You are on page 1of 8

Biomaterials 34 (2013) 1364e1371

Contents lists available at SciVerse ScienceDirect

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

Biomineralization inspired surface engineering of nanocarriers for pH-responsive,


targeted drug delivery
Zhaowei Chen a, b, Zhenhua Li a, b, Youhui Lin a, b, Meili Yin a, Jinsong Ren a, *, Xiaogang Qu a, *
a
State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun,
Jilin 130022, China
b
Graduate School of the Chinese Academy of Sciences, Beijing 1000039, China

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

Article history: Recent insight into the molecular mechanisms of natural biomineralization has enabled biomimetic
Received 8 October 2012 synthesis of functional organic-inorganic hybrid materials under mild reaction conditions. Here, we
Accepted 24 October 2012 describe a novel method to construct organic-inorganic hybrid on mesoporous silica nanoparticles by
Available online 9 November 2012
utilizing electrostatically absorbed hyaluronic acid (HA) as a reaction site for deposition of calcium
phosphate (CaP) minerals. The addition of another layer of HA on the CaP surfaces not only stabilizes the
Keywords:
nanocomposites but also confers target ability toward CD44 overexpressed cancer cells. The nano-
Biomineralization
materials enable controlled release of loaded anticancer drugs in acidic subcellular environments after
Hyaluronic acid
pH-responsive
receptor mediated endocytosis. More importantly, our study demonstrated that the cancer targeting
Targeted drug delivery nanomaterials dramatically enhanced cellular uptake and cytotoxicity toward breast carcinoma cells.
Cancer therapy These results thus open new opportunities for biomineralization guided nanostructure assemblies with
Mesoporous nanomaterials great potential for biomedical applications.
Ó 2012 Elsevier Ltd. All rights reserved.

1. Introduction utilization as coating materials on various implants to promote


bone-implant bonding or as building blocks of 2D/3D scaffolds for
Nature provides spectacular examples of biologically-formed application in tissue engineering [7,11,12]. Clinically, owing to their
minerals (biomineralization), such as the single domain nano- “osteoconductive” properties, biomimetic CaP materials have
magnets of magnetotactic bacteria, the abrasion-resistant enamel been widely used as bone substitutes in dentistry and orthopedics
of teeth and other hard tissues in vertebrates [1e3]. Since bio- [13e15]. Another striking example is that CaP based nanovectors
minerals (frequently calcium phosphate) are formed in biological used for biomedical applications have attracted increasing research
environments under mild reaction conditions (i.e., aqueous solu- attention in the past years because of their non-cytotoxicity and
tion, ambient temperature and pressure, near neutral pH), biomi- pH-dependent biodegradability [2,16,17]. For instance, Epple et al.
metic approaches could provide new means to directly synthesize proposed multi-shell CaP-DNA nanoparticles and CaP-DNA/siRNA
new classes of organic-inorganic functional materials with complex nanohybrids for effective transfection of cells [18e20]. Adair et al.
architectures and versatile properties [2,4e6]. For example, one can developed CaP nanoparticles that encapsulated both fluorophores
take the ordered deposition of calcium phosphate salts on an and chemotherapeutics for in vitro imaging and anticancer drug
organic matrix by means of a heterogeneous nucleation process, in delivery [21,22]. Despite these promises, the exploring of further
which the crystal properties of CaP (such as crystal structures in functions of CaP based functional biomimetic materials still
morphology and thickness) are readily controllable by tuning the remains a big challenge in this field [2,16].
reaction conditions [4,7e10]. In terms of materials design, bio- During the last decades, surface-functionalized, end-capped
inspired CaP hybrid materials have attracted tremendous interest; mesoporous silica nanoparticles (MSPs) have emerged as appealing
this is mainly due to the fact that these hybrids have a wide range of carriers for controlled anticancer drug delivery owing to their low
very useful properties and potential applications. The favorable toxicity, high surface area and large accessible pore volumes [23,24].
aspects of CaP materials have led to their extensive biological One important feature of this system is the controlled release of the
cargo from the host material only at the desired location responding
to specific external stimuli, such as enzymatic activity [25,26],
* Corresponding authors. Tel.: þ86 431 85262625. photo-irradiation [27e29], changes in redox state [30,31], pH
E-mail addresses: jren@ciac.jl.cn (J. Ren), xqu@ciac.jl.cn (X. Qu). [27,32,33] and temperature [25,28,34,35]. Of the various stimuli

0142-9612/$ e see front matter Ó 2012 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.biomaterials.2012.10.060
Z. Chen et al. / Biomaterials 34 (2013) 1364e1371 1365

studied, changing the pH represents an effective strategy for cancer 2. Materials and methods
therapy since extracellular pH in tumor tissue is slightly lower than
2.1. Materials
that in normal tissue, with endosomes and lysosomes exhibiting
even lower pH values. Notwithstanding these advances, many of the Nanopure water (18.2 MU; Millpore Co., USA) was used in all experiments and to
existing pore-blocking species have disadvantages such as poor prepare all buffers. Tetraethylorthosilicate (TEOS), sodium hydroxide, and rhoda-
applicability in aqueous solutions and the toxicity of the capping mine B were purchased from SigmaeAldrich. N-cetyltrimethylammonium bromide
(CTAB), fluorescein isothiocyanate (FITC) and 3-(2-aminoethylamino)-propyl-
agents used. This motivates us to design nanocomposites based on
trimethoxysilane were obtained from Alfa Aesar. Doxorubicin (Dox) was purchased
natural materials capped MSPs that could respond to pH variations from Sangon (ShangHai, China). Sodium hyaluronate (MW z 10 kDa) and calcium
for controlled drug release. Especially, the design and synthesis of nitrate were purchased from Aladdin. All the chemicals were used as received
CaP based hybrid materials gated MSPs with target ability still without further purification.
remains a significant challenge until now. Recently, Lee’s group
2.2. Apparatus and characterization
developed hydroxyapatite (HAp, Ca5(PO4)3OH) covered MSPs by
dispersing urease modified MSPs in acidic solution containing HAp FTIR, XRD, SEM, TEM, N2 adsorptionedesorption, and UVevisible spectroscopy
and urea, in situ CaP mineralization could be triggered on the zeta potential were employed to characterize the materials obtained. FT-IR analyses
surface of MSPs [36]. Although promising, the protocol is compro- were carried out on a Bruker Vertex 70 FT-IR Spectrometer. X-ray measurements
were performed on a Bruker D8 FOCUS. Powder X-ray Diffractometer using Cu Ka
mised by the requirement for tedious processes of the preparation
radiation. SEM images were obtained with a Hitachi S-4800 FE-SEM. N2 adsorptione
of nanomaterials and the lack of target ability which would lead to desorption isotherms were recorded on a Micromeritics ASAP 2020M automated
insufficient uptake at tumor sites and decrease therapeutic effect. sorption analyzer. The samples were degassed at 40  C for 5 h. The specific surface
Herein, a simple but efficient rout is developed for constructing areas were calculated from the adsorption data in the low pressure range using the
CaP hybrid materials end-capped MSPs, in which each particle is BET model and pore size was determined following the BJH method. TEM images
were acquired with a TECNAI G2 transmission electron microscope (Philips Elec-
composed of MSP as the core and natural organic-inorganic hybrid
tronic Instruments Co., The Netherlands) at 200 kV. UVeVis spectroscopy was
as the shell. As shown in Scheme 1, to make the structure, we carried out with Cary 300 UV/Vis spectrophotometer.
firstly electrostatically absorb a layer of HA to the surface of
amino functionalized MSPs (designed as MSN@HA), then utilize the 2.3. Synthesis of MCM-41-type mesoporous silica nanoparticles
carboxyl groups of HA backbone as the nucleation sites for CaP
N-cetyltrimethylammonium bromide (CTAB, 0.50 g) was first dissolved in
heterogeneous nucleation. Thereafter, by precisely controlling the
240 mL of pure water. Sodium hydroxide (1.75 mL, 2 M) was added to CTAB solution,
following growing process, CaP mineral nanoshells with pH tunable followed by adjusting the solution temperature to 80  C. TEOS (2.5 mL) added by
properties could be formed (designed as MSN@CaP). The additional dropwise while stirring was continued. The mixture was allowed to stir for 2 h to
layer of HA to the mineral surfaces via chelation interactions give rise to white precipitates. The solid product was filtered, washed with deionized
water and ethanol, and dried in air. To remove the surfactant template (CTAB), the
produces a colloidal stabilized nanocomposite (designed as
white powder was refluxed for 24 h in a solution of 3.00 mL of HCl (37%) and
MSN@CaP-HA). In addition to providing colloidal stability, the 60.00 mL of ethanol followed by extensively washing with deionized water and
outmost layer of HA confers the nanosystem cancer cell target ethanol. The resulting surfactant-removed MCM-41 was placed under high vacuum
ability through the special interaction with CD44, which is highly to remove the remaining solvent in the mesopores. As for fluorescein-labeled
expressed on various cancer cells [37e42]. Moreover, compared mesoporous silica nanoparticles, fluorescein isothiocyanate (FITC, 2 mg) was reac-
ted with 60 mL of APTES in 1 mL ethanol overnight in the dark. CTAB (0.50 g) was first
with other templates or colloidal stabilizing agents, extracellular
dissolved in 240 mL of pure water. Sodium hydroxide (1.75 mL, 2 M) was added to
matrix HA possesses several advantages which make it an attrac- CTAB solution, followed by adjusting the solution temperature to 80  C. TEOS
tive targeting moiety decorating drug/gene delivery or imaging (2.44 mL) and APTES-modified dye solution added by dropwise while stirring was
vehicles: it is biodegradable, non-immunogenic, non-toxic, and can continued. The mixture was allowed to stir for 2 h to give rise to white precipitates.
Finally, the surfactant template, CTAB, was removed by refluxing in acidic ethanol
be chemically modified [39,43e49]. More importantly, as to the HA
solution to give FITC-labeled MSP.
or HA conjugates endocytosis pathway, they mainly accumulated in
endosomal and lysosomal compartments after CD44 mediated 2.4. Chemical modification of the MSPs surface (surface amine functionalization)
uptake [39,49e51]. Thus, we propose to engineer MSPs with CaP-
HA hybrid shell to overcome one of the biggest challenges in Extracted MCM-41 nanoparticles (500 mg) were added to dry PhMe (50 mL) and
suspended as a result of stirring and sonication. 3-(2-aminoethylamino)-propyl-
MSPs’ applications mentioned above, which should afford a new
trimethoxysilane (250 mL, 0.2 mmol) was added to the mixture using a micropipette.
type of pH-responsive targeted drug delivery platform to enhance The solution was placed under N2 and heated under reflux for 24 h. The nano-
the therapeutic efficiency while minimizing side effects. particles were removed from solution by filtration using a fritted funnel and washed
with PhMe, THF, and EtOH, sequentially. The material was dried for 24 h using
vacuum. The presence of amino groups was confirmed by IR spectroscopy using the
KBr pellet method.

2.5. Dye loading and mineralization experiments

For rhodamine B loading, the MSNs (60 mg) were soaked in a solution of
rhodamine B (10 mg) in pure water for 24 h. The nanoparticles were then centri-
fuged and washed thoroughly with pure water to remove unloaded and adsorbed
molecules. Mineralization of the HA-NPs was carried out in the presence of calcium
nitrate and ammonium phosphate using a method similar to that developed by
Schmidt et al. for the liposomes. In brief, the stock solutions (0.1 M calcium nitrate
and 0.1 M ammonium phosphate) were prepared in deionized water. The HA coated
MSN nanoparticles were prepared as following, 20 mg loaded MSNs were dispersed
into 10 mL 0.1 M NaCl solution. Then, 20 mg HA, which were hydrated in 0.1 M NaCl
solution overnight to allow swelling and complete solubilization, were added into
the above solution stirred for 8 h. In order to induce mineralization, 0.03 mL calcium
nitrate solution was slowly added to the solution containing the MSN@HA and
stirred for 1 h. To the resulting solution, 0.02 mL of the ammonium phosphate
solution was slowly added and stirred for 1 h. This procedure of sequential addition
of the stock solutions was repeated 8 times to achieve effective mineralization. Then,
Scheme 1. Schematic representation of the biomineralization inspired CaP-HA hybrid the suspension was allowed to age for 12 h without stirring. The obtained nano-
coated MSNs. particles were then dispersed into 10 mL HA (2 mg/mL) solution and stirred for 12 h.
1366 Z. Chen et al. / Biomaterials 34 (2013) 1364e1371

The final substrates were lyophilized to obtain HA stabilized mineralization coated electron microscopy (SEM, X-ray diffraction and TEM, respectively).
MSNs. All the washing solutions were collected, and the loading of Rhodamine B The nitrogen adsorptionedesorption isotherms of N2 adsorptione
were calculated from the difference in concentrations of the initial and left dyes to
be approximately 38 mmol/g SiO2.
desorption isotherms of MSNs showed a typical Type IV curve
with a specific surface area of 1320 m2 g1, average pore diameter
2.6. Dye release experiments of 2.9 nm and a narrow pore distribution (Fig. 1D).
The successful mineralization of MSNs was confirmed by
Rhodamine B-loaded MSN@CaP-HA (10 mg) material was dispersed in 5 ml various spectroscopy methods. As shown in Fig. 1A, the emerging
aqueous buffer solutions (pH 7.4 phosphate buffer and pH 4.5 acetate buffer) at
37 C. Aliquots (50 mL) were taken from the suspension at predetermined time
 absorption band at around 1700 cm1 in the sample can be
intervals, and replaced with an equal volume of the fresh medium. The delivery of assigned to C]O stretching of the carboxyl groups contained
rhodamine dye from the pore to the buffer solution was monitored via the absor- within the adsorbed HA molecules. The nucleation of CaP at the
bance band of the dye centered at 553 nm. interfaces was supported by the clear peaks corresponding OePeO
bending modes appearing around 600 and 560 cm1, PeO asym-
2.7. CaP disintegration experiments
metric stretching mode at around 1000 cm1 [12]. Consistently, HA
MSN@CaP-HA (10 mg) material was dispersed in 5 ml aqueous buffer solutions templated mineralization process on MSNs was tracked by zeta
(pH 7.4 phosphate and pH 4.5 acetate buffers) at 37  C, and replaced with an equal potential measurements in deionized water at each step (Fig. 1C).
volume of the fresh medium. The release rate of calcium ions was monitored by The zeta potential of the MSNs decreased from þ16.85 mV to
taking a 50 mL sample and diluting it using Arsenazo III solution (400 mL, 0.2 mM) in
27.92 mV indicating the adsorption of HA on the interfaces which
HBS (HEPES-buffered saline where [HEPES] ¼ 20 mM and [NaCl] ¼ 150 mM at pH 7.4).
The absorbance of Arsenazo III/Ca2þ complex in the solution was measured at is important for the following heterogeneous CaP nucleation. After
656 nm, and the concentration of calcium ions was calculated based on the standard mineralization, the obtained nanoparticles exhibited a zeta
curve. potential of 5.87 mV suggesting that mineral deposition almost
completely shielded anionic carboxyl charges. The final zeta
2.8. Dox loading and mineralization experiments
potential reached 20.18 mV, which manifested the anchoring of
For Dox loading, the MSNs (60 mg) were soaked in a solution of Dox (4 mL, 1 g/L)
another layer of HA. The surrounding of the outmost layer of HA
in nanopure water for 24 h. The nanoparticles were then centrifuged and washed stabilizes the nanocomposite though electrostatic repulsion among
thoroughly with nanopure water to remove unloaded and adsorbed molecules. The the HA chains, which is advantageous for potential oncology
mineralization process was almost the same as above. The loading of Dox was application. TEM investigations also provided direct evidence of the
calculated from the difference in concentrations of the initial and left drugs by UV/
CaP mineral distribution on the HA absorbed MSNs. As shown in
Vis spectroscopy to be approximately 56.4 mmol/g SiO2.
Fig. 2b, mineralized MSNs shows rather rough surfaces and blocked
2.9. Cell culture mesopores, representing the heterogeneous nucleation of CaP
clusters on the exterior surface of MSNs. To confirm the presence of
MDA-MB-231 and NIH-3T3 cells were cultured in 25 cm2 flasks in Isceve’s calcium and phosphate in the shell, energy-dispersive X-ray spec-
Modified Dulbecco’s medium (IMDM) (Gibco) containing 10% (v/v) fetal bovine
troscopy (EDX) was used to analyze the rough surfaces (Fig. 2c).
serum (Gibco) at 37  C in an atmosphere of 5% (v/v) CO2 in air. The media were
changed every three days, and the cells were passaged by trypsinization before TEM-associated selected-area electron diffraction (SAED) result
confluence. (Fig. 2d) demonstrated that no sharp electron diffraction rings were
found, indicating the formation of amorphous CaP which was the
2.10. Confocal laser scanning microscopy (CLSM) characteristic product of template-induced mineralization at the
early stage [8,9]. Furthermore, the change of sorption type, together
FITC exhibits intense green fluorescence under UV light. This property allows the
use of fluorescence to study the distribution of MSN@CaP-HA inside the cells. A with a decrease of surface area and pore size distribution, was
suspension of FITC-MSN@CaP-HA in PBS was introduced to culture medium over- expected due to the capping effect of the nanoparticles (Fig. 1D).
night to mimic the blood circulation process prior to the cellular uptake. MDA-MB- Taken together, these results indicated the successful synthesis of
231 and NIH3T3 cells were then incubated with nanoparticles in medium for 2.5 h, CaP-HA coated MSNs.
and then after washing free nanoparticles, 10 mM lysotracker was added for specific
staining of the lysosomes of cancer cells for further 0.5 h incubation. Finally, the cells
To investigate the pH-responsive gating behavior of the hybrid
were washed three times with PBS and transferred to serum free medium and then nanomaterials, rhodamine B was loaded before CaP mineralization.
examined by CLSM. The resulting particles were then dispersed in buffer at different
pH’s to test their controlled release property. The absorbance
2.11. Cytotoxicity assays maximum of rhodamine B (553 nm) was plotted as a function of
time to generate a release profile. As shown in Fig. 3A, a lower pH
MTT assays were used to probe cellular viability. MDA-MB-231 and NIH3T3 cells
were seeded at a density of 5000 cells/well (90 mL total volume/well) in 96-well (pH 4.5) led to the fast release of the loaded rhodamine B molecules
assay plates. After 24 h, drugs at the indicated concentrations were added and and the release reached a plateau within 24 h with the release
cells were further incubated for 48 h. To determine toxicity, 10 ml of MTT solution amount of about 82%. In contrast, only negligible rhodamine B was
(BBI) was added to each well of the microtiter plate and the plate was incubated in
released from dye loaded MSN@CaP-HA at physiological pH (pH
the CO2 incubator for an additional 4 h. The cells then were lysed by the addition of
100 ml of DMSO. Absorbance values of formazan were determined with Bio-Rad 7.4). The significant difference indicated the induced release in
model-680 microplate reader at 490 nm (corrected for background absorbance at lower pH was due to the degradation of the mineral composite at
630 nm). Six replicates were done for each treatment group. the surface. In a control experiment, monolayer HA absorbed MSNs
exhibited sustained release patterns at both pH 4.5 and 7.4, further
3. Results and discussion supporting the conclusion that the mineral structure has a direct
effect on the accessibility of the pores and confers the superiority
We first synthesized MSPs functionalized with positively merely adsorption of monolayer of organic molecules on MSNs.
charged groups that can electrostatically interact with HA by These results imply that cargo leakage from the biomineralized
treating MSPs with 3-(2-aminoethylamino)-propyltrimethox- system could be significantly reduced during sample storage and
ysilane to afford MSNs. The amines of MSNs exhibited an NeH potential blood circulation before accumulating at the tumor area
bending vibration at 1580 cm1 (Fig. 1A-b). As depicted in by the EPR effect.
Figs. S1 and S2 (see Supporting Information) and Fig. 2a the Next, the disintegration of the mineral coating was investigated
spherical particle shape and hexagonally packed mesoporous by monitoring the concentration of the released calcium ions as
structure of the MSNs are confirmed by scanning and transmission a function of time. As shown in Fig. 3B, at lysosomal pH (pH 4.5),
Z. Chen et al. / Biomaterials 34 (2013) 1364e1371 1367

Fig. 1. (A), (B) FTIR spectra of (a) MSPs, (b) MSNs, (c, c’) MSN@HA, (d, d’) MSN@CaP, and (e, e’) MSN@CaP-HA. (C) Zeta potential measured at each step of the coating process in
deionized water. (D) Nitrogen adsorptionedesorption isotherms for MSN (triangle) and mineralized MSN (circle). Corresponding pore size distributions derived from desorption
isotherm measurements and BJH methods. The error bars represent the standard deviation of three measurements.

most of the calcium ions were released within 2 h, implying the fast indicated that the design renders the system the ability for target
disintegration of the mineral shell. However, only insignificant drug delivery.
calcium ions were released at pH 7.4. These findings suggested that Previous reports suggest that HA nanoconjugates were taken up
upon receptor mediated endocytosis by cells, the CaP mineral into cells via CD44 mediated endocytosis and mainly located in the
coatings could be readily dissolved in acidic intracellular acidic compartments such as endosomal and lysosomal vesicles
compartments, whereas their dissolution elsewhere in the body [39,49,50]. Therefore a co-localization study was carried out to
would be rather slow. determine if the HA coated mineralized nanoparticles also accu-
We next confirmed the targeting efficiency of the nano- mulated within the lysosomes and endosomes after endocytosis.
conjugates. MDA-MB-231 cells, which have a high expression of While incubating cells with FITC doped nanoparticles, the endo-
CD44, were chosen as target cancer cells, whereas NIH3T3 cells somes and lysosomes were labeled with red florescent lysotracker
were used as control cells. It is evident from the flow cytometry (Fig. 5b, f). After 3 h of incubation, a green fluorescence of FITC was
(Fig. 4) that a large shift was observed for MDA-MB-231 cells apparent within MDA-MB-231 cells and co-localized with lyso-
treated with the FITC-labeled nanoconjugate (FITC-MSN@CaP-HA), tracker red fluorescence (Fig. 5d), implying that the FITC-labeled
but no significant shift was observed for NIH-3T3 cells or MDA-MB- nanoparticles had become highly concentrated within endosomes
231 cells treated with FITC-MSN@CaP, verifying that HA conjugated and lysosomes of MDA-MB-231 cells. As mentioned above, CaP
nanoparticles have the capacity to target MDA-MB-231 cells mineral coatings could be readily dissolved in acidic intracellular
effectively. Corresponding confocal fluorescence microscopy compartments, thus the enhanced internalization and low pH
images were consistent with the flow cytometry results: a strong inside lysosomes and endosomes should facilitate the release of the
green fluorescence was observed for MDA-MB-231 cells, but no anticancer drugs, which could improve the therapeutic efficiency.
distinct fluorescence was seen for NIH3T3 cells (Fig. 5a, e). More- To investigate the toxicity of CaP-HA hybrid coated nano-
over, competition experiments, using 5 mg/mL free HA, were per- particles, cell viability was analyzed by the 3-(4, 5-dimethylthiazol-
formed as described previously [43]. As shown in flow cytometry 2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay in the pres-
that the binding and uptake of FITC-MSN@CaP-HA into MDA-MB- ence of MSN@CaP, MSN@CaP-HA, doxorubicin (Dox) loaded
231 cells could be blocked by excess HA (Fig. 4d). These results MSN@CaP-HA (Dox@MSN@CaP-HA) and free Dox (Fig. 6). Due to
could be attributed to the special interaction between HA and CD44 the fact that MSNs, CaP and HA all exhibit low- or non-cytotoxicity,
overexpressed on MDA-MB-231 cells. Since no binding occurs the drug carrier: MSN@CaP and MSN@CaP-HA showed a negligible
between NIH3T3 cells and the HA on the nanoconjugates, neither effect on both kinds of cells, even at high concentrations. However,
shift nor enhancement was observed. Thus, the adsorption of growth inhibition of cells was observed after incubation with
another layer HA afforded the nanoconjugates target ability, which both free Dox and Dox@MSN@CaP-HA, which presented dose-
1368 Z. Chen et al. / Biomaterials 34 (2013) 1364e1371

Fig. 2. TEM micrographs of MSNs (a) and mineralized MSNs (b) EDX spectra (c) and TEM-associated SAED patterns of the mineral coating (d).

dependent cytotoxicity behavior. Killing efficiency of overexpressed on tumor cell surface. Lesley and co-workers [52]
Dox@MSN@CaP-HA on MDA-MB-231 cells was significantly reported that cooperativity is the primary feature of HA binding
increased when increasing the amount of nanoparticles. A similar by cell surface CD44. This cooperativity is the result of multiple
profile, but to a lower extent, was observed on NIH3T3 cells. In binding sites on the repeating disaccharide ligand of HA backbone
addition, as shown in Fig. S3, the MTT results of another control cell and multiple closely arrayed receptors on the cell surface. As
line (HEK 293T cells) were similar with that of NIH3T3 cells. demonstrated above, after receptor mediated endocytosis, the
Moreover, at each concentration, Dox@MSN@CaP-HA showed nanoconjugates mainly accumulated in the endosomal and lyso-
remarkably higher anticancer efficiency than free Dox. The mech- somal compartments, where CaP mineral coatings could be readily
anism involved in the augmentation of anticancer efficiency was dissolved and facilitated drug release. Synthetically, the enhanced
related on one hand to the presence of HA attached on the particle intracellular uptake and the followed drug release resulted in
surface and on the other hand to a large amount of CD44 receptors enhanced killing efficacy as well as improved targeting specificity

Fig. 3. (A) Release profiles of rhodamine B from mineralized MSNs at (a) pH 4.5, (d) pH 7.4 and HA absorbed MSN at (b) pH 7.4 and (c) pH 4.5. (B) Release profiles of calcium ions
from mineralized MSNs in different pH buffers. The error bars represent the standard deviation of three measurements.
Z. Chen et al. / Biomaterials 34 (2013) 1364e1371 1369

Fig. 4. Flow cytometry analysis to monitor the binding of FITC doped nanoparticles with MDA-MB-231 cells (target cells) and NIH3T3 (control cells). (a), (e) cells only, (b), (f) FITC-
MSN@CaP, (c), (g) FITC-MSN@CaP-HA, (g), (h) FITC-MSN@CaP-HA after preincubation for 2 h with free HA (5 mg/mL).

Fig. 5. Confocal laser scanning microscopy investigation of the target ability and localization of FITC-MSN@CaP-HA to MDA-MB-231 cells (A) and NIH3T3 cells (B) each sample was
imaged for (a), (e) FITC, (b), (f) lysotracker, (c), (g) bright filed, and (d), (h) overlay. The images were obtained under magnification of 80.

Fig. 6. In vitro viability of MDA-MB-231 cells (target cells) and NIH3T3 cells (control cells) in the presence of MSN@CaP, MSN@CaP-HA, Dox@MSN@CaP-HA, and free DOX. The error
bars represent the standard deviation of three measurements.
1370 Z. Chen et al. / Biomaterials 34 (2013) 1364e1371

Fig. 7. (a), (b) Flow cytogram representing apoptosis assay based on Annexin V-FITC and PI staining. In the flow cytogram, the cells in Q3 region denotes live cells, Q4: apoptotic, Q2:
late apoptotic and Q1: necrotic cells. (c), (d) Laser scanning confocal microscope images of cancer cells. (a), (c) control cancer cells, (b), (d) Dox@MSN@CaP-HA treated cancer cells.
The images were obtained under magnification of 40.

than conventional chemotherapy using Dox only. As for NIH3T3 4. Conclusion


cells and HEK 293T cells, the relatively low killing efficiency of
Dox@MSN@CaP-HA can be attributed to the minimal internaliza- In summary, we have successfully designed and synthesized
tion of the nanovector owing to two possible reasons: (i) lack of a natural organic-inorganic hybrid capped mesoporous silica
CD44 receptors on the cell surface; (ii) negatively charged surfaces nanocontainers that is based on biomineralization mechanism and
of the nanoparticles. Summarizing the results, we confirmed our responds to pH-stimuli. In preparation process, polyanionic HA was
hypothesis that the CaP-HA hybrid capped MSNs could serve as attached directly to the outlet of the mesopores by the electrostatic
a pH-responsive controlled release system for enhanced cancer interaction and served as a template for CaP heterogeneous
target drug delivery. nucleation to entrap guest molecules within the mesopores. The
It is known that DOX suppresses the growth of cancer cells attachment of another layer of HA on the mineral surface enhanced
through intercalation with cellular DNA, inducing apoptosis. To the colloidal stability of the nanoparticles as well as conferred
evaluate the cell apoptosis after treatment by DOX@MSN@CaP-HA cancer cell target ability. After CD44 mediated cellar uptake, cargo
at DOX concentration of 6 mg/mL, Annexin V FITC and PI assay plus release was triggered by the disintegration of the CaP mineral
flow cytometry were employed. As shown in Fig. 7b, a significantly coating in acidic subcellular compartments. Moreover, we have
higher proportion of cell population treated with DOX@MSN@CaP- successfully demonstrated that the HA nanoconjugates showed
HA is Annexin V FITC positive as compared to the untreated cells a remarkably enhanced efficiency in killing cancer cells while
(Fig. 7a). Fig. 7c, d shows representative morphology and DAPI- sparing normal cells. The low cytotoxicity, targeted cellular uptake
fluorescence photographs of control and treated MDA-MB-231 properties, and efficient intracellular pH-stimuli drug release
cells. In the control group, MDA-MB-231 cells grow exuberantly, provide a basis for potential in vivo controlled release biomedical
with integrated nucleus structure, nucleolus, and clearly distin- applications.
guished karyothecas, and the contents in the nucleus are homo-
geneous. But in the drug group, the majority cells have undergone Acknowledgments
morphological changes to nearly spherical shape and shown
hypercondensed chromatin at the nuclear periphery in most cells. The authors are grateful for the referee’s helpful comments
These results support the fact that DOX@MSN@CaP-HA induces on the manuscript. Financial support was provided by the
MDA-MB-231 cells to undergo apoptosis. National Basic Research Program of China (2011CB936004 and
Z. Chen et al. / Biomaterials 34 (2013) 1364e1371 1371

2012CB720602) and the National Natural Science Foundation of [24] 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 2008;
China (Grants 20831003, 21210002, 20833006, 90913007,
3:197e206.
21072182). [25] Chen C, Geng J, Pu F, Yang X, Ren J, Qu X. Polyvalent nucleic acid/mesoporous
silica nanoparticle conjugates: dual stimuli-responsive vehicles for intracel-
Appendix A. Supplementary data lular drug delivery. Angew Chem Int Ed 2011;50:882e6.
[26] Park C, Kim H, Kim S, Kim C. Enzyme responsive nanocontainers with cyclo-
dextrin gatekeepers and synergistic effects in release of guests. J Am Chem Soc
Supplementary data related to this article can be found at http:// 2009;131:16614e5.
dx.doi.org/10.1016/j.biomaterials.2012.10.060. [27] Aznar E, Marcos MD, Martinez-Manez RN, Sancenon Fl, Soto J, Amoros P, et al.
pH- and photo-switched release of guest molecules from mesoporous silica
supports. J Am Chem Soc 2009;131:6833e43.
References [28] Yang X, Liu X, Liu Z, Pu F, Ren J, Qu X. Near-infrared light-triggered, targeted
drug delivery to cancer cells by aptamer gated nanovehicles. Adv Mater 2012;
[1] Palmer LC, Newcomb CJ, Kaltz SR, Spoerke ED, Stupp SI. Biomimetic systems 24:2890e5.
for hydroxyapatite mineralization inspired by bone and enamel. Chem Rev [29] Mal NK, Fujiwara M, Tanaka Y. Photocontrolled reversible release of guest
2008;108:4754e83. molecules from coumarin-modified mesoporous silica. Nature 2003;421:
[2] Cai Y, Tang R. Calcium phosphate nanoparticles in biomineralization and 350e3.
biomaterials. J Mater Chem 2008;18:3775e87. [30] Liu R, Zhao X, Wu T, Feng P. Tunable redox-responsive hybrid nanogated
[3] Xia F, Jiang L. Bio-inspired, smart, multiscale interfacial materials. Adv Mater ensembles. J Am Chem Soc 2008;130:14418e9.
2008;20:2842e58. [31] Luo Z, Cai K, Hu Y, Zhao L, Liu P, Duan L, et al. Mesoporous silica nanoparticles
[4] Haase NR, Shian S, Sandhage KH, Kröger N. Biocatalytic nanoscale coatings end-capped with collagen: redox-responsive nanoreservoirs for targeted drug
through biomimetic layer-by-layer mineralization. Adv Funct Mater 2011;21: delivery. Angew Chem Int Ed 2011;50:640e3.
4243e51. [32] Chen C, Pu F, Huang Z, Liu Z, Ren J, Qu X. Stimuli-responsive controlled-release
[5] Murphy WL, Mooney DJ. Bioinspired growth of crystalline carbonate apatite system using quadruplex DNA-capped silica nanocontainers. Nucleic Acids
on biodegradable polymer substrata. J Am Chem Soc 2002;124:1910e7. Res 2011;39:1638e44.
[6] Liu K, Jiang L. Multifunctional integration: from biological to bio-inspired [33] Luo Z, Cai K, Hu Y, Zhang B, Xu D. Cell-specific intracellular anticancer drug
materials. ACS Nano 2011;5:6786e90. delivery from mesoporous silica nanoparticles with pH sensitivity. Adv
[7] Yeo MG, Kim GH. Preparation and characterization of 3D composite scaffolds Healthcare Mater 2012;1:321e5.
based on rapid-prototyped PCL/b-TCP struts and electrospun PCL coated with [34] Fu Q, Rao GVR, Ista LK, Wu Y, Andrzejewski BP, Sklar LA, et al. Control of
collagen and ha for bone regeneration. Chem Mater 2011;24:903e13. molecular transport through stimuli-responsive ordered mesoporous mate-
[8] Schmidt HT, Ostafin AE. Liposome directed growth of calcium phosphate rials. Adv Mater 2003;15:1262e6.
nanoshells. Adv Mater 2002;14:532e5. [35] Schlossbauer A, Warncke S, Gramlich PME, Kecht J, Manetto A, Carell T, et al.
[9] Schmidt HT, Gray BL, Wingert PA, Ostafin AE. Assembly of aqueous-cored A programmable DNA-based molecular valve for colloidal mesoporous silica.
calcium phosphate nanoparticles for drug delivery. Chem Mater 2004;16: Angew Chem Int Ed 2010;49:4734e7.
́
4942e7. [36] Rim HP, Min KH, Lee HJ, Jeong ́SY, ̃ Lee SC. pH-tunable
́ ́
calcium phosphate
[10] Nudelman F, Pieterse K, George A, Bomans PHH, Friedrich H, Brylka LJ, et al. covered mesoporous silica nanocontainers for intracellular controlled release
The role of collagen in bone apatite formation in the presence of hydroxy- of guest drugs. Angew Chem Int Ed 2011;50:8853e7.
apatite nucleation inhibitors. Nat Mater 2010;9:1004e9. [37] Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules to signalling
[11] Carlo Reis EC, Borges APB, Araújo MVF, Mendes VC, Guan L, Davies JE. Peri- regulators. Nat Rev Mol Cell Biol 2003;4:33e45.
odontal regeneration using a bilayered PLGA/calcium phosphate construct. [38] Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev
Biomaterials 2011;32:9244e53. Cancer 2004;4:528e39.
[12] Liu H, Xi P, Xie G, Shi Y, Hou F, Huang L, et al. Simultaneous reduction and [39] Qhattal HSS, Liu X. Characterization of CD44-mediated cancer cell uptake and
surface functionalization of graphene oxide for hydroxyapatite mineraliza- intracellular distribution of hyaluronan-grafted liposomes. Mol Pharm 2011;
tion. J Phys Chem C 2012;116:3334e41. 8:1233e46.
[13] LeGeros RZ. Calcium phosphate-based osteoinductive materials. Chem Rev [40] Underhill C. CD44: the hyaluronan receptor. J Cell Sci 1992;103(Pt 2):293e8.
2008;108:4742e53. [41] Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. CD44 is the principal
[14] Park JK, Shim J-H, Kang KS, Yeom J, Jung HS, Kim JY, et al. Solid free-form cell surface receptor for hyaluronate. Cell 1990;61:1303e13.
fabrication of tissue-engineering scaffolds with a poly(lactic-co-glycolic [42] Platt VM, Szoka Jr FC. Anticancer therapeutics: targeting macromolecules and
acid) grafted hyaluronic acid conjugate encapsulating an intact bone nanocarriers to hyaluronan or CD44, a hyaluronan receptor. Mol Pharm 2008;
morphogenetic proteine2/poly(ethylene glycol) complex. Adv Funct Mater 5:474e86.
2011;21:2906e12. [43] Luo Y, Ziebell MR, Prestwich GD. A hyaluronic acidtaxol antitumor bio-
[15] Moreau JL, Xu HHK. Mesenchymal stem cell proliferation and differentiation conjugate targeted to cancer cells. Biomacromolecules 2000;1:208e18.
on an injectable calcium phosphate-chitosan composite scaffold. Biomaterials [44] Pouyani T, Prestwich GD. Functionalized derivatives of hyaluronic acid
2009;30:2675e82. oligosaccharides: drug carriers and novel biomaterials. Bioconjug Chem 1994;
[16] Epple M, Ganesan K, Heumann R, Klesing J, Kovtun A, Neumann S, et al. 5:339e47.
Application of calcium phosphate nanoparticles in biomedicine. J Mater Chem [45] Liu G, Choi KY, Bhirde A, Swierczewska M, Yin J, Lee SW, et al. Sticky nano-
2010;20:18e23. particles: a platform for siRNA delivery by a bis(zinc(ii) dipicolylamine)-
[17] Han S-Y, Han HS, Lee SC, Kang YM, Kim I-S, Park JH. Mineralized hyaluronic acid functionalized, self-assembled nanoconjugate. Angew Chem Int Ed 2012;51:
nanoparticles as a robust drug carrier. J Mater Chem 2011;21:7996e8001. 445e9.
[18] Sokolova VV, Radtke I, Heumann R, Epple M. Effective transfection of cells [46] Lee Y, Lee H, Kim YB, Kim J, Hyeon T, Park H, et al. Bioinspired surface
with multi-shell calcium phosphate-DNA nanoparticles. Biomaterials 2006; immobilization of hyaluronic acid on monodisperse magnetite nanocrystals
27:3147e53. for targeted cancer imaging. Adv Mater 2008;20:4154e7.
[19] Klesing J, Chernousova S, Epple M. Freeze-dried cationic calcium phosphate [47] Choi KY, Min KH, Na JH, Choi K, Kim K, Park JH, et al. Self-assembled hya-
nanorods as versatile carriers of nucleic acids (DNA, siRNA). J Mater Chem luronic acid nanoparticles as a potential drug carrier for cancer therapy:
2012;22:199e204. synthesis, characterization, and in vivo biodistribution. J Mater Chem 2009;
[20] Kozlova D, Chernousova S, Knuschke T, Buer J, Westendorf AM, Epple M. Cell 19:4102e7.
targeting by antibody-functionalized calcium phosphate nanoparticles. [48] Lee H, Lee K, Kim IK, Park TG. Synthesis, characterization, and in vivo diag-
J Mater Chem 2012;22:396e404. nostic applications of hyaluronic acid immobilized gold nanoprobes. Bioma-
[21] Kester M, Heakal Y, Fox T, Sharma A, Robertson GP, Morgan TT, et al. Calcium terials 2008;29:4709e18.
phosphate nanocomposite particles for in vitro imaging and encapsulated [49] Choi KY, Yoon HY, Kim J-H, Bae SM, Park R-W, Kang YM, et al. Smart nano-
chemotherapeutic drug delivery to cancer cells. Nano Lett 2008;8:4116e21. carrier based on PEGylated hyaluronic acid for cancer therapy. ACS Nano
[22] Morgan TT, Muddana HS, Altinoglu EI, Rouse SM, Tabakovic A, Tabouillot T, 2011;5:8591e9.
et al. Encapsulation of organic molecules in calcium phosphate nano- [50] Stern R. Hyaluronan catabolism: a new metabolic pathway. Eur J Cell Biol
composite particles for intracellular imaging and drug delivery. Nano Lett 2004;83:317e25.
2008;8:4108e15. [51] Robert S. Hyaluronidases in cancer biology. Semin Cancer Biol 2008;18:
[23] Kim T-W, Slowing II, Chung P-W, Lin VS-Y. Ordered mesoporous polymer- 275e80.
silica hybrid nanoparticles as vehicles for the intracellular controlled release [52] Lesley J, Hascall VC, Tammi M, Hyman R. Hyaluronan binding by cell surface
of macromolecules. ACS Nano 2010;5:360e6. CD44. J Biol Chem 2000;275:26967e75.

You might also like