You are on page 1of 15

Biomaterials 76 (2016) 87e101

Contents lists available at ScienceDirect

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

Rational design of multifunctional magnetic mesoporous silica


nanoparticle for tumor-targeted magnetic resonance imaging and
precise therapy
Wei-Hai Chen a, 1, Guo-Feng Luo a, 1, Qi Lei a, Feng-Yi Cao a, Jin-Xuan Fan a, Wen-Xiu Qiu a,
Hui-Zhen Jia a, Sheng Hong a, Fang Fang b, Xuan Zeng a, Ren-Xi Zhuo a,
Xian-Zheng Zhang a, *
a
Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
b
State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of
Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China

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

Article history: In this paper, a multifunctional theranostic magnetic mesoporous silica nanoparticle (MMSN) with
Received 15 October 2015 magnetic core was developed for magnetic-enhanced tumor-targeted MR imaging and precise therapy.
Accepted 21 October 2015 The gatekeeper b-cyclodextrin (b-CD) was immobilized on the surface of mesoporous silica shell via
Available online 22 October 2015
platinum(IV) prodrug linking for reduction-triggered intracellular drug release. Then Arg-Gly-Asp (RGD)
peptide ligand was further introduced onto the gatekeeper b-CD via hosteguest interaction for cancer
Keywords:
targeting purpose. After active-targeting endocytosis by cancer cells, platinum(IV) prodrug in MMSNs
Mesoporous silica nanoparticle
would be restored to active platinum(II) drug in response to the innative reducing microenvironment in
Tumor-targeted
Magnetic resonance imaging
cancer cells, resulting in the detachment of b-CD gatekeeper and thus simultaneously triggering the in
Theranostic situ release of anticancer drug doxorubicin (DOX) entrapped in the MMSNs to kill cancer cells. It was
Precise therapy found that with the aid of an external magnetic field, drug loaded MMSNs showed high contrast in MR
imaging in vivo and exhibited magnetically enhanced accumulation in the cancer site, leading to sig-
nificant inhibition of cancer growth with minimal side effects. This multifunctional MMSN will find great
potential as a theranostic nanoplatform for cancer treatment.
© 2015 Elsevier Ltd. All rights reserved.

1. Introduction although the EPR effect can increase the accumulation of nano-
medicines in tumor tissues, the poor cellular internalization and
In conventional cancer therapy, chemotherapeutic drugs insufficient intracellular drug release also hamper the utilization of
without the capability to discriminate between cancerous and anticancer drugs, leading to limited therapeutic effect [8,9]. To
normal cells always result in undesirable side effects and high overcome these barriers, the combination of multiple functional-
systemic toxicity. To achieve satisfactory therapeutic index of drugs, ities into the nanocarrier is highly desired, which relies on the
nanoplatform based drug carriers with passive tumor targeting rational design of sophisticated drug delivery nanosystem [10,11].
ability via the enhanced permeability and retention (EPR) effect Particularly, theranostic nanoplatform with diagnostic and thera-
have attracted increasing attention in cancer therapy [1e4]. peutic capabilities offers great opportunities to revolutionize can-
Nevertheless, the EPR efficacy is always limited by the special tu- cer therapy. The imaging-guided therapy is beneficial to accurately
mor microenvironment, such as dense stroma, mutational extra- destroy cancer and realize precise therapy [12].
cellular matrix, increased interstitial fluid pressure, and Recently, mesoporous silica nanoparticles (MSNs) have
pathophysiological heterogeneity of tumor [5e7]. In addition, garnered prominent research interest as drug nanocarriers due to
their intrinsically favorable biocompatibility, tunable pore sizes,
large loading capacity, and convenient surface functionalization
* Corresponding author. [13e16]. For instance, intelligent molecular machines which
E-mail address: xz-zhang@whu.edu.cn (X.-Z. Zhang). respond to endogenous stimuli, including redox, pH, temperature
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.biomaterials.2015.10.053
0142-9612/© 2015 Elsevier Ltd. All rights reserved.
88 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

and enzymes, have been employed on MSNs to control the drug Multifunctional molecular machine on MMSNs was constituted and
release extensively [17e22]. Despite these encouraging achieve- constructed by platinum(IV) prodrug, b-cyclodextrin, and cancer-
ments, mechanized MSNs have suffered disadvantages since the targeted peptide adamantane-PEG8-glycine-arginine-glycine-
use of stimuli are complicated to apply in vivo or not available aspartic-serine (AD-PEG8-GRGDS) progressively. The b-CD was
innately in the targeted tumor cells as well as lack of diagnostic used as the gatekeeper and immobilized onto mesoporous silica
specificity [23]. To improve the applicability and further function- shell via the linker of platinum(IV) prodrug, which was redox-
alize the MSNs-based materials, integration of MSNs with inorganic sensitive and could be intracellularly activated by reduction to
nanocrystals to form uniform multifunctional nanocomposites has the toxic platinum(II) drug [39,40]. For the first time, platinum(IV)
emerged as one of the most popular research directions recently. prodrug was exploited as the redox-sensitive linker to trigger the
For instance, quantum dots, gold nanoparticles, silver nanoparticles drug release from mesoporous silica as well as chemotherapeutic to
and magnetic nanoparticles, have been successfully incorporated assist the ablation of cancer cells. Finally, the cancer active targeting
within MSNs [24e27]. Of particular significance are magnetic peptide AD-PEG8-GRGDS was assembled on MMSNs as a targeting
mesoporous silica nanoparticles [28e33]. The cooperation of ligand toward cancer cells via hosteguest interaction between AD
magnetic nanoparticles is beneficial for magnetically targeted drug and b-CD.
delivery as well as an excellent contrast agent for magnetic reso- As illustrated in Scheme 1B, after intravenous injection, the
nance imaging (MRI), which provides great opportunity for the multifunctional MMSNs were expected to accumulate at cancer
construction of nanoscaled theranostic platform for cancer therapy. sites as a combined function of the magnetic targeting enhanced
Moreover, as a physical force, the magnetic stimulus is independent EPR effect and the active targeting effect. The magnetic property of
of the complicated tumor physiological microenvironment and the MMSNs would provide great contrasts for magnetic resonance
magnetic targeting mediated EPR effect can increase the accumu- imaging to localize the cancer site and guide the therapy. On the
lation of nanoparticles in the targeted tumor site [34e38], which is other hand, since the RGD motif could targetedly bind avb3 integrin
rewarding to efficiently and specifically deliver the therapeutic overexpressed by cancer cells [41,42], multifunctional MMSNs
agents to the tumor region. would preferentially internalize in cancer cells. After uptaken by
In this study, a multifunctional theranostic platform based on cancer cells, the platinum(IV) prodrug conjugated on the surface of
magnetic mesoporous silica nanoparticles (MMSNs) was inge- MMSN would be reduced by intracellular reductive microenvi-
niously designed for tumor-targeted magnetic resonance imaging ronment and converted to active platinum(II) drug to kill cancer
and precise therapy. As illustrated in Scheme 1, MMSNs with the cells. Importantly, the loaded DOX would be released in situ from
superparamagnetic Fe3O4 core and mesoporous silica shell were mesoporous silica shell since the b-CD gatekeeper was removed in
employed as MRI contrast agent and anticancer drug nano- the progress of reducing platinum(IV) to platinum(II), producing
container simultaneously. Doxorubicin (DOX) as a model anticancer cytotoxicity and inducing the death of cancer cells.
drug was loaded in the porous structure of the MSN shell.

Scheme 1. Schematic illustration of the design and proposed mechanism of the multifunctional MMSNs for tumor-targeted MRI and precise therapy. (A) The functionalization
protocol of the MMSN. (B) The precise treatment strategy by using multifunctional MMSNs. Specifically, the magnetic enhanced EPR effect and the active targeting effect facilitated
the enriching of MMSNs in tumor sites; then MMSNs were selectively uptaken by cancer cells via the receptor-mediated endocytosis; afterward the platinum(IV) prodrug was
reduced by the cellular innate reductive environment and induced the removal of gatekeeper, thereby triggering the in situ drug release for precise therapy.
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 89

2. Materials and methods glycine-aspartic-serine was prepared as our previously reported


method by standard Fmoc solid phase peptide synthesis [44].
2.1. Materials Briefly, peptide chains were grown on 2-chlorotriyl chloride resin.
The coupling of first amino acid conducted 4 equiv (relative to the
N-Fluorenyl-9-methoxycarbonyl (Fmoc) protected L-amino substitution degree of resin) Fmoc-protected amino acid and
acids (Fmoc-Arg(Pbf)-OH, Fmoc-Asp(OtBu)-OH, Fmoc-Ser(tBu)- 6 equiv of DIEA in a DMF solution for 2 h. Other amino acid cou-
OH, and Fmoc-Gly-OH, 2-chlorotrityl chloride resin plings were executed with 4 equiv of Fmoc-protected amino acid,
(100e200 mesh, loading: 0.6 mmol/g, 1%DVB), benzotriazol-1-yl- 4 equiv of HBTU, and 6 equiv of DIEA in a DMF solution for 4 h.
oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), o- During the synthesis, Fmoc protected groups were removed by 20%
benzotriazole-N,N,N0 ,N0 -tetramethyluroniumhexafluorophosphate piperidine/DMF (v/v) for twice and every time for 15 min. At the
(HBTU), 1-hydroxybenzotriazole (HOBt), triisopropylsilane (TIS), end of the synthesis, adamantane chloride was conjugated to the
and piperidine were purchased from GL Biochem. Ltd. (Shanghai, peptide segments as the similar method. Then the resin was
China) and used as received. Diisopropylethylamine (DIEA) was washed with DMF (four times), methanol (four times) and DCM
acquired from GL Biochem. Ltd. (Shanghai, China) and used after (four times), respectively, and then dried under vacuum overnight.
distillation. Fmoc-PEG8-COOH was provided by Zhoubei Technol- Cleavage of the expected peptide and the removal of side chain
ogy Co. Ltd. (Hangzhou, China). Trifluoroacetic acid (TFA), protected groups from the dried resin were performed by sus-
anhydrous ether, ninhydrin, cisplatin (cis-Pt(NH3)2(Cl)2), pending the resin in a cleavage cocktail containing TFA (95%), TIS
hydrogen peroxide (H2O2), succinic anhydride, ferric chloride, so- (2.5%), and H2O (2.5%) for 2 h. The filtration was concentrated to a
dium acetate, 3-aminopropyltriethoxysilane (APTES), cetyl- viscous solution by rotary evaporation. Then viscous filtration was
trimethylammonium bromide (CTAB), tetraethylorthosilicate precipitated in cold ether to obtain the crude product. After vacuum
(TEOS), and ammonium nitrate (NH4NO3) were obtained from drying, the crude product was dissolved in deionized water and
Shanghai Chemical Co. (Shanghai, China) and used directly. Ada- freeze-dried. The molecular weight was determined by ESI-MS
mantane chloride was purchased from Aladdin Reagent Co. Ltd. (Finnigan LCQadvantage). The calculated molecular mass of AD-
(Shanghai, China). N,N-dimethylformamide (DMF), dimethyl sulf- PEG8-GRGDS was 1075.6 and the ESI-MS result found to be 1075.5
oxide (DMSO), methanol and dichloromethane (DCM) were pro- (see Fig. S3).
vided by Shanghai Chemical Co. (Shanghai, China) and distilled
prior to use. Mono-6-ethylenediamine-b-CD was prepared ac- 2.4. Synthesis of the coreeshell type magnetic mesoporous silica
cording to our previous literature procedures [43]. Doxorubicin nanoparticles (MMSNs)
hydrochloride was purchased from Zhejiang Hisun Pharmaceutical
Co. (Zhejiang, China). Fetal bovine serum (FBS), penicillin strepto- The superparamagnetic Fe3O4 core particles were synthesized
mycin, trypsin, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetra- through a solvothermal reaction as previously reported [45]. Then
zoliumbromide (MTT), molecular probe (Hoechst 33342), and the coreeshell type magnetic mesoporous silica nanoparticles were
Dulbecco's phosphate buffered saline (PBS) were purchased from further prepared according to the previous reports with slight
Invitrogen (USA). All other reagents and solvents were of analytical modifications [33]. Briefly, Fe3O4 particles were dispersed in chlo-
grade and used directly. roform with a concentration of 15.6 mg Fe3O4/mL. Firstly, 1.0 mL
Fe3O4 particles were added into 5 mL CTAB solution (0.55 M) and
2.2. Synthesis of the platinum(IV) prodrug the resulted solution was further stirred at 1000 rpm for 20 min.
Then the mixture was heated to 60  C and stirred for another
As shown in Fig. S1, to prepare the platinum(IV) prodrug, the 10 min to evaporate the chloroform. Afterwards, a mixture of 45 mL
cisplatin was firstly oxidized by excess hydrogen peroxide to yield deionized water and 0.3 mL NaOH (0.2 M) was added to the above
Pt(OH)2. In brief, the mixture solution of cisplatin (cis- solution and the resulted solution was heated to 70  C under stir-
Pt(NH3)2(Cl)2, 1.0 g, 3.33 mmol), hydrogen peroxide (H2O2, 30 wt%, ring at 400 rpm. Then 40 mL TEOS was added to the reaction solu-
8 mL) and 90 mL DI water were heated to 50  C and stirred for 6 h in tion drop by drop and repeated one time after 1 h. After stirring
the dark. Then the mixture was cooled to room temperature and another 2 h, the reaction was completed and the MMSN was ob-
further stirred overnight. The crude product was obtained by tained after washing six times with water to remove the unreacted
setting at 4  C for 12 h and filtrating. Then crude product was species.
washed by ice cold water, ethanol and diethyl ether, respectively.
The product (0.85 g) was obtained as a bright yellow powder after 2.5. Synthesis of MMSN-NH2
vacuum dried.
Then Pt(OH)2 reacted with succinic anhydride to afford plati- MMSN (300.0 mg) was dispersed in anhydrous methanol
num(IV) prodrug. In particular, a DMF solution containing Pt(OH)2 (50 mL). Then 3-aminopropyltriethoxysilane (4.5 mL) was added
(0.5 g, 1.5 mmol) and succinic anhydride (1 g, 10.0 mmol) was and the mixture was stirred for 24 h at room temperature. After
heated to 70  C and stirred gently for 12 h in the dark. Then the centrifugation, the nanoparticles were extensively washed with
solvent was removed via vacuum distillation and the product was methanol and the obtained sample was MMSN-NH2.
obtained by recrystallization from acetone at 20  C and isolated
via filtrating, then vacuum dried. The crude product was purified on 2.6. Synthesis of MMSN-NH-Pt-COOH
a silica gel column using ethyl acetate: methanol ¼ 4:1 to afford the
pure product platinum(IV) prodrug (0.61 g). The calculated mo- MMSN-NH2 was conjugated with platinum(IV) prodrug to pre-
lecular mass of platinum(IV) prodrug was 531.2 and the ESI-MS pare MMSN-NH-Pt-COOH using a standing coupling procedure.
result found to be 532.9 (see Fig. S2). Briefly, MMSN-NH2 (200.0 mg), platinum(IV) prodrug (300.0 mg,
0.56 mmol), DIEA (0.98 mL, 5.6 mmol), PyBOP (437.1 mg,
2.3. Synthesis of cancer-targeted peptide adamantane-PEG8- 0.84 mmol) and HOBt (113.5 mg, 0.84 mmol) were dissolved in the
glycine-arginine-glycine-aspartic-serine (AD-PEG8-GRGDS) redistilled DMF (40 mL) and then stirred 24 h at room temperature
in a nitrogen round-bottom flask. The template of CTAB was
The peptide derivative adamantane-PEG8-glycine-arginine- removed by refluxing with a methanol solution of NH4NO3 (60 mL,
90 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

10 mg/mL) through ion exchange for 12 h. Then MMSN-NH-Pt- 2.11. Redox-sensitive drug release of MMSN-NH-Pt-CD/AD-RGD
COOH nanoparticles were obtained by centrifugation and washed
six times with methanol. To investigate redox-sensitive drug release of MMSN-NH-Pt-CD/
AD-RGD under intracellular reductive environment, ascorbic acid
was chose as the reduction agent, which has been demonstrated to
2.7. Synthesis of MMSN-NH-Pt-CD
be a major substance for the reduction of platinum(IV) prodrug
[46,47]. Then 1 mg of DOX loaded MMSN-NH-Pt-CD/AD-RGD
MMSN-NH-Pt-COOH was reacted with mono-6-
nanoparticles were incubated in three different release media at
ethylenediamine-b-CD to prepare MMSN-NH-Pt-CD using a
37  C: (1) PBS (pH 7.4) without ascorbic acid, (2) PBS (pH 7.4) with
standing coupling procedure. In brief, MMSN-NH-Pt-COOH
1 mM ascorbic acid, and (3) PBS (pH 7.4) with 10 mM ascorbic acid,
(100 mg), mono-6-ethylenediamine-b-CD (300.0, mg), NHS
respectively. The drug concentration in the release medium was
(172.5 mg) and 191.7 mg EDC (287.5 mg) were dispersed in 25 mL
detected by RF5301PC spectrofluorophotometer (Shimadzu) at
deionized water and stirred for 48 h in the dark at room temper-
given time intervals. The emission and excitation slit widths were
ature. Then MMSN-NH-Pt-CD was obtained after centrifugation
set at 5 nm with lex ¼ 480 nm.
and washed thoroughly with deionized water, and dried under
vacuum.
2.12. Cell culture

2.8. Synthesis of MMSN-NH-Pt-CD/AD-RGD HeLa (human cervical carcinoma) cancer cells and COS7 (African
green monkey kidney fibroblast cells) normal cells were incubated
MMSN-NH-Pt-CD/AD-RGD was prepared through hosteguest in DMEM medium with 10% FBS and 1% antibiotics (penicillin-
interaction between b-CD and AD. MMSN-NH-Pt-CD (100.0 mg) streptomycin, 10000 U/mL) at 37  C in a humidified atmosphere
was dispersed in 25 mL deionized water and then the peptide AD- containing 5% CO2.
PEG8-GRGDS (50.0 mg) was added. The mixture solution was stir-
red at room temperature for 24 h to obtain MMSN-NH-Pt-CD/AD-
RGD nanoparticles. The nanoparticles were washed thoroughly 2.13. Characterization of in vitro tumor-targeted drug delivery of
with deionized water and dried under vacuum. MMSN-NH-Pt-CD/AD-RGD by CLSM

To confirm the active targeting specificity and magnetically


2.9. Preparation of DOX-loaded MMSNs enhanced drug delivery, we incubated DOX loaded MMSN-NH-Pt-
CD/AD-RGD with HeLa and COS7 cells with or without an external
MMSN-NH-Pt-COOH (100.0 mg) with 25 mg DOX was magnetic field. After the cells treated with DOX loaded MMSN-NH-
dispersed in 10 mL PBS and stirred at room temperature for 24 h, Pt-CD/AD-RGD (The concentration of DOX was 2 mg/mL) in the cell
then further immobilized the gatekeeper b-CD on the surface of culture media for 1 h or 2 h, the culture medias were removed and
nanoparticles for drug loading. To react with mono-6- the cells were washed with PBS three times to remove the unen-
ethylenediamine-b-CD, the above particles, mono-6- docytosed particles. Then the nuclei were stained with Hoechst
ethylenediamine-b-CD (300.0, mg), NHS (172.5 mg) and 33342 at 37  C for 15 min and all cells were observed by CLSM
191.7 mg EDC (287.5 mg) were dispersed in 25 mL deionized water (Nikon C1-si, Japan).
and stirred for 48 h in the dark at room temperature. After that,
drug loaded nanoparticles were collected by centrifugation and
2.14. Analysis of intracellular redox-sensitive drug release by CLSM
washed several times with water to remove the unloaded drug
and quantitative evaluating cellular uptake of DOX by flow
molecules. The supernatant was collected. The DOX loading con-
cytometry
tent was determined by fluorescence measurement (Loading
content ¼ (initial weight of drug - weight of drug in supernatant)/
To further quantitatively evaluate cellular uptake of multifunc-
(weight of drug loaded nanoparticles)). The DOX loaded MMSN-
tional MMSNs and examine the intracellular drug delivery, cells
NH-Pt-CD/AD-RGD was prepared similar to the procedure
treated with DOX loaded MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/
described above.
AD-RGD were measured by CLSM and flow cytometry. HeLa cancer
cells and COS7 normal cells were seeded in a glass bottom dish at a
2.10. Characterizations of the multifunctional MMSNs density of 1  105 cells/well for 24 h, respectively. Then cells were
incubated with DOX loaded MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/
The morphology of Fe3O4 particles and MMSNs were observed AD-RGD (The concentration of DOX was 2 mg/mL) in the cell culture
by transmission electronic microscopy (TEM, JEOL-2100). Zeta po- media for 4 h. After removed the culture media and washed with
tential instrument (Nano-ZS ZEM3600, Malvern Instruments Co. PBS three times, the nuclei were stained with Hoechst 33342 at
Ltd, UK) and Fourier transform infrared spectroscopy (Per- 37  C for 15 min. Then all cells were observed by CLSM (Nikon C1-si,
kineElmer Spectrum One, USA) were employed to monitor the Japan). As the control, the cells were treated with free DOX (2 mg/
reaction processes of MMSNs. The surface area was detected by mL) and observed by CLSM. Quantitative analysis of the intracel-
BrunauereEmmetteTeller (BET) measurements and the pore size lular DOX red fluorescence was determined by flow cytometry (BD
distribution was obtained by Barrete-Joyner-Halenda (BJH) FACSAria TM III, USA). To confirm the internalization of MMSN-NH-
(ASAP2020, Micromeritics) analyses. All the samples were degassed Pt-CD/AD-RGD via the avb3 integrin receptor-mediated endocy-
at 120  C for 6 h under nitrogen before measurements. The content tosis, a competition assay was performed. HeLa cancer cells were
of Fe element and Pt element was determined by inductively pre-treated with the excess free RGD peptide (50 mM) for 4 h in
coupled plasma atomic emission spectrometry (ICP-AES). The DOX advance. Then the culture media were refreshed and incubated
concentration was determined by RF-530/PC fluorescence spec- with MMSN-NH-Pt-CD/AD-RGD (The concentration of DOX was
trofluorophotometer analysis (Shimadzu). All the T2-weighted MR 2 mg/mL) and analyzed by microscopy and flow cytometry just as
images were measured with a 7.0 T MR spectrometer (Bruker Inc., the above procedure. The cells without treatment were as the blank
Billerica, MA, USA). control.
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 91

2.15. Characterization of intracellular distribution of MMSN-NH-Pt- (5-weeks old, Wuhan University Animal Biosafty Level III Lab,
CD/AD-RGD by TEM Wuhan) were used for animal experiments. Subcutaneous tumors
were established by injecting HeLa cancer cells (1  107 cells) into
In this work, TEM was applied to monitor the distribution of subcutaneous of female mice. To demonstrate the magnetic tar-
MMSN-NH-Pt-CD/AD-RGD within HeLa and COS7 cells. HeLa can- geting enhanced MR imaging of tumors, two tumors were inocu-
cer cells and COS7 normal cells were incubated with MMSN-NH-Pt- lated in the left and right back of the hind leg region. For evaluating
CD/AD-RGD (The concentration of DOX was 2 mg/mL) for 12 h. Then the antitumor efficacy of MMSNs, the tumors were inoculated in
the cells were washed, collected by centrifugation and fixed with right back of the hind leg region.
1 mL general fixative (containing 2.5% glutaraldehyde in PBS buffer)
at 4  C and overnight. Cell samples were prepared for TEM obser- 2.20. In vivo MR imaging under magnetic targeting
vation according to standard procedures and then detected by us-
ing a electronic microscopy (TEM, JEOL-2100). When tumors had grown to approximately 200 mm3 in volume,
the tumor-bearing mice were intravenously injected with 200 mL
2.16. Analysis the intracellular uptake amount of Fe by ICP-AES MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/AD-RGD (The concentration
of DOX was 2 mg/kg and the corresponding particle concentration
HeLa cancer cells and COS7 normal cells were incubated with was 3.7 mg/mL). Afterwards, the tumor on the right was exposed to
MMSN-NH-Pt-CD/AD-RGD (The concentration of DOX was 2 mg/ an external magnetic field while the left was not. After injection
mL) for 2 h, 4 h, 6 h, and 8 h, respectively. After washed with PBS 30 min and 90 min, T2-weighted MR images were recorded with a
buffer three times, the cells were counted and collected. Before 7.0 T MR spectrometer (Bruker Inc., Billerica, MA, USA).
measurement, the cell samples were digested with aqua regia
(Extreme caution is required! Aqua regia is highly corrosive and
damaging to skin and eyes!) and diluted with water to 10 mL. 2.21. Biodistribution of nanoparticles in tumor tissues
Triplet parallel samples were performed and the average content of
the intracellular Fe element was analyzed using ICP-AES. The cells When tumors had reached approximately 200 mm3 in volume,
without treatment were as the blank control. the animals were randomized into three groups: MMSN-NH-Pt-CD,
MMSN-NH-Pt-CD/AD-RGD, and MMSN-NH-Pt-CD/AD-RGD with an
2.17. In vitro cell MR imaging external magnetic field. The mice were sacrificed after 24 h tail vein
injection of nanoparticles at a dosage of 2 mg/kg (DOX). Tumor
HeLa cancer cells and COS7 normal cells were respectively tissues and main organs (heart, liver, spleen, lung and kidney) for
seeded in 6-well plates at a density of 5  105 cells/well for 24 h. each group were harvested for analyzing the biodistribution of
Then the cells were incubated with MMSN-NH-Pt-CD/AD-RGD at different nanoparticles. The concentrations of Si in tumor tissues
different Fe concentrations (0.0, 0.1, 0.2, 0.4, 0.6 mM) for 4 h. After and main organs of each group were measured by ICP-AES.
washed with PBS buffer three times, the cells were counted and
collected. Then the cells (1  106) were suspended in gelatin (2%, 2.22. In vivo therapy and histochemistry analysis
300 mL) in plastic vials homogenously and the T2-weighted MR
images were measured with a 7.0 T MR spectrometer (Bruker Inc., When the tumor volume approximately reached 50 mm3, HeLa
Billerica, MA). tumor-bearing mice were studied in five groups (n ¼ 6 per group)
and treated with PBS, free DOX (The concentration of DOX was
2.18. In vitro cytotoxicity test by MTT assay 2 mg/kg), MMSN-NH-Pt-CD (The concentration of DOX was 2 mg/
kg and the corresponding particle concentration was 3.7 mg/mL),
In vitro ablation test was performed with HeLa cancer cells and MMSN-NH-Pt-CD/AD-RGD (The concentration of DOX was 2 mg/kg
COS7 normal cells by MTT assay. Briefly, HeLa cancer cells and COS7 and the corresponding particle concentration was 3.7 mg/mL) and
normal cells were seeded in 96-well plates at a density of 6000 MMSN-NH-Pt-CD/AD-RGD (The concentration of DOX was 2 mg/kg
cells/well, and then cells were incubated in 100 mL DMEM con- and the corresponding particle concentration was 3.7 mg/mL) with
taining 10% FBS and 1% antibiotics for 24 h prior to treating with an external magnetic field, respectively. Mice in five-treated groups
DOX loaded MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/AD-RGD in the were injected every two day until the completion of experiments.
presence or absence of an external magnetic field. After co- The weight of mice and tumor growth was monitored every day.
incubation for 2 h, the culture medium was replaced with 200 mL Tumor size was measured by a caliper and tumor volume was
of fresh medium and further incubated 46 h. Then 20 mL MTT so- determined using the following formula V ¼ W2  L/2, where W
lutions (5 mg/mL) was added to each well and incubated for 4 h. and L were the shortest and longest diameters of tumors, respec-
After that, the medium was removed and 200 mL DMSO was added. tively. On the day of 12, all mice were sacrificed and the tumors
The absorbance was measured at 570 nm using a microplate reader were excised and weighed. Simultaneously, the main organs (heart,
(Bio-Rad, Model 550, USA). The relative cell viability was calculated liver, spleen, lung and kidney) of the mice were also harvested and
as: cell viability ¼ (OD570 (samples)/OD570 (control))  100%, where used for histology analysis.
OD570 (control) was obtained in the absence of MMSN-NH-Pt-CD or The tumor tissues were stained by Prussian blue for evaluating
MMSN-NH-Pt-CD/AD-RGD, and OD570 (samples) was obtained in the the level of iron in tumors. To analyze the levels of apoptosis in
presence of MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/AD-RGD. The tumor cells, tumor tissue sections were stained by terminal deox-
cytotoxicity of DOX unloaded MMSN-NH-Pt-CD or MMSN-NH-Pt- ynucleotidyl transferased dUTP nick end labeling according to the
CD/AD-RGD was also evaluated just like the above procedure. Each manufacturer's protocol (Roche, Penzberg, Germany). The nuclei
value was averaged from four independent experiments. were labeled with 40 ,6-diamidino-2-phenylindole (DAPI) (Invi-
trogen, USA) and then examined by fluorescence microscopy. To
2.19. Animals and tumor model examine the morphology of tumor cells and evaluate the biocom-
patibility of MMSN, the tumor tissues and the main organs (heart,
All animal experiments were agreed with institutional animal liver, spleen, lung and kidney) were stained with hematoxylin and
use and care regulations from Wuhan University. BALB/c nude mice eosin (H&E) for histological examination.
92 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

2.23. Statistics analysis preparation of target products. As shown in Scheme 1A, the
multifunctional machined MMSNs were ultimately prepared after
The quantitative data collected were expressed as mean ± S.D. loading anticancer drug DOX and modifying with b-CD as the
Statistical significance was analyzed by three-sample Student's gatekeeper. Briefly, the detailed synthetic procedure included four
text. Statistical significance was inferred at a value of P < 0.05. steps: (I) MMSNs were first decorated with 3-
aminopropyltrimethoxysilane to obtain MMSN-NH2, (II) MMSN-
3. Results and discussion NH2 was further modified with the redox-sensitive linker plati-
num(IV) prodrug via the amidation reaction, denoted as MMSN-
3.1. Synthesis and characterization of multifunctional MMSNs NH-Pt-COOH, (III) The gatekeeper b-CD was immobilized onto the
surface of MMSN-NH-Pt-COOH by the reaction between mono-6-
The coreeshell type MMSNs were synthesized according to ethylenediamine-b-CD and carboxyl of platinum(IV) prodrug to
literature with slight modifications and the detail procedure was obtain MMSN-NH-Pt-CD, and (IV) The cancer targeting peptide AD-
described in experimental section [33]. As shown in Fig. 1A, the PEG8-GRGDS was functionalized with MMSN-NH-Pt-CD via
monodisperse and uniform MMSNs displayed a typical coreeshell hosteguest interaction between AD and b-CD, denoted as MMSN-
structure. The core of magnetic iron oxide nanoparticles (see NH-Pt-CD/AD-RGD. The successful introduction of multifunctional
Fig. S4) were coated with mesoporous silica shell, demonstrating machines on MMSNs was monitored and characterized by Fourier
that the successful preparation of MMSNs. The hydrodynamic size transform-infrared spectroscopy (FT-IR), size measurement and
of MMSNs was 103.2 ± 0.5 nm, analyzing by dynamic light scat- zeta potential analysis. As shown in Fig. S5, the disappearance of
tering (Fig. 1B). N2 adsorptionedesorption isotherms (Fig. 1C) and three strong bands at 2923, 2853, and 1478 cm1 confirmed that
BJH pore-size distribution curves (Fig. 1D) manifested that the CTAB molecules were removed thoroughly during the refluxing
MMSNs have well-developed pore diameter of 2.8 nm. The BET process in MeOH/NH4NO3 solution, which were the typical CH2
specific surface area and the total pore volume were 593 m2g1 and stretching and bending vibrations of CTAB molecules. The appear-
0.94 cm3g1, respectively. These results demonstrated the obtained ance absorption peak of 1420 cm1 corresponded to the C-N
MMSNs possess the promising potential as the ideal drug nano- stretching vibration in MMSN-NH2 and the absorption peak at
container, which exhibited uniform nanostructure and large ca- 1650 cm1 and 1300 cm1 displayed the typical structure of COOH
pacity for drug delivery. in MMSN-NH-Pt-COOH. Moreover, the hydrodynamic diameter of
Platinum(IV) prodrug was prepared by oxidizing the cisplatin to MMSNs was increased with the stepwise decoration of different
produce Pt(OH)2, followed by reacting with succinic anhydride (see functional groups and the change of zeta potential of MMSNs was
Fig. S1). The cancer targeting peptide AD-PEG8-GRGDS was syn- also related to the incorporation of different moieties (Table 1). The
thesized via the mature solid phase peptide synthesis technique in zeta potential of MMNS-NH2 was þ30.8 mV and decreased
our lab. The product platinum(IV) prodrug and AD-PEG8-GRGDS to 33.9 mV after linking with platinum(IV) prodrug with a
peptide were characterized by electrospray ionization-mass spec- multitude of carboxyl on the surface of MMSN-NH-Pt-COOH. After
trometry (ESI-MS). The theoretical molecular weight was 531.2 for loading DOX and reacting with the gatekeeper b-CD, the zeta po-
platinum(IV) prodrug and 1075.6 for AD-PEG8-GRGDS peptide, tential of MMSN-NH-Pt-CD increased to 9.5 mV and changed to
while the observed molecular weights were 532.9 and 1075.5 (see 3.3 mV since the incorporation of AD-PEG8-GRGDS peptide. As
Fig. S2 and Fig. S3), respectively, confirming the successful shown in Fig. S6 and Table S1, the BET pore volume and BJH pore

Fig. 1. Characterization of multifunctional theranostic magnetic mesoporous nanoparticles. (A) Transmission electron microscope (TEM) image of MMSNs. (B) Hydrodynamic size
distribution of MMSNs measured by dynamic light scattering (DLS). N2 adsorption/desorption isotherms (C) and BJH pore size distribution (D) of MMSNs. (E) Field-dependent
magnetization loop of MMSN-NH-Pt-CD/AD-RGD at 300 K. The absence of a hystersis loop indicated the superparamagnetic property of MMSN-NH-Pt-CD/AD-RGD. (F) T2-
weighted MR images of MMSN-NH-Pt-CD/AD-RGD in aqueous solution at different Fe concentrations. (G) T2 relaxation rate of MMSN-NH-Pt-CD/AD-RGD as a function of Fe
concentrations. (H) The drug release profile of DOX loaded MMSN-NH-Pt-CD/AD-RGD in PBS buffer with different ascorbic acid concentrations. Error bars were based on at least
triplicate measurements.
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 93

Table 1
Monitoring the modification processes of multifunctional theranostic MMSNs by size measurement and zeta potential analysis. The dates were based on triplicate
measurements.

Sample Zeta potential (mV) Hydrodynamic diameter (nm) PDI

MMSNs 15.2 ± 0.4 103.2 ± 0.5 0.06


MMSN-NH2 30.8 ± 0.5 111.2 ± 0.4 0.18
MMSN-NH-Pt-COOH 33.9 ± 0.6 131.1 ± 0.6 0.11
MMSN-NH-Pt-CD 9.5 ± 0.3 148.7 ± 0.5 0.09
MMSN-NH-Pt-CD/AD-RGD 3.3 ± 0.2 163.8 ± 0.5 0.15

diameter decreased gradually during the functionalization process, NH-Pt-CD/AD-RGD at 300 K showed no hysteresis (Fig. 1E), indi-
which further confirmed the successful modification. Moreover, the cating that they have the superparamagnetic property. Further-
solid state 13C NMR spectra (see Fig. S7) further demonstrated the more, T2-weighted MRI of MMSN-NH-Pt-CD/AD-RGD on a 7.0 T MR
functional components were anchored on the surface of MMSNs. spectrometer was investigated and utilized to evaluate their
The stability of MMSN-NH-Pt-CD/AD-RGD at normal physiological contrast enhancement effect. As shown in Fig. 1F, the T2 signal in-
conditions was also monitored by dynamic light scattering (DLS) tensity revealed a concentration-dependent darkening effect and
detection. As shown in Fig. S8, no obvious changes in hydrodynamic decreased significantly with increasing Fe concentration. In addi-
diameter and polydispersity index (PDI) were observed during four tion, the T2 relaxation rate (r2 ¼ 1/T2) of the MMSN-NH-Pt-CD/AD-
days, indicating that MMSN-NH-Pt-CD/AD-RGD was stable at RGD was calculated to be 136.6 mM1S1 (Fig. 1G), suggesting the
normal physiological conditions. These results confirmed the suc- feasibility of applying our designed MMSNs as a contrast agent in
cessful preparation of multifunctional MMSNs. MR imaging.
It is well known that magnetic iron oxide core of MMSNs has To precisely control the drug release intracellularly, the
multiple functionalities, including MRI and magnetic targeting to reduction-sensitive platinum(IV) prodrug was employed as the
direct drugs to localized lesions [48]. In our designed DOX loading linker to connect the gatekeeper and mesoporous silica shell. Here,
MMSN-NH-Pt-CD/AD-RGD, the weight ratio of Fe was 4.7% (w/w) ascorbic acid was selected as a reduction agent to trigger the release
and the weight ratio of Pt was 2.4% (w/w), determining by induc- of drug because it abound in cells and has been demonstrated to be
tively coupled plasma atomic emission spectrometry (ICP-AES). one of the major substance for the reduction of platinum(IV)
According to fluorescence measurement, about 5.4% (w/w) DOX [46,47]. As shown in Fig. 1H, at physiological condition (PBS buffer,
was loaded into the mesoporous silica shell of MMSN-NH-Pt-CD/ pH 7.4), a negligible amount of DOX was leaked from MMSN-NH-
AD-RGD. Field-dependent magnetism loop of drug loaded MMSN- Pt-CD/AD-RGD, indicating that perfect capping efficiency of b-CD-

Fig. 2. In vitro tumor-targeted drug delivery confirmed by confocal laser scanning microscopy (CLSM) observations. CLSM images of HeLa cancer cells incubated with DOX loaded
MMSN-NH-Pt-CD/AD-RGD without (A) and with (B) an external magnetic field for 1 h. CLSM images of HeLa cancer cells incubated with DOX loaded MMSN-NH-Pt-CD/AD-RGD
without (C) and with (D) an external magnetic field for 2 h. CLSM images of COS7 normal cells incubated with DOX loaded MMSN-NH-Pt-CD/AD-RGD without (E) and with (F) an
external magnetic field for 1 h. CLSM images of COS7 normal cells incubated with DOX loaded MMSN-NH-Pt-CD/AD-RGD without (G) and with (H) an external magnetic field for
2 h. The concentration of DOX in each formulation was 2 mg/mL. M represented the magnetic field.
94 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

based machine. However, when the drug loaded MMSN-NH-Pt-CD/ targeting RGD motif can enhance the uptake of particles through
AD-RGD was incubated with 1 mM and 10 mM ascorbic acid (pH specifically binding with cancer cells with avb3 integrin overex-
7.4) in PBS buffer, DOX was rapidly released. This was attributed to pressed [49,50]. With the aid of an external magnetic field,
ascorbic acid induced the cleavage of reduction-sensitive linker in MMSNeNHePt-CD/AD-RGD further enhanced the internalization
the progress of reducing platinum(IV) prodrug to the activated capability via directionally increasing RGDintegrin interactions. In
platinum(II) drug, leading to the removal of b-CD gatekeeper and contrast, few particles were uptaken by COS7 normal cells due to
triggering the in situ drug release. Above results confirmed the the lack of integrin expression. These results suggested that MMSN-
successful construction of multifunctional MMSNs, which exhibi- NH-Pt-CD/AD-RGD can targetedly deliver anticancer drug to cancer
ted uniform nanostructure with suitable surface charge as well as cells instead of normal cells, which showed great promising for
controlled drug release. selectively killing of cancer cells.

3.2. In vitro tumor-targeted drug delivery of MMSN-NH-Pt-CD/AD- 3.3. Quantitative analysis of cellular uptake and intracellular
RGD redox-triggered cytotoxicity of multifunctional MMSNs

To confirm the active targeting specificity and magnetically To further quantitatively evaluate cellular uptake of multifunc-
enhanced drug delivery, we incubated DOX loaded MMSN-NH-Pt- tional MMSNs and examine the intracellular drug delivery, cells
CD/AD-RGD with a cervix carcinoma cell line (HeLa cells, in which treated with DOX loaded MMSN-NH-Pt-CD or MMSN-NH-Pt-CD/
the avb3 integrin is overexpressed) and a normal kidney fibroblast AD-RGD were measured by confocal laser scanning microscopy
cell line (COS7 cells, in which the avb3 integrin expression is (CLSM) and flow cytometry. As shown in Fig. 3A, the red fluores-
negative) with or without magnetic attraction. As shown in Fig. 2, cence of DOX stained the whole HeLa cancer cells even including
red fluorescence of DOX could be clearly observed in HeLa cancer cell nuclei after 4 h incubation with MMSN-NH-Pt-CD/AD-RGD,
cells and the fluorescence intensity increased significantly with the indicating that particles were abundantly uptaken by cells and DOX
time extending (Fig. 2C compared with Fig. 2A). Moreover, with the was released in the cytoplasm effectively. Obviously, RGD targeting
aid of magnetic field, HeLa cancer cells incubated with MMSN-NH- motif on the surface of MMSN-NH-Pt-CD/AD-RGD could facilitate
Pt-CD/AD-RGD showed remarkably enhanced DOX uptake in cellular uptake of nanoparticles through specific receptor-mediated
comparing to cells without magnetic attraction. However, only endocytosis. Afterward the platinum(IV) prodrug was reduced by
weak red fluorescence of DOX could be detected in normal COS7 intracellular reductive agents, leading to removal of the gatekeeper
cells (Fig. 2E and G) and there was a slight increase in the presence and release of the DOX. Finally, the released DOX specifically
of magnetic field (Fig. 2F and H). This was due to the fact that active diffused and accumulated in the nucleus of cancer cells to induce

Fig. 3. Evaluation the intracellular redox-sensitive drug release by CLSM and quantitative analysis of cellular uptake of DOX by flow cytometry. CLSM images of HeLa cancer cells
incubated with DOX loaded MMSN-NH-Pt-CD/AD-RGD for 4 h (A) or 4 h for HeLa cancer cells pre-treated with the excess of free RGD in advance (B). CLSM images of COS7 normal
cells incubated with DOX loaded MMSN-NH-Pt-CD/AD-RGD for 4 h (C). CLSM images of HeLa cancer cells (D) and COS7 normal cells (E) incubated with DOX loaded MMSN-NH-Pt-
CD for 4 h. Flow cytometry measurement of internalized DOX signals in HeLa cancer cells (F) or COS7 normal cells (G). The mean fluorescence intensity (MFI) of HeLa cancer cells (H)
and COS7 normal cells (I) correspond to flow cytometry analysis. The cells without treatment (blank line), treating with MMSN-NH-Pt-CD for 4 h (blue line), 4 h with MMSN-NH-Pt-
CD/AD-RGD (green line), and 4 h with MMSN-NH-Pt-CD/AD-RGD and pre-treated with the excess of RGD in advance (red line). The concentration of DOX in each formulation was
2 mg/mL. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 95

Fig. 4. Evaluating the intracellular MMSNs by TEM observation, ICP-AES analysis and T2-weighted MR examination. (A) TEM image of HeLa cancer cells incubated with MMSN-NH-
Pt-CD/AD-RGD 12 h (B) TEM image of COS7 normal cells incubated with MMSN-NH-Pt-CD/AD-RGD 12 h. (C) Quantification of Fe levels by ICP-AES in HeLa and COS7 cells treated
with MMSN-NH-Pt-CD/AD-RGD for 2 h, 4 h, 6 h, and 8 h, respectively. The cells without treatment were used as the blank control. Statistical significance in difference was analyzed
using student's T-Test: *p < 0.1 and **p < 0.01. (D) T2-weighted MR images of HeLa cells (1  106) incubated with MMSN-NH-Pt-CD/AD-RGD at different Fe concentrations. (E) T2-
weighted MR images of COS7 cells (1  106) incubated with MMSN-NH-Pt-CD/AD-RGD at different Fe concentrations.

apoptosis and cell death. However, when the receptor-mediated fluorescence intensity (MFI) of DOX in HeLa cancer cells was 5.3-
endocytosis was blocked by free RGD peptide, the internalization fold than COS7 cells after incubating with MMSN-NH-Pt-CD/AD-
of particles was drastically decreased on account of the free RGD RGD and 8.2-fold increases compared with that treated with
competition (Fig. 3B). Similarly, limited DOX was detected in COS7 MMSN-NH-Pt-CD (Fig. 3H and I). In contrast, since free DOX has no
normal cells due to the less expression of integrin receptors of selectivity, similar fluorescence intensities in both HeLa and COS7
normal cells (Fig. 3C). As the control, MMSN-NH-Pt-CD without cells were observed (see Fig. S9).
RGD motif showed little uptake of both HeLa and COS7 cells (Fig. 3D Moreover, TEM was utilized to monitor the distribution of
and E), which further confirmed that RGD peptide specifically MMSN-NH-Pt-CD/AD-RGD within HeLa and COS7 cells. As shown
improved the cellular uptake. The same phenomenon could also be in Fig. 4A and B, a multitude of MMSN-NH-Pt-CD/AD-RGD could be
detected from the quantitative flow cytometry analysis. The mean observed in the cytoplasm of HeLa cancer cells (red circle) and only

Fig. 5. Evaluation the anticancer ability of multifunctional MMSNs in vitro. Cytotoxicity of DOX loaded magnetic mesoporous silica nanoparticles against (A) HeLa cancer cells and
(B) COS7 normal cells in the absence or in the presence of a magnetic field for 48 h, respectively. M represented the magnetic field.
96 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

a little was detected in COS7 normal cells, which were consistent Fig. 4D and E, the HeLa cancer cells treated with MMSN-NH-Pt-CD/
with above CLSM observations. The intracellular Fe contents were AD-RGD showed notably decreased signal intensity in the T2-
further examined by ICP-AES quantitative analysis (Fig. 4C). With weighted MR images comparing to COS7 normal cells. With
increased incubation time, the Fe contents enhanced remarkably in increasing Fe concentration, the dark intensity of T2-weighted
the HeLa cancer cells, which were 1.87-fold and 2.6-fold more than images of HeLa cancer cells became more obvious. The above re-
that in the COS7 normal cells for 6 h and 8 h, respectively. Simul- sults verified that the MMSN-NH-Pt-CD/AD-RGD could be inter-
taneously, it is well known that T2 signal intensity increases in a Fe nalized in cancer cells preferentially and used as promising contrast
concentration-dependent manner. The more Fe content in the cell agents for tumor-targeted MRI.
would reveal relatively dark T2-weighted images. As shown in Furthermore, the in vitro cancer inhibition ability of

Fig. 6. In vivo noninvasive tumor-targeted MR imaging. (A) T2-weighted MR images of HeLa tumor-bearing mice before and after intravenous injection of MMSN-NH-Pt-CD/AD-
RGD. (B) The pseudo-color images of the tumor tissues derived from Fig. A. (C) T2-weighted MR images of HeLa tumor-bearing mice before and after intravenous injection of MMSN-
NH-Pt-CD. (D) The pseudo-color images of the tumor tissues derived from Fig. C. The two tumors were inoculated in the left and right back of the hind leg region (white arrow). To
examine magnetic targeting, an external magnet was securely fixed onto the right tumor of each mouse by bandage.
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 97

multifunctional MMSNs was evaluated by MTT assay. At presented


in Fig. 5A, the HeLa cell viability after incubated with DOX loaded
MMSN-NH-Pt-CD/AD-RGD in the absence of a magnetic field was
about 30%. And the cell viability further reduced to 18% in the
presence of an external magnetic field. At the same time, we also
detected that the HeLa cell viability was more than 60% with an
external magnetic field or above 80% without an external magnetic
field after treated with DOX loaded MMSN-NH-Pt-CD. With the aid
of active targeting RGD motif, MMSN-NH-Pt-CD/AD-RGD would
preferentially enter into cancer cells in compared to MMSN-NH-Pt-
CD and significantly enhance cellular uptake. Under the intracel-
lular reductive condition, the platinum(IV) prodrug between the
gatekeeper and MMSNs were cleaved, inducing the accelerated
release of loaded drug to kill cancer cells. In comparison to litera-
ture report using gold nanoparticles as drug nanocarrier [51], the
Fig. 7. The biodistribution of different nanoparticles (Si concentration% ID/g) in various
anticancer therapeutic effect of MMSN-NH-Pt-CD/AD-RGD (IC50 for
organs of mice collected at 24 h after intravenous injection of MMSN-NH-Pt-CD,
HeLa cells was 1.2 mg/mL) was notably better than that of gold MMSN-NH-Pt-CD/AD-RGD, and MMSN-NH-Pt-CD/AD-RGD with an external magnetic
nanoparticles (IC50 for HeLa cells was 1.8 mg/mL). To further confirm field. The statistical significance in difference was analyzed using student's T-Test:
the cell selectivity of MMSNs, the normal cell viability was also *p < 0.05 and **p < 0.01.
investigated. As shown in Fig. 5B, the COS7 normal cell viabilities
were >80% after treated with different formulations of MMSNs. The
low uptake of MMSNs by normal cells resulted in the less cyto- detected (ca. 13.8% ID/g), which was much higher than that of
toxicity. In addition, the unloaded MMSN-NH-Pt-CD/AD-RGD also MMSN-NH-Pt-CD and MMSN-NH-Pt-CD/AD-RGD (ca. 2.3 and 8.9%
exhibited some cytotoxicity to HeLa cancer cells since the plati- ID/g, respectively). To investigate antitumor efficacy of MMSNs
num(IV) prodrug could be reduced to active platinum(II) drug to in vivo, thirty HeLa tumor-bearing mice were randomly divided into
kill cells to some extent (see Fig. S10). Consequently, these results five groups (n ¼ 6) and treated with PBS buffer, free DOX, DOX
illustrated that MMSN-NH-Pt-CD/AD-RGD with cancer targeting loaded MMSN-NH-Pt-CD, DOX loaded MMSN-NH-Pt-CD/AD-RGD
showed the capability to selectively eliminate cancer cells not and DOX loaded MMSN-NH-Pt-CD/AD-RGD with an external
normal cells, which was beneficial to destroy cancer as well as magnetic field every other day, respectively. The injection of
reduce the side effects to normal tissues. different formulations of DOX had the equal dosage of 2 mg/kg and
tumor volume of each mouse was measured daily. As shown in
3.4. In vivo magnetic targeting enhanced MR imaging and precise Fig. 8A, different DOX formulations showed significant therapeutic
therapy effects on suppressing tumor growth in comparison to that of PBS
buffer after intravenous injection. MMSN-NH-Pt-CD/AD-RGD par-
As a noninvasive imaging technique with high spatial and ticles had notably higher inhibition efficacy towards tumor growth
temporal resolution, MRI has been recognized as powerful tool for than the free DOX and MMSN-NH-Pt-CD particles (*p < 0.05),
early detection of cancer and evaluation the therapeutic response. resulting from the active targeting ability of these particles. More
To demonstrate the magnetic targeting enhanced MR imaging of importantly, a remarkably enhanced therapeutic effect in tumor
MMSNs, two tumors were inoculated in the left and right back of suppression for MMSN-NH-Pt-CD/AD-RGD particles with an
the hind leg region and the tumor on the right was in presence of an external magnetic field was observed (**p < 0.01), suggesting that
external magnetic field while the left was not. After intravenous the magnetic attraction increased the tumor accumulation of
injection of MMSN-NH-Pt-CD/AD-RGD or MMSN-NH-Pt-CD on the MMSN-NH-Pt-CD/AD-RGD and the in situ redox-sensitive drug
nude mice bearing HeLa tumors, T2-weighted MR images were release was beneficial to enhance antitumor activity and achieve
taken at a given time. As shown in Fig. 6, signal attenuation at the precise therapy. The superior therapeutic effect of MMSN-NH-Pt-
tumor tissues could be clearly observed in both groups at 30 min CD/AD-RGD particles was also substantiated directly by the average
after injecting, while the signal dropping from MMSN-NH-Pt-CD/ tumor weights (Fig. 8B) as well as the representative tumor images
AD-RGD treated mice was more remarkable than that adminis- (Fig. 8C). Furthermore, the enriching of MMSNs in tumor tissues
trated MMSN-NH-Pt-CD (Fig. 6B comparing to Fig. 6D). Compared was visually evidenced by Prussian blue staining. As shown in
with the simple passive EPR effect of MMSN-NH-Pt-CD, MMSN-NH- Fig. 8EeE4, the significantly increased iron in the tumor treated by
Pt-CD/AD-RGD with the active targeting effect could enhance the MMSN-NH-Pt-CD/AD-RGD with a magnetic field was detected,
accumulation at the tumor site. The tumor targeting and retention which was much more than that in tumors treated by MMSN-NH-
was further increased significantly under the guide of magnetic Pt-CD and MMSN-NH-Pt-CD/AD-RGD without magnetic attraction.
force, leading to the much darker signal in T2-weighted MR images Consistent with the results by MR imaging, Prussian blue staining
in the right tumor (Fig. 6B, the right tumor comparing to the left assay confirmed the targeted tumor accumulation of MMSNs,
tumor). Clearly, the magnetic targeting mediated EPR effect could which contributed to selectively ablating cancer and reducing
overcome tumor heterogeneity and enhance the efficacious accu- adverse effects to normal tissues.
mulation of MMSNs in the tumor site, which also beneficial for The enhanced antitumor effect of MMSNs was further evaluated
tumor-targeted therapy. by Hematoxylin and eosin (H&E) staining and immunohisto-
Furthermore, in vivo tumor targeting efficiency of MMSNs was chemical labeling of terminal deoxynucleotidyl transferased dUTP
evaluated. After 24 h intravenous injection of different MMSNs, the nick end labeling (TUNEL) assay for analysis of apoptosis.
HeLa tumor-bearing mice was sacrificed. Tumor tissues and main Comparing to the control group (Fig. 8F), only a spot of tumor cells
organs (heart, liver, spleen, lung and kidney) for each group were showed apoptosis/necrosis when treated with free DOX (Fig. 8F1)
harvested for analyzing the amounts of Si by ICP-AES. As shown in and MMSN-NH-Pt-CD (Fig. 8F2). Excitingly, a multitude of tumor
Fig. 7, with the aid of an external magnetic field, a considerable cells were destroyed in the MMSN-NH-Pt-CD/AD-RGD treated
amount of MMSN-NH-Pt-CD/AD-RGD accumulation in tumors was groups (Fig. 8F3 and 8F4) and more serious damages were detected
98 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

Fig. 8. Evaluating the therapeutic efficacy of multifunctional MMSNs in vivo. (A) Tumor growth curves in HeLa tumor-bearing mice after intravenous injection of different for-
mulations of DOX. The dose of DOX was 2 mg/kg. (B) The average tumor weights after various treatments. (C) The representative images of the tumors after treatment. (D)
Quantification of the percentage of TUNEL positive apoptotic cells in tumors treated with different DOX formulations. The apoptotic cells in each slide were statistically analyzed by
randomly taking three areas in a large view. *p < 0.05 was determined by a Student's t-test when the group MMSN-NH-Pt-CD/AD-RGD versus the tumors treated with free DOX and
MMSN-NH-Pt-CD, and **p < 0.01 was determined by a Student's t-test when the group MMSN-NH-Pt-CD/AD-RGD with an external magnetic field versus all other treatment groups.
Histological examination of tumor slices after treated with different DOX formulations. (E) Prussian blue staining. (F) H&E staining. (G) TUNEL immunofluorescence staining. The
nuclei were labeled with DAPI, and the apoptotic cells were green. The scale bars were 50 mm. (EeG) The group treated with PBS. (E1-G1) The group treated with free DOX. (E2-G2)
The group treated with DOX loaded MMSN-NH-Pt-CD. (E3-G3) The group treated with DOX loaded MMSN-NH-Pt-CD/AD-RGD. (E4-G4) The group treated with DOX loaded MMSN-
NH-Pt-CD/AD-RGD with an external magnetic field. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 99

under the guide of magnetic force than without. In addition, the slightly decreased due to its systemic toxicity. The H&E staining of
quantitative analysis of apoptotic cells in each group by TUNEL the liver also revealed severe hepatotoxicity in the mice treated
staining assay also confirmed the above observations. As shown in with free DOX (Fig. 9B). The cell nuclei shrinkage and the extensive
Fig. 8D and Fig. 8GeG4, the percentage of apoptotic cells in the apoptotic liver cells were observed (red circle). In contrast, no
tumor treated by MMSN-NH-Pt-CD/AD-RGD with a magnetic field significant change in the body weights was detected and no
were about 68%, which were much more than other groups (PBS: apparent abnormality of major organs in the histopathological
2%, free DOX: 18.9%, MMSN-NH-Pt-CD: 15.7%, and MMSNeNHePt- examination was discovered during the treatment of MMSNs
CD/AD-RGD: 41.2%). These results further confirmed that magnetic (Fig. 9). These results demonstrated that MMSNs were favorably
targeting mediated EPR effect could circulate the heterogeneity of biocompatible and had the excellent performance in eliminating
tumor and increase the enrichment of MMSNs in tumor sites. tumor with minimal side effects.
Subsequently, the in situ redox-sensitive drug release promoted to
increase the therapeutic index and achieve the favorable antitumor 4. Conclusion
effects. However, it is well known that free DOX without any tumor
targeting ability only inhibit tumor growth to some extent and al- In summary, a multifunctional theranostic drug delivery system
ways induce severe side effects on normal organs [52,53]. As shown based on magnetic mesoporous silica nanoparticles was designed
in Fig. S11, the body weights of mice treated with free DOX showed and prepared for tumor-targeted magnetic resonance imaging and

Fig. 9. Evaluating the biocompatibility of multifunctional MMSNs in vivo. The major organs (heart, liver, spleen, kidney and lung) were stained with H&E after different treatments.
The scale bars were 50 mm. (A) The group treated with PBS. (B) The group treated with free DOX. (C) The group treated with DOX loaded MMSN-NH-Pt-CD. (D) The group treated
with DOX loaded MMSN-NH-Pt-CD/AD-RGD. (E) The group treated with DOX loaded MMSN-NH-Pt-CD/AD-RGD with an external magnetic field.
100 W.-H. Chen et al. / Biomaterials 76 (2016) 87e101

precisely controlled therapy. The multifunctional MMSN demon- Res. 44 (2011) 903e913.
[15] Q. He, J. Shi, MSN anti-cancer nanomedicines: chemotherapy enhancement,
strated the ability of selective uptake by cancer cells and induction
overcoming of drug resistance, and metastasis inhibition, Adv. Mater. 26
of the intracellular redox-sensitive release of anticancer drug to (2014) 391e411.
eliminate cancer cells effectively. Moreover, the multifunctional [16] F. Tang, L. Li, D. Chen, Mesoporous silica nanoparticles: synthesis, biocom-
MMSN exhibited high contrast in MRI for locating tumor in vivo and patibility and drug delivery, Adv. Mater. 24 (2012) 1504e1534.
[17] T.D. Nguyen, H.R. Tseng, P.C. Celestre, A.H. Flood, Y. Liu, J.F. Stoddart, et al.,
achieved significant antitumor efficacy with minimal side effects. It A reversible molecular valve, Proc. Natl. Acad. Sci. U. S. A. 102 (2005)
is envisioned that the combination of magnetic nanoparticle and 10029e10034.
mesoporous silica nanoparticle have following advantages: (1) [18] Q. Zhang, F. Liu, K.T. Nguyen, X. Ma, X.J. Wang, B.G. Xing, et al., Multifunctional
mesoporous silica nanoparticles for cancer-targeted and controlled drug de-
favourable biocompatibility and biostability, (2) remarkably livery, Adv. Funct. Mater. 22 (2012) 5144e5156.
improved efficacious accumulation of magnetic mesoporous silica [19] Z. Luo, K. Cai, Y. Hu, B. Zhang, D. Xu, Cell-specific intracellular anticancer drug
nanoparticles in the targeted cancer, (3) stimuli-responsive release delivery from mesoporous silica nanoparticles with pH sensitivity, Adv.
Healthc. Mater. 1 (2012) 321e325.
of therapeutic agents, (4) integrated diagnostic and therapeutic [20] H. Kim, S. Kim, C. Park, H. Lee, H.J. Park, C. Kim, Glutathione-induced intra-
functions. This multifunctional theranostic nanoparticle should cellular release of guests from mesoporous silica nanocontainers with cyclo-
have great potential in the future cancer therapy. dextrin gatekeepers, Adv. Mater. 22 (2010) 4280e4283.
[21] K. Patel, S. Angelos, W.R. Dichtel, A. Coskun, Y.W. Yang, J.I. Zink, et al., Enzyme-
responsive snap-top covered silica nanocontainers, J. Am. Chem. Soc. 130
Acknowledgments (2008) 2382e2383.
[22] Z. Luo, X. Ding, Y. Hu, S. Wu, Y. Xiang, Y. Zeng, et al., Engineering a hollow
nanocontainer platform with multifunctional molecular machines for tumor-
This work was supported by the National Natural Science targeted therapy in vitro and in vivo, ACS Nano 7 (2013) 10271e10284.
Foundation of China (51125014, 51233003 and 51533006), the [23] C. Chen, J. Geng, F. Pu, X. Yang, J. Ren, X. Qu, Polyvalent nucleic acid/meso-
porous silica nanoparticle conjugates: dual stimuli-responsive vehicles for
Ministry of Education of China (20120141130003) and supported by
intracellular drug delivery, Angew. Chem. Int. Ed. Engl. 50 (2011) 882e886.
the Fundamental Research Funds for the Central Universities [24] J. Kim, J.E. Lee, J. Lee, J.H. Yu, B.C. Kim, K. An, et al., Magnetic fluorescent de-
(2014203020201 and 2014203020204). livery vehicle using uniform mesoporous silica spheres embedded with
monodisperse magnetic and semiconductor nanocrystals, J. Am. Chem. Soc.
128 (2006) 688e689.
Appendix A. Supplementary data [25] W. Zhao, J. Gu, L. Zhang, H. Chen, J. Shi, Fabrication of uniform magnetic
nanocomposite spheres with a magnetic core/mesoporous silica shell struc-
ture, J. Am. Chem. Soc. 127 (2005) 8916e8917.
Supplementary data related to this article can be found at http:// [26] R.I. Nooney, D. Thirunavukkarasu, Y. Chen, R. Josephs, A.E. Ostafin, Self-as-
dx.doi.org/10.1016/j.biomaterials.2015.10.053. sembly of mesoporous nanoscale silica/gold composites, Langmuir 19 (2003)
7628e7637.
[27] M. Liong, B. France, K.A. Bradley, J.I. Zink, Antimicrobial activity of silver
References nanocrystals encapsulated in mesoporous silica nanoparticles, Adv. Mater. 21
(2009) 1684e1689.
[1] D. Peer, J.M. Karp, S. Hong, O.C. Farokhzad, R. Margalit, R. Langer, Nanocarriers [28] Y. Wang, H. Gu, Core-shell-type magnetic mesoporous silica nanocomposites
as an emerging platform for cancer therapy, Nat. Nanotechnol. 2 (2007) for bioimaging and therapeutic agent delivery, Adv. Mater. 27 (2015)
751e760. 576e585.
[2] A.Z. Wang, R. Langer, O.C. Farokhzad, Nanoparticle delivery of cancer drugs, [29] M. Liong, J. Lu, M. Kovochich, T. Xia, S.G. Ruehm, A.E. Nel, et al., Multifunc-
Annu. Rev. Med. 63 (2012) 185e198. tional inorganic nanoparticles for imaging, targeting, and drug delivery, ACS
[3] H. Koo, M.S. Huh, I.C. Sun, S.H. Yuk, K. Choi, K. Kim, et al., In vivo targeted Nano 2 (2008) 889e896.
delivery of nanoparticles for theranosis, Acc. Chem. Res. 44 (2011) [30] Z. Li, K. Dong, S. Huang, E. Ju, Z. Liu, M. Yin, et al., A smart nanoassembly for
1018e1028. multistage targeted drug delivery and magnetic resonance imaging, Adv.
[4] W. Dong, Y. Li, D. Niu, Z. Ma, J. Gu, Y. Chen, et al., Facile synthesis of mono- Funct. Mater. 24 (2014) 3612e3620.
disperse superparamagnetic Fe3O4 Core@hybrid@Au shell nanocomposite for [31] W.P. Li, P.Y. Liao, C.H. Su, C.S. Yeh, Formation of oligonucleotide-gated silica
bimodal imaging and photothermal therapy, Adv. Mater. 23 (2011) shell-coated Fe3O4-Au core-shell nanotrisoctahedra for magnetically targeted
5392e5397. and near-infrared light-responsive theranostic platform, J. Am. Chem. Soc. 136
[5] H. Meng, M. Xue, T. Xia, Z. Ji, D.Y. Tarn, J.I. Zink, et al., Use of size and a (2014) 10062e10075.
copolymer design feature to improve the biodistribution and the enhanced [32] P. Yang, Z. Quan, Z. Hou, C. Li, X. Kang, Z. Cheng, et al., A magnetic, luminescent
permeability and retention effect of doxorubicin-loaded mesoporous silica and mesoporous core-shell structured composite material as drug carrier,
nanoparticles in a murine xenograft tumor model, ACS Nano 5 (2011) Biomaterials 30 (2009) 4786e4795.
4131e4144. [33] J. Kim, H.S. Kim, N. Lee, T. Kim, H. Kim, T. Yu, et al., Multifunctional uniform
[6] Z.H. Peng, J. Kope cek, Enhancing accumulation and penetration of HPMA nanoparticles composed of a magnetite nanocrystal core and a mesoporous
copolymer-doxorubicin conjugates in 2D and 3D prostate cancer cells via silica shell for magnetic resonance and fluorescence imaging and for drug
iRGD conjugation with an MMP-2 cleavable spacer, J. Am. Chem. Soc. 137 delivery, Angew. Chem. Int. Ed. Engl. 47 (2008) 8438e8441.
(2015) 6726e6729. [34] Z. Li, S. Yin, L. Cheng, K. Yang, Y. Li, Z. Liu, Magnetic targeting enhanced
[7] D. Ling, W. Park, S.J. Park, Y. Lu, K.S. Kim, M.J. Hackett, et al., Multifunctional theranostic strategy based on multimodal imaging for selective ablation of
tumor pH-sensitive self-assembled nanoparticles for bimodal imaging and cancer, Adv. Funct. Mater. 24 (2014) 2312e2321.
treatment of resistant heterogeneous tumors, J. Am. Chem. Soc. 136 (2014) [35] C. Wang, H. Xu, C. Liang, Y. Liu, Z. Li, G. Yang, et al., Iron oxide @polypyrrole
5647e5655. nanoparticles as a multifunctional drug carrier for remotely controlled cancer
[8] J.Z. Du, X.J. Du, C.Q. Mao, J. Wang, Tailor-made dual pH-sensitive polymer- therapy with synergistic antitumor effect, ACS Nano 7 (2013) 6782e6795.
doxorubicin nanoparticles for efficient anticancer drug delivery, J. Am. Chem. [36] L.S. Lin, Z.X. Cong, J.B. Cao, K.M. Ke, Q.L. Peng, J. Gao, et al., Multifunctional
Soc. 133 (2011) 17560e17563. Fe3O4@polydopamine core-shell nanocomposites for intracellular mRNA
[9] W.H. Chen, G.F. Luo, Q. Lei, H.Z. Jia, S. Hong, Q.R. Wang, et al., MMP-2 detection and imaging-guided photothermal therapy, ACS Nano 8 (2014)
responsive polymeric micelles for cancer-targeted intracellular drug delivery, 3876e3883.
Chem. Commun. 51 (2015) 465e468. [37] D. Ling, N. Lee, T. Hyeon, Chemical synthesis and assembly of uniformly sized
[10] H. Wang, Y. Wu, R. Zhao, G. Nie, Engineering the assemblies of biomaterial iron oxide nanoparticles for medical applications, Acc. Chem. Res. 48 (2015)
nanocarriers for delivery of multiple theranostic agents with enhanced anti- 1276e1285.
tumor efficacy, Adv. Mater. 25 (2013) 1616e1622. [38] Y. Chen, H. Chen, S. Zhang, F. Chen, L. Zhang, J. Zhang, et al., Multifunctional
[11] J. Zhang, Z.F. Yuan, Y. Wang, W.H. Chen, G.F. Luo, S.X. Cheng, et al., Multi- mesoporous nanoellipsoids for biological bimodal imaging and magnetically
functional envelope-type mesoporous silica nanoparticles for tumor-triggered targeted delivery of anticancer drugs, Adv. Funct. Mater. 21 (2011) 270e278.
targeting drug delivery, J. Am. Chem. Soc. 135 (2013) 5068e5073. [39] Y. Yuan, R.T. Kwok, B.Z. Tang, B. Liu, Targeted theranostic platinum(IV) pro-
[12] J. Yu, C. Yang, J. Li, Y. Ding, L. Zhang, M.Z. Yousaf, et al., Multifunctional Fe5C2 drug with a built-in aggregation-induced emission light-up apoptosis sensor
nanoparticles: a targeted theranostic platform for magnetic resonance im- for noninvasive early evaluation of its therapeutic responses in situ, J. Am.
aging and photoacoustic tomography-guided photothermal therapy, Adv. Chem. Soc. 136 (2014) 2546e2554.
Mater. 26 (2014) 4114e4120. [40] A. Kumar, S. Huo, X. Zhang, J. Liu, A. Tan, S. Li, et al., Neuropilin-1-targeted gold
[13] J.L. Vivero-Escoto, I.I. Slowing, B.G. Trewyn, V.S. Lin, Mesoporous silica nano- nanoparticles enhance therapeutic efficacy of platinum(IV) drug for prostate
particles for intracellular controlled drug delivery, Small 6 (2010) 1952e1967. cancer treatment, ACS Nano 8 (2014) 4205e4220.
[14] M.W. Ambrogio, C.R. Thomas, Y.L. Zhao, J.I. Zink, J.F. Stoddart, Mechanized [41] L. Pan, J. Liu, Q. He, J. Shi, MSN-mediated sequential vascular-to-cell nuclear-
silica nanoparticles: a new frontier in theranostic nanomedicine, Acc. Chem. targeted drug delivery for efficient tumor regression, Adv. Mater. 26 (2014)
W.-H. Chen et al. / Biomaterials 76 (2016) 87e101 101

6742e6748. characteristics for in situ monitoring of drug activation, Chem. Commun. 50


[42] R. Haubner, H.J. Wester, W.A. Weber, C. Mang, S.I. Ziegler, S.L. Goodman, et al., (2014) 3868e3870.
Noninvasive imaging of avb3 integrin expression using 18F-labeled RGD- [48] J.R. McCarthy, R. Weissleder, Multifunctional magnetic nanoparticles for tar-
containing glycopeptide and positron emission tomography, Cancer Res. 61 geted imaging and therapy, Adv. Drug Deliv. Rev. 60 (2008) 1241e1251.
(2001) 1781e1785. [49] S. Zitzmann, V. Ehemann, M. Schwab, Arginine-glycine-aspartic acid (RGD)-
[43] G.F. Luo, X.D. Xu, J. Zhang, J. Yang, Y.H. Gong, Q. Lei, et al., Encapsulation of an peptide binds to both tumor and tumor-endothelial cells in vivo, Cancer Res.
adamantane-doxorubicin prodrug in pH-responsive polysaccharide capsules 62 (2002) 5139e5143.
for controlled release, ACS Appl. Mater. Interfaces 4 (2012) 5317e5324. [50] T.L. Kaneshiro, Z.R. Lu, Targeted intracellular codelivery of chemotherapeutics
[44] W.H. Chen, X.D. Xu, G.F. Luo, H.Z. Jia, Q. Lei, S.X. Cheng, et al., Dual-targeting and nucleic acid with a well-defined dendrimer-based nanoglobular carrier,
pro-apoptotic peptide for programmed cancer cell death via specific mito- Biomaterials 30 (2009) 5660e5666.
chondria damage, Sci. Rep. 3 (2013) 3468. [51] W.H. Chen, Q. Lei, G.F. Luo, H.Z. Jia, S. Hong, Y.X. Liu, et al., Rational design of
[45] Y. Deng, D. Qi, C. Deng, X. Zhang, D. Zhao, Superparamagnetic high- multifunctional gold nanoparticles via hosteguest interaction for cancer-
magnetization microspheres with an Fe3O4@SiO2 core and perpendicularly targeted therapy, ACS Appl. Mater. Interfaces 7 (2015) 17171e17180.
aligned mesoporous SiO2 shell for removal of microcystins, J. Am. Chem. Soc. [52] D.R. Kalaria, G. Sharma, V. Beniwal, M.N. Ravi Kumar, Design of biodegradable
130 (2008) 28e29. nanoparticles for oral delivery of doxorubicin: in vivo pharmacokinetics and
[46] N. Graf, S.J. Lippard, Redox activation of metal-based prodrugs as a strategy for toxicity studies in rats, Pharm. Res. 26 (2009) 492e501.
drug delivery, Adv. Drug Deliv. Rev. 64 (2012) 993e1004. [53] A. Gokcimen, A. Cim, H.T. Tola, D. Bayram, A. Kocak, F. Ozgüner, et al., Pro-
[47] Y. Yuan, Y. Chen, B.Z. Tang, B. Liu, A targeted theranostic platinum(IV) prodrug tective effect of N-acetylcysteine, caffeic acid and vitamin E on doxorubicin
containing a luminogen with aggregation-induced emission (AIE) hepatotoxicity, Hum. Exp. Toxicol. 26 (2007) 519e525.

You might also like