structure-property relationships in manganese oxide ...€¦ · structure-property relationships in...

11
Structure-property relationships in manganese oxide - mesoporous silica nanoparticles used for T 1 -weighted MRI and simultaneous anti-cancer drug delivery Yu Chen a , Hangrong Chen a, * , Shengjian Zhang b , Feng Chen a , Shikuan Sun a , Qianjun He a , Ming Ma a , Xia Wang a , Huixia Wu a , Lingxia Zhang a , Linlin Zhang a , Jianlin Shi a, * a State Key of Laboratory of High Performance Ceramic and Superne Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Science, 1295 Ding-xi Road, Shanghai 200050, PR China b Department of Radiology, Cancer Hospital/Institute & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China article info Article history: Received 17 November 2011 Accepted 28 November 2011 Available online 16 December 2011 Keywords: Magnetic resonance imaging Manganese oxide Mesoporous silica Drug delivery Diagnosis abstract The extremely low longitudinal relaxivity (r 1 ) of manganese oxide has severely impeded their substi- tution for cytotoxic gadolinium-based contrast agents for safe clinical magnetic resonance imaging (MRI). Here, we report on a synthetic strategy of chemical oxidation/reduction reaction in-situ in mesopores, followed by hydrogen reduction, for the fabrication of non-toxic manganese oxide/MSNs-based MRI-T 1 contrast agents with highly comparable imaging performance to commercial Gd-based agents. This strategy involves a soft-templatingprocess to prepare mesoporous silica nanoparticles, in-situ reduc- tion of MnO 4 by the soft templatesin mesopores and heat treatment under reducing atmosphere, to disperse manganese oxide nanoparticles within mesopores. This special nanostructure combines the merits of nanopores for maximum manganese paramagnetic center accessibility for water molecules for enhanced MRI performance and encapsulation/sustained release/intracellular delivery of drugs. The synthesized manganese oxide/MSNs were successfully assessed as a high performance contrast agent for MRI-T 1 both in intro and in vivo, and meanwhile, was also demonstrated as an effective anti-cancer drug delivery (doxorubicin) vehicle, therefore, a family of manganese-based theranostics was successfully demonstrated based on the manganese oxide/MSNs composite. Ó 2011 Elsevier Ltd. All rights reserved. 1. Introduction Compared to other representative imaging modalities (e.g., X- ray computed tomography, ultrasound, positron emission tomography, single-photon-emission computed tomography), magnetic resonance imaging (MRI) combines the merits of non- invasive diagnostic mode, superb spatial/anatomical resolution and the capacity for quantitative evaluation of disease patho- genesis [1e9]. However, the limited sensitivity and high toxicity of its contrast agents (CAs) cast a shadow on its farther clinical applications [3,4]. The progress of organic/inorganic synthetic chemistry enables the developments and innovations of CAs for MRI in the past decades, in which T 1 -positive agents of para- magnetic species (producing hyperintense regions on the MR image) and T 2 -negative agents of superparamagnetic materials (producing hypointense regions on the MR image) are the two typical representatives and have been widely studied. Although superparamagnetic nanoparticles, such as iron oxide nano- crystals, give a signal-decreasing effect (T 2 -negative agents), the high susceptibility and low degradability of crystallized nanoparticles, however, still limits their extensive clinical appli- cations, in addition to the limited resolution for certain hypo- intense regions present in many lesions such as hemorrhage and blood clots [8,10,11]. Gd 3þ -based small molecule chelates are the representative and preferential CAs for T 1 -positive MRI because of their high attainable sensitivity, and are playing key roles in disease-diagnosis in current clinical applications (e.g. Magnevist Ò (r 1 z 3.4 mM 1 s 1 )) [3,4]. However, U.S. Food and Drug Administration (FDA) has warned that Gd 3þ -based CAs are associated with nephrogenic systemic brosis (NSF) in patients with impaired kidney function, hypersensitivity reactions and nephrogenic brosing dermopathy (NFD). Therefore, searching for alternatives to Gd 3þ chelates has been urgently rec- ommended and encouraged [12e14]. * Corresponding authors. Tel.: þ86 21 52412712; fax: þ86 21 52413122. E-mail addresses: [email protected] (H. Chen), [email protected] (J. Shi). Contents lists available at SciVerse ScienceDirect Biomaterials journal homepage: www.elsevier.com/locate/biomaterials 0142-9612/$ e see front matter Ó 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.biomaterials.2011.11.086 Biomaterials 33 (2012) 2388e2398

Upload: truongkhuong

Post on 19-Apr-2018

217 views

Category:

Documents


3 download

TRANSCRIPT

at SciVerse ScienceDirect

Biomaterials 33 (2012) 2388e2398

Contents lists available

Biomaterials

journal homepage: www.elsevier .com/locate/biomateria ls

Structure-property relationships in manganese oxide - mesoporous silicananoparticles used for T1-weighted MRI and simultaneous anti-cancer drugdelivery

Yu Chen a, Hangrong Chen a,*, Shengjian Zhang b, Feng Chen a, Shikuan Sun a, Qianjun He a, Ming Ma a,Xia Wang a, Huixia Wu a, Lingxia Zhang a, Linlin Zhang a, Jianlin Shi a,*a State Key of Laboratory of High Performance Ceramic and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Science, 1295 Ding-xi Road,Shanghai 200050, PR ChinabDepartment of Radiology, Cancer Hospital/Institute & Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China

a r t i c l e i n f o

Article history:Received 17 November 2011Accepted 28 November 2011Available online 16 December 2011

Keywords:Magnetic resonance imagingManganese oxideMesoporous silicaDrug deliveryDiagnosis

* Corresponding authors. Tel.: þ86 21 52412712; faE-mail addresses: [email protected] (H. Ch

(J. Shi).

0142-9612/$ e see front matter � 2011 Elsevier Ltd.doi:10.1016/j.biomaterials.2011.11.086

a b s t r a c t

The extremely low longitudinal relaxivity (r1) of manganese oxide has severely impeded their substi-tution for cytotoxic gadolinium-based contrast agents for safe clinical magnetic resonance imaging (MRI).Here, we report on a synthetic strategy of chemical oxidation/reduction reaction in-situ in mesopores,followed by hydrogen reduction, for the fabrication of non-toxic manganese oxide/MSNs-based MRI-T1contrast agents with highly comparable imaging performance to commercial Gd-based agents. Thisstrategy involves a “soft-templating” process to prepare mesoporous silica nanoparticles, in-situ reduc-tion of MnO�

4 by the “soft templates” in mesopores and heat treatment under reducing atmosphere, todisperse manganese oxide nanoparticles within mesopores. This special nanostructure combines themerits of nanopores for maximum manganese paramagnetic center accessibility for water molecules forenhanced MRI performance and encapsulation/sustained release/intracellular delivery of drugs. Thesynthesized manganese oxide/MSNs were successfully assessed as a high performance contrast agent forMRI-T1 both in intro and in vivo, and meanwhile, was also demonstrated as an effective anti-cancer drugdelivery (doxorubicin) vehicle, therefore, a family of manganese-based theranostics was successfullydemonstrated based on the manganese oxide/MSNs composite.

� 2011 Elsevier Ltd. All rights reserved.

1. Introduction

Compared to other representative imaging modalities (e.g., X-ray computed tomography, ultrasound, positron emissiontomography, single-photon-emission computed tomography),magnetic resonance imaging (MRI) combines the merits of non-invasive diagnostic mode, superb spatial/anatomical resolutionand the capacity for quantitative evaluation of disease patho-genesis [1e9]. However, the limited sensitivity and high toxicityof its contrast agents (CAs) cast a shadow on its farther clinicalapplications [3,4]. The progress of organic/inorganic syntheticchemistry enables the developments and innovations of CAs forMRI in the past decades, in which T1-positive agents of para-magnetic species (producing hyperintense regions on the MRimage) and T2-negative agents of superparamagnetic materials

x: þ86 21 52413122.en), [email protected]

All rights reserved.

(producing hypointense regions on the MR image) are the twotypical representatives and have been widely studied. Althoughsuperparamagnetic nanoparticles, such as iron oxide nano-crystals, give a signal-decreasing effect (T2-negative agents), thehigh susceptibility and low degradability of crystallizednanoparticles, however, still limits their extensive clinical appli-cations, in addition to the limited resolution for certain hypo-intense regions present in many lesions such as hemorrhage andblood clots [8,10,11].

Gd3þ-based small molecule chelates are the representative andpreferential CAs for T1-positiveMRI because of their high attainablesensitivity, and are playing key roles in disease-diagnosis in currentclinical applications (e.g. Magnevist� (r1 z 3.4 mM�1s�1)) [3,4].However, U.S. Food and Drug Administration (FDA) has warned thatGd3þ-based CAs are associated with nephrogenic systemic fibrosis(NSF) in patients with impaired kidney function, hypersensitivityreactions and nephrogenic fibrosing dermopathy (NFD). Therefore,searching for alternatives to Gd3þ chelates has been urgently rec-ommended and encouraged [12e14].

Y. Chen et al. / Biomaterials 33 (2012) 2388e2398 2389

Manganese is the essential element for physical metabolismand its homeostasis has been efficiently controlled by biologicalsystems. Importantly, Mn (II) ions could be used as the MRI CAsthanks to the five unpaired electrons with long electronic relax-ation time [15,16]. However, it is very difficult to design andsynthesize highly stable Mn (II) complexes with high sensitivitiesfor clinical applications, which could be partially solved byrecently developed nanotechnologies through buildingmanganese-based nanoparticulate systems, such as MnO, Mn3O4,Mn3O4@SiO2, MnO@mesoporous SiO2, and even hollow MnOnanoparticles [17e23]. Unfortunately, the longitudinal relaxivity(r1) of these nanoparticles were still very low, largely due to thatthe entrapped paramagnetic centers were largely shielded withinthe nanocrystal lattices, resulting in the greatly reduced chancesof interactions between manganese paramagnetic centers andwater molecules [17e23]. Previous results have demonstratedthat the accessibility of the paramagnetic centers to watermolecules plays a key role in improving the efficiency of nano-particles as T1-weighted MRI CAs [24]. It is noteworthy that thetraditional coating of a silica layer on the surface of manganeseoxide nanoparticles could cause the significant reduction of the r1values because of the shielding of the surface active manganeseions from the water molecules by the coating, which areresponsible for the shortening in the T1 relaxation times [19]. Thechallenge now lies in the enhancement of the accessibility of themanganese paramagnetic centers to water molecules, which isthe key issue to be addressed to design highly efficientmanganese-based MRI CAs.

Mesoporous nanomaterial systems have been extensivelyintroduced to nano-biomedicine for controlled drug release, cancerdiagnosis and anti-cancer chemotherapy, etc [25e30]. Previousresults have shown that watermolecules can diffuse anisotropicallyin mesoporous silica with the fastest diffusion component alongmesoporous channel [31]. Importantly, ordered mesoporousmaterials provide an ideal platform to disperse paramagneticcenters as much as possible because of their very large surfaceareas, diverse pore structures, tunable pore sizes and satisfactorybiocompatibility [24]. Considering these two factors, if themanganese paramagnetic centers were distributed evenly withina mesopore system, such a pore system could enable water mole-cules diffuse freely in mesopores with greatly increased accessi-bility to interact withmanganese paramagnetic centers, whichmayresult in the improved longitudinal relaxivity. Furthermore, thenanopores are the ideal drug reservoirs to combine the merits ofchemotherapy and disease-diagnosis in one simultaneously.

Herein, we wish to report on a synthetic strategy (oxidation/reduction reaction combined with heat treatment, O/R-HT) toprepare manganese-based theranostics with high longitudinalrelaxivity for MRI and high surface area/pore volume for chemo-therapy (e.g. doxorubicin) by dispersing manganese oxide nano-particles into mesopores. This synthetic strategy takes theadvantages of the strong oxidation and reduction potentials ofMnO�

4 ions and organic molecules, respectively, for the in-situdispersion of manganese oxide nanoparticles within mesopores.The advantages of this material system are: (1) Highly dispersedmanganese oxides nanoparticles in the penetrating mesoporesystem ensures the high water-accessibility to manganese para-magnetic centers, and (2) Large surface area and pore volume ofmesopores make take up a large amount of therapeutic agentswithin the pore system possible. Importantly, this syntheticstrategy (O/R-HT) could be extended as the general protocol tofabricate diverse manganese-based mesoporous theranostics,which has been demonstrated by introducing OR/-HT process toblock copolymer-templated SBA-15-type ordered mesoporoussilica nanoparticles (MSNs).

2. Materials and methods

2.1. Materials

Sodium hydroxide (NaOH), cetyltrimethyl ammonium bromide (C16TAB), oli-go(ethylene oxide) surfactant Pluroinc P123 (EO20PO70EO20, Mw z 5800), 3-Aminopropyltriethoxysilane (APTES) and fluorescein isothiocyanate (FITC) werepurchased from SigmaeAldrich. Potassium permanganate (KMnO4), tetraethylorthosilicate ðSiðOC2H4Þ4or TEOSÞ, hydrochloric acid (HCl, 36%e38%), ammoniumhydroxide aqueous solution (NH4OH, 25%e28%), absolute ethanol (C2H5OH) wereobtained from Sinopharm Chemical Reagent Co.. Anti-cancer agent doxorubicinhydrochloride ðDOX$HClÞ was purchased from Bejing Huafeng United TechnologyCo., Ltd. Phosphate buffer solution (PBS) was purchased from Shanghai RuichengBio-Tech Co., Ltd. All chemicals were used as received without further purification.Deionized water was used in all experiments.

2.2. Synthesis of Mn-MSNs

The synthetic procedure for Mn-MSNs involves three steps. Firstly, MSNs wereprepared by the typical sol-gel process combined with “soft-templating” method.Briefly, 0.28 g of NaOH was dissolved into 480 mL of deionized water, followed byadding 1.0 g of C16TAB. The mixture was stirred vigorously and the temperature wasraised to 80 �C. 5 mL of TEOS was added dropwise into the solution and the reactionwas continued for another 2 h to give rise to a white precipitation. The product wascollected by centrifugation and washed with plenty of deionized water and ethanolseveral times. The sample was dried under vacuum at the room temperature forfurther use. Secondly, chemical oxidation-reduction reaction was activated by thetreatment of as-prepared MSNs in KMnO4 aqueous solution. Briefly, 100 mg of as-prepared MSNs were dispersed into 10 mL of deionized water under the ultra-sonic treatment. Then 10 mL of KMnO4 aqueous solution (0.1 M) was added intoMSNs dispersions dropwise under the magnetic stirring. The mixture was thentransferred into water bath (40 �C) and the reaction was continued for another 4 hunder the magnetic stirring in the dark. The product was collected by centrifugationand washed with plenty of water under ultrasonic treatment to remove unreactedMnO�

4 ions. After the sample was dried at 80 �C, the remaining surfactant (C16TAB)was removed by calcination in air at 550 �C for 6 h. Finally, the sample was heattreated in reducing atmosphere. Briefly, the as-preparedmanganese oxide dispersedMSNs were treated in mixed H2 (5% volume percentage) and Ar (95% volumepercentage) gases at 500 �C for 4 h.

2.3. Synthesis of Mn-SBA-15

The synthetic strategy was based on the O/R-HT process. Briefly, 0.4 g ofZrOCl2$8H2O and 1.25 g of P123 were dissolved into 100 mL of HCl aqueous solution(2 M) in sequence. The mixture was stirred at 35 �C in water bath for 3 h until thesolution turns clear. 2.8 mL of TEOS was added dropwise into above clear solutionand the reaction was lasted for 24 h. The product was collected by centrifugation,washed thoroughly with water and dried under vacuum at the room temperature.The subsequent KMnO4 treatment (0.005 M KMnO4) was similar to the preparationof Mn-MSNs. After the treatment, manganese oxide loaded SBA-15 nanoparticles(Mn-SBA MSNs) were treated in mixed H2 (5% volume percentage) and Ar (95%volume percentage) gases at 500 �C for 4 h.

2.4. Synthesis of FITC conjugated Mn-MSNs and Mn-SBA-15

Briefly, 15 mg of fluorescein isothiocyanate (FITC) was reacted with 100 mL 3-aminopropyltriethoxysilane (APTES) in 5 mL of absolute ethanol under darkconditions for 24 h. Subsequently, 20 mg of Mn-MSNs (or Mn-SBA-15) were reactedwith FITC-APTES stock solution (1 mL) under dark conditions for 24 h. The FITCgrafted nanoparticles were collected by centrifugation and washed with ethanolseveral times to remove the unreacted FITC-APTES. The product was finally driedunder vacuum at room temperature in the dark.

2.5. Confocal laser scanning microscopy (CLSM) observations of the particle-endocytosis by MCF-7 cells

FITC-Mn-MSNs (or FITC-Mn-SBA-15) were suspended in culture media (RPMI1640) by 30min sonication inwater bath with the concentration of 50 mg/mL. MCF-7cells were seeded in a CLSM-special cell culture dish until the cell density reaches50e60% in 5% CO2 at 37 �C. The RPMI 1640 culture media containing FITC-Mn-MSNs(or FITC-Mn-SBA-15) was applied into the seeded cells and co-incubated for 6 hfollowed by nuclei staining using DAPI (Cell Apoptosis DAPT Detection Kit, KeyGEM).After incubation, the cells were washed three times using PBS. To quench theextracellular FITC fluorescence, trypan blue was added to the culture medium(200 mg/mL) for 10 min co-incubation. After thoroughly washing to remove trypanblue, the cells were viewed with a CLSM (FV 1000, Olympus, Japan). To determinethe intracellular location of the nanoparticles, Lyso-Tracker Red (Lyso-Tracker RedKit, Beyotime Institution of Biotechnology) was employed to stain the lysosomes ofMCF-7 cells after staining nuclei by DAPI. Three-dimensional fluorescence

Y. Chen et al. / Biomaterials 33 (2012) 2388e23982390

reconstruction of nanoparticle-endocytosized cells were conducted by serial layerscanning of cells along Z-axis and 3-D reconstruction of scanned fluorescenceimages.

2.6. CLSM observations of the intracellular release of DOX from DOX-Mn-MSNs

DOX-Mn-MSNswere suspended into RPMI 1640 cell culturemediawith the DOXconcentration of 10 mg/mL, followed by adding the nanoparticle-suspensions intoa CLSM-specific culture dish of seeded MCF-7 cells with a cell density of 60e80%.After the co-incubation for different time intervals (2 h, 4 h and 6 h), the cell waswashed with PBS for three times and stained by DAPI. The fluorescence images ofcells were obtained by a CLSM (FV 1000, Olympus, Japan). Propidium iodide (PI)staining of dead cells was performed to directly observe the therapeutic efficiency ofDOX-loaded Mn-MSNs by CLSM. 100 mL of PI working fluid was diluted into 4 mL ofRPMI 1640 cell culture media, followed by adding 100 mL diluted PI solution into36 h-incubated cells. After the co-incubation for 15 min, the cells were observed byCLSM (FV 1000, Olympus, Japan).

2.7. Bio-TEM images of nanoparticle-endocytosized cells

The Mn-MSNs (or Mn-SBA-15) were suspended into RPMI 1640 cell culture(100 mg/mL) by sonication treatment for 30 min, followed by introducingnanoparticle-suspensions into seeded MCF-7 cells. After incubation for 24 h, thecells were washed by PBS for three times and detached by incubation with 0.25%trypsin for 5 min. The cells were collected by centrifugation at 5000 r/min for 2 min.Then, the cells were fixed by glutaraldehyde at room temperature after the removalof incubation medium. The fixed cells were rinsed with propylene oxide, embeddedin EPOMB12 and polymerized in the oven subsequently at 37 �C for 12 h, 45 �C for12 h and 60 �C for 48 h. Ultrathin sections of approximately 70 nm thick were cutwith a diamond knife on a Leica UC6 ultramicrotome and transferred to the coppergrid. The images were viewed on JEM-1230 electron microscopy.

2.8. In vitro and in vivo MRI assay

The in vitro and in vivo MR imaging experiments were performed on a 3.0 Tclinical MRI instrument (GE Signa 3.0 T). For in vitro MRI test, the manganeseconcentrations of Mn-MSNs dispersed in water were determined by inductivelycoupled plasma atomic emission spectrometry (ICP-AES). T1-weighted Fast-recoveryspin-echo (FR-FSE) sequence is described as follows: TR ¼ 1000, 2000, 3000 and4000, Slice ¼ 3 mm, Space ¼ 0.5 mm, Fov ¼ 20, Phase fov ¼ 0.8,Freq � Phase ¼ 384 � 256, Nex ¼ 2, ETL ¼ 2. For in vivo MRI assay, walk 256 cellswere implanted subcutaneously into SD female mouse (5�106 cell/site) to establisha tumor model. MRI assay were performed when the tumor size reached about2.5 cm � 3 cm � 3 cm (body weightz 210 g). For Mn-MSNs, in vivoMRI was carriedout at selected time intervals before and after intravenous injection of Mn-MSNs PBSsuspensions (dose: 2.8 mg Mn per kg of mouse body); while for Mn-SBA-15, in vivoMRI was conducted via intratumoral injection of Mn-SBA-15 suspensions (dose:0.27 mg Mn per kg of mouse body). Animal procedures were in agreement with theguidelines of the Instituonal Animal Care and Use Committee.

2.9. DOX loading and in vitro release of DOX from DOX-loaded Mn-MSNs underdifferent pH values (pH ¼ 7.4, 6.0 and 5.0)

5mg of Mn-MSNs was dispersed into DOX PBS solution (6 mL, 0.5 mg/mL). Afterstirring for 24 h under dark conditions, the DOX-loaded Mn-MSNs were collected bycentrifugation. To evaluate the DOX loading content, the supernatant DOX solutionwas collected and the residual DOX was measured by UVevis spectra at a wavenumber of 480 nm. For in vitro DOX release assess, 2 mg of DOX-Mn-MSNs wereencapsulated into a dialysis bag and put into 15 mL PBS solutions under different pHvalues (pH ¼ 7.4, 6.0 and 5.0). Then, the releasing process was performed ina shaking table with a shaking speed of 142 rpm at 37 �C, and monitored by UVevisadsorption spectra at a time-course.

2.10. In vitro cytotoxicity

The in vitro cytotoxicity was assessed by the typical 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) reduction assay. To evaluate the cyto-toxicity of DOX-loaded Mn-MSNs against cancer cells, MCF-7 cells were seeded ina 96-well plate at a density of 2 � 103 per well and cultured in 5% CO2 at 37 �C for24 h. Free DOX and DOX-loaded Mn-MSNs were dispersed into the culture media(RPMI 1640) with the equivalent DOX concentration, and the cells were incubated in5% CO2 at 37 �C for 36 h. The concentrations of DOX were 0, 0.16, 0.31, 0.63, 1.25, 2.5,5, 10 and 20 mg/mL, respectively. At the end of the incubation, the culture mediawere removed and 100 mL of MTT solutions (dissolved in RPMI 1640 with a finialconcentration of 0.8 mg/mL) were added. Themediawere then replaced with 100 mLof dimethyl sulfoxide (DMSO) per well, and the absorbance was monitored bya microplate reader (Bio-TekELx800) at the wavelength of 490 nm. The cytotoxicitywas expressed as the percentage of cell viability compared to untreated control cells.The in vitro cytotoxicity of blank Mn-MSNs nanoparticles were also assessed by

similar MTT protocol except for the different initial cell density (1 �104) in 96-wellplate. The concentrations of Mn-MSNs were 0, 6.25, 12.5, 25, 50, 100, 200, 300 and400 mg/mL, respectively.

2.11. In vivo drug distribution experiment

Balb/c nude mice were inoculated with 100 mL of phosphate buffered saline(pH ¼ 7.4) containing 1 �107 MCF-7/ADR cells by subcutaneous injection in the leftflank. Experiments were performed at day 10 after inoculation when the tumorvolume reached approximately 100 mm3. Tumor-bearing mice were randomlyassigned to the following three treatment groups (n ¼ 5): saline control group, freeDOX group (5 mg/kg) or DOX-loaded Mn-MSNs group (5 mg/kg DOX). In eachtreatment group, mice were sacrificed at 24 h after intravenous drug administration(n ¼ 5). Heart, liver, spleen, lung, kidney and tumor were collected and weighed.Following this, one part of the tissues were frozen in GSV 1 embedding medium(SLEE, Germany) at �20 �C. Frozen sections of 20 mm thickness were prepared witha cryotome cryostat (SLEE, MEV, Germany) and stained with 300 nMDAPI (Sigma) atroom temperature. Subsequently, the sections were observed under a confocalmicroscope (FV 1000, Olympus, Japan). The other part of tissues were homogenizedwith lysis buffer. The tissue samples were collected into bottles and centrifuged at15000 rpm for 10min at 4 �C. The amount of DOX in supernatant of each sample wasdetermined using a microplate reader (Infinite F200, TECAN, Austria). The data werenormalized to the tissue weight.

2.12. Characterization

Transmission electron microscopy (TEM)/Scanning transmission electronmicroscopy (STEM) images were aquired on a JEM-2100F electron microscopeoperated at 200 kV. Scanning electron microscopy (SEM) images were obtained ona field emission HITACHI S-4800 microscope. UVevis spectra were recorded ona UV-3101PC Shimadzu UVevis spectroscope. X-ray diffraction (XRD) pattern wascollected using a Rigaku D/Max-2200 PC X-ray diffractometer with Cu target (40 kV,40 mA). Nitrogen adsorption-desorption isotherms at 77 K were measured ona Micrometitics Tristar 3000 system. Electron spin resonance (ESR) spectra wasrecorded on a EMX-8/2.7 spectrometer. Inductively coupled plasma atomic emissionspectrometry (ICP-AES) test was conducted on VISTA AX (Varian company,American).

3. Results and discussion

3.1. Design, synthesis and structural characterization of Mn-MSNs

As shown in Fig. 1, the overall nanostructural fabrication processinvolves the synthesis of MSNs templated by “soft templates” suchas surfactants or block copolymers, oxidation/reduction (O/R)reaction between organic molecules within MSNs and MnO4

� inaqueous solution to in situ form manganese oxide nanoparticleswithin mesopores and meanwhile the removal of the remainingsurfactants, and the heat treatment of nanoparticles underreducing atmosphere. We first chose MCM-41-type ordered MSNs,typically templated by cationic surfactant-cetyltrimethyl ammo-nium bromide (C16TAB), to demonstrate our hypothesis becausetheir pore and particulate sizes can be facilely controlled and tunedfrom 2 to 10 nm and 60e1100 nm, respectively [24]. Most impor-tantly, the biocompatibility of MSNs has been well-demonstratedand their applications in nano-biomedicine have been widelyexplored [32,33]. The synthetic procedure for MSNs was based onthe NaOH-catalyzed hydrolysis/condensation of tetraethyl ortho-silicate (TEOS) in the presence of C16TAB [24]. Then, the surfactant-containing MSNs were dispersed into KMnO4 aqueous solutions(0.05 M) and stirred for 4 h at 40 �C in the dark (See experimentalsection). After the calcination (550 �C, 6 h) to remove the remainingsurfactants (C16TAB), the manganese oxide loaded MSNs (desig-nated asMn-MSNs) were treated in H2/Ar atmosphere (500 �C, 4 h).

The design and synthetic target for this nanoparticulate systemis to disperse manganese paramagnetic centers within mesoporesas thoroughly as possible, making the water-accessibility ofmanganese centers as high as possible. It is known that the typicalsynthetic process for mesoporous materials employs “softtemplates” such as surfactants as the pore-making/directingagents, which are in the form of micelles and evenly distributed

Fig. 1. Schematic representation for the preparation of manganese oxide dispersed MSNs (Mn-MSNs) by the O/R-HT strategy. The “soft templates”, typically employed forsynthesizing mesoporous materials and evenly be restricted within mesopores, could react in-situ with potassium permanganate (KMnO4) by chemical oxidation-reduction (O/R),by which manganese oxide nanoparticles are formed and retained within nanopores (Step A). The relaxivity of manganese oxide within mesopores could be enhanced by heattreatment under reducing atmosphere (Step B). Finally, water molecules could diffuse freely into mesopores, and interact with manganese paramagnetic centers.

Fig. 2. Morphological, structural and in vitroMRI characterizations of Mn-MSNs after H2 reduction. TEM (a) and STEM (a1) images of Mn-MSNs, EDS elemental mappings of Mn (a2),Si (a3) and O (a4) elements on a Mn-MSNs. Plots of b) T�1

1 and c) T�12 versus Mn concentrations for Mn-MSNs before and after H2 reduction (inset: T1- and T2-weighted MRI results

obtained from aqueous suspensions of Mn-MSNs before and after H2 reduction).

Y. Chen et al. / Biomaterials 33 (2012) 2388e2398 2391

Y. Chen et al. / Biomaterials 33 (2012) 2388e23982392

within mesopores before they are removed to create the pores[34,35]. We used potassium permanganate (KMnO4), a cheap andenvironmental-friendly compound with very high oxidationpotential, to oxidize the surfactants in themesopores by a simple O/R reaction under a mild condition, by which manganese speciescould be generated in-situ within mesopores with high dispersity.Importantly, the finial valence of manganese element could befacilely tuned by optimizing synthetic parameters such as H2thermal treatment [36,37].

Fig. 2 shows the morphology, structural and chemical char-acteristics of Mn-MSNs after H2 reduction. The Mn-MSNs exhibitsspherical morphology, high dispersity and two-dimensionalhexagonally ordered pore array (Fig. 2a and Figs. S1, S2 and S4).To further investigate the distribution of manganese

Fig. 3. FITC-Mn-MSNs are internalized by MCF-7 cells. The MCF-7 cells were incubated withwere stained by DAPI (blue), and the extracellular fluorescence was quenched by trypan blCLSM (a1ea3); Three-dimensional fluorescence reconstructions of nanoparticle-endocytosedcells; Three-dimensional fluorescence reconstructions of nanoparticle-endocytosized MCF-cellular location of Mn-MSNs nanoparticles (nuclei (c1): blue fluorescence of DAPI stainingfluorescence of FITC grafting). The yellow fluorescence signal in overlay image (c4), which centered lysosomes while the rests are in the cytoplasm; selected bio-TEM images of MCF-7uptaken by MCF-7 cells by endocytosed process, (d2) Mn-MSNs trapping in vesicles insidpretation of the references to colour in this figure legend, the reader is referred to the web

paramagnetic center within mesopores, EDS elemental mappingof Mn, Si and O elements in Mn-MSNs was conducted(Fig. 2a1ea4 and Fig. S3). The result shows that the manganeseelement is homogeneously distributed in the whole mesoporoussilica matrix without the accumulation at the particle surface,demonstrating the pore-confined formation and deposition ofmanganese oxide nanoparticles within mesopores. Inductivelycoupled plasma atomic emission spectrometry (ICP-AES) resultsshow that the manganese element content in Mn-MSNs is 5.94%(w/w), which could be facilely tuned by varying the initialsynthetic parameters such as MnO4

� concentrations, reaction timeand temperature. The Mn-MSNs nanoparticles exhibit small-angle X-ray diffraction (SAXRD) peaks at 2.5, 4.3 and 5.0� thatare characteristic of (100), (110) and (200) planes of the typical

the nanoparticles (FITC-Mn-MSNs, 50 mg/mL) for 6 h. After incubation, the cell nucleusue and the endocytosed particles with FITC-label (green) inside cells were detected byMCF-7 cells (b1eb3) to demonstrate the internalization of nanoparticles within cancer

7 cells after nucleus and lysosomes double-staining (c1ec4), to demonstrate the intra-; lysosomes (c2): red fluorescence of Lyso-tracker red staining; Mn-MSNs (c3): greenomes from the superposition of the red and green, suggests that some Mn-MSNs havecells after co-incubation with Mn-MSNs nanoparticles for 24 h showing (d1) Mn-MSNse the cell and (d3) endosomal escape of nanoparticles into the cytoplasm. (For inter-version of this article.)

Y. Chen et al. / Biomaterials 33 (2012) 2388e2398 2393

MCM-41-type mesoporous materials, respectively (Fig. S5a). TheSAXRD results indicate that Mn-MSNs possess ordered meso-porous arrays. The wide-angle XRD patterns (WAXRD, Fig. S5b)exhibit no apparent diffraction peaks for crystallized manganeseoxide nanoparticles, demonstrating the absence of largemanganese oxide particles formed on the material matrix duringthe synthetic process. Nitrogen adsorption-desorption techniquewas employed to characterize the pore structures of Mn-MSNsafter the H2 reduction, which shows that the isotherms of Mn-MSNs exhibit the typical Langmuir IV type hysteresis loop, indi-cating the presence of well-defined mesopores (Fig. S6a). Inaddition, Mn-MSNs are highly porous with a surface area of665 m2/g, pore volume of 0.99 cm3/g and pore size of 2.4 nm(Fig. S6b), indicating that the mesopore system of MSNs remainopen and penetrable after the deposition of manganese oxidespecies.

Fig. 4. In vivo MR imaging capability of Mn-MSNs after H2 reduction. A time-course (0, 5, 30and kidney (d1ed4) of a tumor-bearing mouse after intravenous injection of Mn-MSNs; b1e

3.2. Mn-MSNs as the highly efficient theranostics for T1-weightedMR imaging and simultaneous anti-cancer drug delivery

To assess the effectiveness of Mn-MSNs as MRI CAs, themagnetic resonance relaxivity of the nanoparticles was measuredusing a clinical 3.0 T human clinical scanner. The in vitro longitu-dinal relaxation rate (1/T1) and transverse relaxation rate (1/T2) asa function of the manganese ion concentrations of Mn-MSNs beforeand after H2 reduction were evaluated. As shown in Fig. 2b and c,Mn-MSNs have an r1 value of 0.45 mM�1s�1 and r2 value of9.1 mM�1s�1 before the heat treatment. However, the specificrelaxivity of Mn-MSNs was found to significantly increase after H2reduction. The r1 and r2 values of Mn-MSNs after H2 reductionreached 2.28 and 15.9 mM�1s�1, 5.1 and 1.7 times higher than theiroriginal values before H2 reduction, respectively. It can be inferredthat the increased relaxivities of the Mn-MSNs after H2 reduction

, 60 min) of the signal enhancement in T1-weighted MRI of tumor (a1ea4), liver (c1ec4)b4: magnified images of corresponding circled area in a1ea4.

Y. Chen et al. / Biomaterials 33 (2012) 2388e23982394

are mainly due to the lowed valence of manganese ions underreducing atmosphere (e.g. Mn4þ-Mn2þ transformation) [38], liketransforming Fe2O3 into Fe3O4 under H2/Ar atmosphere [27,28],which was further demonstrated by the higher hyperfine splitting(A) value after H2 reduction as indicated in electron spin resonance(ESR) spectra of Mn-MSNs (Fig. S7) [39]. Importantly, this r1 value(2.28 mM�1s�1) is 6.2, 12.7, 17.5 and 19.0 times higher than those of7 nm (0.37 mM�1s�1), 15 nm (0.18 mM�1s�1), 20 nm(0.13 mM�1s�1) and 25 nm (0.12 mM�1s�1) monodispersed MnOnanoparticles, respectively [19]. It could also be directly observedthat the Mn-MSNs nanoparticles decreased both the longitudinalrelaxation time (T1) and the transverse relaxation time (T2) as canbe known from the changes of signal intensity with the increasingmanganese concentrations (image insets of Fig. 2b and c),demonstrating that the prepared Mn-MSNs could functionalize asboth the T1- and T2-weighted MR imaging CAs. The mesoporoussilica channel is responsible for high dispersity of manganese oxidenanoparticles and fast rates in the diffusion of water moleculesinside the mesopores, which plays a significant role in accountingfor the achieved high relaxivity of such a manganese-based Mn-MSNs mesoporous system.

It is known that excellent cell uptake of the CAs guarantees theefficient cell imaging by MRI for diagnosis and intracellular drugdelivery for chemotherapy. The Mn-MSNs nanoparticles werecovalently conjugated with the typical organic fluorescent dye-fluorescein isothiocyanate (FITC-Mn-MSNs) using simple silaneconjugation chemistry, to make them visible by confocal laserscanningmicroscopy (CLSM) for the observations of interaction andintracellular location of nanoparticles. Breast cancer MCF-7 cellswere chosen as the modal cells, which were incubated with fluo-rescent nanoparticles for 6 h at 37 �C at a dose of 50 mg/mL. In orderto distinguish the nanoparticles only attached to the membrane

Fig. 5. (a) The release profiles of DOX from DOX-loaded Mn-MSNs at different pH values (pH(b) CLSM images of MCF-7 cells incubated with DOX-loaded Mn-MSNs (DOX concentration:b31): nuclei stained with DAPI; red fluorescence (b12, c22 and b32): DOX molecules; (b13, b23intensity of paned area in b11, b21 and b31, respectively). (c) Viabilities of MCF-7 cells wheassessed by MTT protocol. (For interpretation of the references to colour in this figure lege

from those internalized by the cells, the extracellular FITC wasquenched by trypan blue [40]. As shown in Fig. 3a1ea3, the greenfluorescent punctuated spots lighting in the cytoplasm of the cellscould be clearly observed, verifying the uptake of the nanoparticlesby cancer cells. Three-dimensional confocal fluorescent imagevolumes of MCF-7 cells further demonstrate that the fluorescentsignal is originated from the cytoplasm, rather than from theaggregates on the cell surface (Fig. 3b1eb3). To verify the intracel-lular location of Mn-MSNs nanoparticles, the lysosomes werelabelled with red fluorescent lysotracker (Fig. 3c1ec4). After 6 hincubation, the green fluorescence of FITC-Mn-MSNs was co-localized with red lysotracker (yellow signal in Fig. 3c4), thusindicating that Mn-MSNs nanoparticles could enter lysosomes ofMCF-7 cells while the rest nanoparticles are within the cytoplasm.However, the nanoparticles could not pass through the nuclearmembrane as demonstrated by the absence of fluorescent signals ofnanoparticles in nucleus. The distribution and possible uptakeprocess of nanoparticles by cancer cells were further observed bybio-TEM after the ultrathin section of the cells. A large number ofnanoparticles could be found in the cytoplasm, which maintainstheir spherical morphology, indicating that the prepared nano-particles could be efficiently uptaken by cancer cells (Fig. 3d2ed3),in consistence with CLSM results. The endocytosis process, a typicalnon-specific procedure for the uptake of nanoparticles by cancercells [41,42], could be observed from Fig. 3d1. Mn-MSNs wereenclosed by the cell membrane to form vesicles, then internalizedby cells (Fig. 3d2). The Mn-MSNs were further processed in endo-somes and lysosomes and eventually released into the cytoplasm(Fig. 3d3).

The effectiveness of Mn-MSNs after H2 reduction as the MRI CAsin vivo was assessed employing a 3.0 T scanner. Fig. 4 shows theMn-MSNs contrast-enhanced T1-weighted MRI of a tumor-bearing

¼ 7.4, 6.0 and 5.0; inset: digital pictures of the releasing media in 600 min of release).10 mg/mL) for different time intervals (2 h, 4 h and 6 h; blue fluorescence (b11, b21 andand b33): merged images of blue and red fluorescence; (b14, b24 and b34): fluorescencen incubated with free DOX and DOX-loaded Mn-MSNs at different concentrations asnd, the reader is referred to the web version of this article.)

Y. Chen et al. / Biomaterials 33 (2012) 2388e2398 2395

mouse. A significant T1-weighted signal enhancement is clearlyvisible in the tumor site after 5 min tail vein injection (Fig. 4a1ea4and b1eb4). The signal enhancement implies that Mn-MSNs couldreach the tumor site, typically by the enhanced permeability andretention (EPR) effect [43], demonstrating the unique potential ofthe prepared nanoparticles for use as the T1-weighted MRI CAs forcancer diagnosis. In addition, the live signal enhancement isprobably due to the phagocytosis of the Mn-MSNs nanoparticles bythe reticuloendothelial systems (RES) (Fig. 4c1ec4). This is benefi-cial for the MR imaging of liver abnormalities such as liver cancer. Atime-course of signal enhancement was clearly observed in T1-weighted imaging of kidney, demonstrating the excretion potentialof nanoparticles by kidney system (Fig. 4d1ed4).

To evaluate the potential of Mn-MSNs as the drug deliverysystem for cancer chemotherapy, a typical anti-cancer chemo-therapeutic agent doxorubicin (DOX) was chosen as themodel drugto investigate the drug loading and intracellular release behavior.The drug loading amount on the support is 382 mg/g, as deter-mined by UVevis characterization. The in vitro drug release prop-erties achieved at different pH values (pH ¼ 7.4,6.0 and 5.0)demonstrated the sustained and pH-responsive DOX release profilefrom DOX-loaded Mn-MSNs (Fig. 5a), which is determined by theelectrostatic interaction between positively charged DOXmoleculesand negatively charged mesopores. This pH-responsive character isvery important because pH values in different tissues and cellular

Fig. 6. (aec) CLSM images of tumor tissue after 24 h intravenous administration of DOX-Marea in a, and Fig. d is the magnified image of corresponding pane area in b); (d) Biodistribuvia intravenous injection (inset: magnified Figure of the DOX concentrations in tumor).

compartments vary significantly. For instance, the tumor extracel-lular environment is significantly more acidic (pH z 6.5) thanblood (pH z 7.4), and the pH values of endosomes and lysosomesare even lower (ca. 5.0e5.5) than cytosolic pH values [44e46].CLSM was used to study the internalization of the DOX-Mn-MSNsinto cancer cells and intracellular release of DOX (Fig. 5b11eb13,b21eb23 and b31eb33). A time-course (2 h, 4 h and 6 h) fluorescentintensity enhancement of DOX in cancer cells indicates the sus-tained and intracellular release of drugs from DOX-loaded Mn-MSNs (Fig. 5b41eb43). The delivery of DOX into the cancer cells ledto growth inhibition and cell death. Mn-MSNs alone show very lowcytotoxic effect (IC50 > 400 mg/mL, Fig. S8), while DOX-loaded Mn-MSNs exhibit a dose-dependent cytotoxic effect, which was slightlyhigher than that observed with equivalent doses of free DOX in therange of high DOX concentration (Fig. 5c). The quantitative growthinhibition of MCF-7 cells was obtained when the cells were treatedwith either free DOX or DOX-Mn-MSNs, demonstrating that Mn-MSNs can be used as a drug delivery vehicle (Fig. S9). Consistentwith the in vitro cytotoxicity and CLSM results, the entrapped DOXfromMn-MSNs seems to exhibit the controlled or sustained releasebehavior in vivo from Mn-MSNs.

The in vivo therapeutic potential was assessed by intravenousinjection of free DOX and DOX-loaded Mn-MSNs (DOX-Mn-MSNs)into tumor-bearing mice at a DOX dose of 5 mg/kg. Mice werescarified 24 h after the injection of drugs, and the major organs

n-MSNs into mice with different magnifications (Fig. b is the magnified image of panetions of DOX in major organs of mice after 24 injection of free DOX and DOX-Mn-MSNs

Fig. 7. T�11 (a) and T�1

2 (b) versus Mn concentrations for Mn-SBA-15 before and after H2 reduction (inset: T1- and T2-weighted MRI results obtained from aqueous suspensions of Mn-SBA-15 before and after H2 reduction); T1- (c, d): and T2- (e, f) weighted MR images of a mouse tumor site before (c, e) and after (d, f) intratumoral injections for 60 min with Mn-SBA-15 (after H2 reduction).

Y. Chen et al. / Biomaterials 33 (2012) 2388e23982396

were harvested and homogenized for DOX extraction. Theconcentration of DOX in each organ was measured by fluorescencespectroscopy. In addition, the presence of red fluorescence fromDOX in tumor tissuewas directly observed by CLSM (Fig. 6aec). Theamount of DOX accumulated in tumor site for DOX-Mn-MSNs wassignificantly higher than that for free DOX, which was likely due tothe accumulation of DOX-Mn-MSNs by the EPR effect (Fig. 6d andinset) [43,47]. The results indicated that Mn-MSNs could inducehigher accumulation of DOX in tumor as the drug delivery systemwith passive tumor targeting property in vivo. However, DOXaccumulation in the RES including liver and spleen is relativelyhigh, implying further surface modification of the nanoparticlessuch as PEGylation is urgently needed for avoiding the capture ofnanoparticles by RES to improve the blood circulation time [48].

3.3. Extending the O/R-HT synthetic strategy for SBA-15-type Mn-MSNs

It is known that a majority of mesoporous materials with diversemorphologies and structures can be synthesized by “soft-templat-ing” strategy using various organic molecules as templates. whichare believed to react with MnO4

� ions similarly with C16TAB togenerate a family of manganese-based ordered mesoporous thera-nostics. To demonstrate the general applicability of the developed O/R-HT process for manganese-based mesoporous theranostics, wechose another typical mesoporous silica system SBA-15, which wasobtained using the oligo(ethylene oxide) surfactant Pluroinc P123

(EO20PO70EO20, Mw z 5800) as the pore-making agents [49]. Toreduce the size of SBA-15-typeMSNs for nanomedical requirements,SBA-15 MSNs were synthesized by our recently developedcomposite liquid crystal templating method for the bottom-uptailoring of nonionic surfactant-templated mesoporous silica nano-materials by ZrIV [50]. Manganese oxide nanoparticles were intro-duced into mesopores of SBA-15 (Mn-SBA-15) employing the O/R-HT process by the oxidation of P123 using MnO4

� ions (detailedstructure characterization, see Fig. S10eS15). The in vitroMRI resultsshow that Mn-SBA-15 MSNs have r1 and r2 values of 1.37 and20.2 mM�1s�1, respectively, before H2 reduction, while these valuesof Mn-SBA-15 after heat treatment are both 2.3 times higher (Fig. 7aand b). In addition, Mn-SBA-15 MSNs also show excellent uptake bycancer cells (CLSM and bio-TEM results, see Fig. S16). The in vivo T1-and T2-weighted MRI images of the mouse tumor revealed brightsignal enhancement and a dark signal void, respectively (Fig. 7cef),in the region where the Mn-SBA-15 MSNs were intratumoral injec-ted. The results verify that Mn-SBA-15 MSNs are uniquely potentialas the dual CAs for both T1- and T2-weighted MR imaging.

4. Conclusions

In summary, we have developed a general synthetic strategy (O/R-HT) to obtain a highly efficient, cytotoxicity-free manganese-based MRI-T1 contrast agent by dispersing manganese oxidespecies within mesopores of mesoporous silica nanoparticles (Mn-MSNs). This process involves the preparation of MSNs and the

Y. Chen et al. / Biomaterials 33 (2012) 2388e2398 2397

reduction of MnO4� in-situ by “soft templates” within mesopores

afterwards for the free water molecule access and interaction withthe highly dispersed manganese oxide species, leading to signifi-cantly increased longitudinal relaxivity (r1 values), highly compa-rable to commercial Gd-based CAs, for MRI, which wasdemonstrated both in vitro and in vivo. Furthermore, the largesurface area and high pore volume of mesopores enable theprepared Mn-MSNs a sustained, pH-responsive and intracellularrelease of anti-cancer agents (doxorubicin) for chemotherapy. Asmost mesoporous materials are fabricated by using organic softtemplates, we can expect that the present O/R-HT strategy can beused to produce various kinds of manganese-based ordered mes-oporous theranostics.

Acknowledgment

We greatly acknowledge financial supports from the NationalBasic Research Program of China (973 Program, Grant No.2011CB707905), Shanghai Rising-Star Program (Grant No.10QH1402800), National Nature Science Foundation of China(Grant No. 51132009, 50823007, 51072212, 51102259), the Scienceand Technology Commission of Shanghai (Grant No.11nm0506500,10430712800), the Science Foundation for Youth Scholar of StateKey Laboratory of High Performance Ceramics and SuperfineMicrostructures (Grant No. SKL201001).

Appendix. Supplementary data

Supplementary data related to this article can be found online atdoi:10.1016/j.biomaterials.2011.11.086.

References

[1] Logothetis NK. What we can do and what we cannot do with fMRI. Nature2008;453:869e78.

[2] Noseworthy JH. Progress in determining the causes and treatment of multiplesclerosis. Nature 1999;399:A40e7.

[3] Terreno E, Castelli DD, Viale A, Aime S. Challenges for molecular magneticresonance imaging. Chem Rev 2010;110:3019e42.

[4] Viswanathan S, Kovacs Z, Green KN, Ratnakar SJ, Sherry AD. Alternatives togadolinium-based metal chelates for magnetic resonance imaging. Chem Rev2010;110:2960e3018.

[5] Mitchell TM, Shinkareva SV, Carlson A, Chang KM, Malave VL, Mason RA, et al.Predicting human brain activity associated with the meanings of nouns.Science 2008;320:1191e5.

[6] Werner EJ, Datta A, Jocher CJ, Raymond KN. High-relaxivity MRI contrastagents: where coordination chemistry meets medical imaging. Angew ChemInt Ed 2008;47:8568e80.

[7] Jun YW, Lee JH, Cheon J. Chemical design of nanoparticle probes for high-performance magnetic resonance imaging. Angew Chem Int Ed 2008;47:5122e35.

[8] Piao Y, Kim J, Bin Na H, Kim D, Baek JS, Ko MK, et al. Wrap-bake-peel processfor nanostructural transformation from beta-FeOOH nanorods to biocompat-ible iron oxide nanocapsules. Nat Mater 2008;7:242e7.

[9] Lee JH, Huh YM, Jun Y, Seo J, Jang J, Song HT, et al. Artificially engineeredmagnetic nanoparticles for ultra-sensitive molecular imaging. Nat Med 2007;13:95e9.

[10] Yu MK, Jeong YY, Park J, Park S, Kim JW, Min JJ, et al. Drug-loaded super-paramagnetic iron oxide nanoparticles for combined cancer imaging andtherapy in vivo. Angew Chem Int Ed 2008;47:5362e5.

[11] Berret JF, Schonbeck N, Gazeau F, El Kharrat D, Sandre O, Vacher A, et al.Controlled clustering of superparamagnetic nanoparticles using block copol-ymers: design of new contrast agents for magnetic resonance imaging. J AmChem Soc 2006;128:1755e61.

[12] Penfield JG, Reilly RF. What nephrologists need to know about gadolinium.Nat Clin Pract Nephrol 2007;3:654e68.

[13] Tromsdorf UI, Bruns OT, Salmen SC, Beisiegel U, Weller H. A highly effective,nontoxic T-1 MR contrast agent based on ultrasmall PEGylated iron oxidenanoparticles. Nano Lett 2009;9:4434e40.

[14] Perez-Rodriguez J, Lai S, Ehst BD, Fine DM, Bluemke DA. Nephrogenic systemicfibrosis: incidence, associations, and effect of risk factor assessment-report of33 cases. Radiology 2009;250:371e7.

[15] Shin JM, Anisur RM, Ko MK, Im GH, Lee JH, Lee IS. Hollow manganese oxidenanoparticles as multifunctional agents for magnetic resonance imaging anddrug delivery. Angew Chem Int Ed 2009;48:321e4.

[16] Huang CC, Khu NH, Yeh CS. The characteristics of sub 10 nm manganese oxideT-1 contrast agents of different nanostructured morphologies. Biomaterials2010;31:4073e8.

[17] Schladt TD, Shukoor MI, Schneider K, Tahir MN, Natalio F, Ament I, et al.Au@MnO nanoflowers: hybrid nanocomposites for selective dual functional-ization and imaging. Angew Chem Int Ed 2010;49:3976e80.

[18] Schladt TD, Schneider K, Shukoor MI, Natalio F, Bauer H, Tahir MN, et al.Highly soluble multifunctional MnO nanoparticles for simultaneous opticaland MRI imaging and cancer treatment using photodynamic therapy. J MaterChem 2010;20:8297e304.

[19] Na HB, Lee JH, An KJ, Park YI, Park M, Lee IS, et al. Development of a T-1contrast agent for magnetic resonance imaging using MnO nanoparticles.Angew Chem Int Ed 2007;46:5397e401.

[20] Yang H, Zhuang YM, Hu H, Du XX, Zhang CX, Shi XY, et al. Silica-coatedmanganese oxide nanoparticles as a platform for targeted magnetic reso-nance and fluorescence imaging of cancer cells. Adv Funct Mater 2010;20:1733e41.

[21] Kim T, Momin E, Choi J, Yuan K, Zaidi H, Kim J, et al. Mesoporous silica-coatedhollow manganese oxide nanoparticles as positive T(1) contrast agents forlabeling and MRI tracking of adipose-derived mesenchyrnal stem cells. J AmChem Soc 2011;133:2955e61.

[22] Peng YK, Lai CW, Liu CL, Chen HC, Hsiao YH, Liu WL, et al. A new and facilemethod to prepare uniform hollow MnO/functionalized mSiO(2) core/shellnanocomposites. ACS Nano 2011;5:4177e87.

[23] Bae KH, Lee K, Kim C, Park TG. Surface functionalized hollow manganese oxidenanoparticles for cancer targeted siRNA delivery and magnetic resonanceimaging. Biomaterials 2010;32:176e84.

[24] Taylor KML, Kim JS, Rieter WJ, An H, Lin WL, Lin WB. Mesoporous silicananospheres as highly efficient MRI contrast agents. J Am Chem Soc 2008;130:2154e5.

[25] Zhu YF, Shi JL, Shen WH, Dong XP, Feng JW, Ruan ML, et al. Stimuli-responsivecontrolled drug release from a hollow mesoporous silica sphere/polyelectrolytemultilayer core-shell structure. Angew Chem Int Ed 2005;44:5083e7.

[26] Kim J, Kim HS, Lee N, Kim T, Kim H, Yu T, et al. Multifunctional uniformnanoparticles composed of a magnetite nanocrystal core and a mesoporoussilica shell for magnetic resonance and fluorescence imaging and for drugdelivery. Angew Chem Int Ed 2008;47:8438e41.

[27] Chen Y, Chen HR, Guo LM, He QJ, Chen F, Zhou J, et al. Hollow/rattle-typemesoporous nanostructures by a structural difference-based selective etchingstrategy. ACS Nano 2010;4:529e39.

[28] Chen Y, Chen HR, Zeng DP, Tian YB, Chen F, Feng JW, et al. Core/shellstructured hollow mesoporous nanocapsules: a potential platform forsimultaneous cell imaging and anticancer drug delivery. ACS Nano 2010;4:6001e13.

[29] Liong M, France B, Bradley KA, Zink JI. Antimicrobial activity of silver nano-crystals encapsulated in mesoporous silica nanoparticles. Adv Mater 2009;21:1684e9.

[30] Chen Y, Chen HR, Zhang SJ, Chen F, Zhang LX, Zhang JM, et al. Multifunc-tional mesoporous nanoellipsoids for biological bimodal imaging andmagnetically targeted delivery of anticancer drugs. Adv Funct Mater 2011;21:270e8.

[31] Stallmach F, Karger J, Krause C, Jeschke M, Oberhagemann U. Evidence ofanisotropic self-diffusion of guest molecules in nanoporous materials ofMCM-41 type. J Am Chem Soc 2000;122:9237e42.

[32] He QJ, Zhang ZW, Gao Y, Shi JL, Li YP. Intracellular localization and cytotoxicityof spherical mesoporous silica nano- and microparticles. Small 2009;5:2722e9.

[33] Lu J, Liong M, Li ZX, Zink JI, Tamanoi F. Biocompatibility, biodistribution, anddrug-delivery efficiency of mesoporous silica nanoparticles for cancer therapyin animals. Small 2010;6:1794e805.

[34] Kresge CT, Leonowicz ME, Roth WJ, Vartuli JC, Beck JS. Ordered mesoporousmolecular-sieves synthesized by a liquid-crystal template mechanism. Nature1992;359:710e2.

[35] Wan Y, Zhao DY. On the controllable soft-templating approach to mesoporoussilicates. Chem Rev 2007;107:2821e60.

[36] Dong XP, Shen WH, Zhu YF, Xiong LM, Shi JL. Facile synthesis of manganese-loaded mesoporous silica materials by direct reaction between KMnO4 and anin-situ surfactant template. Adv Funct Mater 2005;15:955e60.

[37] Dong XP, Shen WH, Zhu YF, Xiong LM, Gu JL, Shi JL. Investigation on Mn-loaded mesoporous silica MCM-41 prepared via reducing KMnO4 with insitu surfactant. Microporous Mesoporous Mater 2005;81:235e40.

[38] Badalyan AG, Syrnikov PP, Azzoni CB, Galinetto P, Mozzati MC, Rosa J, et al.Manganese oxide nanoparticles in SrTiO3: Mn. J Appl Phys 2008;104:4.

[39] Clavel G, Willinger MG, Zitoun D, Pinna N. Manganese-doped zirconia nano-crystals. Eur J Inorg Chem; 2008:863e8.

[40] Rosenholm JM, Meinander A, Peuhu E, Niemi R, Eriksson JE, Sahlgren C, et al.Targeting of porous hybrid silica nanoparticles to cancer cells. ACS Nano 2009;3:197e206.

[41] Conner SD, Schmid SL. Regulated portals of entry into the cell. Nature 2003;422:37e44.

[42] Marsh M, McMahon HT. Cell biology - the structural era of endocytosis.Science 1999;285:215e20.

[43] Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeabilityand the EPR effect in macromolecular therapeutics: a review. J Control Release2000;65:271e84.

Y. Chen et al. / Biomaterials 33 (2012) 2388e23982398

[44] Du JZ, Sun TM, Song WJ, Wu J, Wang JA. Tumor-acidity-activated charge-conversional nanogel as an intelligent vehicle for promoted tumoral-celluptake and drug delivery. Angew Chem Int Ed 2010;49:3621e6.

[45] Ganta S, Devalapally H, Shahiwala A, Amiji M. A review of stimuli-responsivenanocarriers for drug and gene delivery. J Control Release 2008;126:187e204.

[46] Lee ES, Gao ZG, Bae YH. Recent progress in tumor pH targeting nanotech-nology. J Control Release 2008;132:164e70.

[47] Liu Z, Fan AC, Rakhra K, Sherlock S, Goodwin A, Chen XY, et al. Supramolecularstacking of doxorubicin on carbon nanotubes for in vivo cancer therapy.Angew Chem Int Ed 2009;48:7668e72.

[48] He QJ, Zhang JM, Shi JL, Zhu ZY, Zhang LX, Bu WB, et al. The effect of PEGy-lation of mesoporous silica nanoparticles on nonspecific binding of serumproteins and cellular responses. Biomaterials 2010;31:1085e92.

[49] Zhao DY, Feng JL, Huo QS, Melosh N, Fredrickson GH, Chmelka BF, et al. Tri-block copolymer syntheses of mesoporous silica with periodic 50 to 300angstrom pores. Science 1998;279:548e52.

[50] He QJ, Shi JL, Zhao JJ, Chen Y, Chen F. Bottom-up tailoring of nonionicsurfactant-templated mesoporous silica nanomaterials by a novelcomposite liquid crystal templating mechanism. J Mater Chem 2009;19:6498e503.