Anda di halaman 1dari 8

www.advmat.

de
www.MaterialsViews.com

Core–Shell MnSe@Bi2Se3 Fabricated via a Cation Exchange


COMMUNICATION

Method as Novel Nanotheranostics for Multimodal


Imaging and Synergistic Thermoradiotherapy
Guosheng Song, Chao Liang, Hua Gong, Meifang Li, Xianchuang Zheng, Liang Cheng,
Kai Yang,* Xiqun Jiang, and Zhuang Liu*

Radiation therapy (RT), which is an extensively used cancer light induced photothermal therapy (PTT), to realize combina-
treatment strategy accompanying with surgery and chemo- tion therapy of cancer for highly effective cancer treatment.[6,11]
therapy, uses ionizing radiation to destruct tumors primarily Despite those encouraging achievements reported in the past
through the generation of oxygen radicals to attack biomole- 1–2 years, how exactly the photothermal effect would enhance
cules of importance (e.g., DNA) inside cancer cells.[1] However, the efficacy of RT via a synergistic manner merits careful inves-
external RT using high-energy radiation (X-ray or γ-ray) to kill tigation. Moreover, the synthesis of nanostructures with the
cancer cells would in the meantime cause severe damages to multiple imaging and therapy functions via simplified methods
normal tissues which are penetrated by the radiation rays.[2] are still desired for the development of imaging-guided, NIR-
Moreover, due to the hypoxic nature inside the tumor micro- triggered thermoradiotherapy.
environment, tumor cells containing insufficient oxygen are Herein, MnSe@Bi2Se3 core–shell nanostructures are ration-
more resistant to ionizing radiation than normal cells, resulting ally designed and successfully fabricated by a partial cation
in the resistance or failure of RT.[3] exchange method, using MnSe nanocrystal as a template,
Nanomedicine to improve the efficacy and reduce toxic effects whose outer layer of manganese is replaced by bismuth to
of conventional cancer therapeutics has attracted tremendous form a Bi2Se3 shell. In this system, the paramagnetic MnSe
attentions in recent years.[4] As far as RT is concerned, tumor- core offers contrasts for both T1- and T2-weighted magnetic
homing nanoagents with high-Z elements such as gold,[5] rare resonance (MR) imaging, whereas the Bi2Se3 shell endows the
earth elements,[6] and bismuth[7] as radiosensitizers can effec- nanostructure with strong absorbance of both X-ray and NIR
tively concentrate a greater local radiation dose within the light, useful for computed tomography (CT) imaging, enhanced
tumor, thus enhancing the RT efficacy to cancer while reducing RT, as well as PTT. It is found that the combination of RT and
radiotoxicity to surrounding normal tissues.[5b,6,8] On the other PTT using MnSe@Bi2Se3 nanoparticles can bring a strong syn-
hand, new approaches with the help of nanotechnology have ergistic effect for the treatment of a mouse tumor model. It is
also been proposed to improve the oxygenation level in the then discovered that such a synergistic treatment outcome is
tumor,[3b,9] such as by using manganese dioxide nanoparticles owing to the remarkably increased oxygenation resulted from
to trigger the production of O2 from H2O2 which is enriched the mild hyperthermia during PTT to boost the blood flow into
in tumor microenvironment, so as to surmount hypoxia-associ- the tumor, enhancing the efficacy of RT for effective cancer
ated radioresistance.[10] Using carefully engineered nanoagents killing. Our work not only presents a simple cation exchange
as multifunctional platforms, RT may be combined with other method to fabricate core–shell nanostructures with highly inte-
types of therapeutic strategies, including near-infrared (NIR) grated imaging and therapy functions but also demonstrates a
promise concept to enhance conventional RT by: i) using X-ray
Dr. G. Song, C. Liang, H. Gong, Prof. L. Cheng,
absorbing agents to locally concentrate radiation energy and
Prof. Z. Liu ii) employing NIR triggered PTT to overcome hypoxia-associated
Institute of Functional Nano and Soft Materials radioresistance.
(FUNSOM) There are numerous examples for syntheses of core–shell
Collaborative Innovation Center of Suzhou nanoparticles by sequential growth methods, which, however,
Nano Science and Technology
Soochow University often contains tedious synthetic procedures and would result in
Suzhou, Jiangsu 215123, China structures obviously larger than the original cores.[4e,6,12] Here
E-mail: zliu@suda.edu.cn a cation exchange method was used to produce MnSe@Bi2Se3
M. Li, Prof. K. Yang core–shell nanostructures from pre-made MnSe nanoparticles
School of Radiation Medicine and Protection and School by simply adding a controlled amount of Bi source into a solu-
for Radiological and Interdisciplinary Sciences (RAD-X)
Medical College of Soochow University
tion of MnSe (Figure 1a). Compared to manganese chalcoge-
Suzhou, Jiangsu 215123, China nides, bismuth chalcogenides have a much lower solubility
E-mail: kyang@suda.edu.cn (higher pKsp).[13] This will be a thermodynamic driving force
Dr. X. Zheng, Prof. X. Jiang favorable for the cation exchange reaction of manganese by
College of Chemistry and Chemical Engineering bismuth.[14]
Nanjing University The as-prepared MnSe nanoparticles showed a bullet-like
Nanjing 210093, China
morphology as revealed by transmission electron microscopy
DOI: 10.1002/adma.201503006 (TEM) imaging (Figure 1b and inset), with an average length

6110 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015, 27, 6110–6117
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
Figure 1. Synthesis and characterization of MnSe@Bi2Se3 core–shell nanoparticles. a) A scheme showing the fabrication of MnSe@Bi2Se3 core–shell
nanostructure by cation exchange and the subsequent PEGylation. TEM images of as-synthesized MnSe b) and MnSe@Bi2Se3 c) nanoparticles. Inset:
The corresponding magnified TEM images (scale bar = 50 nm). d) HAADF-STEM images of an individual MnSe@Bi2Se3 core–shell nanostructure.
The element maps showed the distribution of Mn (red), Se (yellow), and Bi (green). e) Line scan profiles recorded from a MnSe@Bi2Se3 nanoparticle.
f) Powder XRD patterns of MnSe and MnSe@Bi2Se3 nanoparticles. g) UV–vis–NIR absorbance spectra of MnSe and MnSe@Bi2Se3 nanoparticles
dispersed in chloroform at the same concentration (60 µg mL−1).

of 132 ± 19 nm and width of 105 ± 25 nm. After the bismuth the as-prepared MnSe@Bi2Se3 sample (Figure 1f). The above
source was introduced into the reaction solution of as-made results unambiguously reveal that those nanoparticles are com-
MnSe templates, the solution temperature was slowly increased posed by the MnSe core together with the Bi2Se3 shell and
to activate the cation exchange. The resulted MnSe@Bi2Se3 show a bullet-like morphology with clear boundary of core–
core–shell nanostructures showed well-maintained morphology shell nanostructure. Moreover, after coating with the Bi2Se3
and size (length = 132 ± 21 and width = 111 ± 21 nm) (Figure 1c shell, the obtained MnSe@Bi2Se3 core–shell nanoparticles
and inset). The precise control/preservation of morphology and exhibited greatly enhanced optical absorption from visible to
sizes is an obvious advantage of the cation exchange method in NIR region, in marked contrast to bare MnSe core nanoparti-
comparison to the conventional sequential growth method to cles which showed light yellow color with little absorbance at
prepare core–shell nanoparticles.[13–15] longer wavelengths (Figure 1g). As determined by inductively
The successful fabrication of MnSe@Bi2Se3 core–shell coupled plasma atomic-emission spectroscopy (ICP-AES) meas-
nanostructures was evidenced by the high-resolution TEM urement, the exact molar ratio of Mn and Bi was 1:1.97 for our
imaging (Figure S1, Supporting Information), energy disper- MnSe@Bi2Se3 nanoparticles. Note that for MnSe nanoparticles
sive spectra (EDX) (Figure S2, Supporting Information), and with the current size, even if excess Bi source was added, the
elemental mapping by the high-angle annular dark-field scan- internal MnSe core still could not be fully replaced by Bi2Se3
ning TEM (HAADF-STEM) (Figure 1d,e). As expected, char- (Figure S3, Supporting Information).
acteristic X-ray diffraction (XRD) peaks of both rhombohedral Next, we would like to functionalize as-made oleylamine
Bi2Se3 and hexagonal MnSe crystal planes were observed in capped MnSe@Bi2Se3 nanoparticles to make them soluble in

Adv. Mater. 2015, 27, 6110–6117 © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 6111
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

water and stable in physiological solutions. An amphiphilic University Laboratory Animal Center. As revealed by in vivo CT
polymer, PEG-grafted poly(maleicanhydride-alt-1-octadecene) imaging (Figure 2b), a strong contrast showed up in the tumor
(C18PMH-PEG), was introduced to coat those nanoparticles upon injection of nanoparticles, with the HU value increased
by hydrophobic interaction.[16] The resulted PEG modified from ≈50.4 to ≈126.2 HU, suggesting the great CT contrasting
MnSe@Bi2Se3 (MnSe@Bi2Se3-PEG) exhibited excellent dis- efficiency of MnSe@Bi2Se3-PEG.
persity in various solutions including water, phosphate buff- Magnetic resonance (MR) imaging possesses excellent spa-
ered saline (PBS), and cell culture medium, without showing tial resolution and is more sensitivity than CT imaging.[19]
any agglomeration as evidenced by the consistent diameters Mn2+ with five unpaired 3d electrons is known to be a contrast
(≈140 nm) measured by dynamic light scattering (DLS) (Figure S4, agent in T1-weighted MR imaging.[20] Encouragingly, with the
Supporting Information). The zeta potential of MnSe@Bi2Se3- increase of MnSe@Bi2Se3-PEG concentrations, the acquired
PEG nanoparticles was measured to be −3.25 ± 1.32 mV in cell MR images became brighter under the T1-weighted mode,
culture medium. while gradually turned darker under the T2-weighted mode
The X-ray attenuation agents usually containing high-Z (Figure S5, Supporting Information). The corresponding lon-
elements are able to produce contrasts in CT imaging.[17] Bis- gitudinal relaxivity (r1) and transverse relaxivity (r2) of MnSe@
muth has a higher X-ray attenuation coefficient compared to Bi2Se3-PEG were measured to be r1 = 5.44 mM−1 s−1, and r2 =
several stable nonradioactive heavy elements such as I, W, Au, 27.6 mM−1 s−1, respectively (Figure 2c), which were comparable
and Ta used in CT imaging.[7,18] The CT images of MnSe@ to manganese oxide nanoparticles.[21] Although being encapsu-
Bi2Se3-PEG aqueous solutions thus showed a sharp signal lated inside the Bi2Se3 shell, the paramagnetic MnSe core still
enhancement as the increase of Bi concentrations (Figure S5, could induce the effective shortening of T1 relaxation of sur-
Supporting Information). The slope of Hounsfield units (HU) rounding water protons, similar to many other magnetic nano-
for MnSe@Bi2Se3-PEG (≈5.85 HU mM−1) appeared to be particles shelled by Au, SiO2, CuS2−x, or TaOx coatings.[6,12a,22]
higher than that of a clinically used CT contrast agent (Iohexol, Then, the contrast enhancing effect in vivo was evaluated in
≈4.48 HU mM−1) (Figure 2a). The excellent CT signal enhance- Balb/c mice bearing 4T1 tumors before and after injection of
ment ability of MnSe@Bi2Se3-PEG encouraged us to further MnSe@Bi2Se3-PEG (10 mg kg−1) (Figure 2d). For T1-weighted
use it for in vivo CT imaging. In our experiments, 4T1 murine MR imaging, the tumor became brighter and showed marked
breast tumors growing on Balb/c mice were injected with a positive enhancement, while the T2-weighted images showed
solution of MnSe@Bi2Se3-PEG (10 mg kg−1). Animal experi- negative contrast in the tumor after injection of nanoparticles,
ments were carried out under protocols approved by Soochow demonstrating the capability of MnSe@Bi2Se3-PEG to serve as

Figure 2. CT and MR imaging with MnSe@Bi2Se3-PEG. a) HU values of MnSe@Bi2Se3-PEG or Iohexol solutions at different concentrations under CT
imaging. b) CT images of a 4T1 tumor-bearing mouse before (up) and after (down) injection with MnSe@Bi2Se3-PEG. The CT contrast was obviously
enhanced in the mouse tumor after nanoparticle injection. c) T1 relaxation rate (1/T1) and T2 relaxation rate (1/T2) of MnSe@Bi2Se3-PEG solutions
measured as the function of Mn2+ concentrations. d) T1-weighted MR images (up) and T2-weighted MR images (down) of a 4T1 tumor-bearing mouse
before (left) and after (right) injection with MnSe@Bi2Se3-PEG. Our nanoparticles offered strong contrasts in the tumor under both T1-weighted and
T2-weighted MR imaging modes.

6112 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015, 27, 6110–6117
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
Figure 3. In vitro PTT and enhanced RT. a) Confocal fluorescence images of 4T1 cells treated with PBS, NIR, MnSe@Bi2Se3-PEG, and MnSe@Bi2Se3-
PEG + NIR. Live and dead cells were stained by Calcein AM and PI and presented in green and red colors in those images, respectively. b) Imaging
of the marker (γ-H2AX) for double-strand DNA breaks in 4T1 cells treated with PBS, RT (4 Gy), MnSe@Bi2Se3-PEG, and MnSe@Bi2Se3-PEG + RT
(4 Gy). Cells exposed to nanoparticles after RT exhibited the highest level of DNA damage (positive in γ-H2AX). c) Clonogenic survival assay of 4T1
cells treated with and without MnSe@Bi2Se3-PEG under series of radiation doses at 0, 2, 4, and 6 Gy. The error bars represent the standard error of
mean of at least three replicates. P values: *P < 0.05, **P < 0.01.

the contrast agent for both T1- and T2-weighted MR imaging. showed much lower percentages of viable cell colonies than that
Notably, the integration of T1- and T2-weighted MR modes in without nanoparticle treatment, at the same X-ray irradiation
a single nanoagent may potentially enhance the ability of dis- doses. By using a linear-quadratic model to analyze enhance-
criminating hypointense tissue in MR imaging, to overcome ment of radiation effects,[8] the sensitizing enhancement ratio
the intrinsic shortcoming of the T2-weighted mode.[12a] of MnSe@Bi2Se3-PEG was calculated to be ≈1.16, which is
Our MnSe@Bi2Se3-PEG exhibited no obvious adverse effects comparable to gold nanoparticles as a radiosensitizer.[8,23] These
to the viabilities of 4T1 and HeLa cells at tested concentrations results confirmed that MnSe@Bi2Se3-PEG could increase X-ray
from 1.56 to 100 µg mL−1 for 24 h, as evaluated by the methyl induced DNA damage and thus enhance the efficacy of RT to
thiazolyl tetrazolium (MTT) assay (Figure S6, Supporting Infor- inhibit the proliferation of cancer cells, likely due to MnSe@
mation). With strong NIR absorbance contributed by the topo- Bi2Se3-PEG nanoparticles inside cells to absorb and concentrate
logical insulator Bi2Se3 shell,[18a] effective photothermal heating the radiation dose toward those cells.[6,8]
of MnSe@Bi2Se3-PEG could be induced upon irradiation by After demonstrating the capability of MnSe@Bi2Se3-PEG to
the 808 nm NIR laser (Figures S7 and S8, Supporting Informa- trigger PTT and enhance RT in vitro, we then would like to use
tion). Those MnSe@Bi2Se3-PEG nanoparticles also possessed these nanoparticles for in vivo cancer treatment in a mouse tumor
excellent photothermal stability without significantly losing model. Balb/c mice bearing subcutaneous 4T1 tumors were
their absorbance after multiple rounds of NIR laser irradiation intratumorally injected with MnSe@Bi2Se3-PEG (4 mg kg−1),
(Figure S9, Supporting Information). Therefore, as expected, and then irradiated with the 808 nm laser (0.6 W cm−2). As
4T1 cancer cells incubated with MnSe@Bi2Se3-PEG could monitored by a IR thermal camera, after 20 min of NIR irra-
be effectively ablated by the NIR laser (808 nm, 1 W cm−2) diation, the temperature of tumors injected with MnSe@Bi2Se3-
(Figure 3a). PEG increased to ≈45 °C, while those injected with PBS were
Next, we wondered whether MnSe@Bi2Se3-PEG with strong not notable heated (final temperature ≈ 36 °C) (Figure 4a,b).
X-ray attenuation ability would be able to enhance RT trig- To study the in vivo efficacy of PTT, enhanced RT, and the
gered by ionizing radiation such as X-ray. 4T1 cells were incu- combination of PTT and RT induced by MnSe@Bi2Se3-PEG, a
bated with either phosphate buffered saline (PBS) or MnSe@ total of seven groups of mice were used in our experiments:
Bi2Se3-PEG (40 µg mL−1) and then exposed to X-ray radiation Group 1: PBS; Group 2: PBS + NIR; Group 3: MnSe@Bi2Se3-
at a dose of 4 Gy. Cells were then immunofluorescently labeled PEG; Group 4: X-ray irradiation without nanoparticle injection
for γ-H2AX, a marker of double-strand DNA breaks, to evaluate (RT alone); Group 5: MnSe@Bi2Se3-PEG + NIR (PTT); Group 6:
DNA damage in cancer cells induced by RT (Figure 3b). While MnSe@Bi2Se3-PEG + X-ray (enhanced RT); Group 7: MnSe@
our nanoparticles by themselves induced no detectable DNA Bi2Se3-PEG + NIR + X-ray (PTT + enhanced RT). The nano-
damage to cells, a remarkably enhanced DNA damage level particle dose was (4 mg kg−1) in all groups. The tumor growth
induced by X-ray was observed for cells treated with MnSe@ curves and representative photographs of mice from different
Bi2Se3-PEG in comparison to the PBS control, suggesting the groups were shown in Figure 4c and Figure S10 (Supporting
strong RT enhancement effect of those Bi-containing nanopar- Information), respectively. As expected, neither NIR irradiation
ticles. In addition to the DNA damage assay, the clonogenic sur- nor MnSe@Bi2Se3-PEG injection showed appreciable effect
vival assay was also conducted to evaluate the in vitro efficacy on tumor growth. The mild PTT treatment with injection of
of RT (Figure 3c). The group treated with MnSe@Bi2Se3-PEG MnSe@Bi2Se3-PEG followed by NIR irradiation (0.6 W cm−2)

Adv. Mater. 2015, 27, 6110–6117 © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 6113
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Figure 4. In vivo combined cancer treatment with MnSe@Bi2Se3-PEG. a) IR thermal images of 4T1 tumor-bear mice with injection of PBS or MnSe@
Bi2Se3-PEG (dose = 4 mg kg−1), under the 808 nm laser irradiation (0.6 W cm−2). b) Temperature changes of tumors monitored by the IR thermal camera
during laser irradiation. c) Tumor volume growth curves of mice after various treatments (five mice for each group). NIR irradiation was conducted
by the 808 nm laser at 0.6 W cm−2 for 20 min and the radiation dose used for RT was 4 Gy. Statistical analysis was performed using the Student's
two-tailed t-test (*P < 0.05 and **P < 0.01).

(Group 5) was able to partially inhibit the tumor growth, with by oxygen in normal tissues but could be turned on when the
the overall tumor growth inhibition (TGI) ratio determined to oxygen content is reduced, providing hypoxia specificity during
be ≈38%. Compared with RT alone (Group 4) (TGI ≈ 23%), fluorescence imaging.[25] The mixture of Ir-PVP + MnSe@
injection of MnSe@Bi2Se3-PEG plus X-ray irradiation (Group Bi2Se3-PEG showed no obvious fluorescence change after NIR
6) induced more significant tumor growth inhibition (TGI ≈ irradiation, indicating the great stability of this fluorescent
41%, P < 0.05), suggesting the ability of MnSe@Bi2Se3-PEG to probe during PTT (Figure S12, Supporting Information). Mice
enhance the efficacy of in vivo RT. Most importantly, a remark- bearing subcutaneous 4T1 tumors were intratumorally injected
able tumor growth inhibition efficiency (TGI ≈ 90%) was with Ir-PVP or a mixture of Ir-PVP + MnSe@Bi2Se3-PEG. From
observed for the tumors treated by MnSe@Bi2Se3-PEG + NIR + the whole body imaging (Figure 5a,b), the tumors treated with
RT (Group 7), demonstrating the synergistically enhanced ther- MnSe@Bi2Se3-PEG + NIR showed obviously decreased fluores-
apeutic effect by such combined PTT and RT therapy. Micro- cence intensity post-NIR irradiation. In contrast, the fluores-
scopy images of hematoxylin and eosin (H&E) stained and ter- cence intensities in tumors treated with MnSe@Bi2Se3-PEG
minal deoxynucleotidyl transferase-mediated dUTP-biotin nick without NIR irradiation or NIR irradiation alone remained
end labeling (TUNEL) stained tumor slices also uncovered that unchanged. Therefore, the oxygenation of tumor hypoxia was
the combined PTT and RT triggered by MnSe@Bi2Se3-PEG improved immediately after NIR irradiation.
(Group 7) exerted the most significant damages to tumor cells To further confirm the ability of mild PTT to overcome
(Figure S11, Supporting Information). hypoxia in vivo, hypoxyprobe (pimonidazole) immuno-histo-
The presence of hypoxic tumor cells in tumor micro- chemical assay was performed for tumor slices post different
environment is one of the main reasons of resistance to treatments.[3a,26] Images of immunoperoxidase stained tumor
RT.[3] The effect of ionizing radiation can be largely increased slices revealed that PTT treatment induced by MnSe@Bi2Se3-
under well-oxygenated conditions compared with treatment PEG + NIR could largely reduce the hypoxic level in almost the
of hypoxic tumors.[3b,9a,24] To examine the effect of mild PTT whole tumor (Figure 5c). Immunofluorescence staining assay
on the tumor hypoxia, the in vivo imaging of tumor hypoxia was further carried out by staining the cell nuclei, blood vessels,
was performed with a recently reported fluorescence probe, and hypoxia areas with 2-(4-amidinophenyl)-6-indolecarbami-
poly(N-vinylpyrrolidone)-conjugated iridium (III) complex (Ir- dine dihydrochloride (DAPI) (blue), anti-CD31 antibody (red),
PVP).[25] The phosphorescence emission of Ir-PVP is quenched and antipimonidazole antibody (green), respectively (Figure 5d).

6114 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015, 27, 6110–6117
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
Figure 5. Effect of NIR-induced PTT on tumor hypoxia. a) Whole-body fluorescence imaging of mice bearing subcutaneously 4T1 tumors with i.t.
injection of tumor hypoxia fluorescence probe Ir-PVP, or a mixture of Ir-PVP + MnSe@Bi2Se3-PEG. The images were taken before and after NIR irradia-
tion. b) The relative fluorescence intensities of Ir-PVP in tumors for different groups of mice at different time points after NIR irradiation. P values: *P
< 0.05. c) Immunoperoxidase staining by the hypoxyprobe for tumor slices without (left) or with (right) MnSe@Bi2Se3-PEG induced PTT treatment.
d) Representative immunofluorescence images of tumor slices. The nuclei, blood vessels, and hypoxia areas were stained with DAPI (blue), anti-CD31
antibody (red), and antipimonidazole antibody (green), respectively.

Consistent to immunoperoxidase staining results, the tumor metabolized by the liver.[7] To investigate whether MnSe@Bi2Se3-
treated with MnSe@Bi2Se3-PEG + NIR showed obviously weak- PEG would result in any side effect, blood chemistry, complete
ened pimonidazole-stained (green) hypoxic area compared to blood panel analysis, and histology examination were carried out
the control group, indicating that the tumor hypoxia was effec- for healthy mice after intravenous injection of MnSe@Bi2Se3-PEG
tively reduced by mild hyperthermia. More interestingly, for (10 mg kg−1), at the 1st, 7th, 14th, and 28th day post-injection
tumor tissues nearby blood vessels identified by CD31 staining (Figure S13, Supporting Information). Encouragingly, most of
(red), the hypoxia situation was greatly reversed, suggesting measured parameters are rather close to those of untreated healthy
that the mild PTT may enhance blood flow into the tumor and mice and fell within the normal reference ranges. Moreover, no
then improve the tumor oxygenation.[3,27] Therefore, the overall noticeable tissue damage and adverse effect to major organs was
tumor oxygenation status could be improved immediately after observed from the H&E stained organ slices (Figure S14, Sup-
PTT, decreasing the hypoxia-associated radioresistance,[1b,27a,28] porting Information). Our preliminary data showed no obvious
and thus resulting in excellent synergistic therapeutic effects clue of toxicity induced by MnSe@Bi2Se3-PEG to the treated mice.
achieved by the combined PTT and RT. Nevertheless, further careful studies are still required to system-
Bismuth is considered to have low toxicity among heavy ele- atically look into the long-term biodistribution, excretion, and toxi-
ments.[7] Some Bi-containing compounds could be gradually cology behaviors of those nanoparticles in animals.

Adv. Mater. 2015, 27, 6110–6117 © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 6115
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

In conclusion, we have used the cation exchange method Adv. Mater. 2013, 25, 4452; d) G. Song, J. Shen, F. Jiang, R. Hu,
for the synthesis of multifunctional MnSe@Bi2Se3 core–shell W. Li, L. An, R. Zou, Z. Chen, Z. Qin, J. Hu, ACS Appl. Mater. Inter.
nanostructures. During cation exchange, those core–shell nano- 2014, 6, 3915; e) G. Song, Q. Wang, Y. Wang, G. Lv, C. Li, R. Zou,
particles show well-preserved size and morphology without size Z. Chen, Z. Qin, K. Huo, R. Hu, J. Hu, Adv. Funct. Mater. 2013,
23, 4281; f) G. Song, C. Li, J. Hu, R. Zou, K. Xu, L. Han, Q. Wang,
expansion and interior composition mixing. This nanoscale
J. Yang, Z. Chen, Z. Qin, K. Ruan, R. Hu, J. Mater. Chem. 2012,
cation exchange reaction is able to produce novel nanomaterials 22, 17011; g) Q. He, S. Guo, Z. Qian, X. Chen, Chem. Soc. Rev.
with multicomponent compositions and structures that are 2015, DOI: 10.1039/C4CS00511B; h) J. Ge, Q. Jia, W. Liu, L. Guo,
not easily accessed by traditional sequential growth methods. Q. Liu, M. Lan, H. Zhang, X. Meng, P. Wang, Adv. Mater. 2015, 27,
In such MnSe@Bi2Se3 system, the paramagnetic MnSe core 4169.
offers contrasts for both T1- and T2-weighted MR imaging, [5] a) M. G. Castro, D. Y. Joh, L. Sun, M. Stangl, A. Al Zaki, S. Murty,
while the Bi2Se3 shell endows the nanostructure with the ability P. P. Santoiemma, J. J. Davis, B. C. Baumann, M. Alonso-Basanta,
of absorbing ionizing radiation useful for CT imaging and D. Bhang, G. D. Kao, A. Tsourkas, J. F. Dorsey, PLoS One 2013,
enhanced RT as well as high NIR optical absorbance to enable 8, e62425; b) X.-D. Zhang, Z. Luo, J. Chen, X. Shen, S. Song,
PTT. Using MnSe@Bi2Se3-PEG as the multifunctional agent, Y. Sun, S. Fan, F. Fan, D. T. Leong, J. Xie, Adv. Mater. 2014, 26,
4565.
the efficacy of RT could be enhanced not only by the nanopar-
[6] Q. Xiao, X. Zheng, W. Bu, W. Ge, S. Zhang, F. Chen, H. Xing,
ticles in the tumor to concentrate radiation energy but also by Q. Ren, W. Fan, K. Zhao, Y. Hua, J. Shi, J. Am. Chem. Soc. 2013, 135,
the mild PTT triggered by the NIR laser to improve the oxy- 13041.
genation level in the tumor to overcome hypoxia-associated [7] X.-D. Zhang, J. Chen, Y. Min, G. B. Park, X. Shen, S.-S. Song,
radioresistance. Our work, on the one hand, presents an inter- Y.-M. Sun, H. Wang, W. Long, J. Xie, K. Gao, L. Zhang, S. Fan,
esting cation exchange approach to construct multifunctional F. Fan, U. Jeong, Adv. Funct. Mater. 2014, 24, 1718.
theranostic nanostructures, and on the other illustrates a prom- [8] A. Al Zaki, D. Joh, Z. Cheng, A. L. B. De Barros, G. Kao, J. Dorsey,
ising concept to enhance the efficacy of RT using nanoagents A. Tsourkas, ACS Nano 2014, 8, 104.
absorbing both ionizing radiation and NIR optical light, so as to [9] a) B. J. Moeller, R. A. Richardson, M. W. Dewhirst, Cancer Metast.
achieve synergistic therapeutic outcomes via the combination Rev. 2007, 26, 241; b) B. A. Teicher, T. S. Herman, S. M. Jones,
Cancer Res. 1989, 49, 2693.
therapy.
[10] P. Prasad, C. R. Gordijo, A. Z. Abbasi, A. Maeda, A. Ip, A. M. Rauth,
R. S. DaCosta, X. Y. Wu, ACS Nano 2014, 8, 3202.
[11] a) S. Wang, X. Li, Y. Chen, X. Cai, H. Yao, W. Gao, Y. Zheng, X. An,
Supporting Information J. Shi, H. Chen, Adv. Mater. 2015, 27, 2775; b) J. F. Hainfeld, L. Lin,
D. N. Slatkin, F. A. Dilmanian, T. M. Vadas, H. M. Smilowitz, Nano-
Supporting Information is available from the Wiley Online Library or medicine: Nanotechnol. Biol. Med. 2014, 10, 1609.
from the author. [12] a) N. Lee, H. R. Cho, M. H. Oh, S. H. Lee, K. Kim, B. H. Kim,
K. Shin, T.-Y. Ahn, J. W. Choi, Y.-W. Kim, S. H. Choi, T. Hyeon, J. Am.
Chem. Soc. 2012, 134, 10309; b) M. Chen, S. Tang, Z. Guo, X. Wang,
Acknowledgements S. Mo, X. Huang, G. Liu, N. Zheng, Adv. Mater. 2014, 26, 8210.
[13] S. Gupta, S. V. Kershaw, A. L. Rogach, Adv. Mater. 2013, 25, 6923.
This work was partially supported by the National Natural Science [14] B. J. Beberwyck, Y. Surendranath, A. P. Alivisatos, J. Phys. Chem. C
Foundation of China (Grant Nos. 51302180, 51222203, 51002100, 2013, 117, 19759.
and 51132006), the National “973” Program of China (Grant Nos. [15] a) L. De Trizio, H. Li, A. Casu, A. Genovese, A. Sathya,
2011CB911002 and 2012CB932601), a Project Funded by the Priority G. C. Messina, L. Manna, J. Am. Chem. Soc. 2014, 136,
Academic Program Development of Jiangsu Higher Education 16277; b) D. Zhang, A. B. Wong, Y. Yu, S. Brittman, J. Sun, A. Fu,
Institutions, and Postdoctoral science foundation of China (Grant Nos. B. Beberwyck, A. P. Alivisatos, P. Yang, J. Am. Chem. Soc. 2014,
2014M561706 and 2013M531400). 136, 17430; c) R. D. Robinson, B. Sadtler, D. O. Demchenko,
C. K. Erdonmez, L. W. Wang, A. P. Alivisatos, Science 2007, 317,
Received: June 22, 2015
355.
Revised: July 15, 2015
[16] X. Liu, H. Tao, K. Yang, S. Zhang, S.-T. Lee, Z. Liu, Biomaterials
Published online: September 2, 2015
2011, 32, 144.
[17] O. Rabin, J. M. Perez, J. Grimm, G. Wojtkiewicz, R. Weissleder, Nat.
Mater. 2006, 5, 118.
[18] a) J. Li, F. Jiang, B. Yang, X.-R. Song, Y. Liu, H.-H. Yang, D.-R. Cao,
[1] a) X. Y. Su, P. D. Liu, H. Wu, N. Gu, Cancer Biol. Med. 2014, 11, W.-R. Shi, G.-N. Chen, Sci. Rep. 2013, 3, 1998; b) A. L. Brown,
86; b) E. L. Jones, L. R. Prosnitz, M. W. Dewhirst, P. K. Marcom, P. C. Naha, V. Benavides-Montes, H. I. Litt, A. M. Goforth,
P. H. Hardenbergh, L. B. Marks, D. M. Brizel, Z. Vujaskovic, Clin. D. P. Cormode, Chem. Mater. 2014, 26, 2266.
Cancer Res. 2004, 10, 4287. [19] E. Terreno, D. D. Castelli, A. Viale, S. Aime, Chem. Rev. 2010, 110,
[2] C. Zhang, K. Zhao, W. Bu, D. Ni, Y. Liu, J. Feng, J. Shi, Angew. Chem. 3019.
Int. Ed. 2015, 54, 1770. [20] a) S. Viswanathan, Z. Kovacs, K. N. Green, S. J. Ratnakar,
[3] a) X. Sun, X.-F. Li, J. Russell, L. Xing, M. Urano, G. C. Li, A. D. Sherry, Chem. Rev. 2010, 110, 2960; b) N. Moloto,
J. L. Humm, C. C. Ling, Radiother. Oncol. 2008, 88, 269; M. J. Moloto, M. Kalenga, S. Govindraju, M. Airo, Opt. Mater. 2013,
b) M. R. Horsman, J. Overgaard, Clin. Oncol. 2007, 19, 36, 31.
418. [21] a) Y. Lu, L. Zhang, J. Li, Y.-D. Su, Y. Liu, Y.-J. Xu, L. Dong, H.-L. Gao,
[4] a) L. Cheng, C. Wang, L. Feng, K. Yang, Z. Liu, Chem. Rev. 2014, 114, J. Lin, N. Man, P.-F. Wei, W.-P. Xu, S.-H. Yu, L.-P. Wen, Adv. Funct.
10869; b) E.-K. Lim, T. Kim, S. Paik, S. Haam, Y.-M. Huh, K. Lee, Mater. 2013, 23, 1534; b) H. B. Na, J. H. Lee, K. An, Y. I. Park,
Chem. Rev. 2015, 115, 327; c) K. Dong, Z. Liu, Z. Li, J. Ren, X. Qu, M. Park, I. S. Lee, D.-H. Nam, S. T. Kim, S.-H. Kim, S.-W. Kim,

6116 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015, 27, 6110–6117
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
K.-H. Lim, K.-S. Kim, S.-O. Kim, T. Hyeon, Angew. Chem. Int. Ed. [26] C. R. Gordijo, A. Z. Abbasi, M. A. Amini, H. Y. Lip, A. Maeda, P. Cai,
2007, 46, 5397. P. J. O’Brien, R. S. DaCosta, A. M. Rauth, X. Y. Wu, Adv. Funct.
[22] a) Q. Tian, J. Hu, Y. Zhu, R. Zou, Z. Chen, S. Yang, R. Li, Q. Su, Mater. 2015, 25, 1858.
Y. Han, X. Liu, J. Am. Chem. Soc. 2013, 135, 8571; b) Z. Li, S. Yin, [27] a) C. W. Song, A. Shakil, J. L. Osborn, K. Iwata, Int. J. Hyperthermia
L. Cheng, K. Yang, Y. Li, Z. Liu, Adv. Funct. Mater. 2014, 24, 2312. 2009, 25, 91; b) P. W. Vaupel, D. K. Kelleher, Int. J. Hyperthermia
[23] S. Jain, J. A. Coulter, A. R. Hounsell, K. T. Butterworth, 2010, 26, 211; c) E. Y. Chen, K. S. Samkoe, S. Hodge, K. Tai, H. Hou,
S. J. McMahon, W. B. Hyland, M. F. Muir, G. R. Dickson, A. A. Petryk, R. Strawbridge, P. J. Hoopes, N. Khan, Oxygen Transport
K. M. Prise, F. J. Currell, J. M. O’Sullivan, D. G. Hirst, Int. J. Radiat. To Tissue XXXVI, Advances in Experimental Medicine and Biology, (Eds:
Oncol. 2011, 79, 531. H. M. Swartz, D. K. Harrison, D. F. Bruley), Springer, New York,
[24] H. J. Feldmann, Lung Cancer 2001, 33, S77. 2014, p. 87; d) M. Hurwitz, P. Stauffer, Semin. Oncol. 2014, 41, 714.
[25] X. Zheng, X. Wang, H. Mao, W. Wu, B. Liu, X. Jiang, Nat. Commun. [28] W. Song, H. J. Park, C. K. Lee, R. Griffin, Int. J. Hyperthermia 2005,
2015, 6, 5834. 21, 761.

Adv. Mater. 2015, 27, 6110–6117 © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 6117

Anda mungkin juga menyukai