t
are weights of the loaded
drug, nanoparticles, unloaded drug, and initially added
drug, respectively.
To study the release prole of DOX and CDDP from
the nanocarriers, a dialysis process was performed. All
release studies were performed under sink conditions
(amount of drug <10% of the solubility). To this end, 1
mg of drug-loaded nanoparticles were dispersed in 5 mL
of PBS and dialyzed against 30 mL of PBS using a 10 kDa
dialysis bag (Sigma) in two different pH values of 7.4 and
5.5 at 37°C under constant shaking at 150 rpm. In order to
simulate the photothermal condition, a separate group of
each pH value was subjected to a 30 min temperature
increase to 42°C. At predetermined time intervals, 5 mL
of the solution outside the dialysis bag was withdrawn for
analysis and replaced with the same volume of fresh PBS
solution.
Statistical Analysis
Statistical package for social science (SPSS v. 25.0) soft-
ware was used for statistical analysis of all the experi-
ments. Multiple samples were analyzed by one-way
ANOVA. To compare the control samples with the treated
ones, the StudentNewmanKeuls post hoc test was done.
Measures are reported as means ± standard deviations and
statistically signicant difference was reported at p < 0.05.
The XRD data were analyzed using XPert HighScore
Pluse software. All the graphs were plotted by Microsoft
Excel 2010 software.
Results And Discussion
Synthesis Of PS-IONs
Iron oxide nanoparticles (IONs) were rst synthesized
through the sonochemical method and then coated with a
silica shell to improve their biocompatibility, thermal sta-
bility, and dispersibility in aqueous solutions.
34
This rela-
tively simple method also enables the functionalization of
the particlessurface for further applications. APTMS was
used to both halt the increase-in-thickness of the silica
shell on the IONs and to functionalize the surface of silica
coated-IONs with amine groups. The zeta potential of the
NH
2
modied silica-coated IONs was 27.7 mV. The pre-
sence of amine groups on the surface not only enhances
the stability of particles due to their positive charge but
Table 1 Characteristics Of Eleven Groups For Cytotoxicity Assessment By MCF-7 Breast Cancer Cells
Group No. Control DOX (4.2 µg/mL) CDDP (2.8 µg/mL) PS-IONs (200 µg/mL) Drug-Loaded PS-IONs Laser
1
2
3
4
5
6
7✓✓
8
9✓✓
10
11 ✓✓
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8773
also enables the attachment of PEG molecules from their
carboxylic acid ends.
After preparation of the silica-coated IONs, their sur-
faces were modied with PEG to increase their circulation
time in the bloodstream and enhance their drug loading
ability for targeted drug delivery application. After pegy-
lation, the zeta potential of nanoparticles decreased to
2.27 mV which conrms the presence of the COOH
groups on the surface of PS-IONs instead of NH
2
ones.
The detailed procedure for the preparation of the PS-IONs
and the drug loading on them is illustrated in Scheme 1.
Figure 1a and bshow the TEM images of the PS-IONs.
It could be observed that the nanoparticles were spherical
in shape with a narrow size distribution and an average
size of 20±3 nm. The IONs could be distinguished from
the silica through the difference in their contrasts.
Scheme 1 Illustration of the synthesis procedure of PS-IONs and drug loading mechanism.
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8774
According to TEM images, the silica shell was thin
enough to limit the adverse effect on magnetization satura-
tion. The severe aggregations of the PS-IONs were pre-
sumably due to the hydrogen bonding of PEG molecules
among the nanoparticles.
X-ray diffraction (XRD) experiments were also per-
formed to investigate the crystalline structure of IONs and
PS-IONs (Figure 1c). The characteristic diffraction peaks in
both spectra could be assigned to the (220), (311), (400),
(422), (511) and (440) planes of the spinel structure of
magnetite (Fe
3
O
4
) according to the JCPDS 190629.
Furthermore, the broad peak at around 2θ= 20° indicates
the presence of amorphous silica in the synthesized
nanoparticles.
35
The Scherer equation was employed to cal-
culate the average crystallite size from the (311) peak in each
spectrum. The calculated particle size for both samples was
15 nm which is in agreement with the observations of the
TEM images indicating that the silica coating process does
not induce major unfavorable effects such as dissolution or
growth of the crystals on the Fe
3
O
4
nanocrystals.
Considering that the size of PS-IONs was less than a
magnetic domain for these components (25 nm for Fe
3
O
4
),
it was expected that they possess a superparamagnetic
behavior.
34
With this in mind, VSM analysis was performed
on PS-IONs to evaluate their magnetic behavior (Figure 1d).
The results show that the magnetization saturations of the
IONs and PS-IONs were 66.6 emug
1
and 16.75 emug
1
,
respectively. These data were consistent with the previous
observations which indicate that silica coating decreases the
magnetization saturation of IONs by at least four folds.
36
The
magnetic residue of both IONs and silica-coated ones were
4mT, hence both samples were classied as superparamag-
netic nanoparticles.
34
The superparamagnetic behavior in
silica-coated particles possibly results from bipolar-bipolar
interactions between the magnetic cores. Therefore, since the
bipolar-bipolar magnetic interactions had a reverse relation-
ship with the distance, the interactions were intensied as a
result of theentrapment ofmore than one ION in a silica shell
and/or the physical attachment by the hydrogen bonding of
PEG molecules.
19,34
FT-IR spectroscopy was employed to verify the forma-
tion of the silica shell surrounding the Fe
3
O
4
nanoparticles
and also to investigate the success of the PEGylation
process. Figure 1e shows the IR spectra of IONs (black),
Figure 1 (a, b) TEM images of PS-IONs (scale bars are 150 nm and 40 nm, respectively). (c) XRD patterns [the yellow, green, pink, blue, violet, light brown and orange
squares relate to (220), (311), (400), (422), (511), (440) and (530) planes of the spinel structure of magnetite, respectively], (d) hysteresis loops, (e) FT-IR spectra, and (f)
DLS particles size distribution proles of IONs (black line) and PS-IONs (red line).
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8775
PS-IONs (red), and PEG (blue). The presence of silica,
magnetite, and PEG in the PS-IONs was clearly shown by
the observation of the characteristic bands of all three,
magnetite (560, 1400 cm
1
, Fe-O band vibration), silica
(1080 cm
1
asymmetric Si-O-Si stretching), and PEG
(2925 cm
1
CH
2
stretching vibration, 1150 cm
1
C-O-C
stretching), in the PS-IONsspectrum (red line). There was
also an evidence of bonds formation between silica and
Fe
3
O
4
(1190 cm
1
, Fe-O-Si vibration) conrming the for-
mation of the desired structure. Unlike the IONs, dissol-
ving the PS-IONs even in hot hydrochloride acid (37%
w/v) was not possible except after prewashing in a hydro-
uoric acid to remove the silica coating, endorsing the full
coverage of silica on the nanoparticles.
The hydrodynamic size of the nanoparticles in aqueous
solution was determined by DLS (Figure 1f). The results
showed a single peak with a small polydispersity index
which points to the negligible aggregation of PS-IONs in
aqueous solution. The average diameters in a hydrated state
were 58.8 (PDI=0.171) and 122 nm (PDI=0.156) for IONs
and PS-IONs, respectively. The measured size of IONs by
the DLS method was signicantly higher compared to the
TEM measurements. According to previous reports, this may
be occurred due to the fast occulation of bare magnetic
nanoparticles, the presence of water molecules on their sur-
face, and/or the magnetic dipole-dipole attraction between
magnetic nanoparticles.
3739
Also, the measured size for PS-
IONs was approximately six times higher than that measured
by TEM. This increase may be due to the fact that more than
one ION was trapped in the same silica coat and/or irrever-
sible aggregates formed through thePEG bridges. In addition
the hydrophilic PEG chains were extended in the aqueous
solution or/and dynamic association in the liquid.
8,34
In view
of the fact that the nanocarrierssize plays an important role
in their biodistribution and accumulation in the organs and
cells, the synthesized PS-IONs are absolutely ideal as nano-
carriers for cancer therapy applications.
In Vitro Cytocompatibility
The most important characteristic of a nanocarrier in a given
drug delivery application is its negligible toxicity to normal
cells. The cytotoxic effects of PS-IONs on L929 broblast
cells were evaluated by conducting a MTT assay. Figure 2
shows that the presence of nanoparticles did not induce any
signicant detrimental effect on the viability of normal cells at
24, 48, and 72 h time points. The viability of cells was more
than 88±5.2% even after exposure to 500 μg/mL of nanopar-
ticles for 72 h, endorsing the success of the employed surface
modication in the improvement of the nanoparticles
biocompatibility.
40
Unlike some of the previous observations,
the increase in the cell density in comparison to the control in
some samples is attributed to their metabolic changes after
being exposed to nanoparticles rather than the dissolution of
IONs.
19
Light microscopy images of the cells exposed to
different concentrations of PS-IONs for dened times indi-
cated that the presence of the nanoparticles in the culture
medium had no adverse effect on the attachment and morphol-
ogy of the cells (Figure S1). This signicant improvement in
the biocompatibility was attributed to both the silica coating
and the PEGylation which makes the prepared particles sui-
table candidates for a drug delivery application.
Cellular Uptake
It is essential for the nanoparticles to internalize into target cells
in the highest amount in order to be considered as effective
drug delivery vehicles. The cellular uptake of PS-IONs by
MCF7 breast cancer cells was studied both quantitatively and
qualitatively. According to Figure 3a, the PS-IONs could be
obviously visualized in the cytoplasm of the cells as brown
spots indicating that their accumulation in the cells did not
affect the cellsnormal morphology. In addition, the cumula-
tive cellular uptake of PS-IONs was linearly increased as their
concentration in the medium increased up to 200 µg/mL. The
percentage of the uptaked PS-IONs increased linearly at low
Figure 2 Viability of L929 normalbroblast cells after 24, 48, and 72 hrs of incubation
with different concentrations of PS-IONs (ranging from 10 to 500 µg/mL). There was
no signicant difference between the control and other groups.
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8776
Figure 3 (a) Light microscopy images of the cells (the total magnication was 400 times) exposed to 100 (left) and 200 µg/mL (right) of PS-IONs for 24 hrs. The brown dots
show the uptaken nanoparticles. (b) Concentration dependent cellular uptake percentage. The inset shows the amount of PS-IONs uptake (µg/mL) versus initial PS-IONs
concentration (µg/mL) in the culture media.
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8777
concentrations (up to 50 µg/mL), while further increase of the
PS-IONs concentration in the culture medium caused their
uptake rate to decelerate so that for concentrations above 100
µg/mL the uptake percentage declined indicating the cells were
approaching their saturated uptake capacity (Figure 3b). This
excellent cellular uptake was related not only to the optimum
particle size and the spherical morphology of PS-IONs but also
to their surface characteristics. By altering the serum protein
adsorption on the nanoparticles surface, the coating affects the
nanoparticles-cells interactions and thus improved the cellular
uptake.
41,42
So, the presence of PEG on the particles surface
improved the cellular uptake and viability through prevention
of the attachment of proteins to their surface.
43,44
In Vitro Hemocompatibility
Another determinant factor in the clinical applicability of
nanocarriers is their means of interaction with the blood
cells. A proper drug delivery system should not induce any
adverse effect on the morphology and activity of the blood
cells.
45
Accordingly, the hemocompatibility of the as-
synthesized nanoparticles was investigated by studying
their effect on blood coagulation and the morphology of
red blood cells (RBCs).
13,46,47
The coagulation times in
the presence of PBS (control) and 200 μg/mL of PS-IONs
were investigated. The results showed that the PT and
APTT have not been affected in the presence of such a
high concentration of nanoparticles and still remained in
the normal reference range (Table 2).
Normally RBCs have a biconcave disk shape which
easily changes in interaction with foreign substances.
13,46
The investigation of RBCs morphology, when exposed to a
foreign material, is a routine method to evaluate the new
biomaterials hemocompatibility. Hence, the peripheral
blood smear test was performed to study the inuence of
the PS-IONs on the shape of RBCs. It can be observed from
Figure 4 and Figure S2 that the RBCs had undergone
neither deformation nor aggregation after exposure to the
200 µg/mL of PS-IONs even after 4 hrs of incubation when
compared to the controls. This excellent hemocompatibility
was attributed to the negligible electrostatic interaction
between negatively charged RBCs and PEGylated nanopar-
ticles, resulting from the presence of non-reactive methoxy
groups on their surface.
In Vitro Drug Loading And Release
The ability of nanoparticles to carry and deliver a suf-
cient amount of anticancer drug to the tumor tissues is of
great importance. Among the various anticancer drugs,
CDDP and DOX are widely used for solid cancerous
tumor treatment, therefore loading each of them on nano-
carriers has been extensively investigated.
48-50
As the var-
ious anticancer drugs have different action mechanisms, it
has been proven that cancerous cells could be killed more
effectively by the use of dual-drug chemotherapy.
8,9
Therefore, the potency of the PS-IONs was examined as
magnetically targeted dual-drug carriers by loading both
DOX and CDDP on them. The loading efciency of DOX
in the presence of a constant amount of CDDP (15 ppm)
was investigated. As shown in Figure 5a the DLC was
raised by increasing the initial DOX concentration. At the
optimum conditions (30 ppm DOX), the obtained DLE
was 69.3±1.4% with DLC of 21±0.42 µg DOX/mg PS-
IONs. At the normal pH, DOX has a positive charge while
the carboxyl groups on the particles surface were nega-
tively charged.
51
Therefore, DOX could be efciently
loaded into PEG chains through electrostatic interactions,
as well as the formation of amide and hydrogen bonds.
11,22
To investigate the loading of CDDP on the nanoparti-
cles, different concentrations of CDDP were added to the
solutions containing constant amounts of DOX loaded PS-
IONs (1 mg/mL) and DOX (15 ppm) (Figure 5b). The
amount of loaded CDDP obtained in the optimum condi-
tion (30 ppm CDDP) was 14±0.4 μg CDDP/mg PS-IONs
with a DLE of 46.6±1.4%, resulting in a total DLC of
3.5% w/w. In the absence of DOX, the DLC of CDDP was
increased by 100% while the DLE remained unchanged.
Hence, the DLE of CDDP is not a function of its initial
concentration. It can be concluded that the molecules of
both drugs compete against each other to seize the avail-
able carboxyl groups on the surface of the particles. So,
the higher DLC of DOX in comparison to CDDP is attrib-
uted to the higher reaction rate of hydrogen or electrostatic
bonding of DOX rather than coordination bonding of
CDDP with the carboxyl groups of the particlessurface.
After the drug loading, the zeta potential of PS-IONs was
changed positively to reach +1.25 mV. According to pre-
vious reports, attachment of CDDP and DOX slightly
Table 2 Blood Coagulation Times After Dilution With The Same
Volumes Of PBS And PS-IONs Suspension (nal Concentration
Of 200 μg/mL)
Normal
Range
Diluted
With PBS
Diluted With PS-
IONs (200 µg/mL)
APTT (s) 28 to 38 39.0 38.0
PT (s) 11 to 14 13.0 13.2
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8778
increase the zeta potential. So the zeta potential changes
conrmed the successful drug loading.
52,53
IR spectra of
the drug-loaded PS-IONs were also employed to conrm
the presence of both drugs on them. As it can be seen in
Figure S3, the characteristic peaks of both DOX (3512,
632 cm
1
) and CDDP (3296, 1319 cm
1
) were presented
in the spectrum of the drug-loaded PS-IONs, demonstrat-
ing the reliability of the loading method.
Having a controlled drug release in the body and tumor
tissues is an indispensable characteristic of a drug delivery
system for cancer therapy. Minimizing the drug release in
the normal tissues not only reduces the side effects of the
drugs, it also maximizes the drug dosage at the tumor site
as well, resulting in a more efcient and favorable treat-
ment. To study the in vivo release behavior of drugs
from the as-synthesized PS-IONs, the pH conditions of
normal and cancerous tissues were simulated in vitro.
Furthermore, the effect of photothermal treatment using
nanoparticles was investigated by imposing an initial tem-
perature increase to 42°C for 30 min. As shown in
Figure 5c only about 23±2.2% of DOX released from the
PS-IONs after 24 h at the physiological condition, while
the release reached to 41±2.7% and 45±2.6% by decreas-
ing the pH to 5.5 and by applying the photothermal con-
dition, respectively. This indicates that both the
temperature increase and the pH decrease stimulate the
drug release. Applying the photothermal and acidic pH
conditions simultaneously increased the release rate three
folds. This pH- and thermo-responsive drug release could
be explained according to the drug-PEG interactions. The
interaction intensity between DOX molecules (pK
a
= 8.2)
and PEG chains weakens when decreasing the pH of the
media from neutral or weak alkaline to acidic condition.
Therefore DOX could remain loaded on the nanoparticles
during the blood circulation and could escape from them
more easily within the tumor cells. Similarly, through
increasing the systems energy level, the temperature rise
has the same effect on the interaction intensity between
DOX and PEG.
11,22
As Figure 5D shows, the CDDP release was more
temperature-dependent than pH-dependent. As mentioned
before, CDDP molecules were loaded through the coordi-
nation of interactions between the hydrolyzed drug mole-
cules and the terminated carboxyl groups of PEG. By
decreasing the pH, the terminated carboxyl groups were
protonated which attenuated the interaction between the
drug molecules and the PS-IONs. In addition, according to
the equilibrium hydrolysis reaction of CDDP (Figure S4)
in acidic conditions and in presence of excess amount of
chlorine ions the rate of mono- and dichloro-cisplatin
formation increases which results in a faster release of
drug molecules. Moreover, through supplying the coordi-
nation dissociation energy, higher temperature leads to an
increase in CDDP release rate by more than two folds.
54,55
Applying the photothermal treatment and acidic pH con-
ditions simultaneously increased the release rate by 2.7
folds.
The release rate of CDDP in all situations was more
than that of DOX (Figure S5) which indicated the differ-
ence in drug-PS-IONs interactions. Unlike the CDDP, the
DOX molecules could entrap between the PEG chains by
hydrogen bonding in addition to the terminal attachment
which resulted in the higher DLC.
11,18
This was a reason for
the slower release of DOX, because some of the DOX
molecules had to diffuse through the PEG chains in order
to enter the solution while the terminally-loaded CDDP
molecules could directly be released into the solution.
These data indicate that the designed dual stimuli-triggered
Figure 4 Light microscopy images (the total magnication was 400 times) of blood smear prepared samples from EDTA-anticoagulated blood without dilution (control) and
after addition of PS-IONs (nal concentration 200 μg/mL) and PBS after 4 h. The RBCs showed neither deformation nor aggregation when compared to the controls.
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8779
drug release system provides a favorable drug delivery
platform for combination with photothermal therapy.
PS-IONs As MRI Contrast Agent
The ability of PS-IONs to serve as MRI contrast agents
was investigated using a 3T clinical instrument. The
recorded images showed proton relaxation enhancement
compared to the deionized water control (Figure 6a). The
calculated r
2
for the PS-IONs was 127.31 mM
1
s
1
(Figure 6b) which is comparable with that obtained for
PEGylated or silica coated IONs.
40,56
The obtained trans-
verse relaxivity was almost twice the amount of its bare
IONs.
57,58
This enhancement could be explained according
to the structural characteristics of PS-IONs. As reported
before, the eld gradients generated by magnetized nano-
particles resulted in dephasing of the magnetic moment in
Figure 5 Loading capacity of (a)DOXand(b) CDDP on PS-IONs as a function of their initial concentation in the solutions containing 15 ppmof CDDP and DOX, respectively. In
vitro release proles of (c)DOXand(d) CDDP from PS-IONs at different temperature and pH conditions. Asterisks (*) indicate signicant difference (P < 0.05).
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8780
waters protons. Therefore magnetic nanoparticles induced
spin-spin relaxation as the proton passes through such eld
gradients. By increasing the iron concentration, the mag-
netic moment becomes stronger resulting in distortion of
spin coherence of water protons in transverse relaxation.
56
The transverse relaxivity is related to the hydrody-
namic diameter of the aggregated nanostructure and the
magnetic saturation. Although the magnetic saturation of
PS-IONs was reduced comparing to its initial value, the
signicant increase in the PS-IONs hydrodynamic dia-
meter was the leading factor in the MRI contrast enhance-
ment. The PEGylated nanoparticles could make small
reversible aggregates through the hydrogen bonding.
Formation of these cluster-like structures not only
increased the effective hydrodynamic diameter of nano-
particles several times but also shortened the distance
among magnetic particles which, in turn, induced a more
inhomogeneous local magnetic eld. Finally, improved
hydrophilicity of PEGylated nanoparticles facilitated the
diffusion of water molecules near the surface of magnetic
particles, immobilizes them by hydrogen bonding, and
extended the interaction between the magnetic eld and
protons.
56,59
Cytotoxicity Effect Of Multiple Treatment
Method
The efciency of the multiple-treatment-method was stu-
died on MCF7 breast cancer cells in vitro and was com-
pared with individual chemotherapy and photothermal
therapy according to Table 1 . The obtained results are
summarized in Figure 7. As was expected, the sole laser
treatment (0.5 W/cm
2
, 808nm) neither had a signicant
effect on the cellsviability nor exerts an effective interac-
tion with the CDDP and DOX drugs. The viability of the
cells exposed to DOX (4.2 µg/mL) and CDDP (2.8 µg/mL)
for 48 h, were 20.5±4% and 57±5%, respectively. Although
the free drugs have signicantly decreased the cell viability,
these data indicate the inefcacy of the drugs at such low
concentrations. This was the reason for the extensive side
effects of traditional cancer chemotherapy, as it requires
Figure 7 MTT viability assay of MCF7 cells after exposure to 0.5 W/cm
2
near-IR
laser irradiation for 10 min, incubation with 200 μg/mL of PS-IONs, chemotherapy
with DOX (4.2 μg/mL) or CDDP (2.8 μg/mL), dual-drug chemotherapy, photother-
mal treatment, and multiple treatment method. Values are mean±SD. Double
asterisks (**) indicate nonsignicant difference (P>0.05).
Figure 6 (a)T
2
-weighted MR images of PS-IONs in aqueous media at various concentrations and different echo times. (b)T
2
relaxation rate (R
2
) versus iron concentration
in PS-IONs.
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8781
administration of a high dosage of drugs in the whole body
to reach a desirable cytotoxicity for tumor cells.
60
DOX, an anthracycline antibiotic, restrains the DNA
remodeling through intercalating between DNA nucleotides
and inhibiting the activity of topoisomerase II (TOP2).
61
CDDP, a powerful DNA chelating agent, induces cancer
cells apoptosis by covalently bonding with DNA purine
bases to form DNA adducts which could inhibit cellular
transcription and replication.
62
These two drugs have inde-
pendent action mechanisms, and consequently, DOX could
further impede the repairing of CDDP-damaged DNA by
TOP2. Therefore, the combination chemotherapy by DOX
and CDDP could disrupt the cellular repair mechanisms. A
mixed use of cancer drugs with different mechanisms of
action is a clinical method to overcome the cells resistance.
It would also reduce the side effects of treatment by
decreasing the administered dosage and improving the treat-
ment efciency.
8,9,63
As it is shown in Figure 7,dual-drug
chemotherapy had signicantly enhanced the cytotoxicity
so that the cellsviability declined to 10.65±0.9%. This
result conrmed the synergetic effect of chemotherapy
drugs on the cancer cells.
As it was qualitatively shown in Figure 3, PS-IONs had
no unfavorable effect on the cancer cellsmorphology.
They also demonstrated very good biocompatibility
(Figure 2) and excellent cellular uptake. By loading the
drugs on these nanocarriers, they would escape the drug
resistance by entering the cells through endocytosis enhan-
cing the cytotoxicity.
64,65
The results show that although
almost half of each drug were remained loaded on the
nanocarriers (50±3% of CDDP and 45±2.6% of DOX,
according to in vitro simulation), they resulted in the
same cytotoxicity as the free drugs. This was most likely
due to the effective cell membrane penetration and accu-
mulation of the drug-loaded PS-IONs into MCF7 cells by
endocytosis and phagocytosis. Consequently, this phenom-
enon increased the accumulated drugs within the cells
since they would release more rapidly due to the intracel-
lular acidic condition of cancer cells.
66,67
In addition to drug delivery, the PS-IONs show remark-
able ability as photothermal agents. Free PS-IONs demon-
strated superior cytotoxicity over CDDP (2.8 µg/mL) by
reducing the cell viability to 30±4%, after an exposure to
low power laser irradiation (0.5 W/cm
2
, 10 min) which
increased the temperature from 37°C to 42°C. In contrast
to previous studies, a low power laser irradiation, close to
the maximum permissible exposure (MPE) for skin (0.33
W/cm
2
), had been used for photothermal therapy.
6,16,24
By
emitting the NIR light beam to the drug-loaded PS-IONs, a
signicant enhancement in cytotoxicity had been obtained.
The multiple-treatment-method increased the cancer cell
killing efciency to 97.3±0.8% in a single dose therapy.
This result surpasses the best of previously reported out-
comes for SPION-based nanocarriers despite the fact that
very low drug concentration and laser power intensity is
simultaneously employed in our study (Tab le 3). This excel-
lent result was partly attributed to the higher accumulated
free drugs content in the cells as a result of the photothermal
and acidic condition in comparison to the non-photothermal
condition (see Figure 5). Furthermore, besides the photo-
thermal-induced cytotoxicity, the therapeutic index of
CDDP was enhanced by the increase in temperature.
68
Therefore, as a result of cellular internalization, controlled
intracellular drug release, dual drug chemotherapy, and
photothermal therapy, PS-IONs were an outstanding candi-
dates for cancer therapy applications.
Conclusion
The superparamagnetic PS-IONs were prepared by a rela-
tively simple and efcient sonochemical method. The par-
ticles have favorable chemical stability and water
dispersibility and also demonstrated excellent bio- and
hemocompatibility. The SiO
2
and Fe
3
O
4
nanoparticles
were known as in vivo biodegradable nanomaterials with
non-toxic byproducts.
69
The PS-IONs could act as dual-
drug nanocarriers. DOX was loaded on particles by both
hydrogen bonding and electrostatic interactions
11,18,22
with
DLE and DLC of 63.9% and 2.1%, respectively, while
CDDP could efciently conjugate with carboxyl groups
of PEG molecules through coordination interaction result-
ing in DLE and DLC of 46.6% and 1.4%, respectively.
Both drugs had a slow release prole in normal blood
conditions. Only 31% of CDDP and 22% of DOX were
released during 30 h under normal conditions which sig-
nicantly decrease the drug accumulation and toxicity in
the normal tissues. These drug carriers revealed a dual
stimuli-triggered release behavior. High release rates of
69% and 84% were obtained for DOX and CDDP, respec-
tively, during 30 h in a simulated photothermal condition
in an acidic cancerous environment. Synergistic photother-
mal therapy and DOX/CDDP combination chemotherapy
could be achieved through the use of PS-IONs nanocar-
riers. PS-IONs efciently delivered both DOX and CDDP
into the MCF-7 cells and demonstrated potent antitumor
activity in vitro which was signicantly intensied by
exposure to a low power near-IR laser irradiation. As the
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8782
Table 3 Summary Of The Parameters And Outcomes Of Some SPION-Based Chemo-Photothermal Therapy Studies
Nanostructure Concentration
(µg/mL)
Laser [(W/cm
2
);
(λ(nm); t(min)]
Drug;
Concentration
(µg/mL)
Cell
Line
Relaxivity
(mM
1
s
1
)
Cyto-toxicity
(%)
Comments
This study 200 0.5; 808; 10 DOX; 4.2
CDDP; 2.8
MCF-7 127.31 97.3 ± 0.8
rGO-Fe
2
O
3
@Au NPs
30
50 2; 808; 5 DOX; 50 HeLa 54.99 ~96
GO-PEG-γ-Fe
2
O
329
Not reported 2; 808; 5 DOX; 6 HeLa 33.15 ~93 Loading factor was 1.18
Porous hollow Fe
3
O
423
150 µg Fe/mL 1.5; 808; 10 DOX; Not
reported
4T1 119.66 ~75
Lactobionic acid/Fe
3
O
4
@polydopamine/PEG
73
125 1; 808; 30 DOX; ~46 HepG2 Not studied 75.4 Cytotoxicity increased to 83.7% after
exposure to magnetic eld
Fe
3
O
4
@mSiO
2
-PO*-folic acid
26
125 2; 808; 30 DOX; ~5.6 HeLa Not studied 81
Fe
3
O
4
@MnO
2
@PPy
27
600 1; 638; 10 DOX; 420 HepG2 Not studied 91.6 Samples were placed in a magnetic eld
for 30 min
IONPs@AuNPs@SiNWs
15
300 1; 808; 20 DOX; 50 MCF-7/
ADR
Not studied 90 Samples were placed in a magnetic eld
for 3 hrs
Cu
9
S
5
@mSiO
2
@Fe
3
O
4
-PEG
74
200 0.76; 980; 10 DOX; 25 HeLa 13.583 83
Fe
3
O
4
@MoS
2
/PEG/2-deoxy-D-glucose
28
200 0.5; 808; 3 DOX; ~101 MDA-
MB-
231
48.86 ~70 The cell viability decreased to 1.4% after
the second treatment
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8783
drug-loaded nanoparticles can enter the cells via endocy-
tosis, they could overcome the P-gp drug resistance
mechanism. Moreover, only the free drugs were pumped
out by resistance mechanisms of the cells.
64,65,70,71
Also,
the Fe
3
O
4
nanoparticles may act as the P-gp inhibitor in
some drug resistance cancer cells.
72
Therefore, this deliv-
ery system can potentially be used for other combination
therapies and holds a great promise as an effective tool to
treat drug-resistance cancer tumors.
Acknowledgments
The authors gratefully thank Prof. M. Reaz Hormozi-nez-
had from the Department of Chemistry of the Sharif
University of Technology for his help and support, and
Dr. Rabeei and Mr. Shahbazi for doing the blood tests at
Vardavard Medical Laboratory.
Author Contributions
All authors contributed to data analysis, drafting and revising
the article, gave nal approval of the version to be published,
and agree to be accountable for all aspects of the work.
Disclosure
The authors report no conicts of interest in this work.
References
1. Caricati-Neto A, Errante PR, Menezes-Rodrigues FS. Emerging a new
strategy for the antitumor immunotherapy : pharmacological modula-
tion of the Ca 2 +/Camp signaling interaction. Arch Microbiol
Immunol.2017;1(3):8997. doi:doi:10.26502/ami.93650012
2. Desantis CE, Fedewa SA, Sauer AG, Kramer JL, Smith RA, Jemal A.
Breast cancer statistics, 2015 : convergence of incidence rates between
black and white women. A Cancer J Clin.2016;66(1):3142. doi:
doi:10.3322/caac.21320.
3. Prados J, Melguizo C, Ortiz R, et al. Doxorubicin-loaded nanoparti-
cles: new advances in breast cancer therapy. Anticancer Agents Med
Chem.2012;12(9):10581070. doi:doi:10.2174/187152012803529646
4. Huang J, Qian W, Wang L, et al. Functionalized milk-protein-coated
magnetic nanoparticles for MRI-monitored targeted therapy of pancreatic
cancer. Int J Nanomedicine.2016;11:30873099. doi:10.2147/IJN.S92722
5. Meirow D, Nugent D. The effects of radiotherapy and chemotherapy
on female reproduction. Hum Reprod Update.2001;7(6):535543. doi:
doi:10.1093/humupd/7.6.535
6. Espinosa A, Di Corato R, Kolosnjaj-Tabi J, Flaud P, Pellegrino T,
Wilhelm C. Duality of iron oxide nanoparticles in cancer therapy:
amplication of heating efciency by magnetic hyperthermia and
photothermal bimodal treatment. ACS Nano.2016;10(2):24362446.
doi:10.1021/acsnano.5b07249
7. Foglia S, Ledda M, Fioretti D, et al. In vitro biocompatibility study of
sub-5 nm silica-coated magnetic iron oxide uorescent nanoparticles
for potential biomedical application. Sci Rep.2017;7(April):113.
doi:10.1038/srep46513
8. Wu H, Jin H, Wang C, et al. Synergistic cisplatin/doxorubicin combi-
nation chemotherapy for multidrug-resistant cancer via polymeric
nanogels targeting delivery. ACS Appl Mater Interfaces.2017;9
(11):94269436. doi:doi:10.1021/acsami.6b16844
9. Betancourt T, Brown B, Brannon-Peppas L. Doxorubicin-loaded
PLGA nanoparticles by nanoprecipitation: preparation, characteriza-
tion and in vitro evaluation. Nanomedicine.2007;2(2):219232. doi:
doi:10.2217/17435889.2.2.219
10. Liu X, Huang N, Li H, Wang H, Jin Q, Ji J. Multidentate polyethy-
lene glycol modi Fi ed gold nanorods for in vivo near-infrared
photothermal cancer therapy. Appl Mater Interfaces.2014;6:5657
5668. doi:doi:10.1021/am5001823
11. Zhang X, Clime L, Roberge H, et al. pH-triggered doxorubicin
delivery based on hollow nanoporous silica nanoparticles with free-
standing superparamagnetic Fe 3 O 4 cores. J Phys Chem C.
2011;115:14361443. doi:doi:10.1021/jp1075498
12. Pandey S, Thakur M, Mewada A, Anjarlekar D, Mishra N, Sharon M.
Carbon dots functionalized gold nanorod mediated delivery of dox-
orubicin: tri-functional nano-worms for drug delivery, photothermal
therapy and bioimaging. J Mater Chem B.2013;1(38):49724982.
doi:doi:10.1039/c3tb20761g
13. Khafaji M, Vossoughi M, Hormozi-Nezhad MR, Dinarvand R,
Börrnert F, Irajizad A. A new bifunctional hybrid nanostructure as
an active platform for photothermal therapy and MR imaging. Sci
Rep.2016;6. doi:10.1038/srep27847
14. Elbialy NS, Fathy MM, Reem A-W, et al. Multifunctional magnetic-
gold nanoparticles for efcient combined targeted drug delivery and
interstitial photothermal therapy. Int J Pharm.2018;554:256263.
15. Guo D, Ji X, Wang H, et al. Silicon nanowire-based multifunctional
platform for chemo-photothermal synergistic cancer therapy. J Mater
Chem B.2018;6(23):38763883. doi:10.1039/C7TB02907A
16. Sivakumar B, Aswathy RG, Romero-Aburto R, et al. Highly versatile
SPION encapsulated PLGA nanoparticles as photothermal ablators of
cancer cells and as multimodal imaging agents. Biomater Sci.2017;5
(3):432443. doi:doi:10.1039/c6bm00621c
17. Oh Y, Je JY, Moorthy MS, Seo H, Cho WH. pH and NIR-light-respon-
sive magnetic iron oxide nanoparticles for mitochondria-mediated apop-
totic cell death induced by chemo-photothermal therapy. Int J Pharm.
2017;531(1):113. doi:doi:10.1016/j.ijpharm.2017.07.014
18. Liang P, Chen Y-C, Chiang C, et al. Doxorubicin-modied magnetic
nanoparticles as a drug delivery system for magnetic resonance
chemotherapy. Int J Nanomedicine.2016;11:20212037. doi:
doi:10.2147/IJN.S94139
19. Hurley KR, Ring HL, Etheridge M, et al. Predictable heating and
positive MRI contrast from a mesoporous silica-coated iron oxide
nanoparticle. Mol Pharm.2016;13(7):21722183. doi:doi:10.1021/
acs.molpharmaceut.5b00866
20. Yang Y, Liu J, Sun X, et al. Near-infrared light-activated cancer cell
targeting and drug delivery with aptamer-modied nanostructures.
Nano Res.2016;9(1):139148. doi:doi:10.1007/s12274-015-0898-4
21. Kossatz S, Grandke J, Couleaud P, et al. Efcient treatment of breast
cancer xenografts with multifunctionalized iron oxide nanoparticles
combining magnetic hyperthermia and anti-cancer drug delivery.
Breast Cancer Res.2015;17(1):117. doi:10.1186/s13058-015-0576-1
22. Benyettou F, Ocadiz Flores JA, Ravaux F, et al. Mesoporous γ-iron
oxide nanoparticles for magnetically triggered release of doxorubicin
and hyperthermia treatment. Chem - A Eur J.2016;22(47):17020
17028. doi:10.1002/chem.201602956
23. Hu Y, Hu H, Yan J, et al. Multifunctional porous iron oxide nanoagents
for MRI and photothermal/chemo synergistic therapy. Bioconjug Chem.
2018;29(4):12831290. doi:10.1021/acs.bioconjchem.8b00052
24. Lee S, George Thomas R, Ju Moon M, et al. Near-infrared hepta-
methine cyanine based iron oxide nanoparticles for tumor targeted
multimodal imaging and photothermal therapy. Sci Rep.2017;7(1):1
14. doi:doi:10.1038/s41598-017-01108-5
25. Wu M, Zhang D, Zeng Y, Wu L, Liu X, Liu J. Nanocluster of
superparamagnetic iron oxide nanoparticles coated with poly (dopa-
mine) for magnetic eld-targeting, highly sensitive MRI and photo-
thermal cancer therapy. Nanotechnology.2015;26(11):115102. doi:
doi:10.1088/0957-4484/26/11/115102
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8784
26. Luo X, Wang Y, Lin H, Qu FDOX. Fe 3 O 4@ mSiO 2-PO-FA
nanocomposite for synergistic chemo-and photothermal therapy. RSC
Adv.2016;6(113):112232112240. doi:10.1039/C6RA23292B
27. Yang Y, Wang C, Tian C, Guo H, Shen Y, Fe ZM. 3 O 4@ MnO 2@ PPy
nanocomposites overcome hypoxia: magnetic-targeting-assisted con-
trolled chemotherapy and enhanced photodynamic/photothermal therapy.
J Mater Chem B.2018;6(42):68486857. doi:10.1039/C8TB02077A
28. Xie W, Gao Q, Wang D, et al. Doxorubicin-loaded Fe 3 O 4@ MoS
2-PEG-2DG nanocubes as a theranostic platform for magnetic reso-
nance imaging-guided chemo-photothermal therapy of breast cancer.
Nano Res.2018;11(5):24702487. doi:10.1007/s12274-017-1871-1
29. Chen M-L, Gao Z-W, Chen X-M, Pang S-C ZY. Laser-assisted in situ
synthesis of graphene-based magnetic-responsive hybrids for multi-
modal imaging-guided chemo/photothermal synergistic therapy.
Talanta.2018;182:433442. doi:10.1016/j.talanta.2018.02.030
30. Chen H, Liu F, Lei Z, Ma L, Wang Z. Fe 2 O 3@ Au core@ shell
nanoparticlegraphene nanocomposites as theranostic agents for bioi-
maging and chemo-photothermal synergistic therapy. RSC Adv.
2015;5(103):8498084987. doi:10.1039/C5RA17143A
31. Seabra AB, Paula AJ, de Lima R, Alves OL, Duran N. Nanotoxicity
of graphene and graphene oxide. Chem Res Toxicol.2014;27(2):159
168. doi:10.1021/tx400385x
32. Li R, Guiney LM, Chang CH, et al. Surface oxidation of graphene
oxide determines membrane damage, lipid peroxidation, and cyto-
toxicity in macrophages in a pulmonary toxicity model. ACS Nano.
2018;12(2):13901402. doi:10.1021/acsnano.7b07737
33. Lele BS, Kulkarni MG. Single step room temperature oxidation of
poly(ethylene glycol) to poly(oxyethylene)-dicarboxylic acid. J Appl
Polym Sci.1998;70(5):883890. doi:10.1002/(ISSN)1097-4628
34. Morel A-L, Nikitenko SI, Gionnet K, et al. Sonochemical approach to
the synthesis of Fe3O4@SiO2 coreshell nanoparticles with tunable
properties. ACS Nano.2008;2(5):847856. doi:10.1021/nn800091q
35. Zhu Y, Ikoma T, Hanagata N, Kaskel S. Rattle-type Fe3O4@SiO2
hollow mesoporous spheres as carriers for drug delivery. Small.
2010;6(3):471478. doi:doi:10.1002/smll.200901403
36. Ding HL, Zhang YX, Wang S, Xu JM, Xu SC, Li GH.
Fe3O4@SiO2core/shell nanoparticles: the silica coating regulations
with a single core for different core sizes and shell thicknesses. Chem
Mater.2012;24(23):45724580. doi:10.1021/cm302828d
37. Fan H-L, Zhou S-F, Qi G-S, Liu Y-Z. Continuous preparation of
Fe3O4 nanoparticles using impinging stream-rotating packed bed
reactor and magnetic property thereof. J Alloys Compd.
2016;662:497504. doi:10.1016/j.jallcom.2015.12.025
38. Fidale LC, Nikolajski M, Rudolph T, Dutz S, Schacher FH, Heinze T.
Hybrid Fe3O4@ amino cellulose nanoparticles in organic mediahet-
erogeneous ligands for atom transfer radical polymerizations. J Colloid
Interface Sci.2013;390(1):2533. doi:10.1016/j.jcis. 2012.09.019
39. Lee J, Isobe T, Senna M. Preparation of ultrane Fe3O4Particles by
precipitation in the presence of PVA at high pH. J Colloid Interface
Sci.1996;177(2):490494. doi:10.1006/jcis.1996.0062
40. Zhang C, Wängler B, Morgenstern B, et al. Silica-and alkoxysilane-
coated ultrasmall superparamagnetic iron oxide particles: a promising
tool to label cells for magnetic resonance imaging. Langmuir.
2007;23(3):14271434. doi:10.1021/la061879k
41. Pozzi D, Colapicchioni V, Caracciolo G, et al. Effect of polyethyleneglycol
(PEG) chain length on the bio-nano- interactions between PEGylated lipid
nanoparticles and biological uids: from nanostructure to uptake in cancer
cells. Nanoscale.2014;6(5):27822792. doi:doi:10.1039/c3nr05559k
42. Murugan K, Choonara YE, Kumar P, Bijukumar D, Du Toit LC,
Pillay V. Parameters and characteristics governing cellular internali-
zation and trans-barrier trafcking of nanostructures. Int J
Nanomedicine.2015;10:2191.
43. Prijic S, Scancar J, Romih R, et al. Increased cellular uptake of
biocompatible superparamagnetic iron oxide nanoparticles into
malignant cells by an external magnetic eld. J Membr Biol.
2010;236(1):167179. doi:doi:10.1007/s00232-010-9271-4
44. Silva AH, Lima E, Mansilla MV, et al. Superparamagnetic iron-oxide
nanoparticles mPEG350- and mPEG2000-coated: cell uptake and
biocompatibility evaluation. Nanomed Nanotechnol Biol Med.
2016;12(4):909919. doi:doi:10.1016/j.nano.2015.12.371
45. Zhao Y, Sun X, Zhang G, Trewyn BG, Slowing II, Lin VS-Y.
Interaction of mesoporous silica nanoparticles with human red
blood cell membranes: size and surface effects. ACS Nano.2011;5
(2):13661375. doi:doi:10.1021/nn103077k
46. Hu Q, Zhang Y, Wang C, et al. Hemocompatibility evaluation in vitro
of methoxy polyethyleneglycol-polycaprolactone copolymer solu-
tions. J Biomed Mater Res - Part A.2016;104(3):802812. doi:
doi:10.1002/jbm.a.35594
47. Bender EA, Adorne MD, Colomé LM, Abdalla DSP, Guterres SS,
Pohlmann AR. Hemocompatibility of poly(ɛ-caprolactone) lipid-core
nanocapsules stabilized with polysorbate 80-lecithin and uncoated or
coated with chitosan. Int J Pharm.2012;426(1):271279.
doi:10.1016/j.ijpharm.2012.01.051
48. Zhang Z, Wang J, Nie X, et al. Near infrared laser-induced targeted
cancer therapy using thermoresponsive polymer encapsulated gold
nanorods. J Am Chem Soc.2014;136(20):73177326. doi:10.1021/
ja412735p
49. Kumar CSSR, Mohammad F. Magnetic nanomaterials for hyperther-
mia-based therapy and controlled drug delivery. Adv Drug Deliv Rev.
2011;63(9):789808. doi:10.1016/j.addr.2011.03.008
50. Ma P, Xiao H, Yu C, et al. Enhanced cisplatin chemotherapy by iron oxide
nanocarrier-mediated generation of highly toxic reactive oxygen species.
Nano Lett.2017;17(2):928937. doi:10.1021/acs.nanolett.6b04269
51. Wang X, Teng Z, Wang H, et al. Increasing the cytotoxicity of
doxorubicin in breast cancer MCF-7 cells with multidrug resistance
using a mesoporous silica nanoparticle drug delivery system. Int J
Clin Exp Pathol.2014;7(4):13371347.
52. Shrikhande SS, Jain DS, Athawale RB, et al. Evaluation of anti-
metastatic potential of Cisplatin polymeric nanocarriers on B16F10
melanoma cells. Saudi Pharm J.2015;23(4):341351. doi:10.1016/j.
jsps.2014.08.004
53. Byeon HJ, Thao LQ, Lee S, et al. Doxorubicin-loaded nanoparticles
consisted of cationic- and mannose-modied-albumins for dual-tar-
geting in brain tumors. J Control Release.2016;225:301313.
doi:10.1016/j.jconrel.2016.01.046
54. Lau JKC, Deubel DV. Hydrolysis of the anticancer drug cisplatin:
pitfalls in the interpretation of quantum chemical calculations. J
Chem Theory Comput.2006;2(1):103106. doi:10.1021/ct050229a
55. Štarha P, Smola D, Tuček J, Trávníček Z. Efcient synthesis of a
maghemite/gold hybrid nanoparticle system as a magnetic carrier for
the transport of platinum-based metallotherapeutics. Int J Mol Sci.
2015;16(1):20342051. doi:10.3390/ijms16012034
56. Thapa B, Diaz-Diestra D, Beltran-Huarac J, Weiner BR, Morell G.
Enhanced MRI T2Relaxivity in contrast-probed anchor-free
PEGylated iron oxide nanoparticles. Nanoscale Res Lett.
2017;12:113. doi:10.1186/s11671-017-2084-y
57. Bian X, Song ZL, Qian Y, et al. Fabrication of graphene-isolated-au-
nanocrystal nanostructures for multimodal cell imaging and photo-
thermal-enhanced chemotherapy. Sci Rep.2014;4:2932.
doi:10.1038/srep06093
58. Li L, Nurunnabi M, Naujjaman M, Jeong YY, Lee YK, Huh KM. A
photosensitizer-conjugated magnetic iron oxide/gold hybrid nanopar-
ticle as an activatable platform for photodynamic cancer therapy. J
Mater Chem B.2014;2(19):29292937. doi:10.1039/c4tb00181h
59. Lin Y, Wang S, Zhang Y, et al. Ultra-high relaxivity iron oxide
nanoparticles conned in polymer nanospheres for tumor MR
imaging. JMaterChemB.2015;3(28):57025710. doi:10.1039/
c5tb00593k
60. Di H, Wu H, Gao Y, Li W, Zou D, Dong C. Doxorubicin- and
cisplatin-loaded nanostructured lipid carriers for breast cancer com-
bination chemotherapy. Drug Dev Ind Pharm.2016;42(12):2038
2043. doi:10.1080/03639045.2016.1190743
Dovepress Khafaji et al
International Journal of Nanomedicine 2019:14 submit your manuscript | www.dovepress.com
DovePress 8785
61. Tahover E, Patil YP, Gabizon AA. Emerging delivery systems to
reduce doxorubicin cardiotoxicity and improve therapeutic index:
focus on liposomes. Anticancer Drugs.2015;26(3):241258.
doi:10.1097/CAD.0000000000000182
62. Ma P, Xiao H, Li C, et al. Inorganic nanocarriers for platinum drug
delivery. Mater Today.2015;18(10):554564. doi:10.1016/j.mattod.
2015.05.017
63. Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail che-
motherapy by combination drug delivery systems. Adv Drug Deliv
Rev.2016;98:1934. doi:10.1016/j.addr.2015.10.022
64. Unsoy G, Khodadust R, Yalcin S, Mutlu P, Gunduz U. Synthesis of
Doxorubicin loaded magnetic chitosan nanoparticles for pH respon-
sive targeted drug delivery. Eur J Pharm Sci.2014;62:243250.
doi:10.1016/j.ejps.2014.05.021
65. Wang F, Wang Y-C, Dou S, Xiong M-H, Sun T-M, Wang J.
Doxorubicin-tethered responsive gold nanoparticles facilitate intra-
cellular drug delivery for overcoming multidrug resistance in cancer
cells. ACS Nano.2011;5(5):36793692. doi:10.1021/nn200007z
66. Swietach P, Vaughan-Jones RD, Harris AL, Hulikova A. The chemistry,
physiology and pathology of pH in cancer. Philos Trans R Soc B. 2014.
67. Alkilany AM, Nagaria PK, Hexel CR, Shaw TJ, Murphy CJ, Wyatt
MD. Cellular uptake and cytotoxicity of gold nanorods: molecular
origin of cytotoxicity and surface effects. Small.2009;5(6):701708.
doi:10.1002/smll.200801546
68. Quinto CA, Mohindra P, Tong S, Bao G. Multifunctional superpar-
amagnetic iron oxide nanoparticles for combined chemotherapy and
hyperthermia cancer treatment. Nanoscale.2015;7(29):1272812736.
doi:10.1039/c5nr02718g
69. Ehlerding EB, Chen F, Biodegradable CW. Renal Clearable Inorganic
Nanoparticles. Adv Sci.2016;3(2):1500223. doi:10.1002/advs.201500223
70. Kievit FM, Zhang M. Surface engineering of iron oxide nanoparticles
for targeted cancer therapy. Acc Chem Res.2011;44(10):853862.
doi:10.1021/ar2000277
71. Kapse-Mistry S, Govender T, Srivastava R, Yergeri M. Nanodrug
delivery in reversing multidrug resistance in cancer cells. Front
Pharmacol.2014;5:159.
72. Wang X, Zhang R, Wu C, et al. The application of Fe3O4 nanopar-
ticles in cancer research: a new strategy to inhibit drug resistance. J
Biomed Mater Res Part A.2007;80A(4):852860. doi:10.1002/jbm.
a.30901
73. Chen Y, Zhang F, Wang Q, et al. The synthesis of LA-Fe 3 O 4@
PDA-PEG-DOX for photothermal therapychemotherapy.Dalt
Trans.2018;47(7):24352443. doi:10.1039/C7DT04080F
74. Liu B, Zhang X, Li C, et al. Magnetically targeted delivery of
DOX loaded Cu 9 S 5@ mSiO 2@ Fe 3 O 4-PEG nanocomposites
for combined MR imaging and chemo/photothermal synergistic
therapy. Nanoscale.2016;8(25):1256012569. doi:10.1039/C5NR0
6322A
International Journal of Nanomedicine Dovepress
Publish your work in this journal
The International Journal of Nanomedicine is an international, peer-
reviewed journal focusing on the application of nanotechnology in
diagnostics, therapeutics, and drug delivery systems throughout the
biomedical eld. This journal is indexed on PubMed Central,
MedLine, CAS, SciSearch
®
, Current Contents
®
/Clinical Medicine,
Journal Citation Reports/Science Edition, EMBase, Scopus and the
Elsevier Bibliographic databases. The manuscript management system
is completely online and includes a very quick and fair peer-review
system, which is all easy to use. Visit http://www.dovepress.com/
testimonials.php to read real quotes from published authors.
Submit your manuscri pt here: https:// www.dovepress.com/inte rnational-jou rnal-of-nanom edicine-journal
Khafaji et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
International Journal of Nanomedicine 2019:14
8786
... Co-administered systems are advantageous because the individual drug dosages and administration timing can be regulated independently (36), as seen in our PEG-PC nanoparticles containing SCH772984 and GEM. Furthermore, a plausible reason for co-administration to work better than free form of the drugs is that the nanoparticles could be taken up by the cells via the endosomal pathway where the drug is released under acidic pH in the endosome (8,37,38). ...
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with a poor response to the limited treatment options currently available. Hence, there is a need to identify new agents that could enhance the efficacy of existing treatments. This study investigated a combination therapy using gemcitabine (GEM) and SCH772984, an extracellular signal-regulated kinase (ERK) inhibitor, in both free form and nanoparticle-encapsulated form for PDAC treatment. Cell viability and Matrigel growth assays were used to determine the anti-proliferative and cytotoxic effects of GEM and SCH772984 on PDAC cells. Additionally, western blotting was used to determine the degree to which SCH772984 engaged ERK in PDAC cells. Lastly, immunohistochemistry and hematoxylin and eosin (H&E) staining were used to determine how GEM and SCH772984 affected expression of Ki-67 cell proliferation marker in PDX (patient derived xenograft) PDAC tissues. PDAC cell lines (MIA PaCa-2 and PANC-1) treated with the combination of free GEM and SCH772984 showed reduction in cell viability compared to cells treated with free GEM or SCH772984 administered as a single agent. Encapsulated forms of GEM and SCH772984 caused a greater reduction in cell viability than the free forms. Interestingly, co-administration of GEM and SCH772984 in separate nanoparticle (NP) systems exhibited the highest reduction in cell viability. Western blotting analysis confirmed ERK signaling was inhibited by both free and encapsulated SCH772984. Importantly, GEM did not interfere with the inhibitory effect of SCH772984 on phosphorylated ERK (pERK). Collectively, our studies suggest that combination therapy with GEM and SCH772984 effectively reduced PDAC cell viability and growth, and co-administration of NP encapsulated GEM and SCH772984 in separate NP systems is an effective treatment strategy for PDAC.
Article
Combination chemotherapy is a relatively recent and preferred method for cancer treatment. Sustained delivery of dual drugs can be achieved with a suitable matrix. In the present work, a pH-responsive polyelectrolyte complex (PEC) of trimethylchitosan and carboxymethylkaraya gum containing silver nanoparticles (SNps) has been developed as a matrix material for co-delivery of the drugs, 5-fluorouracil (5-Fu) and curcumin (Cur). The experimental conditions have been optimized for high yield and high swelling of the PEC nanocomposite. 1H-NMR, FT-IR, FE-SEM, P-XRD, HR-TEM, EDS, TGA techniques and zeta potential measurements have been employed in the physico-chemical characterization of the nanocomposite material. The presence of SNps with an average diameter of 16.57 ± 1.25 nm influenced the surface structure and hydrophilicity of the PEC. The swelling study indicated higher swelling at pH 7.4 than at pH 1.2. The two drugs, 5-Fu and Cur, were successfully entrapped and released from the nanocomposite in a sustained manner. Cytotoxicity studies performed with the MCF-10A cell line confirmed the biocompatibility of the nanocomposite and those with the MCF-7 cell line indicated the synergistic effect of the dual drugs in controlling cancer cell growth. The overall study indicates the usefulness of the PEC nanocomposite made from modified polysaccharides, chitosan and karaya gum as a promising material for the development of a dual drug delivery system for cancer treatment.
Article
In response to the pressing need for more efficient and targeted cancer therapies, this study presents the development of biodegradable hydroxyapatite/polyaniline (HAp/PANI) nanocomposite drug carriers for near-infrared (NIR)-induced drug delivery. The synthesis involved loading polyaniline onto mesoporous hydroxyapatite spheres, resulting in high drug loading capacity and tunable NIR responsiveness. The HAp/PANI spheres exhibited superior photothermal properties compared to pristine HAp under NIR irradiation, along with excellent biocompatibility. Importantly, the drug release behavior could be precisely controlled by adjusting NIR power and irradiation time, leading to enhanced anticancer efficacy against HCT-116 colorectal cancer cells. These findings highlight the potential of HAp/PANI mesoporous spheres as promising drug carriers for NIR-responsive cancer therapy.
Article
Phototherapy, including photothermal therapy (PTT) and photodynamic therapy (PDT), has the advantages of spatiotemporal selectivity, non-invasiveness, and negligible drug resistance. Phototherapy has been approved for treating superficial epidermal tumors. However, its therapeutic efficacy is limited by the hypoxic tumor microenvironment and the highly expressed heat shock protein. Moreover, poor tissue penetration and focused irradiation laser region in phototherapy make treating deep tissues and metastatic tumors challenging. Combination therapy strategies, which integrate the advantages of each treatment and overcome their disadvantages, can significantly improve the therapeutic efficacy. Recently, many combination therapy strategies have been reported. Our study summarizes the strategies used for combining phototherapy with other cancer treatments such as chemotherapy, immunotherapy, sonodynamic therapy, gas therapy, starvation therapy, and chemodynamic therapy. Some research cases were selected to analyze the combination therapy effect, delivery platform feature, and synergetic anticancer mechanisms. Moreover, additional research cases are summarized in the tables. This review provides strong evidence that phototherapy-based combination strategies can enhance the anticancer effect compared with phototherapy alone. Additionally, the challenges and future perspectives associated with these combinational therapies are discussed.
Article
Full-text available
Combination therapies can enhance the sensitivity of cancer to drugs, lower drug doses, and reduce side effects in cancer treatment. However, differences in the physicochemical properties and pharmacokinetics of different therapeutic agents limit their application. To avoid the above dilemma and achieve accurate control of the synergetic ratio, a nanoscale co-delivery system (NCDS) has emerged as a prospective tool for combined therapy in cancer treatment, which is increasingly being used to co-load different therapeutic agents. In this study, we have summarized the mechanisms of therapeutic agents in combination for cancer therapy, nanoscale carriers for co-delivery, drug-loading strategies, and controlled/targeted co-delivery systems, aiming to give a general picture of these powerful approaches for future NCDS research studies.
Article
The utilization of Platinum (Pt)-based drugs has significantly transformed the landscape of cancer chemotherapy, serving as highly effective anticancer agents. However, the adverse effects and amplification of resistance to drugs in cancer cells compromise their therapeutic usefulness. In order to overcome the constraints, platinum-based chemotherapeutic agents are often administered in combination with other therapeutic modalities as the primary course of treatment for diverse cancer types, yielding notable therapeutic benefits. Nonetheless, in certain cases, the effectiveness of these combination treatments is diminished due to differing pharmacokinetic properties of the combined drugs, short circulation half-lives, and severe side effects. To this end, considerable resources have been dedicated to the development of nanoparticle-mediated drug delivery systems for Pt-based drugs. This is partly because of their adaptability in terms of drug delivery, capability to affect the tumor's microenvironment, and ability to include more genes, proteins, and small molecules to fight chemotherapy resistance in cancer. This paper summarizes the structure and cellular mechanisms of Pt(II) and Pt(IV) compounds, the loading of Pt(II) and Pt(IV) drugs into various delivery systems by employing physical encapsulation and conjugation techniques, recent strategies involving various types of nanoparticle-based platinum drugs and multifunctional approach of nanotechnology-enabled platinum-based cancer therapies including chemo-chemotherapy combination, chemo-photodynamic therapy, chemo–photothermal therapy, chemo–immunotherapy, chemoradiotherapy, electrochemotherapy, ultrasound-mediated delivery, triple multiple functionalities, and other combination therapies, and discussed pharmacokinetics (PK) of Pt-based drugs. Furthermore, we delve into the current challenges related to these approaches and offer insights into their potential future advancements. Our comprehensive summary and perspective aim to assist researchers in generating innovative ideas for developing highly potent nanomedicine specifically designed for anticancer applications, incorporating Pt-based drugs.
Article
Full-text available
Cancer is a major public health problem and the second leading cause of mortality around the world. Antitumor immunotherapy using monoclonal antibodies is considered selective and efficient in the treatment of different types of tumors, but its cost and toxic effects limit its application. Many tumor microenvironments, including lymphoma and carcinoma, are enriched in immune suppressive cells that contribute to immune exhaustion by means expression of inhibitory ligands, suppressive cytokines, and tumor-promoting factors. Antitumor therapies targeted to reduce the induction, recruitment, or suppressive activities of the immune cells have been investigated. New antitumor strategies using drugs targeted to intracellular signaling involved in cell proliferation and survival, angiogenesis, and metastasis have become promising in recent years. Thus, our discovery of the role of functional interaction between intracellular signaling pathways mediated by calcium ions (Ca 2+) and cyclic adenosine monophosphate (cAMP) (Ca 2+ /cAMP signaling interaction) in these cellular responses, opened a great avenue for the development of new antitumor therapeutic strategies. Here, we discuss how the combined use of monoclonal antibodies with drugs that modulate the Ca 2+ /cAMP signaling interaction to reduce tumor growth could be a potential strategy in the antitumor immunotherapy due to the increment of antitumor efficacy and reduction of adverse effects.
Article
Abundant efforts have recently been made to design potent theranostic nanoparticles, which combine diagnostic and therapeutic agents, for the effective treatment of cancer. In this study, we developed multifunctional magnetic gold nanoparticles (MGNPs) that are able to (i) selectively deliver the drug to the tumor site in a controlled-release manner, either passively or by using magnetic targeting; (ii) induce photothermal therapy by producing heat by near-infrared (NIR) laser absorption; and (iii) serve as contrast agents for magnetic resonance imaging (MRI) (imaging-guided therapy). The prepared MGNPs were characterized by different physical techniques. They were then coated and conjugated with polyethylene glycol (PEG) and doxorubicin (DOX) to form MGNP-DOX conjugates. The high efficacy of MGNP-DOX for combined chemo-photothermal therapy was observed both in vitro and in vivo. The effectiveness of MGNP-DOX as theranostic nanoparticles was confirmed by histopathological examination and immunohistochemical studies. Moreover, MGNP-DOX showed good potential as MRI contrast agents for guided chemo-photothermal synergistic therapy.
Article
Considering traditional treatment methods such as chemotherapy with side effects and lack of targeting, photodynamic therapy (PDT) and photothermal therapy (PTT) as innovative, less by-effect, and light-controlled treatments have received extensive attention, but the efficacy of PDT is seriously hindered by hypoxic tumor microenvironment. Here, a multifunctional core-shell structure nanocomposite composed by a core of iron oxide (Fe3O4) and two shells of manganese dioxide (MnO2) and polypyrrole (PPy) was successfully prepared and used to solve these issues. PPy, simultaneously as photothermal agent and photosensitizer, were first combined with MnO2 to coat magnetic Fe3O4 for achieving the increase of the intracellular O2 concentration and improving the singlet oxygen generation upon laser irradiation, so that the enhanced PDT/PTT could be achieved. After the efficient loading of doxorubicin (DOX) on the Fe3O4@MnO2@PPy nanocomposite, an acid controlled-release behavior for DOX as well as magnetic targeting assisted synergistic effects of chemotherapy and improved PDT/PTT for tumor cells could be realized, which also could protect normal cells from damage. All these features may render our nanocomposite a promising platform to reverse the hypoxia-triggered PDT resistance and the chemotherapy-caused side effects in cancer therapy and other biomedical applications.
Article
Combination therapies for cancer have been promising strategies to reverse multidrug resistance, improve treatment efficiency, and minimize side effects. Herein, a new type of multifunctional silicon nanowires (MFSiNWs)-based system, i.e., iron oxide nanoparticles and gold nanoparticles decorated silicon nanowires (IONPs@AuNPs@SiNWs), is prepared through a facile synthetic strategy. The resultant nanohybrids could load therapeutic drug molecules (e.g., doxorubicin, DOX) with high drug loading capacity (2000 mg g⁻¹), and be simultaneously responsive to pH/NIR (near-infrared) light/magnetism, thus enabling high-efficacy synergistic treatment of drug-resistant cancer cells. This current work provides the pioneering demonstration of silicon nanomaterials-based combinational cancer treatment with high therapeutic efficiency.
Article
Magnetic graphene-based hybrids are being increasing recognized as an effective nanotheranostic agent in biomedicine. Conventional technologies for their synthesis have drawbacks not only from a synthetic standpoint, mainly requiring high temperatures and multi-step processes, but also from a biological perspective, chemical precursors or surfactants involved in the chemical process are toxic to cells. Herein, we report a novel approach for one-step fabricating magnetic graphene hybrid nanocomposites based on laser irradiation of an Fe target in GO-PEG aqueous solution at room temperature without using any other chemical reagent. TEM, XPS, FT-IR, XRD, Mossbauer spectrum and VSM observation reveal that γ-Fe2O3 nanoparticles were directly grown on the surface of GO-PEG with uniform morphology and superior dispersibility. These GO-PEG-γ-Fe2O3 nanocomposites (labeled as GPF) showed low cytotoxicity in vitro compared to chemically synthesized nanoparticles since the pulsed-laser-ablation-in-liquid (PLAL) process is free of toxic agents. After tail vein injection of the nanotheranostics, the tumor was clearly mapped by T2-weighted magnetic resonance of γ-Fe2O3, photothermal imaging of graphene and fluorescence imaging of loaded antitumor DOX. Meanwhile the tumor cells both in vitro and in vivo achieved highly superior inhibition by the synergistic chemo/photothermal therapeutic effect which provided an intense heating effect and enhanced DOX release upon 808 nm NIR light exposure. The results revealed that the magnetic graphene-based hybrids prepared by PLAL is competent for future multi-modal imaging assisted tumor targeted chemo/photothermal synergistic therapy of cancer.
Article
Nanoagents of integrating multiple imaging and therapeutic modalities have attracted tremendous attention for biomedical applications. Herein, we synthesize porous hollow Fe3O4 as a theranostic agent for combined photothermal/chemo cancer therapy and MRI. The as-prepared porous iron oxide nanoagents allow for T2-weighted MR imaging. Interestingly, we demonstrate that the porous structure endows the nanoagents an outstanding photothermal property for cancer cell killing, in comparison with other types of iron oxide nanomaterials. Under the exposure of NIR laser, the heat produced by porous Fe3O4 can accelerate the release of the loaded drug (e.g. DOX) to enhance chemotherapeutic efficacy, promoting the ablating of cancer cells with synergistic photothermal/chemo therapy.
Article
While 2-dimensional graphene oxide (GO) is used increasingly in biomedical applications, there is uncertainty on how specific physicochemical properties relate to biocompatibility in mammalian systems. Although properties such as lateral size and the colloidal properties of the nanosheets are important, the specific material properties that we address here is the oxidation state and reactive surface groups on the planar surface. In this study, we used a GO library, comprised of pristine, reduced (rGO), and hydrated GO (hGO), in which quantitative assessment of the hydroxyl, carboxyl, epoxy and carbon radical contents were used to study the impact on epithelial cells and macrophages, as well as in the murine lung. Strikingly, we observed that hGO, which exhibits the highest carbon radical density, was responsible for the generation of cell death in THP-1 and BEAS-2B cells as a consequence of lipid peroxidation of the surface membrane, membrane lysis, and cell death. In contrast, pristine GO had lesser effects while rGO showed extensive cellular uptake with minimal effects on viability. In order to see how these in vitro effects relate to adverse outcomes in the lung, mice were exposed to GOs by oropharyngeal aspiration. Animal sacrifice after 40h demonstrated that hGO was more prone than other materials in generating acute lung inflammation, accompanied by the highest lipid peroxidation in alveolar macrophages, cytokine production (LIX, MCP-1) and LDH release in bronchoalveolar lavage fluid. Pristine GO showed less toxicity while rGO had minimal effects. In summary, we demonstrate that the surface oxidation state and carbon radical content play major roles in the induction of toxicity by GO in mammalian cells and the lung.
Article
A facile methodology is presented to construct a multifunctional nanocomposite, which integrates photothermal therapy and specific drug release into a single nanostructure. Firstly, the magnetic Fe3O4@polydopamine core-shell nanoparticles (Fe3O4@PDA) were synthesized via reversed-phase microemulsion approach. By varying the amount of DA, Fe3O4@PDA with particle size of 28-38 nm can be obtained. To further make sure the monodispersity, biocompatibility and specific uptake, PEG and lactobionic acid (LA) were modified on to Fe3O4@PDA (LA-Fe3O4@PDA-PEG), whose fast photothermal conversion is derived the combination of Fe3O4 and PDA with high near infrared (NIR) absorption. And then, doxorubicin hydrochloride (DOX) was adopted as the typical anticancer drug, which was loaded on to LA-Fe3O4@PDA-PEG via electrostatic and π–π stacking interaction. The release kinetics investigation further demonstrated the acid/heat-triggered DOX release. HepG2 cells (hepatocellular cell line) were used as the target cancer cells, and the fast uptake was owing to the nanoparticle size and abundant asialoglycoprotein receptor on HepG2 cells. Besides, the external magnetic field also can improve the uptake, especially, when the magnet was placed at the bottom of the cell disk. The enhanced specific cytotoxicity toward HepG2 cells was also ascribed to the synergistic effect of chemo- and photothermal therapy. Based on the novel properties, LA-Fe3O4@PDA-PEG-DOX nanocomposite showed the potential application on hepatocyte therapy.
Article
Molybdenum disulfide (MoS2), a typical transition-metal dichalcogenide, has attracted increasing attention in the field of nanomedicine because of its preeminent properties. In this study, magnetic resonance imaging (MRI)-guided chemo-photothermal therapy of human breast cancer xenograft in nude mice was demonstrated using a novel core/shell structure of Fe3O4@MoS2 nanocubes (IOMS NCs) via the integration of MoS2 (MS) film onto iron oxide (IO) nanocubes through a facile hydrothermal method. After the necessary PEGylation modification of the NCs for long-circulation purposes, such PEGylated NCs were further capped by 2-deoxy-D-glucose (2-DG), a non-metabolizable glucose analogue to increase the accumulation of the as-prepared NCs at the tumor site, as 2-DG molecules could be particularly attractive to resource-hungry cancer cells. Such 2-DG-modified PEGylated NCs (IOMS-PEG-2DG NCs) acted as drug-carriers for doxorubicin (DOX), which could be easily loaded within the NCs. The obtained IOMS-PEG(DOX)-2DG NCs exhibited a T2 relaxivity coefficient of 48.86 (mM)−1·s−1 and excellent photothermal performance. 24 h after intravenous injection of IOMS-PEG(DOX)-2DG NCs, the tumor site was clearly detected by enhanced T2-weighted MRI signal. Upon exposure to an NIR 808-nm laser for 5 min at a low power density of 0.5 W·cm−2, a marked temperature increase was noticed within the tumor site, and the tumor growth was efficiently inhibited by the chemo-photothermal effect. Therefore, our study highlights an excellent theranostic platform with great potential for targeted MRI-guided precise chemo-photothermal therapy of breast cancer. Open image in new window