You are on page 1of 12

Biomaterials 38 (2015) 10e21

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Hyaluronic acid-modified Fe3O4@Au core/shell nanostars for


multimodal imaging and photothermal therapy of tumors
Jingchao Li a, c, 1, Yong Hu c, 1, Jia Yang b, 1, Ping Wei c, Wenjie Sun c, Mingwu Shen c, ***,
Guixiang Zhang b, **, Xiangyang Shi a, c, *
a
State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University,
Shanghai 201620, PR China
b
Department of Radiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, PR China
c
College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Development of multifunctional theranostic nanoplatforms for diagnosis and therapy of cancer still re-
Received 25 August 2014 mains a great challenge. In this work, we report the use of hyaluronic acid-modified Fe3O4@Au core/shell
Accepted 19 October 2014 nanostars (Fe3O4@Au-HA NSs) for tri-mode magnetic resonance (MR), computed tomography (CT), and
Available online
thermal imaging and photothermal therapy of tumors. In our approach, hydrothermally synthesized
Fe3O4@Ag nanoparticles (NPs) were used as seeds to form Fe3O4@Au NSs in the growth solution. Further
Keywords:
sequential modification of polyethyleneimine (PEI) and HA affords the NSs with excellent colloidal
Fe3O4@Au nanostars
stability, good biocompatibility, and targeting specificity to CD44 receptor-overexpressing cancer cells.
Hyaluronic acid
CT imaging
With the Fe3O4 core NPs and the star-shaped Au shell, the formed Fe3O4@Au-HA NSs are able to be used
MR imaging as a nanoprobe for efficient MR and CT imaging of cancer cells in vitro and the xenografted tumor model
Tumors in vivo. Likewise, the NIR absorption property enables the developed Fe3O4@Au-HA NSs to be used as a
Photothermal therapy nanoprobe for thermal imaging of tumors in vivo and photothermal ablation of cancer cells in vitro and
xenografted tumor model in vivo. This study demonstrates a unique multifunctional theranostic nano-
platform for multi-mode imaging and photothermal therapy of tumors, which may find applications in
theranostics of different types of cancer.
© 2014 Elsevier Ltd. All rights reserved.

1. Introduction toxicity [15e17]. Another promising nanoplatform is gold NPs


(AuNPs). On one hand, with a higher atomic number than that of
The past decade has seen a myriad of interest in using various iodine, AuNPs have been extensively employed as a contrast agent
inorganic or organic nanoparticles (NPs) or microparticles for a for CT imaging of different biological systems due to their better X-
wide variety of biomedical applications because of their unique ray attenuation property than that of Omnipaque (a conventional
structural features and functionalities [1e7]. In particular, for iodine-based CT contrast agent) [18e23]. On the other hand, AuNPs
magnetic iron oxide (Fe3O4) NPs, besides their uses in magnetic with particular shapes such as nanorods [24,25], nanoshells
separation [8,9], hyperthermia [10,11], catalysis [12], and drug/gene [26,27], nanoflowers [28], nanocages [29,30], or nanostars (NSs)
delivery [13,14], Fe3O4 NPs have been used as negative contrast [31,32] display strong surface plasmon resonance (SPR) absorption
agents for T2-weighted magnetic resonance (MR) imaging due to intensity in near infrared (NIR) region [33], enabling their uses for
their high relaxivity, excellent contrast enhancement, and low thermal imaging and photothermal therapy of cancer or other
biological systems.
For diagnosis and therapy of cancer, it is essential to develop a
* Corresponding author. College of Chemistry, Chemical Engineering and theranostic platform that is able to integrate both diagnosis ele-
Biotechnology, Donghua University, Shanghai 201620, PR China. Tel.: þ86 21 ments and therapeutic agents [34,35]. In particular, for accurate
67792656; fax: þ86 21 67792306 804. cancer imaging applications, it is meaningful to design a multi-
** Corresponding author. Tel.: þ86 21 63240090 4166; fax: þ86 21 63240825. functional platform affording dual or multi-mode imaging because
*** Corresponding author. Tel.: þ86 21 67792750; fax: þ86 21 67792306 804.
each imaging modality has its own limitations and advantages [36].
E-mail addresses: mwshen@dhu.edu.cn (M. Shen), guixiangzhang@sina.com
(G. Zhang), xshi@dhu.edu.cn (X. Shi). For instance, Cai et al. prepared Fe3O4@Au nanocomposite particles
1
Authors contributed equally to this work. for MR/CT dual mode imaging [37]. Tian et al. reported the

http://dx.doi.org/10.1016/j.biomaterials.2014.10.065
0142-9612/© 2014 Elsevier Ltd. All rights reserved.
J. Li et al. / Biomaterials 38 (2015) 10e21 11

Fe3O4@Cu2xS core/shell NPs for both MR and thermal imaging of modified by partially thiolated PEI (PEI-SH) via AueS bond, fol-
tumors [38]. To exert a therapeutic effect to tumors, an anticancer lowed by modification with HA via 1-ethyl-3-[3-
drug is usually encapsulated within a designed nanoparticulate dimethylaminopropyl] carbodiimide hydrochloride (EDC)
system [39e42] or conjugated onto the surface of NPs [43e46]. coupling reaction with the PEI amines on the surface of the NSs
Alternatively, cancer cells can also be ablated under laser irradia- (Scheme 1). The formed Fe3O4@Au-HA NSs were characterized via
tion in the presence of NPs with strong NIR absorption property, different techniques. Their stability, biocompatibility including
such as tungsten oxide (WO2.9) nanorods [47], Cu7.2S4 nanocrystals hemocompatibility and cytocompatibility, targeting specificity to
[48], Au nanorods [49], and Au NSs [50]. Therefore, for accurate CD44 receptor-overexpressing cancer cells, and potentials to be
theranostics of cancer or imaging-guided cancer therapy, it is used as T2-weighted MR and CT contrast agents for dual mode MR/
essential to integrate multi-mode imaging elements and thera- CT imaging of cancer cells in vitro and xenografted tumor model
peutic agents within one nanoparticulate system. Although some of in vivo were investigated in detail. Furthermore, the developed
the above NP systems have been demonstrated to be able to exert Fe3O4@Au-HA NSs were used for photothermal ablation of cancer
both dual mode imaging (e.g., CT/thermal or MR/thermal imaging) cells in vitro and xenografted tumor model in vivo, as well as
and photothermal therapeutic efficacy of cancer [35,38,47], these thermal imaging of the tumor model in vivo.
NP systems are lack of targeting specificity presumably due to the
technical difficulty of surface biofunctionalization. Development of 2. Experimental section
various multifunctional nanoplatforms that enable targeted multi- 2.1. Materials
mode imaging and photothermal therapy of cancer still remains a Hyaluronic acid (HA, Mw ¼ 31,200) was purchased from Zhenjiang Dong Yuan
great challenge. Biotechnology Corporation (Zhenjiang, China). EDC, N-hydroxysuccinimide (NHS),
Our previous work has shown that dendrimer-entrapped AuNPs cetyltrimethyl-ammoniumbromide (CTAB), and methyl thioglycolate (MTG) were
[19e23,51,52] and Fe3O4 NPs prepared via either controlled co- supplied by J&K Chemical Ltd (Shanghai, China). Branched polyethyleneimine (PEI,
Mw ¼ 25,000) and sodium borohydride (NaBH4) were purchased from Aldrich (St.
precipitation [53,54] or hydrothermal [15e17,55] approaches are Louis, MO). HAuCl4$4H2O, ferrous chloride tetrahydrate (FeCl2$4H2O > 99%),
able to be used as CT and MR contrast agents, respectively for ammonia (25e28% NH3 in water solution), silver nitrate, ascorbic acid (AA) and all
molecular imaging applications. Dual mode MR/CT imaging func- other chemicals and solvents were from Sinopharm Chemical Reagent Co., Ltd
tionality can be easily realized by assembly of dendrimer- (Shanghai, China). All chemicals were used as received. HeLa cells (a human cervical
carcinoma cell line) and U87MG cells (a human glioblastoma carcinoma cell line)
entrapped AuNPs onto preformed Fe3O4 NPs [37] or by hydro-
were obtained from Institute of Biochemistry and Cell Biology, the Chinese Academy
thermal synthesis of Fe3O4/Au composite NPs [56,57]. In another of Sciences (Shanghai, China). Modified eagle medium (MEM), Dulbecco's modified
work, Liu and coworkers have shown that Au NSs having a strong eagle medium (DMEM), fetal bovine serum (FBS), penicillin, and streptomycin were
surface plasmon resonance (SPR) band at NIR region can be pre- from Hangzhou Jinuo Biomedical Technology (Hangzhou, China). 3-(4,5-
pared from Au seeds in the growth solution using silver ion com- Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was purchased
from Shanghai Sangon Biological Engineering Technology & Services Co., Ltd
plexes as growth inhibitors [58]. Likewise, Au NSs with magnetic (Shanghai, China). Water used in all experiments was purified using a Milli-Q Plus
Fe3O4 or Fe cores formed by exposing the core/shell Fe3O4@Au NPs 185 water purification system (Millipore, Bedford, MA) with a resistivity higher than
(seeds) or Fe seeds into the Au growth solution can be used for 18.2 MU$cm. Regenerated cellulose dialysis membranes with molecular weight cut-
gyromagnetic imaging of cells or fluorescence imaging/photo- off (MWCO) of 14,000 were acquired from Fisher.
thermal destruction of cancer cells [59,60], while Fe3O4@Au
2.2. Synthesis of partially thiolated PEI
nanostars (NSs) formed by the reduction of Au(III) onto the
dextran-coated Fe3O4 NPs with hydroxylamine as a seeding agent The partially thiolated PEI (PEI-SH) was synthesized according to protocols
described in the literature [61]. In brief, MTG (108 mL) was added to a freshly pre-
displayed five distinct functions (aptamer-based targeting, MR
pared PEI aqueous solution (10 mL, 1.0 g), and the mixture was continuously stirred
imaging, optical imaging, photothermal therapy and chemo- at 60e70  C in a water bath for 24 h to complete the reaction. After that, the reaction
therapy) [34]. However, these studies have not completely mixture was dialyzed against water (6 times, 2L) using a dialysis membrane with
demonstrated the potentials to use the developed nanoplatforms MWCO of 14,000 for 3 days, followed by lyophilization to obtain the purified PEI-SH.
for CT/MR dual mode imaging and photothermal therapy of cancer
cells in vitro and in vivo. Our previous successes in the preparation 2.3. Synthesis of Fe3O4@Ag seeds

of Fe3O4/Au composite NPs lead us to hypothesize that the hydro- Fe3O4@Ag seeds were synthesized according to our previous work with some
thermally synthesized Fe3O4@Au or Fe3O4@Ag seeds may further modifications [56]. Firstly, PEI was used as a stabilizer to synthesize Ag NPs at the
PEI/Ag salt molar ratio of 1:20. Namely, a silver nitrate aqueous solution (68.0 mg,
grow to form star-shaped Au shells onto the Fe3O4 core NPs,
2 mL) was added into a PEI aqueous solution (0.05 g/mL, 10 mL) under vigorous
thereby affording the creation of Fe3O4@Au NSs for multi-mode magnetic stirring. After 30 min, an icy cold NaBH4 aqueous solution (75.66 mg, 1 mL)
imaging and photothermal therapy of cancer. Our prior work has was rapidly added into the above mixture and the mixture was continuously stirred
also shown that in the presence of branched polyethyleneimine for 2 h to complete the reaction. The obtained PEI-Ag NPs were then purified via
(PEI), hydrothermally formed Fe3O4 NPs are able to be afforded dialysis as described above and finally redispersed in 5 mL water for further use.
Then, Fe3O4@Ag seed particles were synthesized using a one-pot hydrothermal
with amine functionality [17]. Hence, Fe3O4 NPs can be easily approach as described in our previous work [56]. FeCl2$4H2O (1.25 g) dissolved in
modified with targeting ligand folic acid (FA) or hyaluronic acid 7.75 mL water was mixed with ammonium hydroxide (6.25 mL) under vigorous
(HA) for targeted MR imaging of FA receptor- and CD44 receptor- magnetic stirring. The mixture was kept in air for about 10 min while stirring to
overexpressing tumors, respectively [15,16]. Logically, the ensure iron (II) to be oxidized. Then the mixture was transferred into a 50-mL
autoclave (KH-50 Autoclave, Shanghai Yuying Instrument Co., Ltd., Shanghai,
Fe3O4@Au NSs to be designed in this work may also be modified
China) and the obtained suspension of PEI-Ag NPs (5 mL) was also added into the
with PEI for further modification of targeting ligands, thereby autoclave. The mixture was stirred thoroughly and then autoclaved in a sealed
generating a multifunctional nanoplatform for targeted thera- pressure vessel at 134  C for 3 h. Subsequently, the autoclave was cooled down to
nostics of cancer. room temperature, and the product was collected via magnetic separation. The
In this present study, we report the formation of HA-targeted formed Fe3O4@Ag seeds were further purified by rinsing with water for 3 times and
finally redispersed in 15 mL water.
Fe3O4@Au NSs for tri-mode (MR/CT/thermal) imaging and photo-
thermal therapy of cancer. First, Fe3O4@Ag seeds were synthesized 2.4. Synthesis of Fe3O4@Au NSs
via a facile one-pot hydrothermal route according to our previous
To prepare a gold growth solution, HAuCl4 (30 mg/mL, in 580 mL water) was
work with some modifications [56]. Then the Fe3O4@Ag seeds were added to an aqueous solution of CTAB (381 mg, 10 mL), followed by successive
added into the Au growth solution to form Fe3O4@Au NSs with the addition of AgNO3 (1.1 mg) and ascorbic acid (12 mg) under vigorous magnetic
help of silver nitrate. The formed Fe3O4@Au NSs were then surface stirring. Then 0.1 mL Fe3O4@Ag seeds (10 times diluted with water from the above
12 J. Li et al. / Biomaterials 38 (2015) 10e21

AgNO3 Hydrothermal synthesis

NaBH4 Fe(II) salt


PEI PEI-Ag NPs Fe3O4@Ag seeds

Growth
HA PEI-SH

EDC/NHS

Fe3O4@Au-HA NSs Fe3O4@Au-PEI NSs Fe3O4@Au NSs

Scheme 1. Schematic illustration of the synthesis of Fe3O4@Au-HA NSs.

suspension) were added and the mixture solution changed to blue within a few and Imaging system (Shanghai Niumag Corporation, Shanghai, China). The param-
minutes, indicating the formation of Fe3O4@Au NSs. After additional stirring for 1 h, eters were set as following: CPMG sequence, 0.5 T magnet, point
the product was purified by 3 cycles of centrifugation/redispersion in water to resolution ¼ 156 mm  156 mm, section thickness ¼ 0.6 mm, TR ¼ 6000 ms,
remove CTAB, and the obtained Fe3O4@Au NSs were redispersed in 10 mL water. TE ¼ 80 ms, number of excitation ¼ 1. The linear fitting of the inverse T2 relaxation
times (1/T2) as a function of the Fe concentration was used to calculate the T2
2.5. Formation of Fe3O4@Au-PEI NSs relaxivity (r2). CT imaging of the samples dispersed in water with different Au
concentrations (0.01e0.08 M) was performed using a GE LightSpeed VCT imaging
PEI-SH (0.1 g) dissolved in 1 mL water was added into the above aqueous so- system (GE Medical Systems) with 100 kV, 80 mA, and a slice thickness of 0.625 mm.
lution of Fe3O4@Au NSs (10 mL). The mixture was sonicated for 30 min, and then The X-ray attenuation intensity in Hounsfield units (HU) was evaluated by loading
stirred at room temperature for 24 h. After removing the non-absorbed PEI-SH the digital CT images in a standard display program and then selecting a uniform
through 3 cycles of centrifugation (5000 rpm, 5 min)/redispersion in water, the round region of interest on the resultant CT image for each sample. To determine the
Fe3O4@Au-PEI NSs were obtained and redispersed in 10 mL water. photothermal property of the Fe3O4@Au-HA NSs, an aqueous suspension of
Fe3O4@Au-HA NSs with different Au concentrations (0.32e24 mM, 0.3 mL),
2.6. Formation of Fe3O4@Au-HA NSs Fe3O4@Ag seeds (with Fe concentration similar to that of the Fe3O4@Au-HA NSs with
Au concentration of 24 mM), or water was put into a quartz cuvette, and illuminated
HA (520 mg, in 10 mL water) was mixed with EDC (128 mg) and NHS (80 mg)
by a 915 nm laser (Shanghai Xilong Optoelectronics Technology Co. Ltd, Shanghai,
under vigorous magnetic stirring for 3 h. Then, the activated HA solution was
China) with a power density of 1.2 W/cm2 for 300 s. The temperature of different
dropped into the above aqueous suspension of Fe3O4@Au-PEI NSs (10 mL) under
samples was recorded by an online DT-8891E thermocouple thermometer (Shenz-
magnetic stirring. After 3 days, the HA-modified Fe3O4@Au NSs (Fe3O4@Au-HA NSs)
hen Everbest Machinery Industry Co., Ltd., Shenzhen, China) every 5 s.
were subjected to multiple cycles of centrifugation/redispersion in water to remove
the small molecular impurities and the non-reacted HA. Finally, the purified
Fe3O4@Au-HA NSs were redispersed in water and/or phosphate buffered saline (PBS) 2.8. Hemolysis and cytocompatibility assay
before further use.
Hemolysis assay of Fe3O4@Au-HA NSs was carried out according to protocols
described in the literature [55]. Briefly, fresh human blood (kindly provided by
2.7. Characterization techniques
Shanghai First People's Hospital with approval by the ethical committee of Shanghai
Fourier transform infrared (FTIR) spectra were collected using a Nicolet Nexus First People's Hospital) was centrifuged, purified, and 10 times diluted with PBS to
670 FTIR spectrophotometer (Thermo Nicolet Corporation, USA). A Bruker AV400 obtain human red blood cells (HRBCs). Then, 0.1 mL diluted HRBC suspension was
nuclear magnetic resonance spectrometer was used to record the 1H NMR spectra of added to 0.9 mL water (as a positive control), 0.9 mL PBS (as a negative control), and
PEI and PEI-SH samples. The samples were dissolved in D2O before measurements. A 0.9 mL PBS containing Fe3O4@Au-HA NSs at different Au concentrations (0.25, 0.5,
Malvern Zetasizer Nano ZS model ZEN3600 (Worcestershire, U.K.) equipped with a 1.0, 2.0, and 4.0 mM, respectively). The mixtures were gently shaken, and then kept
standard 633 nm laser was used to analyze the hydrodynamic sizes and zeta po- still at room temperature for 2 h. After that, the samples were centrifuged
tentials of the samples. Thermal gravimetric analysis (TGA) was carried out using a (10,000 rpm, 1 min) and the absorbance of the supernatants (hemoglobin) was
TG 209 F1 (NETZSCH Instruments Co., Ltd., Germany) thermal gravimetric analyzer measured by UVevis spectrophotometer. The hemolysis percentage was calculated
at a heating rate of 20  C/min under N2 atmosphere. UVevis spectroscopy was based on the absorbance at 541 nm according to the literature [56].
performed using a Lambda 25 UVevis spectrophotometer (PerkinElmer, Boston, HeLa cells were continuously cultured and passaged in 25 cm2 plates with
MA) and the samples were dispersed in water before measurements. Transmission DMEM supplemented with 10% heat-inactivated FBS, 100 U/mL penicillin, and
electron microscopy (TEM, JEOL 2010F, Japan) was performed at an operating 100 mg/mL streptomycin under 37  C and 5% CO2. For MTT assay, HeLa cells were
voltage of 200 kV. TEM samples were prepared by dropping an aqueous particle seeded into 96-well plates with 200 mL fresh medium at a density of 1  104 cells/
suspension (6 mL) onto a carbon-coated copper grid and air dried before measure- well. After incubation for 12 h to bring the cells to confluence, the medium was
ments. X-ray diffraction (XRD) measurements were performed on a D/max 2550 PC replaced with 200 mL fresh medium containing Fe3O4@Au-HA NSs with different
X-ray diffractometer (Rigaku Cop., Japan) with Cu Ka radiation (l ¼ 0.154056 nm). final Au concentrations (0.2, 0.4, 0.6, 0.8, 1.0, 1.5, and 2.0 mM, respectively) and the
The Fe and Au concentrations of the samples dispersed in water or PBS were cells were incubated for 24 h at 37  C and 5% CO2. Thereafter, MTT solution (20 mL,
analyzed using Leeman Prodigy inductively coupled plasma-optical emission spec- 5 mg/mL in PBS buffer) was added into each well and the cells were incubated for
troscopy (ICP-OES, Hudson, NH). The T2 relaxometry measurements and T2- another 4 h. The assays were carried out according to the manufacturer's instruction.
weighted MR imaging of the samples dispersed in water at different Fe concentra- The absorbance at 570 nm of each well was measured using a Thermo Scientific
tions (0.005e0.08 mM) were performed using an NMI20-Analyst NMR Analyzing Multiskan MK3 ELISA reader (Thermo Scientific, USA). The background subtraction
J. Li et al. / Biomaterials 38 (2015) 10e21 13

at 570 nm was applied to eliminate the influence of the added materials. For each intratumorally injected with PBS (0.1 mL) were used as control. After 10 min, the
sample, mean and standard deviation of five parallel wells were recorded. tumor site was exposed to a 915 nm laser with a power density of 1.2 W/cm2 for
5 min. During the process of laser radiation, a photothermal medical device (GX-300,
2.9. Targeted MR and CT imaging of cancer cells in vitro Shanghai Infratest Electronics Co., Ltd, Shanghai, China) with an infrared camera was
used to obtain the whole-body infrared thermal images at different time points.
HeLa cells seeded in 6-well plates at a density of 2  106 cells/well in DMEM
were brought to confluence after overnight culture. Then the medium was replaced
2.14. In vivo photothermal ablation of HeLa tumors
with fresh medium containing Fe3O4@Au-HA NSs at different Au concentrations (0,
1.25, 2.5, 3.75, and 5.0 mM, respectively) and the cells were incubated at 37  C and 5% HeLa tumor-bearing nude mice were randomly divided into four groups (n ¼ 4
CO2 for 6 h. U87MG cells cultured with MEM were used as control and were treated for each group). The mice were intratumorally injected with 0.1 mL PBS without
in the same manner [16]. Thereafter, the cells were washed 3 times with PBS, laser irradiation (Control group), 0.1 mL PBS and then the tumor site was exposed to
trypsinized, centrifuged, and resuspended in 0.5 mL PBS (containing 0.5% agarose) in a 915 nm laser with a power density of 1.2 W/cm2 for 10 min (Laser group), 0.1 mL
1.5-mL Eppendorf tubes before MR and CT imaging. T2-weighted MR imaging was PBS containing Fe3O4@Au-HA NSs ([Au] ¼ 32 mM) without laser irradiation (NSs
carried out using a 1.5 T Signa HDxt superconductor clinical MR system (GE Medical group), and 0.1 mL PBS containing Fe3O4@Au-HA NSs ([Au] ¼ 32 mM) with laser
Systems, Milwaukee, WI) under the following parameters: point irradiation under similar power density and time period (NSs þ Laser group). The
resolution ¼ 156 mm  156 mm, section thickness ¼ 0.6 mm, TR ¼ 3000 ms, similar treatments were carried out again after the next day (at day 3), but the
TE ¼ 90 ms, and number of excitation ¼ 1. CT scanning was performed using GE volume of PBS or PBS solution containing Fe3O4@Au-HA NSs at the same Au con-
LightSpeed VCT imaging system (GE Medical Systems) under the conditions similar centration was decreased to 0.05 mL. The tumor size and body weight of all mice
to those used to analyze the X-ray attenuation intensity of samples as described were measured and pictures of mice were taken at pre-determined time points. The
above. length and width of the tumors were measured by using a digital vernier caliper and
the tumor volumes were calculated according to the formula of (tumor
2.10. In vitro cellular uptake assay length  (tumor width)2)/2. The survival rate of the mice in each group was
calculated according to the formula of N1/N  100%, where N1 and N represent the
The specific uptake of Fe3O4@Au-HA NSs by HeLa cells overexpressing CD44
number of surviving mice and the number of total mice in each group, respectively.
receptors was investigated by ICP-OES. U87MG cells without CD44 receptor
expression [16] were used as control. Briefly, 5  105 HeLa or U87MG cells per well
were seeded in 12-well plates at 37  C and 5% CO2 the day before the experiment. 2.15. H&E and TUNEL staining
The next day, the medium was replaced with fresh medium containing Fe3O4@Au- Four groups of HeLa tumor-bearing nude mice were treated according to the
HA NSs at the Au concentrations of 1.0 or 2.0 mM. After 6 h incubation, the medium above protocols (n ¼ 1 for each group). After 2 h, the mice were euthanized and the
was discarded and the cells were washed with PBS for 3 times, trypsinized, tumors were removed, fixed in 4% paraformaldehyde, and embedded in paraffin for
centrifuged, and resuspended in 1 mL PBS. A portion of cells (100 mL cell suspension) H&E staining and TUNEL staining according to standard protocols described in our
was counted, and the remaining cells were centrifuged, collected, and lysed using previous work [44]. The morphology of tumor sections after different treatments
aqua regia solution (1.0 mL, nitric acid/hydrochloric acid, v/v ¼ 1:3). Then the was observed using a Leica DM IL LED inverted phase contrast microscope. The
samples were diluted with 1.0 mL PBS, and the Au concentration in different cells number and percentage of TUNEL-positive cells in each sample were counted and
was measured by ICP-OES. determined from five random selected fields.

2.11. In vivo MR and CT imaging of tumors 2.16. Statistical analysis


All animal experiments were performed according to the guidelines of the One-way ANOVA statistical analysis was performed to evaluate the significance
institutional committee for animal care, and also in accordance with the policy of the of the experimental data. 0.05 was selected as the significance level, and the data
National Ministry of Health. Male 4- to 6-week-old BALB/c nude mice (15e20 g) were indicated with (*) for p < 0.05, (**) for p < 0.01, and (***) for p < 0.001,
were purchased from Shanghai Slac Laboratory Animal Center (Shanghai, China). To respectively.
establish a xenografted tumor model, HeLa cells (2  106/mouse) were subcutane-
ously implanted into the back of the nude mouse. When the tumor nodules reached
a volume of 0.12e0.30 cm3 after 10 days, the tumor-bearing mice were used. 3. Results and discussion
For MR imaging of tumors, HeLa tumor-bearing nude mice were first anes-
thetized by intraperitoneal injection of pentobarbital sodium (40 mg/kg), then a PBS 3.1. Synthesis and characterization of Fe3O4@Au-HA NSs
solution of Fe3O4@Au-HA NSs ([Fe] ¼ 5.0 mM, 0.1 mL) was intratumorally injected
into the tumor site. After 10 min, the mice were placed inside a custom-built rodent
receiver coil (Chenguang Med Tech, Shanghai, China) and MR imaging was per- In our previous work, we synthesized Fe3O4@Au composite
formed using a 1.5 T Signa HDxt superconductor clinical MR system. T2-weighted nanoparticles via a one-pot hydrothermal route for in vivo dual
MR images of the mice before and after 10 min post injection were obtained using a mode MR/CT imaging applications [56]. By virtue of the same hy-
conventional spin-echo sequence under the parameters similar to those used for MR
drothermal approach, Fe3O4@Ag seed particles were formed, fol-
imaging of cancer cells in vitro.
For tumor CT imaging, HeLa tumor-bearing nude mice were anesthetized as lowed by exposure to Au growth solution to form the Fe3O4@Au
mentioned above and intratumorally injected with a PBS solution of Fe3O4@Au-HA NSs. To render the Fe3O4@Au NSs with targeting specificity, the
NSs ([Au] ¼ 123.5 mM, 0.1 mL). CT scans were performed before and at 10 min post Fe3O4@Au NSs were first reacted with PEI-SH via AueS bond, and
injection using a GE LightSpeed VCT clinical imaging system with 100 kV, 80 mA, and then reacted with HA via EDC chemical crosslinking of the PEI
a slice thickness of 0.625 mm.
amines onto the surfaces of Fe3O4@Au NSs (Scheme 1).
2.12. In vitro photothermal ablation of HeLa cells
The formation of PEI-SH (Fig. S1, Supporting Information) was
first confirmed by FTIR spectroscopy (Fig. S2, Supporting
HeLa cells were seeded into 96-well plates with 200 mL fresh DMEM at a density
of 1  104 cells/well and incubated for 12 h to allow the cells to be attached before
Information). By comparison with PEI, an obvious peak emerging
photothermal experiments. Then the medium was carefully removed and fresh at 1640 cm1 in the spectrum of PEI-SH is attributed to the char-
medium (200 mL) containing 20 mL Fe3O4@Au-HA NSs at different final Au concen- acteristic peak of amido linkage (Fig. S1). In addition, the appear-
trations (0, 0.1, 0.2, 0.3, or 0.4 mM, respectively) was added into each well. After ance of another weak band at 2570 cm1 suggests the existence of
incubation for another 6 h, the cells were irradiated by a 915 nm laser with an output
eSH in the formed PEI-SH, in agreement with the literature [61]. 1H
power density of 1.2 W/cm2 for 5 and 10 min, respectively. The cell viability was then
measured via MTT assay according to the procedures described above. Mean and NMR was also carried out to confirm the structure of PEI-SH and to
standard deviation for the triplicate wells were reported. In parallel, the morphology quantify the degree of PEI thiolation (Fig. S3, Supporting
of cells treated with PBS or Fe3O4@Au-HA NSs at the Au concentration of 0.4 mM for Information). We can see that the PEI and PEI-SH show similar
6 h, followed by irradiation with a 915 nm laser (at an output power density of 1.2 W/ characteristic peaks except for the peak at 3.4 ppm, which can be
cm2) for 10 min and then rinsing with PBS for 3 times was observed by Leica DM IL
LED inverted phase contrast microscope.
attributed to the proton signal of eCOeCH2eSe in MTG. Based on
the NMR integration, the average number of SH coupled to each PEI
2.13. In vivo photothermal imaging was estimated to be 16.9. Both FTIR and NMR results suggest the
HeLa tumor-bearing nude mice were first anesthetized by intraperitoneal in-
successful formation of PEI-SH.
jection of pentobarbital sodium (40 mg/kg), then a PBS solution of Fe3O4@Au-HA To form the Fe3O4@Ag seed particles, PEI-stabilized Ag NPs (PEI-
NSs ([Au] ¼ 32 mM, 0.1 mL) was intratumorally injected into the mice. The mice Ag NPs) with a mean diameter of 5.3 nm (Fig. S4aeb, Supporting
14 J. Li et al. / Biomaterials 38 (2015) 10e21

Information) were first formed via NaBH4 reduction chemistry, were also recorded to evaluate their long-term colloidal stability
similar to our previous work related to the formation of PEI- (Fig. S6, Supporting Information). It is clear that the hydrodynamic
stabilized Au NPs [56]. In the presence of PEI-stabilized Ag NPs, a size does not have any appreciable changes within a time period of
hydrothermal approach [17,56] was used to synthesize Fe3O4@Ag 2 weeks, indicating their good colloidal stability. Furthermore, the
seeds with a size of 13.1 nm (Fig. S4ced, Supporting Information) colloidal stability of Fe3O4@Au-HA NSs was also checked by
using FeCl2$4H2O as Fe precursor. Then, the formed Fe3O4@Ag exposing them to water, PBS, and cell culture medium (DMEM) for
seeds were developed into Fe3O4@Au NSs by dropping them into at least one month. We show that the NSs are still stable and no
the Au growth solution containing silver ions (Fig. S4eef, precipitation occurs (Fig. S7, Supporting Information), further
Supporting Information). The selected volume and concentration confirming their excellent colloidal stability in different aqueous
of Au growth solution have been optimized to enable the formed media.
NSs with a desirable Fe/Au molar ratio to ensure their subsequent The grafting of PEI and subsequent conjugation of HA onto the
effective multimode imaging and therapy applications. To render surface of Fe3O4@Au NSs were also confirmed by TGA (Fig. S8,
the Fe3O4@Au NSs with good colloidal stability in aqueous solution Supporting Information). Due to the fact that at 700  C, most of
and amine functionality, Fe3O4@Au NSs were washed with water to the organic components have been burned off, we selected 700  C
remove the capping agent CTAB in the solution, followed by to calculate the weight loss of NSs after each step of surface
modification with PEI-SH via AueS bond. The aminated Fe3O4@Au- modification. Compared with Fe3O4@Au NSs just showing a weight
PEI NSs were then modified with HA to be afforded with targeting loss of 0.94%, the PEI grafting via AueS bond formation rendered
specificity to CD44 receptor-overexpressing cancer cells. the NSs with a weight loss of 14.23%. Further conjugation of HA via
Zeta potential measurements were employed to confirm the EDC chemistry resulted in an increased weight loss of 24.36% for
successful modification of HA onto the surface of NSs (Fig. S5a, Fe3O4@Au-HA NSs. The grafting percentages of PEI and HA were
Supporting Information). Fe3O4@Au-PEI NSs dispersed in water calculated to be 13.29% and 10.13%, respectively.
had a positive potential of þ32.7 mV because of the surface UVevis spectroscopy was used to investigate the optical prop-
modification of PEI with abundant amines. After modification with erty of the Fe3O4@Au-HA NSs (Fig. 1a). It is clear that Fe3O4@Au-HA
HA, the zeta potential of Fe3O4@Au-HA NSs was reversed to be NSs exhibit an obvious surface plasmon resonance (SPR) peak at
negative (27.7 mV), suggesting the successful conjugation reac- 870 nm, which is amenable for photothermal therapy applications
tion [16,62,63]. The hydrodynamic sizes of NSs in aqueous solution under NIR laser irradiation [47]. In contrast, Fe3O4@Ag seeds do not
before and after HA modification were measured to be 298.8 and show any obvious absorption features in the same region. It should
339.4 nm (Fig. S5b, Supporting Information), respectively by dy- be noted that the solution of Fe3O4@Au-HA NSs became blue
namic light scattering (DLS). This suggests that the HA coating because of the surface coating of Au shells, which is different from
enlarged the periphery of Fe3O4@Au-PEI NSs, further confirming that of the Fe3O4@Ag seeds (Fig. 1a, inset).
the successful HA conjugation. What's more, the hydrodynamic The morphology and size of the Fe3O4@Au-HA NSs were
sizes of the Fe3O4@Au-HA NSs at different storage time periods investigated by TEM imaging (Fig. 1bee). It can be seen that star-

1.5
a Fe3O4@Ag seeds
b c
Fe3O4@Au-HA NSs
Absorbance

1.0

0.5

0.0
400 600 800 1000
Wavelength (nm)
60
* (111)

d Mean diameter = 119.4 nm


σ = 19.4 nm e f
50 * Au
Fe3O4
Intensity (a.u.)
Frequency (%)

40
(400)

30
* (311)
*(220)
(220)

* (200)

20
* (222)
(311)

(511)
(422)

(440)

10

0 20 30 40 50 60 70 80 90
60 80 100 120 140 160 180
Diameter (nm) 2θ(degree)

Fig. 1. (a) UVevis spectra of Fe3O4@Ag seeds and Fe3O4@Au-HA NSs (inset is the photograph of Fe3O4@Ag seeds and Fe3O4@Au-HA NSs dispersed in water), (b, c) TEM image, (d) size
distribution histogram, (e) high-resolution TEM image, and (f) XRD pattern of the Fe3O4@Au-HA NSs.
J. Li et al. / Biomaterials 38 (2015) 10e21 15

shaped Au shells are coated onto the surface of clustered Fe3O4 NPs, 3.2. T2 MR relaxometry and X-ray attenuation property
and the Fe3O4@Au-HA NSs have a quite uniform size distribution
(Fig. 1b). A close observation of a single Fe3O4@Au-HA NS reveals Fe3O4 NPs have been known to be able to shorten the T2
that several spike-like gold shell crystals are densely and discon- relaxation time of water protons, resulting in MR contrast
tinuously surrounding a cluster of Fe3O4 NPs (Fig. 1c), in agreement enhancement. The transverse relaxivity (r2, the transverse relaxa-
with the literature [34]. The mean diameter of the internal sphere tion rate per mM of Fe) is usually used to quantify the efficiency to
(Fig. 1c) was estimated to be 119.4 ± 19.4 nm (Fig. 1d). High- use Fe3O4 NPs as contrast agents. T2-weighted MR imaging data
resolution TEM image confirmed the crystal structure of the star- show that the developed Fe3O4@Au-HA NSs are able to weaken the
shaped Au shells, as lattices of the crystals can be clearly signal intensity of the MR images with the Fe concentration
observed (Fig. 1e). In addition, the NSs were also found to be sur- (Fig. 2a). By plotting T2 relaxation rate (1/T2) as a function of Fe
rounded with a transparent polymer shell on the outer surface, concentration (Fig. 2b), a linear relationship between the relaxation
which is associated with the PEI coating and HA modification rate and the Fe concentration (R2 ¼ 0.9999) can be found with a
(Fig. 1e). The crystalline structure of the Fe3O4@Au-HA NSs was also slope of 144.39 mM1 s1, which is identified to be the r2 value of
characterized by XRD (Fig. 1f). The diffraction peaks well match the the Fe3O4@Au-HA NSs. It seems that the star-shaped Au shell
planes of Fe3O4 and Au crystals, indicating the formation of crys- coating and the further conjugation of PEI and HA do not appre-
talline Fe3O4@Au composite structures. Meanwhile, due to the core/ ciably weaken the r2 relaxivity when compared to Fe3O4@Au
shell structure, some peaks related to Fe3O4 are not prominent. The composite NPs reported in our previous work [56]. The relatively
elemental composition of the Fe3O4@Au-HA NSs dispersed in water high r2 value of Fe3O4@Au-HA NSs may be due to the fact that water
or PBS was quantitatively measured using ICP-OES, and the Fe/Au protons are accessible to the surface of clustered Fe3O4 NPs in the
molar ratio was estimated to be 1:24.7. core of the NSs via the interstitial spaces between the Au spikes.

a H c

Fe (mM) 0.005 0.01 0.02 0.04 0.08 Au (M) 0.01 0.02 0.04 0.06 0.08
L
600
b d
r2=144.39 mM-1s-1, R2=0.9999
12
450
Hounsfield Unit (HU)
1/T2 (s-1)

8 300

4 150

0
0.00 0.02 0.04 0.06 0.08 0.00 0.02 0.04 0.06 0.08
Fe concentration (mM) Au concentration (M)
e 90 f 70
water seeds 0.32 mM 0.8 mM
1.6 mM 3.2 mM 16 mM 24 mM
80 60
70 50
Temperature (°C)

60 40
ΔT (°C)

50
30
40
20
30
10
20
0
0 50 100 150 200 250 300 0 1 2 3 16 24
Time (s) Au concentration (mM)

Fig. 2. (a) Color T2-weighted MR images and (b) linear fitting of 1/T2 of Fe3O4@Au-HA NSs at different Fe concentrations (The color bar from red to blue indicates the gradual
decrease of MR signal intensity). (c) CT images and (d) X-ray attenuation intensity of the Fe3O4@Au-HA NSs with different Au concentrations. (e) Temperature elevation of water and
the aqueous solution of Fe3O4@Ag seeds ([Fe] ¼ 0.972 mM) or Fe3O4@Au-HA NSs at different Au concentrations (0.32, 0.8, 1.6, 3.2, 16 and 24 mM, respectively) under the irradiation of
a 915 nm laser with a power density of 1.2 W/cm2 as a function of irradiation time. (f) The temperature change (DT) of an aqueous suspension of Fe3O4@Au-HA NSs at different Au
concentrations over a period of 300 s. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
16 J. Li et al. / Biomaterials 38 (2015) 10e21

Our data suggest a great potential to use the Fe3O4@Au-HA NSs as a When the Au concentration increases to 2.0 mM, the Fe3O4@Au-HA
T2 negative contrast agent for MR imaging applications. NSs start to display cytotoxicity and the cell viability can still reach
On the other hand, the potential to use Fe3O4@Au-HA NSs as a 66.5%. Taken together with the results from hemolytic assay, we can
CT contrast agent was confirmed by X-ray attenuation intensity safely conclude that the developed Fe3O4@Au-HA NSs have a good
measurement (Fig. 2c). We show that the CT image of Fe3O4@Au- biocompatibility in the studied concentration range, which is
HA NSs becomes brighter with the Au concentration, correlating essential for their further biomedical applications.
well with the quantitative analysis of the attenuation intensity
change of the Fe3O4@Au-HA NSs as a function of Au concentration 3.5. In vitro targeted MR and CT imaging of cancer cells
(Fig. 2d). Since AuNPs are known to be a good CT contrast agent
[20], there is no doubt to conclude that the Fe3O4@Au-HA NSs can We next investigated the potential to use the developed
be potentially used as a contrast agent for CT imaging applications Fe3O4@Au-HA NSs as a nanoprobe for targeted dual mode MR/CT
due to the integrated Au component, similar to our previous study imaging of cancer cells in vitro. In this study, we selected HA as a
related to Fe3O4@Au composite NPs [56,57]. targeting ligand. HA, a member of the glycosaminoglycan family
composed of repeating disaccharide units of D-glucuronic acid and
3.3. Photothermal property of Fe3O4@Au-HA NSs N-acetyl-D-glucosamine, has been recognized as an attractive tar-
geting ligand that can bind to CD44 receptor-overexpressing cancer
The strong SPR absorption of Fe3O4@Au-HA NSs in the NIR re- cells [16,62,64]. U87MG cells without CD44 receptor over-
gion drove us to explore their photothermal property. The tem- expression were used as control. After incubation with the NSs at
perature change of an aqueous suspension of the Fe3O4@Au-HA NSs different Fe or Au concentrations for 6 h, the cells were subjected to
as a function of Au concentration (0.32e24 mM) under laser irra- T2-weighted MR imaging and CT imaging, respectively (Fig. 3aeb).
diation for 300 s was monitored (Fig. 2e). It is clear that the It can be seen that the MR signal intensity of both HeLa and U87MG
Fe3O4@Au-HA NSs with higher Au concentration result in a more cells decreases with the Fe concentration, however the decreasing
prominent temperature increase, and the solution temperature trend of U87MG cells is much less than that of HeLa cells under
reaches 81.2  C at the Au concentration of 24 mM. In contrast, the similar conditions (Fig. 3a). This can be further confirmed by
water and the aqueous suspension of Fe3O4@Ag seeds (with the quantitative analysis of the signal intensity of the cells (Fig. 3c),
same Fe concentration as the Fe3O4@Au-HA NSs at the Au con- where the signal intensity of the treated HeLa cells was much lower
centration of 24 mM) do not have obvious temperature increase than that of the U87MG cells under a given Fe concentration
under similar experimental conditions. The plot of temperature (p < 0.01).
change (DT) over a time period of 300 s versus Au concentration For CT imaging, due to the fact that it is difficult to visually
shows that the temperature only increase 5.0 and 10.1  C for water differentiate the brightness of the CT images of the cells treated
and Fe3O4@Ag seeds, respectively. With the increase of Au con- with the NSs at different Au concentrations [19,65], it is essential to
centration (from 0.32 to 24 mM), the aqueous suspension has a perform quantitative analysis of the CT signal intensity using the
temperature increase of 15.4, 24.8, 34.8, 47.1, 57.0, and 62.8  C, manufacturer's standard display program (Fig. 3d). It can be seen
respectively (Fig. 2f). Our results indicate that Fe3O4@Au-HA NSs that the CT values of both HeLa and U87MG cells treated with the
are able to generate heat rapidly and efficiently upon laser irradi- Fe3O4@Au-HA NSs increase with the Au concentration. Apparently,
ation. It should be noted that the developed Fe3O4@Au-HA NSs still at a relatively high Au concentration, the CT value of HeLa cells with
have a very good colloidal stability during and after the laser irra- CD44 receptor-overexpression is much higher than that of U87MG
diation process, which is amenable for their effective photothermal cells at the same Au concentration (p < 0.05). These results suggest
therapy applications. that the developed Fe3O4@Au-NSs have a high affinity to CD44
receptor-overexpressing cancer cells, thereby enabling targeted
3.4. Hemolytic assay and cytotoxicity assay dual mode MR/CT imaging of cancer cells via receptor-mediated
active targeting pathway.
For biomedical applications, hemocompatibility and cyto-
compatibility of the developed Fe3O4@Au-HA NSs should be eval- 3.6. In vitro cellular uptake assay
uated. Hemolytic assay was used to assess the hemocompatibility
of the Fe3O4@Au-HA NSs (Fig. S9, Supporting Information). We can The targeted uptake of the Fe3O4@Au-HA NSs by HeLa cells was
see that the Fe3O4@Au-HA NSs at the Au concentration ranging further quantitatively confirmed by ICP-OES analysis of the Au
from 0.25 to 4.0 mM do not cause any obvious hemolysis effect uptake (Fig. 3e). It is clear that for both HeLa and U87MG cells, the
when compared with the negative control (PBS). In contrast, the treatment of Fe3O4@Au-HA NSs with a higher concentration leads
positive control of water induces a significant hemolysis of HRBCs. to a higher Au uptake within the cells. Importantly, at the same Au
Based on the absorbance of the supernatant at 541 nm that is concentration (1.0 and 2.0 mM), the Au uptake in HeLa cells over-
associated with the absorption of the released hemoglobin from expressing CD44 receptors is significantly higher than that in
HRBCs, the hemolysis percentages of HRBCs in the presence of U87MG cells without CD44 receptor overexpression. This further
Fe3O4@Au-HA NSs at the Au concentrations of 0.25, 0.5, 1.0, 2.0, and confirmed the role played by HA-mediated targeting that enables
4.0 mM were calculated to be 0.26%, 0.62%, 0.79%, 1.39%, and 1.65%, specific uptake the Fe3O4@Au-HA NSs, in agreement with our
respectively (Fig. S9, inset), which are all less than the threshold previous work [16].
value of 5% [19]. This suggests that the developed Fe3O4@Au-HA
NSs have a good hemocompatibility in the studied concentration 3.7. In vivo MR and CT imaging of a xenografted tumor model
range.
The cytotoxicity of the Fe3O4@Au-HA NSs was evaluated by MTT With the excellent targeting specificity of the Fe3O4@Au-HA NSs
viability assay of HeLa cells treated with the particles (Fig. S10, for in vitro MR/CT imaging of cancer cells, we next explored the
Supporting Information). It can be seen that the viability of HeLa potential to use the NSs as a contrast agent for MR/CT imaging of a
cells does not have any appreciable changes after incubation with xenografted tumor model. The tumor-bearing mice intratumorally
the Fe3O4@Au-HA NSs at the Au concentrations of 0.2, 0.4, 0.6, 0.8, injected with the Fe3O4@Au-HA NSs before and at 10 min post in-
1.0, and 1.5 mM, respectively, when compared with the PBS control. jection were imaged by MR and CT, respectively (Fig. 4aeb). We can
J. Li et al. / Biomaterials 38 (2015) 10e21 17

a [Fe]/mM 0 0.05 0.10 0.15 0.20 b [Au]/mM 0 1.25 2.50 3.75 5.00
U87MG cells U87MG cells

HeLa cells HeLa cells

c 1600 d 120
U87MG cells ***
** HeLa cells U87MG cells
*** HeLa cells
1200 90

Hounsfield unit (HU)


*** **
Signal intensity

*** *
800 60

400 30

0 0
0 0.05 0.10 0.15 0.20 0 1.25 2.50 3.75 5.00
Fe concentration (mM) Au concentration (mM)
e 420 f 120
0 min 5 min 10 min
***
Cellular Au uptake (pg/cell)

U87MG cells
*** *** *** ***
HeLa cells 100 *** *** *** ***
*** *** ***
Cell viability (%)

280 80
***
60

140 40

20
0
0
Control 1.0 2.0 0.0 0.1 0.2 0.3 0.4
Au concentration (mM) Au concentration (mM)

Fig. 3. (a) T2-weighted MR images, (b) CT images, (c) MR signal intensity, and (d) CT values of HeLa and U87MG cells after treated with Fe3O4@Au-HA NSs at different Fe or Au
concentrations for 6 h. (e) The Au uptake by HeLa and U87MG cells treated with Fe3O4@Au-HA NSs at the Au concentration of 1.0 and 2.0 mM for 6 h. (f) MTT viability assay of HeLa
cells after treatment with the Fe3O4@Au-HA NSs at different Au concentrations and different laser irradiation time periods.

clearly see that the tumor region darkens obviously in a typical T2- potential to use these NSs for photothermal ablation of cancer cells
weighted MR image at 10 min post injection (Fig. 4a), when in vitro. The viability of HeLa cells treated with the Fe3O4@Au-HA
compared to before injection. The quantitative signal intensity NSs at different Au concentrations under laser irradiation was
analysis reveals that the MR signal intensity of the tumor region assessed by MTT assay (Fig. 3f). In all cases, HeLa cells without
before injection (641.0) dramatically decreases to 41.1 at 10 min treatment with Fe3O4@Au-HA NSs under laser irradiation (5 or
post-injection. Likewise, in the CT images, the intratumoral 10 min), or HeLa cells treated with Fe3O4@Au-HA NSs at different
administration of the Fe3O4@Au-HA NSs makes the tumor region Au concentrations (0.1e0.4 mM) without laser irradiation do not
much brighter at 10 min post injection (Fig. 4b). To quantify the CT display any appreciable viability changes when compared to the
contrast enhancement, the CT values of the tumor region before and PBS control. In contrast, when HeLa cells treated with the
at 10 min post injection were measured. The CT value of the tumor Fe3O4@Au-HA NSs were irradiated by a 915 nm NIR laser for 5 min,
region was estimated to be 2303.3 HU at 10 min post injection, much the cell viability started to have a significant decrease at the Au
higher than that before injection (157.4 HU). This suggests that the concentration of 0.2 mM (p < 0.001). Further extension of the laser
intratumoral injection of the NSs leads to a quite uniform distribu- irradiation time to 10 min led to more prominent cell death under
tion of the particles within the tumor region, allowing for effective all the studied Au concentrations (p < 0.001). The cells treated with
MR/CT imaging of the whole tumor. These results suggest that the Fe3O4@Au-HA NSs at a higher Au concentration under a longer time
formed Fe3O4@Au-HA NSs have a great potential to be used as a of laser irradiation have more decreased viability. Around 62.2%
contrast agent for in vivo tumor MR/CT imaging. HeLa cells treated with the Fe3O4@Au-HA NSs at an Au concen-
tration of 0.4 mM were able to be killed under laser irradiation for
3.8. In vitro photothermal ablation of cancer cells 10 min. Our results suggest a great potential to use Fe3O4@Au-HA
NSs for photothermal ablation of cancer cells.
The high photothermal conversion efficiency and the targeting The photothermal therapeutic efficacy of Fe3O4@Au-HA NSs was
specificity of the Fe3O4@Au-HA NSs drove us to investigate the further evaluated by optical microscopic observation of cell
18 J. Li et al. / Biomaterials 38 (2015) 10e21

a b

0 min 10 min 0 min 10 min

c (d) 70
Region 11 Region 12

60

Temperature (°C)
50

40

30

20
0 50 100 150 200 250 300
Time (s)

Fig. 4. (a) T2-weighted MR images and (b) CT images of the tumors before injection and at 10 min post intratumoral injection of 0.1 mL PBS solution containing Fe3O4@Au-HA NSs
([Fe] ¼ 5.0 mM, [Au] ¼ 123.5 mM). (c) Photothermal images of two tumor-bearing mice injected with 0.1 mL PBS (the left mouse, indicated region 11) or 0.1 mL PBS containing
Fe3O4@Au-HA NSs ([Au] ¼ 32 mM, the right mouse, indicated region 12), respectively, followed by irradiation with a 915 nm laser (1.2 W/cm2) at a time point of 0, 1.5 min, and 5 min,
respectively. (d) The temperature profiles in Regions 11 and 12 as a function of the laser irradiation time.

morphology (Fig. S11, Supporting Information). The cells treated contrast, Region 12 injected with Fe3O4@Au-HA NSs has a rapid
with laser alone or with Fe3O4@Au-HA NSs without laser irradia- temperature increase from 32.8 to 58.9  C after 90 s laser irradia-
tion have similar attachment morphology, with cell numbers tion, and then remains above 50  C for the following 205 s. Our data
approximately similar to the PBS control (Fig. S11aec). In contrast, suggest a great potential to use the developed Fe3O4@Au-HA NSs
when the cells were incubated with Fe3O4@Au-HA NSs and then for thermal imaging of tumors.
irradiated by the laser for 10 min, most of the cells died and only a The high local temperature in the tumor region after treatment
small portion of adherent cells left after the washing step. Our re- with the Fe3O4@Au-HA NSs under laser irradiation is believed to be
sults suggest that the treatment of laser or Fe3O4@Au-HA NSs alone able to kill the tumor cells. We then investigated the photothermal
does not exert any therapeutic efficacy to the cancer cells, and the therapeutic efficacy of Fe3O4@Au-HA NSs by measuring the vol-
developed Fe3O4@Au-HA NSs are able to effectively ablate cancer umes of the tumors after different treatments (Fig. 5a). It is obvious
cells under laser irradiation, corroborating the MTT results. that the volumes of tumors treated with laser alone or Fe3O4@Au-
HA NSs alone increase with the time, similar to the control group,
3.9. In vivo photothermal imaging and ablation of a xenografted suggesting that the laser irradiation alone or injection of
tumor model Fe3O4@Au-HA NSs without laser irradiation does not have any
impact on the tumor growth. In contrast, the tumors treated with
We next explored the feasibility to use the developed Fe3O4@Au-HA NSs under laser irradiation are able to be completely
Fe3O4@Au-HA NSs for photothermal imaging and ablation of a inhibited. On day 19, the tumor tissue almost completely dis-
xenografted tumor model in vivo. Two HeLa tumor-bearing mice appeared in the mice after the photothermal therapy, which is
were intratumorally injected with 0.1 mL PBS or 0.1 mL PBS con- significantly different from other groups (Fig. S12, Supporting
taining Fe3O4@Au-HA NSs with Au concentration of 32 mM, Information). Furthermore, mice in different treatment groups
respectively (Regions 11 and 12 in Fig. 4c). After 10 min, Regions 11 maintained their weights during the experimental time period
and 12 were irradiated under a 915 nm laser (1.2 W/cm2) for 300 s. (Fig. 5b), implying that the laser irradiation alone, the injection of
The full-body thermal images of the mice were captured using an the Fe3O4@Au-HA NSs alone, or the combination of the above two is
infrared camera. It is clear that only a slight temperature change unable to generate toxicity to the mice. To further investigate the
was detected at Region 11. In contrast, Region 12 displays a sig- photothermal therapeutic efficacy of the Fe3O4@Au-HA NSs, the
nificant temperature increase due to the injection of the Fe3O4@Au- survival rate of the mice in the four groups was evaluated (Fig. 5c).
HA NSs. The tumor temperature was also monitored as a function of It is obvious that the mice treated with the Fe3O4@Au-HA NSs un-
the laser irradiation time (Fig. 4d). It can be observed that Region 11 der laser irradiation maintain a 100% survival rate after 60 days,
injected with PBS only has a slight temperature increase of 4.5  C which is significantly higher than the mice in the other three
and remains below 36.8  C during the laser irradiation. In sharp groups. The survival rate of the mice without treatment, treated
J. Li et al. / Biomaterials 38 (2015) 10e21 19

a 12 b 35 c
Control Control
Laser Laser 100
Relative tumor volume

NSs NSs
9

Body weight (g)


30 NSs + Laser

Survival rate (%)


NSs + Laser
75
Control
Laser
6 25 NSs
50 NSs + Laser

3 20 25

0 15 0
0 4 8 12 16 20 0 4 8 12 16 20 0 10 20 30 40 50 60
Time (day) Time (day) Time (day)

Fig. 5. The relative tumor volume (a), body weight (b), and survival rate (c) of HeLa tumor-bearing mice as a function of time post treatment.

with Fe3O4@Au-HA NSs only, and treated with laser only is 25%, 0% Information). These results demonstrate that the developed
and 0%, respectively. Our results suggest that the developed Fe3O4@Au-HA NSs are able to be used as an efficient nanoplatform
Fe3O4@Au-HA NSs have a great potential to be used for photo- for photothermal ablation of tumors in vivo.
thermal therapy of tumors.
4. Conclusion
3.10. H&E and TUNEL staining
In summary, we developed a convenient approach to synthe-
The photothermal ablation of tumors using Fe3O4@Au-HA NSs sizing Fe3O4@Au-HA NSs with a quite uniform morphology for
was further confirmed by histological examination of tumor sec- multi-mode imaging and photothermal therapy of tumors. The
tions after different treatments (Fig. 6). H&E staining of the tumor hydrothermally synthesized Fe3O4@Ag seeds are able to be
sections shows that the tumors treated with laser alone and with deposited with star-shaped Au shells that can be modified with PEI
the Fe3O4@Au-HA NSs alone display well-shaped tumor cells via AueS bond. Likewise, the PEI-mediated reaction can be used for
without the appearance of necrosis region, similar to the control HA conjugation onto the surface of the NSs to render them with
group treated with PBS. However, obvious necrosis area can be seen targeting specificity to CD44 receptor-overexpressing cancer cells.
in the tumor tissue treated with Fe3O4@Au-HA NSs under laser The formed Fe3O4@Au-HA NSs are water dispersible, colloidally
irradiation (Fig. 6a). The photothermal ablation of tumors using stable, and biocompatible in the given concentration range.
Fe3O4@Au-HA NSs was further characterized by TUNEL staining Importantly, the Fe3O4@Au-HA NSs are able to be used as a multi-
(Fig. 6b). It is obvious that only sparse positive staining of apoptotic functional nanoplatform for MR/CT imaging of cancer cells in vitro
cells appear in the tumor sections treated with PBS, laser alone, or and xenografted tumor model in vivo. The NIR absorption property
Fe3O4@Au-HA NSs alone. In contrast, a large area of positive stained of the NSs enables them to be used as a platform for photothermal
apoptotic cells can be seen after the tumors were treated with therapy of cancer cells in vitro and the xenografted tumor model
Fe3O4@Au-HA NSs under laser irradiation. Additional quantitative in vivo, as well as additional thermal imaging of tumors. The
analysis of TUNEL-stained tumor sections reveals that the apoptosis developed Fe3O4@Au-HA NSs may be used as a multifunctional
rate of the tumors in the Control, Laser, NSs, and NSs þ Laser groups nanoplatform for efficient theranostics of different types of CD44
is 6.4%, 12.3%, 7.8%, and 88.6%, respectively (Fig. S13, Supporting receptor-overexpressing cancer.

Control Laser NSs NSs + Laser


a

Fig. 6. Representative H&E staining images (a) and TUNEL assay images (b) of xenografted HeLa tumors with different treatments. The scale bars in each panel of (a) and (b)
represent 50 and 100 mm, respectively.
20 J. Li et al. / Biomaterials 38 (2015) 10e21

Acknowledgments [23] Wen S, Li K, Cai H, Chen Q, Shen M, Huang Y, et al. Multifunctional dendrimer-
entrapped gold nanoparticles for dual mode CT/MR imaging applications.
Biomaterials 2013;34:1570e80.
This research is financially supported by the National Natural [24] Huang X, El-Sayed IH, Qian W, El-Sayed MA. Cancer cell imaging and photo-
Science Foundation of China (21273032, 81101150, and 81371623), thermal therapy in the near-infrared region by using gold nanorods. J Am
the Fund of the Science and Technology Commission of Shanghai Chem Soc 2006;128:2115e20.
[25] Seo S-H, Kim B-M, Joe A, Han H-W, Chen X, Cheng Z, et al. NIR-light-induced
Municipality (11nm0506400 for X. S. and 12520705500 for M. S.), surface-enhanced Raman scattering for detection and photothermal/photo-
the Sino-German Center for Research Promotion (GZ899), and the dynamic therapy of cancer cells using methylene blue-embedded gold
Program for Professor of Special Appointment (Eastern Scholar) at nanorod@SiO2 nanocomposites. Biomaterials 2014;35:3309e18.
[26] Ji X, Shao R, Elliott AM, Stafford RJ, Esparza-Coss E, Bankson JA, et al.
Shanghai Institutions of Higher Learning. Bifunctional gold nanoshells with a superparamagnetic iron oxide-silica core
suitable for both MR imaging and photothermal therapy. J Phys Chem C
2007;111:6245e51.
Appendix A. Supplementary data [27] Ma Y, Liang X, Tong S, Bao G, Ren Q, Dai Z. Gold nanoshell nanomicelles for
potential magnetic resonance imaging, light-triggered drug release, and
photothermal therapy. Adv Funct Mater 2013;23:815e22.
Supplementary data related to this article can be found online at [28] Han J, Li J, Jia W, Yao L, Li X, Jiang L, et al. Photothermal therapy of cancer cells
http://dx.doi.org/10.1016/j.biomaterials.2014.10.065. using novel hollow gold nanoflowers. Int J Nanomed 2014;9:517e26.
[29] Yang J, Shen D, Zhou L, Li W, Li X, Yao C, et al. Spatially confined fabrication of
coreeshell gold nanocages@mesoporous silica for near-infrared controlled
photothermal drug release. Chem Mater 2013;25:3030e7.
References [30] Gao L, Fei J, Zhao J, Li H, Cui Y, Li J. Hypocrellin-loaded gold nanocages with
high two-photon efficiency for photothermal/photodynamic cancer therapy
[1] Taylor-Pashow KM, Della Rocca J, Huxford RC, Lin W. Hybrid nanomaterials for in vitro. ACS Nano 2012;6:8030e40.
biomedical applications. Chem Commun 2010;46:5832e49. [31] Yuan H, Fales AM, Vo-Dinh T. TAT peptide-functionalized gold nanostars:
[2] Shen M, Shi X. Dendrimer-based organic/inorganic hybrid nanoparticles in enhanced intracellular delivery and efficient NIR photothermal therapy using
biomedical applications. Nanoscale 2010;2:1596e610. ultralow irradiance. J Am Chem Soc 2012;134:11358e61.
[3] Leung KC-F, Xuan S, Zhu X, Wang D, Chak C-P, Lee S-F, et al. Gold and iron [32] Wang S, Huang P, Nie L, Xing R, Liu D, Wang Z, et al. Single continuous wave
oxide hybrid nanocomposite materials. Chem Soc Rev 2012;41:1911e28. laser induced photodynamic/plasmonic photothermal therapy using
[4] Du C, Zhao J, Fei J, Gao L, Cui W, Yang Y, et al. Alginate-based microcapsules photosensitizer-functionalized gold nanostars. Adv Mater 2013;25:3055e61.
with a molecule recognition linker and photosensitizer for the combined [33] Hwang S, Nam J, Song J, Jung S, Hur J, Im K, et al. Sub 6 nanometer plasmonic
cancer treatment. Chem Asian J 2013;8:736e42. gold nanoparticle for pH-responsive near-infrared photothermal cancer
[5] Qi W, Duan L, Li J. Fabrication of glucose-sensitive protein microcapsules and therapy. New J Chem 2014:918e22.
their applications. Soft Matter 2011;7:1571e6. [34] Li C, Chen T, Ocsoy I, Zhu G, Yasun E, You M, et al. Gold-coated Fe3O4 nano-
[6] Li D, He Q, Yang Y, Mo €hwald H, Li J. Two-stage pH response of poly (4- roses with five unique functions for cancer cell targeting, imaging, and ther-
vinylpyridine) grafted gold nanoparticles. Macromolecules 2008;41:7254e6. apy. Adv Funct Mater 2013;24:1772e80.
[7] Qi W, Wang A, Yang Y, Du M, Bouchu MN, Boullanger P, et al. The lectin [35] Tian Q, Wang Q, Yao KX, Teng B, Zhang J, Yang S, et al. Multifunctional pol-
binding and targetable cellular uptake of lipid-coated polysaccharide micro- ypyrrole@Fe3O4 nanoparticles for dual-modal imaging and in vivo photo-
capsules. J Mater Chem 2010;20:2121e7. thermal cancer therapy. Small 2013;10:1063e8.
[8] Liu HL, Sonn CH, Wu JH, Lee K-M, Kim YK. Synthesis of streptavidin-FITC- [36] Zhou T, Wu B, Xing D. Bio-modified Fe3O4 core/Au shell nanoparticles for
conjugated coreeshell Fe3O4-Au nanocrystals and their application for the targeting and multimodal imaging of cancer cells. J Mater Chem 2012;22:
purification of CD4þ lymphocytes. Biomaterials 2008;29:4003e11. 470e7.
[9] Bao J, Chen W, Liu T, Zhu Y, Jin P, Wang L, et al. Bifunctional Au-Fe3O4 [37] Cai H, Li K, Shen M, Wen S, Luo Y, Peng C, et al. Facile assembly of Fe3O4@Au
nanoparticles for protein separation. ACS Nano 2007;1:293e8. nanocomposite particles for dual mode magnetic resonance and computed
[10] Samanta B, Yan H, Fischer NO, Shi J, Jerry DJ, Rotello VM. Protein-passivated tomography imaging applications. J Mater Chem 2012;22:15110e20.
Fe3O4 nanoparticles: low toxicity and rapid heating for thermal therapy. [38] Tian Q, Hu J, Zhu Y, Zou R, Chen Z, Yang S, et al. Sub-10 nm Fe3O4@Cu2exS
J Mater Chem 2008;18:1204e8. coreeshell nanoparticles for dual-modal imaging and photothermal therapy.
[11] Shi D, Cho HS, Chen Y, Xu H, Gu H, Lian J, et al. Fluorescent polystyreneeFe3O4 J Am Chem Soc 2013;135:8571e7.
composite nanospheres for in vivo imaging and hyperthermia. Adv Mater [39] Wang Y, Guo R, Cao X, Shen M, Shi X. Encapsulation of 2-methoxyestradiol
2009;21:2170e3. within multifunctional poly (amidoamine) dendrimers for targeted cancer
[12] Wang C, Daimon H, Sun S. Dumbbell-like PtFe3O4 nanoparticles and their therapy. Biomaterials 2011;32:3322e9.
enhanced catalysis for oxygen reduction reaction. Nano Lett 2009;9:1493e6. [40] Zhang X-y, Chen J, Zheng Y-f, Gao X-l, Kang Y, Liu J-c, et al. Follicle-stimulating
[13] Pan B, Cui D, Sheng Y, Ozkan C, Gao F, He R, et al. Dendrimer-modified hormone peptide can facilitate paclitaxel nanoparticles to target ovarian
magnetic nanoparticles enhance efficiency of gene delivery system. Cancer carcinoma in vivo. Cancer Res 2009;69:6506e14.
Res 2007;67:8156e63. [41] Feng W, Zhou X, He C, Qiu K, Nie W, Chen L, et al. Polyelectrolyte multilayer
[14] Chen Y, Chen H, Zeng D, Tian Y, Chen F, Feng J, et al. Core/shell structured functionalized mesoporous silica nanoparticles for pH-responsive drug de-
hollow mesoporous nanocapsules: a potential platform for simultaneous cell livery: layer thickness-dependent release profiles and biocompatibility.
imaging and anticancer drug delivery. ACS Nano 2010;4:6001e13. J Mater Chem B 2013;1:5886e98.
[15] Li J, Zheng L, Cai H, Sun W, Shen M, Zhang G, et al. Polyethyleneimine- [42] Li D, Li C, Wang A, He Q, Li J. Hierarchical gold/copolymer nanostructures as
mediated synthesis of folic acid-targeted iron oxide nanoparticles for in vivo hydrophobic nano tanks for drug encapsulation. J Mater Chem 2010;20:
tumor MR imaging. Biomaterials 2013;34:8382e92. 7782e7.
[16] Li J, He Y, Sun W, Luo Y, Cai H, Pan Y, et al. Hyaluronic acid-modified hy- [43] Zheng Y, Fu F, Zhang M, Shen M, Zhu M, Shi X. Multifunctional dendrimers
drothermally synthesized iron oxide nanoparticles for targeted tumor MR modified with alpha-tocopheryl succinate for targeted cancer therapy. Med
imaging. Biomaterials 2014;35:3666e77. Chem Commun 2014;5:879e85.
[17] Cai H, An X, Cui J, Li J, Wen S, Li K, et al. Facile hydrothermal synthesis and [44] Zhu J, Zheng L, Wen S, Tang Y, Shen M, Zhang G, et al. Targeted cancer
surface functionalization of polyethyleneimine-coated iron oxide nano- theranostics using alpha-tocopheryl succinate-conjugated multifunctional
particles for biomedical applications. ACS Appl Mater Interfaces 2013;5: dendrimer-entrapped gold nanoparticles. Biomaterials 2014;35:7635e46.
1722e31. [45] Zhu J, Shi X. Dendrimer-based nanodevices for targeted drug delivery appli-
[18] Guo R, Wang H, Peng C, Shen M, Pan M, Cao X, et al. X-ray attenuation cations. J Mater Chem 2013;1:4199e211.
property of dendrimer-entrapped gold nanoparticles. J Phys Chem C [46] Zheng L, Zhu J, Shen M, Chen X, Baker Jr JR, Wang SH, et al. Targeted cancer
2010;114:50e6. cell inhibition using multifunctional dendrimer-entrapped gold nanoparticles.
[19] Peng C, Li K, Cao X, Xiao T, Hon W, Zheng L, et al. Facile formation of Med Chem Commun 2013;4:1001e5.
dendrimer-stabilized gold nanoparticles modified with diatrizoic acid for [47] Zhou Z, Kong B, Yu C, Shi X, Wang M, Liu W, et al. Tungsten oxide nanorods:
enhanced computed tomography imaging applications. Nanoscale 2012;4: an efficient nanoplatform for tumor CT imaging and photothermal therapy.
6768e78. Sci Rep 2014;4:3653.
[20] Peng C, Zheng L, Chen Q, Shen M, Guo R, Wang H, et al. PEGylated dendrimer- [48] Li B, Wang Q, Zou R, Liu X, Xu K, Li W, et al. Cu7.2S4 nanocrystals: a novel
entrapped gold nanoparticles for in vivo blood pool and tumor imaging by photothermal agent with a 56.7% photothermal conversion efficiency for
computed tomography. Biomaterials 2012;33:1107e19. photothermal therapy of cancer cells. Nanoscale 2014;6:3274e82.
[21] Wang H, Zheng L, Peng C, Guo R, Shen M, Shi X, et al. Computed tomography [49] Mackey MA, Ali MR, Austin LA, Near RD, El-Sayed MA. The most effective gold
imaging of cancer cells using acetylated dendrimer-entrapped gold nano- nanorod size for plasmonic photothermal therapy: theory and in vitro ex-
particles. Biomaterials 2011;32:2979e88. periments. J Phys Chem B 2014;118:1319e26.
[22] Wang H, Zheng L, Peng C, Shen M, Shi X, Zhang G. Folic acid-modified den- [50] Chen H, Zhang X, Dai S, Ma Y, Cui S, Achilefu S, et al. Multifunctional gold
drimer-entrapped gold nanoparticles as nanoprobes for targeted CT imaging nanostar conjugates for tumor imaging and combined photothermal and
of human lung adenocarcinoma. Biomaterials 2013;34:470e80. chemo-therapy. Theranostics 2013;3:633e49.
J. Li et al. / Biomaterials 38 (2015) 10e21 21

[51] Peng C, Qin J, Zhou B, Chen Q, Shen M, Zhu M, et al. Targeted tumor CT im- [59] Wei Q, Song H-M, Leonov AP, Hale JA, Oh D, Ong QK, et al. Gyromagnetic
aging using folic acid-modified PEGylated dendrimer-entrapped gold nano- imaging: dynamic optical contrast using gold nanostars with magnetic cores.
particles. Polym Chem 2013;4:4412e24. J Am Chem Soc 2009;131:9728e34.
[52] Chen Q, Li K, Wen S, Liu H, Peng C, Cai H, et al. Targeted CT/MR dual mode [60] Fan Z, Senapati D, Singh AK, Ray PC. Theranostic magnetic core-plasmonic
imaging of tumors using multifunctional dendrimer-entrapped gold nano- shell star shape nanoparticle for the isolation of targeted rare tumor cells
particles. Biomaterials 2013;34:5200e9. from whole blood, fluorescence imaging, and photothermal destruction of
[53] Wang SH, Shi X, Van Antwerp M, Cao Z, Swanson SD, Bi X, et al. Dendrimer- cancer. Mol Pharm 2013;10:857e66.
functionalized iron oxide nanoparticles for specific targeting and imaging of [61] Pan B, Gao F, Ao L, Tian H, He R, Cui D. Controlled self-assembly of thiol-
cancer cells. Adv Funct Mater 2007;17:3043e50. terminated poly(amidoamine) dendrimer and gold nanoparticles. Colloid
[54] Shi X, Wang SH, Swanson SD, Ge S, Cao Z, Van Antwerp ME, et al. Dendrimer- Surf A-Physicochem Eng Asp 2005;259:89e94.
functionalized shell-crosslinked iron oxide nanoparticles for in-vivo magnetic [62] Yu M, Jambhrunkar S, Thorn P, Chen J, Gu W, Yu C. Hyaluronic acid modified
resonance imaging of tumors. Adv Mater 2008;20:1671e8. mesoporous silica nanoparticles for targeted drug delivery to CD44-
[55] Shen M, Cai H, Wang X, Cao X, Li K, Wang SH, et al. Facile one-pot preparation, overexpressing cancer cells. Nanoscale 2013;5:178e83.
surface functionalization, and toxicity assay of APTS-coated iron oxide [63] Choi KY, Min KH, Na JH, Choi K, Kim K, Park JH, et al. Self-assembled hyal-
nanoparticles. Nanotechnology 2012;23:105601. uronic acid nanoparticles as a potential drug carrier for cancer therapy:
[56] Li J, Zheng L, Cai H, Sun W, Shen M, Zhang G, et al. Facile one-pot synthesis of synthesis, characterization, and in vivo biodistribution. J Mater Chem
Fe3O4@Au composite nanoparticles for dual-mode MR/CT imaging applica- 2009;19:4102e7.
tions. ACS Appl Mater Interfaces 2013;5:10357e66. [64] Cho H-J, Yoon I-S, Yoon HY, Koo H, Jin Y-J, Ko S-H, et al. Polyethylene glycol-
[57] Hu Y, Li J, Shen M, Shi X. Formation of multifunctional Fe3O4/Au composite conjugated hyaluronic acid-ceramide self-assembled nanoparticles for tar-
nanoparticles for dual mode MR/CT imaging applications. Chin Phys B geted delivery of doxorubicin. Biomaterials 2012;33:1190e200.
2014;23:078704. [65] Liu H, Xu Y, Wen S, Chen Q, Zheng L, Shen M, et al. Targeted tumor computed
[58] Baginskiy I, Lai T-C, Cheng L-C, Chan Y-C, Yang K-Y, Liu R-S, et al. Chitosan- tomography imaging using low-generation dendrimer-stabilized gold nano-
modified stable colloidal gold nanostars for the photothermolysis of cancer particles. Chem Eur J 2013;19:6409e16.
cells. J Phys Chem C 2013;117:2396e410.

You might also like