Você está na página 1de 21

1166 Current Drug Targets, 2011, 12, 1166-1186

Multifunctional Mesoporous Silica Nanoparticles for Combined


Therapeutic, Diagnostic and Targeted Action in Cancer Treatment
Jessica M. Rosenholm1,2, Cecilia Sahlgren*,3 and Mika Lindén*,1

1
Center for Functional Materials, Department of Physical Chemistry, Åbo Akademi University, Porthansgatan 3-5, FI-
20500, Turku, Finland
2
Nano Biomedical Research Center, Med-X Institute, Shanghai Jiao Tong University, 1954 Huashan Rd., 200030
Shanghai, P.R. China
3
Department of Biology, Åbo Akademi University, Artillerigatan 6A, FI-20520 Turku, Finland, and Turku Centre for
Biotechnology, University of Turku and Åbo Akademi University, P.O. Box 123, FI-20521, Turku, Finland

Abstract: The main objective in the development of nanomedicine is to obtain delivery platforms for targeted delivery of
drugs or imaging agents for improved therapeutic efficacy, reduced side effects and increased diagnostic sensitivity. A
(nano)material class that has been recognized for its controllable properties on many levels is ordered mesoporous
inorganic materials, typically in the form of amorphous silica (SiO 2). Characteristics for this class of materials include
mesoscopic order, tunable pore dimensions in the (macro)molecular size range, a high pore volume and surface area, the
possibility for selective surface functionality as well as morphology control. The robust but biodegradable ceramic matrix
moreover provides shelter for incorporated agents (drugs, proteins, imaging agents, photosensitizers) leaving the outer
particle surface free for further modification. The unique features make these materials particularly amenable to modular
design, whereby functional moieties and features may be interchanged or combined to produce multifunctional
nanodelivery systems combining targeting, diagnostic, and therapeutic actions. This review covers the latest developments
related to the use of mesoporous silica nanoparticles (MSNs) as nanocarriers in biomedical applications, with special
focus on cancer therapy and diagnostics.
Keywords: Mesoporous silica nanoparticles (MSNs), drug delivery systems, multimodal imaging, gene delivery, multidrug
resistance, photodynamic therapy, theragnostic agents, targeted cancer therapy.

1. INTRODUCTION monitoring (imaging) – often referred to as theragnostics. In


the beginning of the millennium, the research field of meso-
The application of nanotechnology in medicine is anti-
scopically ordered nanoporous silica-based materials entered
cipated to meet the therapeutic and diagnostic challenges that
the drug delivery scene. The pore size and mesoscopic order
today limit the therapeutic window and clinical applicability
of these materials can be controlled using supramolecular
of many drugs and hamper early diagnosis. A promising in surfactant aggregates as structure-directing agents during the
this field of research is the development of nanoparticle-
synthesis, resulting in a highly porous, amorphous silica
based systems for targeted both diagnostic and therapeutic
material. The pore dimensions can be adjusted to fit the size
agents. Incorporation of drugs into the delivery platform
of payload molecules. A nanotechnology approach is used
protects the drug from premature degradation and elimina-
for engineering of the Sol-gel derived, amorphous silica,
tion and is important for clinical translation of poorly soluble
(SiO2), is known to be biocompatible and is suitable for
therapeutic agents, which are associated with numerous for- delivery of many types of biologically active agents,
mulation-related complications [1]. Novel drug delivery
including hydrophobic drugs, nucleic acids (DNA, siRNA)
technology is thus expected to improve bioavailability,
and proteins, including hydrophobic drugs, nucleic acids
pharmacokinetics and tissue distribution of existing and
(DNA, siRNA) and proteins [3-15]. Due to their unique
novel drugs.
properties, which include a narrow pore size distribution,
Consequently, drug delivery is a central part of drug high specific surface area and pore volume along with
development. New technologies are constantly being deve- tunable pore sizes and mesoscopic order, mesoporous silica
loped to meet the requirements of ‘smart’ drug delivery [2]. particles have attracted attention after the first full reports of
In cancer therapy, nanoparticles are expected to bring for- the synthesis and characterization of this interesting class of
ward multifunctional capabilities within the same system, i.e. materials appeared in the early 90´s [16-18]. With the
simultaneously function in targeted delivery of drugs to advances in morphology control in the 2000’s, enabling
cancer cells, in combination with concurrent real-time in vivo fairly straightforward synthesis of mesoporous silica nano-
particles (MSNs), not only the nanoscopic structure of
mesoporous silica but also the particles themselves could be
*Address correspondence to this author at the Center for Functional utilized as a nanoscale object. This development boosted the
Materials, Department of Physical Chemistry, Åbo Akademi University, interest for MSNs in biotechnological and biomedical
Porthansgatan 3-5, FI-20500, Turku, Finland;
E-mails: cecilia.sahlgren@abo.fi, mika.linden@abo.fi applications.

1389-4501/11 $58.00+.00 © 2011 Bentham Science Publishers Ltd.


Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1167

Due to the large surface area and the controllable surface usually on the micrometer scale, although MSNs has been
functionality of MSNs, they can be loaded with large prepared successfully also under acidic [20], neutral [21],
amounts of drugs and coupled to molecules of choice for and close to neutral conditions [22]. The addition of particle
targeting purposes, simultaneously carrying traceable (fluo- growth quenchers is another useful adjunct for morphology
rescent or magnetically active) modalities. Thus, MSNs are control on the nanoscale. Some crucial issues such as upscal-
truly multifunctional and can be used not only to specifically ing, redispersibility after all production (functionalization)
deliver drugs (therapeutic action) but also for medical steps as well as hydrolytic stability in physiological media,
imaging (diagnostic action). Targeted action that minimize remain to be properly addressed on the material design side
interactions with healthy tissue and enhances the cellular before MSNs are to be clinically applied, however the
uptake of the drugs in diseased cells, in combination with the diverse functionality and flexibility of MSNs as (potential)
high drug load potential of MSNs will have a significant drug carriers have been vastly demonstrated since the
impact on the therapeutic efficacy of drugs with limited beginning of the 2000’s.
clinical applicability. The high drug load potential of MSNs
makes them efficient tools for multidrug delivery, a desirable 2.2. Core-Shell Particles
feature in cancer therapy given the complexity of oncogenic
activities. Furthermore, as the particles can be internalized In order to increase the functionality level of all-silica
by cells, their use as carriers for contrast agents can sig- MSNs, integration of mesoporous silica with nanocrystals of
nificantly prolong the imaging time-frame, as the half-lives other materials to form uniform core-shell composite nano-
of many currently commercially available contrast agents are structures with great potential in various biotechnological
extremely short and many are toxic. Thus, tissue specificity and biomedical fields, especially medical imaging, have
is desired in order to improve the clinical diagnosis and also emerged as a popular approach also outside the mesoporous
because it is possible to reduce the dose, since the contrast silica community. Mesoporous silica shells have thus been
agent would become concentrated near the target. Conse- successfully deposited on solid cores consisting of different
quently, such drug delivery systems to target the delivery of materials such as platinum [23, 24], gold [25-29], single
drugs or imaging agents to specific tissues and cells of the NaCl crystals [30], quantum dots [27, 31], and silver [32].
body will not only have enormous impact on human health, The mesoporous silica-coated composite material subject to
but will also be of great interest for medical imaging. In the most extensive research is, however, beyond doubt magnetic,
context of nanomedicine, many critical challenges related to typically ferro- or superparamagnetic iron oxide (SPIO) to
the development of medical technology for improved yield them magnetically active (for external control via an
diagnostic and therapeutic action are met by MSNs, which applied magnetic field to achieve magnetically controlled
will be outlined in the following. drug delivery, hyperthermia, or magnetic field-assisted
uptake of MSNs by cells) as well as suitable for magnetic
resonance imaging (MRI). The magnetic nanoparticles can
2. MODULAR DESIGN OF MESOPOROUS SILICA be incorporated/encapsulated into the mesoporous silica via
NANOPARTICLES many fabrication strategies [33]. A frequently applied
synthesis route for production of materials with nanoscale
2.1. Particle Synthesis
morphology is coating a silica shell around the magnetic iron
The exponential growth in reports covering applications oxide nanocrystals to form a magnetic-core/silica-shell
of mesoporous silica in biological systems is due to recent structure. Hereby, the mesoporous layer can be deposited
advances in morphology control of the materials, enabling either directly onto the magnetic cores [31, 34, 35], or then
efficient particle size tailoring in the nanometer range [19]. the magnetic crystals can be pre-coated [36-39] with a dense
Many mesoporous nanoparticulate fabrication routes are silica layer via the modified Stöber process [40, 41],
dilute versions of the synthesis of MCM-41 (Mobil Com- generally used to coat metal/metal oxide crystals with silica,
position of Matter No. 41), the by far most extensively whereas the subsequent mesoporous coating process would
studied mesoporous material. The synthesis of MCM-41 be equivalent to that of coating of silica surfaces [42-44].
relies on a cooperative self-assembly of supramolecular The thus formed solid silica layer both protects the
aggregates of alkylammonium surfactants with polymerizing underlying iron oxide from acidic environments (in which it
silica species under alkaline (basic) conditions, eventually would dissolve) as well as facilitate the self-assembly of the
leading to an organic-inorganic composite material consist- cationic surfactant templates on the negatively charged silica
ing of hexagonally ordered rod-shaped micelles surrounded surface [36]. However, we note that the synthesis of uniform
by an amorphous silica (SiO2) network. When the organic magnetic cores with outstanding magnetization and relaxi-
portion is removed by either thermal treatment or solvent vity properties is probably the most challenging part in these
extraction (ion exchange), an inorganic (ceramic) material syntheses. Similarly as their all-siliceous counterparts,
with hexagonally ordered voids (pores) with diameters of obtaining still separable nanoparticles after all coating steps,
approximately 3 nm is obtained. The pore size can be tuned especially in the case of single crystal coating, can be a
by for example changing the surfactant, the surfactant chain challenge [34]. However, as the particles are magnetically
length, or by adjusting the synthesis conditions. The reason separable, sequential centrifugation and redispersion steps
for why most nanoparticulate syntheses are based on this are avoided, which affects the single particle integrity in
particular synthesis is due to the fact that morphology terms of redispersability in a favorable way. Two examples
control is generally easier to achieve under basic conditions of mesoporous silica coated magnetite (Fe3O4) particles of
than under acidic conditions. Spherical morphologies are, different size are shown in Fig. (1).
however, readily achievable under acidic conditions albeit
1168 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

a.

b.

Fig. (1). Examples of core-shell magnetite-mesoporous silica nanoparticles. a) A schematic representation of MR-imagable MSNs with a
SPIO nanoparticle core covered by a protective non-porous silica layer and a second layer of mesoporous silica. Drugs, fluorophores,
molecular MRI agents, and (responsive) coatings can be introduced by rational synthesis design. b) Transmission electron micrographs
(TEM) of SPIO nanoparticles-mesoporous silica particles of different types. Note that the overall particle size, the thickness of the
mesoporous silica layer, and the diameter of the SPIO nanoparticle core can be independently controlled. The micrographs were obtained by
a JEM 2010 (JEOL, Japan) instrument with 200 KV accelerated voltage. Synthesis and images Jixi Zhang, Nano Biomedical Research
Centre, Med-X Research Institute, Shanghai Jiao Tong University.

2.3. Flexible Surface Functionalization Strategies is readily accomplished and fairly straightforward, including
methods such as surface grafting of functional silanes, poly-
The flexibility and modularity in the further design of
mer coating, covalent linking of complexes, and co-
MSNs relies both on the mode of formation, structure, and
condensation. Mesoporous silica-specific functionalizations
surface chemistry. Firstly, the modification of silica surfaces have been also frequently reviewed in the literature [45].
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1169

Secondly, the synthesis regime allows for in situ incur- should not leak out during this conjugation step. However, as
poration of functional groups into the silica matrix by the MSN should at this point anyhow be designed not to
addition of functional silanes into the synthesis solution, thus release its cargo immediately upon immersion in aqueous
acting as part of the silica source. This mode-of-action result media (i.e. when exposed to physiological conditions), it is
in functional groups distributed fairly homogeneously in line with rational processing of the MSNs for drug
throughout the silica network, partly embedded in the pore delivery. As standard bioconjugation reactions such as 1-
walls but also exposed on the pore wall surfaces, thus ethyl-3-(3-dimethylaminopropyl)-carbodiimide- (EDC) or
accessible for guest molecules such as drugs. In synthesis N,N'-Dicyclohexylcarbodiimide- (DCC) based coupling (or
strategies where the functional silane acts also as a co- other peptide synthesis reactants) are mostly utilized for the
structure directing agent [46] the fraction of functional attachment, reactive groups should be readily accessible on
groups present on the pore walls can be enhanced. the particle surface. Most often amino groups are utilized for
Functional groups introduced via these co-condensation this kind of conjugation, but also -OH, -COOH, or -SH
strategies are frequently utilized to enhance interactions with groups may be utilized. Fluorochromes are attached to the
incorporated drug molecules (see below) or to couple particle system using similar chemistry, generally via amine-
fluorochromes (or gadolinium-chelate complexes) to render specific reactions, but in the case of small-molecular ligands
the MSNs imagable by optical (or MRI) methods. Thirdly, the fluorophores may benefit from being embedded “deeper”
the particle (outer) surface can subsequently be separately into the particle structure, in order not to have several bulky
functionalized in order to tune the particle surface charge (to molecules onto the particle surface which may hamper or
promote cellular uptake, reduce opsonization of plasma affect the binding (targeting) ability of the biospecific
proteins, direct the particles to specific intracellular molecule. In the case of a “middle layer”, either a responsive
compartments, etc.), enhance particle suspension stability, polymer layer or PEG terminal functional groups of the
improve the ‘stealth’ properties of or introduce gate-keeping polymer chains may thus be essential. In many cases the
properties to the nanoparticulate system, and to attach targeting ligand can be attached to the polymer chains before
targeting ligands to enable active targeting of the MSNs. In conjugated to the particles, whereas bifunctional polymers
order to protect the (inner) pore wall surfaces, reactive would be advantageous in order to attach the ligand to one
functional groups can be attached to the particle surface end of the polymer chain and “reserve” the other end for
before surfactant removal in case the function in question particle conjugation (or use protective groups) to attain
can withstand the conditions applied for extraction [47-49]. chemoselectivity. PEG is readily commercially available
However, as the modifications used for further functionaliza- with different (bi)functionalities for this purpose. When the
tion of MSNs are often macromolecular such as polymers ligand is attached as the last step, depending on the func-
(polyethylene glycol (PEG) for stealthiness [50], poly(D,L- tional groups of the ligand, bridging between particles (self-
lactide-co-glycolide) (PLGA) as release retardant [51], conjugation) should be avoided. This is also crucial if a poly-
poly(allylamine) hydrochloride/poly(sodium-4-styrene sulfo- mer chain is conjugated to the particles with monofunctional
nate) (PAH/PSS) multilayers for pH-responsiveness [52, 53], terminal ends (the same functional group in both ends of the
poly(N-isopropylacrylamide) (PNIPAAm) for thermo-res- chain) or in the use of bifunctional linker molecules.
ponsiveness [54, 55]) or proteins (antibodies or (poly)pep- Importantly, as the ligand-receptor binding is conformation-
tides for targeting) the functionalization is mainly restricted defined, it is crucial the ligand is attached to the particle in a
to the outer particle surface in any case, as the pore sizes for way that it retains its binding activity. Thus, for proteinous
MSNs are generally around 3 nm and thus cannot accom- ligands, also the conditions applied in the conjugation step
modate macromolecular substances. It has also suggested must prevent denaturation of the protein. As bioconjugation
that the most kinetically available Si-OH groups are those on protocols are normally adapted for biomolecules, this
the outside surfaces of the silica particles, and hence also procedure is quite straightforward and the challenge rather
kinetically more accessible for functionalization [56] and lies in playing with the chemistries in a modular fashion to
thus preferential modification of the particle surface can be have a reproducible functionalization protocol sequence
achieved also after extraction of the surfactant template. To compatible with the drug loading process.
keep the beneficiaries of a MSN system, a key issue is that
the pore network is still available after these functionaliza-
tions, as the loading of cargo (drugs) is generally performed 3. MSNS AS DRUG DELIVERY VEHICLES
after these steps. 3.1. Loading of Biologically Active Cargo into Mesopo-
rous Silica
2.4. Attachment of Targeting Moieties
One of the main strengths of MSNs in delivery applica-
The attachment of cell-specific targeting ligands to the tions is the possibility to carry large amounts of payload due
outer surface of the cells is a common method for enhancing to their high intrinsic specific surface areas and pore
MSN cell-specificity. If the targeting moiety is a small- volumes. Drug loading degrees exceeding 30 wt% have been
molecular ligand, such as sugars or folic acid, the attachment reported for mesoporous silica, and keeping in mind that the
of the ligand can be performed before drug loading and in density of amorphous silica is about 2.2 g/cm3, this value
either organic or aqueous solvent, of which organic solvent would correspond to loading degrees exceeding 50 wt% for
might be preferential to not alter the silica matrix. In the case polymeric particles assuming a polymer density of 1 g/cm3.
of proteinous targeting moieties (such as polypeptides or Furthermore, the drug loading levels can be rationally
antibodies), on the other hand, the conjugation step is most controlled, which means that both the drug loading as well as
likely to be performed last and moreover in aqueous solvent, the particle concentration can be used as parameters to
to preserve the activity of the protein, and thus the drug control the total drug levels in vitro and in vivo. This is
1170 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

important, as the cell selectivity in the targeting process as efficient pH-dependent adsorption, as was recently shown
well as the particle toxicity are particle concentration- for the adsorption of the highly water soluble anti-
dependent. inflammatory drug salicylic acid [59]. High amounts of
salicylic acid could be adsorbed from aqueous solvent onto
As mentioned above, the loading of drugs into and/or
onto MSNs is typically carried out as a separate step amino-functionalized mesoporous silica at pH = 3, up to 16
µmol/m2 when the silica surface was functionalized by
following particle synthesis. Separation of material synthesis
surface hyperbranching polymerization of polyethyleneimine
and drug incorporation allows independent optimization of
(PEI), while negligible amounts salicylic acid was adsorbed
structural and physicochemical properties of the carrier and
on native silica under the same conditions. The high surface
the conditions used for the drug loading. Incorporation of
concentration of amino groups lead to strongly positive net-
cargo into silica can be achieved by different means; the
three most important being physical adsorption from solution surface charges even under low pH-conditions, and also
provide the possibility for specific amine-carboxylic acid
into the mesopores, physical adsorption from solution onto
interactions. Under neutral conditions the extent of adsorp-
the outer surface of the MSNs, and covalent linking. Fur-
tion was much lower due to the presence of negatively
thermore, also impregnation by incipient wetness, as well as
charged silanol groups, suggesting that the pH-swing
loading from a drug melt has been studied. The molecular
methodology could be an attractive means for drug adsorp-
structure as well as dimensions of the cargo with respect to
the diameter of the mesopores, its water solubility (and tion from aqueous solution. The adsorption of famotidine, a
histamine H2-receptor antagonist that inhibits stomach acid
cytotoxicity), and the chemical stability of the cargo deter-
production, into mesoporous silica from 50/50 v/v methanol/
mine which of these methods is the preferred one. In the
water solutions at pH = 7 was studied by Tang et al. [60].
following we will briefly discuss the different means of
Famotidine contains four amino-groups in its molecular
cargo incorporation into mesoporous silicas. The discussion
structure, and the adsorbed amount could be directly
is based on selected examples highlighting the mechanistic
aspects related to each method. As most of the published correlated with the amount of carboxylic acid groups present
on the functionalized mesoporous silica. No adsorption of
mechanistic studies have been performed on microparticles
famotidine was observed for corresponding native silica
rather than on MSNs, much of the discussion is based on
materials, although the drug and the silica should be
microparticles. However, the mechanistic aspects are largely
oppositely charged under the studied conditions. These
valid for both particle size ranges, and results obtained for
results further stress the importance of adequate surface
MSNs are included whenever considered relevant. The same
argumentation holds for the cargo, as studies directly aimed modifications to optimize the adsorbate-adsorbent interac-
tions. The adsorption process could be satisfactorily des-
at cancer therapy represent only a fraction of the published
cribed as Langmuir adsorption under the used constant pH-
papers related to controlled adsorption-desorption (loading-
conditions, suggesting monolayer adsorption to equivalent
release) of bioactive cargo from mesoporous silica.
adsorption sites, and that there was little interaction between
3.1.1. Adsorption from Aqueous Solution: Small-Molecular the adsorbed molecules. The monolayer capacity of the
Drugs mesoporous silica with the highest carboxylic acid group
content was 340 mg famotidine/g silica. The fact that the
The most often used method for the loading of small
adsorption can be satisfactorily modeled also means that the
drugs into the pores of MSNs is physical adsorption from
drug loading degree can be rationally fine-tuned to be any
solution. The surface silanol groups, ≡Si-OH, always present
loading degree between zero and the maximum loading.
on the silica surface, and which typically serve as adsorption
sites, are present at surface concentrations of at least 2-4 3.1.2. Adsorption from Aqueous Solution: Macromolecular
groups/nm2 [57]. Therefore, the surface concentration of Species
adsorption sites is normally not a limiting factor for the
The loading of biomacromolecules is, naturally, normally
extent of adsorption. The silanol groups are amphoteric, and
carried out from aqueous solution in order to maintain the
can be present in both a protonated, ≡Si-OH2+, and a
biological activity. Many studies have been published related
deprotonated form, ≡Si-O-, in aqueous media depending on
to the adsorption of proteins and enzymes into the pores of
the pH [58]. The point of zero charge for silica, i.e. the pH at
mesoporous silica. Most of them are aimed at permanent
which the net-surface charge is zero, is about 2-3, which
means that in the absence of specific ion adsorption, the immobilization of enzymes to create bionanoreactors, and
thus fall outside the scope of the present review. The
silica surface is negatively charged under biologically
interested reader is referred to two recent reviews dedicated
relevant conditions. Therefore, electrostatic adsorption of
to this topic [61, 62]. In the following, we will briefly cover
positively charged adsorbates (i.e. in this case, drugs) is an
some mechanistic aspects of the incorporation of biomacro-
attractive method for incorporating water-soluble cargo into
molecular species into and onto mesoporous silica.
siliceous MSNs. The extent of electrostatic adsorption can be
further increased by introducing functional groups, very 3.1.3. Adsorption of Proteins
often weak acids and bases like carboxylic acids or amines,
onto the silica surface, which can be used to tune the Balkus and co-workers were the first to publish a work
effective surface charge under given pH conditions, and also on the immobilization of globular proteins (cytochrome C,
provide the possibility for additional specific adsorbate- papain, and trypsin) on ordered mesoporous silica with pore
adsorbent interactions. The extent to which the effective sizes in the range of 3 nm [63]. Deere et al., on the other
surface charge at a given pH is altered is naturally a function hand, were the first to report adsorption isotherms at 25 °C
of both the chemical nature of the introduced functions and of cytochrome C on different mesoporous silica molecular
their surface concentration. This fact can be utilized for sieves [64, 65]. The isotherms were recorded at a pH of 6.5
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1171

and are of the Langmuir type. Release of cytochrome C by pairs has been successfully incorporated into the mesopores
washing could be achieved only if the pH was raised to 10, of mesoporous silica by adsorption from aqueous solution
which is close to its pI, i.e. the pH value in solution at which [71]. It was shown that pre-incorporation of divalent metal
the sum of the charges on the protein is zero. A maximum ions into the pores enhanced the adsorption of DNA, as the
loading of 10.2 µmol/g was achieved under the conditions metal ions served as charge mediators between the nega-
employed in this study. Hartmann et al. showed that the tively charged silica surface and the negatively charged
extent of adsorption of both cytochrome C [66] and lyso- DNA. The adsorption of DNA could be further enhanced by
zyme [67] onto mesoporous silica (the studied mesoporous functionalization of the pore walls with amino groups, and
silica supports had pore diameters in the range 3-11 nm) is DNA loading degrees of 15.7 mg/g silica could be reached
strongly pH dependent, and a maximum loading degrees for mesoporous silica materials with pore dimensions of 10
were shown to occur close to the pI of each protein. This nm. For native silica materials with mesopore dimensions of
observation was attributed to a minimum of electrostatic about 5 and 10 nm the adsorption process could be described
repulsion between adsorbed proteins, favoring protein close as Langmuir adsorption, while the adsorption process was
packing inside the mesopores. Furthermore, under these better described by the Freundlich isotherm for silicas with 3
“charge-neutralized” conditions, hydrophobic adsorbent- nm mesopores. This suggests that the intermolecular
adsorbate interactions increase in importance, and the protein interactions cannot be neglected in the latter cases.
is also compact due to lacking internal charge repulsion However, in many cases the pore diameters of MSNs are
effects. Hydrogen bonding and hydrophobic effects can be
in the range of 2-3 nm, which severely limits the possibility
also utilized for efficient adsorption of macromolecular
for adsorption of macromolecules into the mesopores. There-
active agents into mesoporous silicas, as recently shown for
fore, adsorption of genes or proteins onto the outer surface of
the incorporation of lipase into mesoporous silica func-
MSNs has been also studied. The underlying driving forces
tionalized by alkyl-groups [68]. The lysozyme and cyto-
for adsorption to the outer surface of the particles are largely
chrome C adsorption data could be generally fitted using a the same as for adsorption into mesopores, although the
Langmuir adsorption isotherm [66, 67]. Introduction of Al
influences of confinement effects are naturally absent. Nel et
into the silica network lead to increased levels of adsorption,
al. [72] adsorbed siRNA and DNA constructs to MSNs that
and this was ascribed to the presence of additional negative
has been firstly coated with a layer of PEI, a well-known
charges at the Al-sites. The maximum loadings were about
gene transfection agent. Particle/nucleic acid ratios of 10-100
680 mg lysozyme/g silica and about 500 mg cytochrome
were needed for complete adsorption. As expected, the high
C/silica g under the studied conditions. It was concluded that positive charge density of the PEI layer was beneficial for
the proteins were mainly located into the mesopores in all
adsorption, and corresponding particles coated with an
cases, but that differences in the pore volume of the supports
organophosphate layer exhibited a much lower nucleic acid
accounted for the observed differences in adsorption capa-
adsorption capacity.
city. Stucky et al. [69] reported the adsorption and desorp-
tion at pH = 7 of conalbumin (MW 77 kD, pI 6.0), chicken 3.1.5. Adsorption from Non-Aqueous Solution – Poorly
egg ovalbumin (MW 44 kD, pI 4.9), and soybean trypsin Water Soluble Small-Molecular Drugs
inhibitor protein (MW 14 kD, pI 5.2) onto/from amino-func-
Many attractive drug candidates have low water solu-
tionalized mesoporous silicas with different pore diameters
bility, and therefore, in these cases loading of the active
(5.9 nm, and 16 nm). While the smaller pore material mainly
component by adsorption from aqueous solution is typically
adsorbed the smallest protein, the trypsin inhibitor, the three
not an option. Furthermore, as MSNs are inherently
proteins studied were adsorbed by the larger pore material.
biodegradable, silica dissolution during the loading process
The proteins were tightly associated with the support due to
attractive electrostatic interactions, but could be released by under aqueous conditions could lead to unwanted structural
changes of the carrier. Therefore, drug incorporation by
increasing the ionic strength of the solution. All the studies
adsorption from non-aqueous solvents has been widely
discussed above have been conducted using microparticles
applied. In these cases, electrostatic interactions are typically
of mesoporous silica. However, high loading degrees of
less important, and the driving forces for adsorption are
cytochrome C has been also reported for MSNs when the
normally hydrogen-bonding or polar interactions. Also in
adsorption was performed from aqueous solution into MSNs
having pore diameters around 5 nm [70]. A maximum this case, the strength of these interactions can be enhanced
by introducing surface groups onto the mesoporous support
loading of 415 mg cytochrome C/g silica could be achieved,
which have a strong affinity for the adsorbate (drug), like
but at the expense of structural integrity of the MSNs. A
amino groups in the case of adsorption of carboxylic acids.
loading degree of 95 mg cytochrome C/g silica could be
The choice of solvent is mainly determined by the solubility
reached without loss of structural order. No reason for the
and stability of the biologically active component. However,
loss of order at high loading degrees was given, but we
tentatively ascribe it to concurrent hydrolytic degradation of the solvent properties also have a great influence on the
adsorption process itself, as the solvent molecules compete
the MSNs due to silica dissolution, which would be more
for adsorption sites on the silica surface. Water molecules
rapid/pronounced from MSNs than for micron-sized par-
naturally also compete for the adsorption sites under aqueous
ticles.
conditions, but in those cases the electrostatic adsorbate-
3.1.4. Adsorption of Genes adsorbent interactions are typically dominant. In addition,
solvent-adsorbate interactions could play an important role
The adsorption of genes into mesoporous silicas has also for the extent of adsorption, as the driving force for adsorp-
been studied. For example, linear DNA with a green fluore-
tion is lower in good solvents. For example, more than 30
scent protein (GFP) sequence with approximately 760 base-
wt% of the anti-inflammatory agent ibuprofen can be incur-
1172 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

porated into mesoporous silica by adsorption from hexane if process. However, we note that in many studies related to
the pore size is not limiting the extent of adsorption [73, 74] MSNs, amino-functionalized MSNs have been used. CO 2
while the corresponding value is 7 wt % if the adsorption is interacts specifically with amino-groups, why any possible
carried out from methanol [75]. Methanol is a very good alterations of the surface chemistry of amino-functionalized
solvent for ibuprofen, and is also capable of forming MSNs due to CO2 chemisorption to amino-groups should be
hydrogen bonds with the surface silanols. Furthermore, the taken in to the account when using liquid CO2 as the solvent
maximum amount of ibuprofen adsorbed from the aromatic for drug loading into amino-functionalized MSNs.
solvent toluene to bare mesoporous silica is about 0.2
µmol/m2, whereas the corresponding value from the aliphatic 3.1.6. Covalent Linking of Cargo to the Support
solvent hexane is 1.6 µmol/m2 [59]. In aromatic solvents, Covalent linking of the cargo to the mesoporous silica
attractive π-π interactions between the solvent and aromatic carrier is an attractive alternative to physical adsorption,
adsorbents can occur, decreasing the tendency towards especially when the cargo has non-negligible water solubility
adsorption. Probably even more importantly, there are and/or the cargo is highly cytotoxic, in order to prevent
attractive interactions between the π electrons in the aromatic premature leaching of drug. Furthermore, the loading
ring of toluene and the protons in the carboxylic acid group degrees observed for MSNs are often lower than those
as well as in surface silanols, decreasing the tendency obtained for corresponding microparticles. This may be
towards adsorption even further. The solvent-adsorbate related to the fact that large amounts of physically adsorbed
interactions are much weaker and dominated by dispersion cargo may be leached out during the washing step, as for 100
forces in many aliphatic solvents, like n-hexane and cyclo- nm particles about 50 % of the pore volume is contained in
hexane, and the lower achievable loading degree of ibupro- the outermost 10 nm of the particle [80]. There are, however,
fen from toluene as compared to hexane can be therefore not so many studies published to date where covalent linking
understood. It has also been shown that for the adsorption of of the cargo to mesoporous silica has been performed with
ibuprofen from hexane, for supports of a given pore size, the the aim of being able to subsequently release the cargo. This
surface area rather than the pore volume is determining the may be connected to the fact that it in most cases is essential
maximum loading capacity [73]. that the biologically active molecules are reformed into their
Furthermore, a marked influence of the solvent polarity original chemical structure upon detachment. Furthermore,
on the extent of drug adsorption to mesoporous silica has there has to be a mechanism by which the cargo is released
been observed. Kuroda and co-workers studied the adsorp- when the carrier particle has reached the target site. The
tion of the anticancer agent taxol into mesoporous silica covalent linking of ibuprofen to amino-functionalized meso-
from different solvents, and found that this chemotherapy porous silica has been demonstrated, but no release data was
agent was adsorbed from dichloromethane or toluene, but shown [81]. In a recent interesting study by Lin and co-
not from methanol or acetone [76]. This finding was related workers [82], a cysteine residue was covalently attached to
to the different polarities of the solvents, and especially to thiol-functionalized MSNs, which were effectively interna-
the proton-acceptor solubility parameter, which is clearly lized by HeLa cells. Covalent attachment of the cysteine by
lower for toluene and dichloromethane than for methanol S-S-linkage to the silica nanoparticle prevented premature
and acetone. Taxol contains both hydroxyl groups and carbo- release in the extracellular media, but cysteine was released
nyl groups that are possible acceptors for hydrogen bonding. inside the cells due to reduction of the S-S bond. Due to high
Therefore, it has a good affinity with solvents having a high intracellular concentrations of gluthatione, the intracellular
proton acceptor capacity, and attractive solvent-adsorbate redox-potential is much higher compared to the outside
interactions, together with preferential solvent-adsorbent mileu, thereby providing an inbuilt differential release
interactions through hydrogen bond formation with surface mechanism. Another example for successful covalent linking
silanols, decrease the extent of taxol adsorption. of the biologically active agent where subsequent release has
been demonstrated is the covalent linking of the chemother-
In most studies, the adsorption of poorly water-soluble apeutic agent methotrexate (MTX) to amine-functionalized
drugs into mesoporous silica has been carried out using a MSNs [83]. It has previously been shown that MTX does not
fixed high concentration of the drug in solution, as a high adsorb to silica, and that alumina has to be introduced into
drug loading degree has been aimed for. However, some the silica framework to enhance the drug adsorption degree
detailed adsorption studies have been also reported. The [84]. However, covalent linking of MTX to silica was suc-
adsorption isotherms for ibuprofen adsorption to mesoporous cessfully accomplished using standard EDC coupling linking
silicas from n-hexane have been determined. Here, it is a carboxylic acid group present in the MTX molecular struc-
shown that the adsorption can satisfactorily be described ture to amino groups present on the mesoporous silica. The
using the Langmuir adsorption isotherm, suggesting a amino-groups were incorporated either by co-condensation
monolayer adsorption [74]; which is why the adsorption or by surface hyperbranching-polymerization of PEI. Pre-
process is dependent on the surface area rather than the total mature (extracellular) release could be prevented, and the
pore volume, for situations where the pore diameter is not intracellular detachment of MTX from the support in vitro
limiting the extent of adsorption. was mainly ascribed to enzymatic degradation of the amide
The main drawback associated with impregnation from bond in the lysosomes.
organic solvents is the possibility for remaining solvent 3.1.7. Additional Methods for Loading of Cargo
residues after solvent evaporation [77]. Thus, the use of
other fluids, like supercritical [78] or near-critical [79] CO2, Incipient wetness has also been used for the loading of
as the solvent for the biologically active component is a drugs into mesoporous silica [85]. Here, the drug is dis-
promising alternative to organic solvents in the drug loading solved into a suitable solvent and just as much liquid volume
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1173

that can be accommodated in the available pore volume of ponding microparticles. However, the release rate and also
the mesoporous silica support is used in the loading process. the release mechanism are strongly influenced by the mag-
Both itraconazole and ibuprofen were loaded into meso- nitude of attractive cargo-silica interactions. Bergström et al.
porous silica with pore dimensions of 8.4 nm using this [89] studied the release of cationic and anionic dyes from
technique. In comparison with physical adsorption from mesoporous silica spheres into PBS by confocal microscopy
dichloromethane, the incipient wetness impregnation method on single particles, an approach originally developed by the
favored the positioning of itraconazole molecules inside group of Landry for the characterization of differential
intra-wall micropores, while the solvent method favored functionalization of mesoporous silica [90]. For the anionic
their positioning on mesopore walls. Furthermore, the same dye, which does not interact strongly with the silica, the
group also studied the loading of both model drugs from a release was controlled by simple diffusion. The release of the
melt of the drug [85]. While the high viscosity of the cationic dye, on the other hand, followed a distinctively
itraconazole melt prevented loading, ibuprofen was different pattern, as a significant fraction of the remained dye
successfully incorporated at lower loading degrees using this permanently adsorbed onto the pore walls of the negatively
method, and was shown to be preferentially located in intra- charged mesoporous silica spheres, predominantly near the
wall micropores. At higher loading levels, formation of surface of the spheres. This formation of a semipermeable
larger particles of ibuprofen located outside the silica “skin” close to the particle surface led to delayed release of
mesopore system was observed. the dye molecules, and consequently the release event could
not be described by standard diffusion models. Although the
3.1.8. Concurrent Loading of Several Active Cargos results were obtained for spherical mesoporous particles
It is also possible to combine the means described above several micrometers in diameter and the effects may be
for the loading of two or more biologically active com- smaller for MSNs, these interesting results highlight the
ponents into MSNs. This is an extremely important feature, interdependency between the adsorbate-adsorbent interact-
as efficient next-generation cancer therapies based on che- tions and the release profiles. Furthermore, the pore size and
motherapy in combination with controlled release systems number of available adsorption sites with respect to the
will most likely involve the co-delivery of two or more molecular size of the cargo has been recently shown to
active agents. The most straightforward means is, if possible, define the relative influence of attractive adsorbent-adsorbate
to adsorb several active components in parallel, as recently interactions, and also on the subsequent release kinetics.
demonstrated for the loading of two different hydrophobic Bräuchle et al. [91] studied the mobility of the DOX inside
model drugs from cyclohexane [86]. In another recent study, dried, mesoscopically ordered silica-surfactant films, and
the antitumor drug doxorobucin (DOX) was first adsorbed found that this anticancer drug was immobile in the
onto cyanato-functionalized MSNs from a methanol/water cylindrical channels of films templated by the non-ionic
mixed solvent, after which a second generation polyamido- surfactant brij® 56, while it was highly mobile inside corres-
amine dendrimer was covalently attached to the outer surface ponding channels templated by the relatively bulky non-
of the MSN particles by urea linkage to cap the pore open- ionic surfactant pluronic® P-123. Furthermore, mobility was
ings [87]. A DOX loading as high as 40 wt % was obtained. also observed inside films templated by the smaller cationic
siRNA was then adsorbed to the dendrimer-functionalized surfactant cetyltrimethylammonium bromide (CTAB) which
outer surface of the DOX-loaded MSN particles. In another has a molecular dimension comparable to that of brij® 56.
recent study, Lin and co-workers first synthesized boronic These findings were attributed to the blocking of silanol sur-
acid-functionalized MSNs, after which cyclic adenosine face groups by the cationic surfactant, hindering hydrogen-
monophosphate (AMP) was adsorbed into the mesopores bonding between oxygen atoms present in the molecular
from phosphate buffered saline (PBS) [88]. Gluconic acid- structure of DOX and the silica surface, while free silanols
modified insulin was subsequently covalently linked to the were present in the films templated by the non-ionic
boronic acid groups present on the outer surface of the surfactants. The difference between the DOX mobility inside
MSNs. The loadings of cyclic AMP and gluconic acid- the pluronic® P-123 and Brij® 56 templated films, on the
functionalized insulin were reported to be 27 and 64 µmol/g, other hand, was attributed to a reduced influence of
respectively. hydrogen bonding in larger channels. The level of DOX
mobility was directly reflected in the rate of diffusion-
3.2. Cargo Release from Mesoporous Silica controlled DOX release from the films.
3.2.1. Non-functionalized Mesoporous Silica: Simple However, in many cases a slower drug release than
(Matrix) Diffusion expected for first-order release kinetics is observed at longer
times, even in the absence of strong attractive adsorbate-
In most cases, the release of small-molecular cargo incur- silica interactions. This has been attributed to differences in
porated into the pores of mesoporous silica is diffusion- cargo-support interactions between the inner and outer
controlled and follows first order kinetics in the absence of portion of the surface. Other suggested reasons for this
strong adsorbate-adsorbent interactions. Important parame- observation has been the deposition of hydroxyapatite
ters influencing the release rate are the effective pore size, crystals onto the silica surface leading to partial pore
the polarity of the pore system, the water solubility of the blocking, when ibuprofen release from mesoporous silica
cargo, and state of the cargo in the pores (amorphous or was studied in a simulated body fluid [92] as well as for
crystalline) which also has an influence on the cargo empty MSNs [93], in agreement with previous considera-
dissolution kinetics. Furthermore, the particle size will have tions. However, recently an alternative explanation was
an important influence on the release rate, and faster overall brought forward to account for the clearly slower ibuprofen
release rates can be expected from MSNs than for corres- release from MSNs at longer times. It is based on silica
1174 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

Fig. (2). Release of ibuprofen from pristine (non-functionalized) mesoporous silica (left) in simulated body fluid (SBF) along with its square-
root of time plot (right), indicating a diffusion-controlled release process. (From: Rosenholm JM. Modular design of mesoporous silica
materials: towards multifunctional drug delivery systems. DSc(Tech) thesis. Turku: Abo Akademi University 2008).

dissolution and preferential re-deposition close to the pore tration-dependent effective diffusion constants. In another
entrances leading to partial pore blocking [94]. Indeed, silica recent study, the release of the antibiotic vancomycin from
does dissolve under physiological conditions, and this is a mesoporous silicas with different pore diameters was studied
much wanted property of the material in drug delivery [96]. For the pore size range studied, 4-9 nm, variations in
applications using MSNs. However, this is often overlooked the pore diameter had little effect on the drug release rate.
in release studies and typically the silica dissolution kinetics However, the pore morphology and the effective length of
is not reported. The dissolution kinetics may be different for the pores, which naturally is directly related to the diffu-
silicas prepared under different experimental conditions, and sional path length, had clear influences on vancomycin
is especially dependent on if the silica has been thermally release rate, as expected for a diffusion-controlled release.
treated or not, as the degree of silica condensation is en- Again, these results highlight that the release rate of macro-
hanced by thermal treatment. The dissolution-reprecipitation molecules from MSNs may be faster than that of corres-
mechanism is supported by the fact that a marked decrease in ponding microparticles. This observation is also perfectly in
pore volume was observed, although the mesostructure line with diffusion theory, which furthermore is charac-
appeared intact as determined by X-ray diffraction (XRD) terized by a fast release in the beginning with gradually
for particles with a fairly broad size-distribution of 200-800 (square-root of time dependent) decreasing rate of diffusion
nm. One may expect that this is another parameter that leads (release), see Fig. (2). As diffusion-controlled systems are
to faster kinetics from smaller particles than from larger furthermore often accompanied by an initial “burst” release,
particles with otherwise similar structural and surface this has raised the need for means to minimize early release
chemical properties. of the active component from MSNs, and in the best case
completely hinder a pre-mature release before the cargo has
The molecular-level interactions influencing the release
reached its site of action. This will be discussed in the next
of macromolecular cargo from mesoporous silica is naturally
more complex due to the presence of a multitude of potential section.
binding sites and also due to the possibility for molecular 3.2.2. Stimuli-Responsive Release Systems Based on MSNs
weight- or loading degree-dependent conformational and
polarity differences. Very few mechanistic studies related to The value of having the most selective targeting system
the release of macromolecular cargo from mesoporous silicas is of course lost if the cargo is released from the carrier
have been reported. However, in a recent study, the release before it reaches the target site. For chemotherapy applica-
of fluorescently labeled dextrans, a commonly used model tions it is desirable that no release of cargo should occur in
for proteins, from microparticles of mesoporous silica with a the extracellular environment, while a fast release of the
pore size of 6.5 nm was described [95]. Two dextrans with cargo should occur once the carrier particle has reached its
different molecular weights, 3 and 10 kD, were used. As point of action. As discussed above, the release of cargo
expected, the release rate of the high-molecular weight from mesoporous silica is often diffusion-controlled and
dextran was slower than that of the low-molecular weight typically associated with a pronounced initial burst. There-
dextran, but the release curves had a similar shape fore, there is a strong current interest in the development of
suggesting that the release mechanism was similar in both means to prevent a fast initial release of the cargo, which in
cases. It was concluded that dextran release was not dif- the optimal case would be sensitive to the local environment
fusion-limited. The release followed a logarithmic time of the carrier particles in a way that release of the cargo is
dependency normally observed for chemisorption events, only triggered inside the cells or within specific cellular
and the dextran release process was discussed in terms of compartments after internalization. In addition to the
parallel adsorption and desorption events coupled to concen- reversible covalent linking of drug to the mesopore surface
already discussed above, many other inherent or stimuli-
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1175

responsive systems have been reported. The development in system is the adsorption of poly(dimethyldiallylammonium
this field up to 2009 was recently reviewed [97, 98], and in chloride) to carboxylic acid-modified mesoporous silica
the following we will therefore only give some selected [104]. Vancomycin was used as the model drug, and
examples of stimuli-responsive systems which could operate pronounced release of entrapped vancomycin was observed
under biologically relevant conditions. These can be divided at pHs ≤ 4.5, while little drug release was observed at pH
into two main classes; a) systems which can be triggered by 6.5. On a related note, chitosan has been also shown to pro-
external stimuli, such as ultraviolet light, and b) systems vide pH-sensitive molecular gate properties, as demonstrated
which should react to the differences in chemical conditions by the release of insulin from mesoporous silica/silicon films
between the extra-cellular and intra-cellular environments, [105]. Zero-release was observed under pH-neutral con-
such as differences in pH, redox potential, or enzymatic ditions, while insulin was released at pH 6 due to swelling of
activity. We will mainly focus on systems related to the the chitosan hydrogel. Furthermore, a pH-dependency was
latter class in the following. recently demonstrated using a molecular machine-concept
based on [2] pseudorotaxane which was covalently linked to
3.2.2.1. pH-Responsive Systems MSNs. A model fluorescent dye remained inside the
The means of MSN administration determines which mesopores under pH-neutral conditions, while a release of
type of functionality is needed. For example, for pH-res- cargo was observed upon decreasing the pH below 5, which
ponsive systems, the desired pH-response is in most cases is close to the pH in lysosomes [106]. Another example of
opposite for oral and intravenous administration. In order to corresponding pH-responsive systems is gold nanoparticle-
prevent premature release in the gastric tract, the mesopores capped mesoporous silica, where the gold nanoparticles were
should remain closed at under acidic conditions and open covalently linked to the outer surface of the mesoporous
under more pH neutral conditions, for instance by some gate- silica through an acid-labile acetal linker [107]. Zero-release
keeping moieties. Such a pH-response has been demons- of a dye used as the model cargo was observed under neutral
trated for MSNs onto which polyamines in the form of conditions, while the dye diffused out of the pores upon
silanes were covalently linked to act as gate-keepers [99]. lowering of the pH. Almost complete release of the cargo
The mesopores remained closed under acidic conditions due was observed within 10 hours at a pH of 4.0.
to the “swollen” conformation of the polyamines when
3.2.2.2. Redox-Sensitive Systems
charged, thus hindering the release of the cargo; but release
of vitamin B was observed under pH neutral conditions to an Another often used means for stimuli-responsive release
extent which was dependent on the ionic composition of the of cargo from MSNs is to use diffusion barriers linked to the
medium [100]. Both pH-induced changes in the extent of silica by redox sensitive bonds, typically S-S bonds, as it is
hydrogen-bonding interaction between amines (neutral pH, generally accepted that the redox potential as much lower in
open gate) and Coulombic repulsions (acidic pH, closed intracellular compartments than in the extracellular milieu.
gate) and the formation of anion-amine complexes were In this case, the stimuli-response under biological conditions
suggested to account for this observation. Also electrostatic is built upon linker bond-cleavage by cell-produced reducing
assembly of poly(acrylic acid) to amino-functionalized me- agents. The first demonstration of redox-sensitive stimuli-
soporous silica has been shown to effectively entrap proteins responsive cargo release based on MSNs was the capping of
under acidic conditions, while about 35 % of the encap- the mesopores by CdS nanoparticles, where the nanoparticles
sulated protein was released at pH 7.4 within 10 h [101]. were anchored to the silica surface by a silane containing an
Another example of a polymer-based coating showing a S-S bond in its structure [108]. As a demonstrator,
corresponding pH-response is the pH-dependent release of vancomycin- and adenosine triphosphate (ATP) was loaded
ibuprofen from mesoporous silica spheres coated with a co- into the mesopores followed by CdS capping. Release of
polymer layer consisting of poly(methacrylic acid-co-vinyl cargo occurred upon reduction of the S-S bond. Similar
triethoxylsilane) [102]. Here, the silane function links the results were later reported using other nanoparticles for
polymer covalently to the silica, while the pH-dependent capping of the mesopores, like gold [109, 110] and iron
protonation of the carboxylic acid groups in the metharylate oxide [111]. Other redox-responsive systems utilizing the
control the degree of polymer swelling. Recently, a MSN redox sensitivity of the S-S bond include the grafting of a
system with charge-convertible, pH-hydrolyzable groups on disilane-disulfide compound preferentially to the outer sur-
the pore walls was also reported [103]. The employed face of mesoporous silica [112] or cross-linking of a poly(N-
citraconic amide functionality possesses a negative charge at acryloxysuccinimide)-grafted mesoporous silica using a
pH 7.4 (extracellular conditions) but undergoes charge con- disulfide-based bifunctional primary amine [113]. Layer-by-
version at acidic pH 5 (endosomal conditions). The feasi- layer assembly of oppositely charged polyelectrolyte
bility of this mechanism was demonstrated by release of multilayers followed by S-S crosslinking has been also
cytochrome C, which was more pronounced at pH 5 due to recently demonstrated to show redox-triggerable molecular
the proteins effective positive charge at this pH. gate properties [114].
On the other hand, for intravenous administration, which 3.2.2.3. Stimuli-Responsive Systems Based on Enzymatic
is likely to be the most frequently used administration route Degradation
for MSN-based chemotherapy the pores should preferentially
remain closed under the neutral pH conditions prevailing in Enzymatic degradation is a very attractive route towards
the blood stream, while the pores should be open to the promoting intracellular release, albeit to date only demons-
outside under the more acidic pH conditions prevailing in trated under “in sink” conditions. The first demonstration of
tumor sites and intracellular compartments like endosomes an enzyme-triggered MSN-based release system was based
and lysosomes. One early example of such a pH-responsive on the attachment of a 2-rotaxane onto the silica surface and
1176 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

an ester-linked α-cyclodextrine stopper [115]. The ester bond transfected into HeLa and Chinese hamster ovarian (CHO)
could then be cleaved by the use of porcine liver esterase, cells [121]. The positive charge was provided by second
which led to release of entrapped dye from the mesopores. generation polyamidoamine (G2-PAMAM) dendrimers caps,
On a related note, α-cyclodextrine was also detached by α- which thus simultaneously acted as gatekeepers for the
amylase-induced hydrolysis of the ester bond, leading to fast loaded Texas red dye. Even though in this case the dye was
release of cargo [116]. Another demonstration of an enzyme- only used to make the MSNs fluorescently visible and hence
responsive triggered release system is the attachment of no release was demonstrated, this system constituted an early
amidine-units to biotinyl-functionalized MSNs [117]. In this MSN model for a double-delivery system able of simulta-
case, the release of a dye used as the model cargo could be neously delivering drugs and genes intracellularly. Later, the
induced by addition of trypsin. Complete release of the dye same group demonstrated successful delivery of both DNA
was reached in 3 h. In another interesting development, and chemicals into plant cells using MSNs [110]. A similar
enzyme-triggered release has been also demonstrated for approach was recently demonstrated by Chen et al. [87],
lactose-functionalized MSNs, where the lactose was cova- where they fabricated MSNs encapsulated with DOX inside
lently linked to the outer surface of the MSNs. Complete the pores, which in turn were also capped with G2-PAMAM
release of cargo occurred within 3 h in the presence of β-D- dendrimers but complexed with siRNAs targeted against
galactosidase under physiological conditions. It is clear that mRNA encoding Bcl-2 protein, which is responsible for non-
enzyme-triggered release is a very promising route for site- pump multidrug resistance in cancer cells. In this case also
specific drug delivery of anticancer drugs, and one can DOX release was observed under sink conditions in a
anticipate rapid development in this field during the next glutathione solution, whereas release in PBS was almost
years. negligible; and the cytotoxic activity of loaded DOX was
also demonstrated in vitro on human ovarian A2780/AD
3.3. Additional Selected Diffusion Barrier Concepts cells.
In a recent development, stimuli-responsive capping of Other modifications used to enhance the transfection
the pores of mesoporous silica was demonstrated (under sink efficiency of MSNs, as they are not as such effective in
conditions) using an antibody as the capping agent [118]. It delivering nucleic acids, have included both modified
was shown that addition of an antigen (complementary to the (mannosylated [122]) and non-modified PEI [72, 123]. PEI
antibody) induces the release of the caps, which resulted in a is a cationic polymer exhibiting the highest positive charge
clearly faster release of the entrapped cargo. Although the density when fully protonated in aqueous solution and is thus
capping did not appear to be as tight as in many of the ideal to form nanocomplexes with negatively charged
systems discussed above, the concept is very exciting, as it nucleic acids. Additionally, its success in gene delivery lies
can allow more specific biological release systems to be in its buffering capacity at pH 4-6 which consequently leads
developed. to its characteristic ability to destabilize lysosomal mem-
branes and thus promote endosomal escape of the genes
Another very interesting concept with high biological [120]. This property can also lead to cytotoxicity in high
potential relies on “protocells”, i.e. MSN supported lipid doses, but when coated onto MSNs, this cytotoxicity can be
bilayers [119]. The protocells were prepared by fusion of circumvented while the high transfection efficiency of PEI
positively charged liposomes onto negatively charged can be retained. Moreover, this cationic modification of the
MSNs. The lipid bilayer served as a diffusion barrier for MSNs was shown to enhance the cellular uptake of MSNs,
encapsulated cargo, as demonstrated for capsin. The lipo- in line with the common notion that cationic nanoparticles
some fusion process could be also used to load MSNs with are internalized more effectively than negatively charged
cargo, which in other cases typically is carried out in a (connected to the fact that cellular membranes are negatively
separate step. Thus, this approach has the advantage of being charged). This is in accordance with the observation of the
fairly straightforward in that the number of particle uptake of amino-functionalized co-condensed MSNs as
preparation steps is low, and also in that the MSN surface is compared to MSNs functionalized by surface-hyperbranch-
inherently biocompatible. Furthermore, it would probably be ing polymerization of PEI, where negligible uptake was seen
also possible to introduce targeting ligands at the same time, after 2 h incubation with HeLa cells of the co-condensed
which would make such particles highly interesting for drug MSNs while considerable uptake was seen for the PEI-
delivery applications. The capability of the particles to MSNs at the same time point [86]. Also for G2-PAMAM
release entrapped cargo inside cells was demonstrated in dendrimers functionalized core-shell magnetite-MSNs the
vitro using calcein as the model drug, and the release was uptake in glioma cells was more efficient for G2-PAMAM-
suggested to be pH-triggered in nature. MSNs (at both 1 h and 4 h) than when these were esterified
to yield a lower surface charge of the particles [124].
3.4. Gene Delivery Systems
Since nucleic acids alone are not able to penetrate the cell 3.5. MSN-Based Imaging Systems
wall, the therapeutic success of gene therapy is largely Of the bioimagible MSN systems presented in literature,
dependent on the development of efficient delivery systems multifunctional SPIO-based nanodevices are of particular
for DNA, which is typically condensed into nanoparticles by interest for cancer diagnosis and therapy [125] as diagnostic
a cationic polymer to produce non-viral gene delivery sys- tools, e.g., for early tumor detection. Other applications that
tems [120]. There have been also some successful attempts would greatly benefit of efficient in vivo imaging contrast
in using MSNs as gene delivery systems. For instance, agents include cellular tracking. Methods that monitor the
cationically modified MSNs were shown to be able to fate and distribution of transplanted stem cells non-inva-
complex with plasmid DNA (pEGFP-C1) and successfully
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1177

sively are for instance a key issue for developing successful potentially be used to carry therapeutic cargo – carrying
stem cell therapies [126]. MRI is an ideal imaging modality MSNs into the burgeoning field of theragnostics.
for the biodistribution of magnetically labeled cells, whereby
in the case of MSNs, either SPIO nanoparticles [34, 127-
129] or paramagnetic gadolinium (Gd)-complexes [130-133] 4. MSNS AS THERAGNOSTIC AGENTS IN CANCER
can be encapsulated into the particles to render them MRI 4.1. MSNs in Cancer Medicine
active. Optical imaging techniques can be also utilized,
whereby the MSNs should comprise optically active func- Successful combat against cancer requires: 1) early
tions such as fluorescent dyes [134, 135] or near infra-red detection of cancer or malignant lesions; 2) identification of
(NIR) contrast agents [136]; as well as combinations of the biomarkers and oncogenic signals fueling cancer prog-
optical and magnetic properties to provide multimodal ression for the choice of therapy; and, 3) efficient delivery of
imaging [23, 125-128, 130, 137, 138]. While fluorescence is the drug(s) of choice to the lesion to enhance therapeutic
an invaluable tool for the in vitro biological evaluation of efficacy and reduce unwanted side effects. MSNs meet these
MSNs, the in vivo applicability of fluorescent (only) MSNs demands by their ability to carry different types of
is limited. A prerequisite for successful in vivo imaging therapeutic and diagnostic entities, even simultaneously,
include a stable signal and in the case of cellular tracking, their high drug load capacity, the possibility for tunable
that the contrast agents (in this case, the MSNs) remain load/release mechanisms and their chemical flexibility,
internalized long enough to be monitored. On this topic, allowing for modifications of size, charge and surface func-
Mou and co-workers have investigated the influence of both tionalizations which determine their stability, pharmaco-
surface charge [139] and size [140] on as well as the route kinetics, and biodistribution (see Fig. 3). In addition to
[133] of the cellular uptake of MSNs. On the MRI side, improved therapy, this nanoplatform will offer diagnostic
incorporating contrast agents into MSNs benefits in the sense benefits through targeted delivery of imaging agents for
of extended half-lives (in the case of Gd-based contrast enhanced signal detection. Further, an interesting application
agents) and enhanced cellular uptake/ labeling efficiency (in is the possibility to utilize MSNs for targeted delivery of
the case of SPION) provided by the silica layer, which signaling/biomarker sensors to enhance the detection of the
furthermore can be easily further functionalized as well as normally weak oncogenic bioactivity. As this drug delivery
platform can carry multiple entities they provide excellent

Fig. (3). Schematic presentation of the topics of this review addressing the aims of nanomedicine for improved cancer therapy and diagnosis
(blue arrows), the specific characteristics of mesoporous silica particles that make them especially suitable as tools to meet these aims (red
arrow to the right) and the existing challenges (red arrow to the left), when it comes to detailed understanding of the biobehavior of the drug
delivery platform that need to be attended, before this system can be considered for clinical application.
1178 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

tools for multidrug delivery and combined diagnostic and MSNs on isolated cells. Several in vitro studies demonstrate
therapeutic action (theragnostics). Despite recent advances that differently modified MSNs in the size range of 100-200
highlighting MSNs as promising drug delivery platforms, the nm are generally well tolerated by several cell types at
development of MSNs for cancer therapeutics and diag- different concentrations (typically up to 0.1 mg/mL) [141-
nostics is still at an early stage and MSNs for targeted cancer 144], whereas unmodified MSNs show higher toxicity [145].
therapy has yet to enter clinical trials. The great challenge for The influence of size, shape, and surface-charge and -
translation of this technology into medical applications is to functionalizations on cellular toxicity has been discussed by
understand the biological behavior of MSNs. the authors in a recent review [146]. As this section deals
with medical applications we will concentrate on the in vivo
Biology presents complex problems for material develop-
demonstrations of biocompatibility and the readers are
ment. Particle size, charge and surface functionalizations are
critical parameters that clearly influence the biodistribution referred to the other review for more detailed information on
how particle characteristics influence cellular toxicity. In line
and pharmacokinetics of drug delivery systems. Upon
with several in vitro investigations, a recent study showed
administration and before reaching the site of action the drug
that non-functionalized mesoporous silica materials of
delivery system will encounter a multitude of biomolecules,
different size where lethal when injected intravenously and
cellular components and biological membranes. The system
intraperitoneally, whereas no lethality was observed when
needs to stay intact, preferentially exhibit zero premature
release of the carried therapeutics, and show minimal the particles where injected subcutaneously [147]. Post-
mortem analyses indicated that death might have been due to
interaction with and impact on the functions of biological
thrombosis. Although subcutaneous injections did not lead to
systems other than the targeted tissues. Interactions with
death, they did cause severe systemic toxicity. The authors
healthy cells may disturb their normal functions and lead to
concluded that the toxicity might be mitigated by modifica-
vehicle-induced side effects. Protein adsorption to the drug
tions of the materials. Indeed, several in vivo studies using
carriers may affect recognition of the particles by the
immune system and influence particle life time in the functionalized MSNs have revealed no signs of acute toxi-
city in animals [147-149]. Further, a recent report demons-
circulation and toxicity. Protein coverage of the particle may
trated lack of acute toxicity within 6 h after intravenous
also cover active targeting molecules on the surface and
injections of positively charged mesoporous silica nano-
hinder recognition. The clearance mechanisms for each drug
particles in the size range of 50-100 nm [148]. These part-
delivery system need to be clarified to avoid problems due to
icles were shown to be targeted to the liver but the prolonged
long term accumulation in healthy organs. Detailed under-
standing on how modifications/functionalizations influence effect of particle administration was not addressed in this
study. Non-porous silica particles (mean size: 50-200 nm)
the degradation rate of the particles will be necessary to
were recently shown to undergo renal and hepatic clearance
optimize the life time of the drug delivery systems according
(urine and bile) over time in a size dependent manner [148].
to the application. The rate of biodegradation, necessary for
Smaller particles were preferably cleared, whereas larger
the elimination from the body, should be matched to the
particles were trapped by macrophages and accumulated in
circulation time needed for the drug delivery vehicle to carry
out its function. Longer circulation times will facilitate the liver and spleen where they remained up to 4 weeks after
injection [148]. Subsequent imaging of the major organs also
efficient accumulation in the target tissues whereas a more
further supported that the particles mainly accumulated in
rapid degradation will be beneficial for diagnostic action, as
the liver, even though some particles were observed also in
that will reduce unspecific background signal from non-
kidneys, lung, and spleen.
targeted tissue. Biodistribution of delivery vehicle upon
intravenous administration will be also affected by the Although the studies described above did not address
vascular integrity and the degree of vascularization of organs long term toxicity of accumulated particles and detailed
and tumors in particular. Active targeting to tumors requires understanding on how size, charge and surface modifications
detailed knowledge of the protein make-up of the cancer of MSNs influence in vivo toxicity and biodistribution is still
cells and the molecular function of the targeted molecule. lacking, they suggest that these drug delivery platforms
Due to the heterogeneous nature of cancer, it is difficult to exhibit sufficient biocompatibility to be suited for
envision the discovery of a universal targeting ligand but biomedical applications.
rather the optimal delivery vehicle will need to be a generic
system where ligands can be easily interchanged, based on 4.3. Diagnostic Applications
"targeting diagnostics" of each tumor. Detailed under-
standing on how size, charge and different surface modifica- Medical imaging rely on positron emission tomography
tions influence interactions with the immune surveillance (PET) and MRI. The latter is a highly sensitive technique for
system, molecular and cellular functions of interacting the detection of ultrastructural changes, whereas PET offers
tissues, biodistribution and clearance from the body is the detection of a biological activity. Early detection of the
needed before the particles can advance to the clinic. Below lesion is important for a more successful treatment. The
we discuss recent advances within MSNs research and how biological signal in early lesions is normally weak and small
MSNs can meet the above mentioned challenges for tumors might go undetected due to lack of specificity or poor
successful application in cancer medicine. sensitivity. Hence delivery of imaging agents to the site of
the lesion to enhance the signal and increase specificity is of
key interest. MSNs are flexible imaging agents that can be
4.2. Biocompatibility of MSNs
conjugated to imaging agents suitable for PET, MRI and
Safety and toxicity of the drug delivery vehicle is of fluorescence imaging (see above). Although extremely
central importance and has been addressed primarily for useful for quantification of biological behavior in vitro
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1179

[150], fluorescent dyes are not the preferred option for was an ideal platform for efficient MRI due to their ability to
clinical imaging due to problems of tissue autofluorescence carry large payloads of Gd centers, along with a good water
and deep tissue photon penetration. accessibility of the Gd-chelates inside the porous matrix.
Carniato et al. [133] carried out an investigation of pore size
4.3.1. Magnetic Resonance Imaging dependence (MCM-41, dp = 33 Å vs. SBA-15, dp = 82 Å) on
On the scarcity of in vivo studies on MSNs, most can be the water accessibility in terms of their 1H nuclear magnetic
found within papers dealing with imaging. Thus, most in resonance (NMR) relaxometric properties as a function of
vivo MSN evaluations rely on MRI techniques, often applied temperature and magnetic field strength. They found a 4-fold
for core-shell type particles consisting of a magnetic core difference in the relaxivity values by the different accessi-
and a mesoporous shell. For instance, Mou and co-workers bility of water molecules to Gd centers localized inside or
investigated the long-term applicability/contrast enhance- outside the pores. Thus, the Gd-complexes grafted outside
ment of their developed tumberlike so-called Mag- the pores, i.e. mostly on the particle surface, contributed to
Dye@MSNs [123], with both optical (fluorescent) and the observed relaxivity to the largest extent, an effect which
magnetic properties, both after intravenous administration was especially pronounced for the MCM-41 structure for
via eye vein injection [125], and implantation of Mag- which accessibility of the Gd-centers inside the pores was
Dye@MSN-labeled human mesenchymal stem cells more limited. However, when comparing Gd-labeled MCM-
(hMSCs) at the olfactory cortex of the brain [124] by MRI. 41 to the commercial contrast agent ProHance®, the former
In the previous case, the Mag-Dye@MSNs were found to phantoms (3.0 T) showed a remarkable 1H R1 enhancement
accumulate into organs belonging to the reticuloendothelial at increasing Gd concentrations compared to ProHance®
system (RES), mainly spleen and liver. They were able to phantoms. Also in vivo, Taylor et al. [131] found that much
monitor the liver for 3 months, whereby it was found that the smaller doses of their Gd-MSNs were needed as compared to
signal-to-noise ratio recovered after 90 days, indicating that typical Gd doses for a significant T1-weighted enhancement
the Mag-Dye@MSNs were resistant to decomposition and (9.4 T) of the aorta of a DBA/1J mouse 15 min post-injection
not easily excreted from the body, and thus a potential into the tail vein. They concluded that Gd-MSNs could be
candidate for long-term liver/spleen MRI monitoring [125]. suitable for intravascular MRI. In line with the findings of
In the latter case, the animal was imaged by MRI 9 days Mou and co-workers described above, after injecting higher
after hMSC implantation, i.e. after long-term growth and doses into the tail vein, a significant loss of MR signal was
differentiation, and the Mag-Dye@MSN-labeled hMSCs observed in the liver 1 h post-injection. This indicated that
were still visualized [124]. Moreover, in this study, the Gd-labeled MSNs could be used as contrast enhancers also
labeled cells can be visualized in a clinical 1.5 T MRI system for T2-weighted MR images. They postulated that liver
with an in vitro minimal detectable cell number of about signal loss was a result of phagocytosis of the particles by
104, and an in vivo detection quantity of 1.25 × 105 cells, liver macrophage cells, which they demonstrated in vitro
whereas in the former study a 7 T MRI was used for animal with related monocyte cells in the same study [131].
imaging. Hyeon and co-workers also applied dually active
4.3.2. Biosensing
(fluorescent and magnetic) PEGylated MSNs and examined
their passive tumor accumulation after intravenous injection Detection of biological activity is an attractive diagnostic
as well as visibility after subcutaneous injections of MCF-7 application [149, 150]. The large surface area of MSNs
cells labeled with Fe3O4@mSiO2 [34]. After 2 and up to 24 h allowing for conjugation with a high concentration of sen-
post-injection, the accumulation of the MSNs was detected sing molecules, and the optical transparency which permits
in the T2-weighted MR images. Recently, the same group high detection rate of the signal, render MSNs highly suita-
repeated a similar protocol but based on a MSN system with ble for biosensing. Drugs, enzymes, antibodies, and nucleo-
surface-bound Fe3O4 nanocrystals, also rendering them tides, all suitable for MSNs incorporation, can serve as
magnetically active for MRI [34]. Also these MSNs were recognition systems in biosensing applications. Although not
passively targeted to shoulder-implanted MCF-7 tumors. demonstrated for in vivo activity, MSNs have been
Thus, they also attempted to deliver DOX to the tumor sites successfully synthesized for the detection of glucose [151],
with the aid of the MSNs as delivery vehicles, and were H2O2 [152], and NO2 [153]. An example of a molecular
indeed able to observe DOX-originating fluorescence as well recognition system capable of distinguishing between struct-
as apoptosis in the harvested tumor cells 48 h post-injection urally similar neurotransmitters utilizing different functiona-
sacrifice. However, since no free DOX was used as control, lizations of the exterior and the interior particle surfaces was
it remains slightly unclear if drug delivery was in fact shown by Radu et al. [154]. The functionalizations affected
particle-mediated or due to free (leached) drug. However, the diffusion rate, and hence detection of the analytes into
this study constitutes a promising step towards the use of the particles based on their charge. Martínez and co-workers
MSNs as theragnostic agents. developed a MSN based system for the recognition of anions
MSNs have also shown to be feasible candidates as T1 that were capable to respond to different degrees to AMP,
adenosine diphosphate (ADP), and ATP [155]. Although the
contrast agents. For instance, Mou and co-workers have
above described systems have been not applied for cancer
fabricated MSNs tagged with both the FITC fluorophore and
diagnostics, they demonstrate the flexibility of the tech-
a paramagnetic T1 contrast agent, Gd-DTPA, and implanted
nology platform and the potential for MSNs in biosensing.
Gd-Dye@MSN-labeled hMSCs into the basal ganglions of
MSN-mediated delivery of signaling sensors or antibodies
nude mice, which could be visualized under a clinical 1.5 T
MR system even 14 days post-implantation [130]. Likewise, recognizing specific biomarkers for cancer might provide an
additional value in the diagnosis of the oncogenic activity,
Taylor et al. [131] grafted MSNs with a Gd-DTTA (diethyl-
and facilitate the choice of therapeutic modality.
enetriaminetetraacetic acid) chelate. It was found that MSNs
1180 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

4.4. Therapeutic Applications drug resistance include induction of cellular repair systems,
reduced apoptotic sensitivity, and/or increasing drug efflux
4.4.1. Enhanced Therapeutic Efficacy through Efficient
in tumor cells. The latter, also denoted as pump mechanisms
Drug Delivery
of drug resistance, is mediated by membrane glycoproteins
The therapeutic window of many drugs is very narrow that function as drug transporters. Nanoparticles can bypass
due to frequent occurrence of serious side effects, arising pump mechanisms by efficient intracellular drug delivery
from a non-specific drug uptake by healthy cells. Further which allows for obtaining high intracellular drug concentra-
therapeutic efficacy of many drugs is hampered by low tions, counteracting the activity of multidrug resistance
active concentrations due to poor drug solubility, drug pumps [149]. Other cellular resistance mechanisms can be
instability, and poor cellular uptake of the active agent. overcome by specific targeting of the responsible molecules.
Therefore, the development of delivery systems that can Such an approach was recently demonstrated by Chen et al.
carry a high drug payload, protect the drug from degradation, [87]. They used DOX-loaded MSNs carrying siRNAs tar-
target specific cell populations, and facilitate cellular uptake geted against the antiapoptotic Bcl-2 protein. The MSN
of the active agent is necessary for clinical applicability of complex was delivered into multidrug-resistant A2780/AD
many drugs. Mesoporous silica structures has been utilized human ovarian cancer cells to induce significantly enhanced
for intracellular delivery of both hydrophobic anticancer cell death, compared to free DOX. They attributed this effect
agents such as camptothecin [135, 156] and paclitaxel [70, to the knockdown silencing of Bcl-2 mRNA, leading to
157, 158], and the more hydrophilic DOX [34, 120]. Due to enhanced cytotoxicity of the drug. Recently, it has been
the enhanced cellular uptake of MSNs (and hence the more reported the successful delivery of a siRNA construct cap-
efficient drug delivery), the intracellular drug concentrations able of knocking down GFP expression on PANC-1, BxPC3,
and the subsequent biological effects were enhanced. Such and HEPA-1 cell lines [87]. Moreover, thanks to the en-
enhanced and furthermore selective drug action by MSN- hanced cellular uptake of the cationically modified MSNs,
mediated drug delivery was also recently shown, where the the delivery of paclitaxel to the pancreatic cancer cell line
level of apoptosis induced by MTX-conjugated nanoparticles (PANC-1) was increased. This study postulated the simulta-
on cancer cells was significantly higher than that of corres- neous delivery of siRNA (to knock down the expression of
ponding amounts of free drug, whereas the non-cancerous the P-glycoprotein drug exporter) and chemotherapy agents
cell line remained unaffected. Additionally, successful deliv- that are actively exported by this glycoprotein, as an
ery of nucleic acids [70, 117, 121, 159] highlights the effective strategy in the treatment of drug resistant cancers
potential of MSNs in gene therapy. The high drug load based on pump mechanisms. Thus, the multimodality of
capacity and flexible drug coupling chemistry of MSNs also MSNs allows for intelligent design in providing synergistic
permits multidrug delivery, which is a critical issue in cancer capabilities, which could be also utilized to overcome both
therapy as tumors exhibit a complex cross talk between pump and non-pump multidrug resistances in cancer cells
oncogenic signals and often develop resistance to a given (see Fig. 4).
drug. Rosenholm et al. recently showed successful co- Poor vascularizations that restrict access of the drug
delivery of two hydrophobic model drugs, the fluorescent delivery system to the tumor site, high interstitial pressure
molecules 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbo- and low microvascular pressure counteracting extravasation
cyanine perchlorate (DiI, red color), and 3,3'-dioctadecyloxa- of the drug carrier from the tumor mass comprise cell
carbocyanine perchlorate (DiO, green color), which were extrinsic mechanisms of drug resistance. Such mechanisms
simultaneously loaded into the pore network, further are harder to overcome especially for larger vehicles. They
emphasizing the potential for multidrug delivery [86]. may require local instead of intravenous administration,
Despite the large number of recent reports demonstrating combined with mechanisms that enhance cellular interact-
the capability of MSNs to deliver drugs under in vitro con- tions and uptake.
ditions, the therapeutic efficiency of drug-containing MSNs
4.4.3. Targeted Therapy
has been not demonstrated under in vivo conditions. To the
best of our knowledge, the only in vivo study involving bio- A core problem of current anticancer drugs is the very
logically active mesoporous silica is the recent investigation high incidence of severe unwanted side effects in healthy
by Dai et al. [160]. They showed that mesoporous silica tissues. The ultimate goal in cancer therapy is to restrict the
doped with calcium and silver significantly activate the int- therapeutic effect exclusively to cancerous cells whereas the
rinsic pathway of coagulation cascade, induce platelet adher- healthy tissue should be minimally affected. Nanoparticles
ence, promote the blood clotting, and achieve hemorrhage have taken the center stage as drug carriers for targeted
control in femoral artery and liver, when administered as a delivery. Tumor targeting can be principally achieved
powder form directly onto an open wound in a rabbit model. through the combined effect of passive targeting and active
In an earlier study, Mellaerts et al. loaded the poorly water- targeting strategies. Due to the leaky vasculature and poorly
soluble drug itraconazole into mesoporous silica to enhance operational lymph system of tumors, nanoparticles accu-
the oral availability of the drug [161]. However, neither of mulate into the tumor site by passive targeting, i.e. via the
these studied were focused on nanoparticulate formulations. enhanced permeability and retention (EPR) effect, whereas
the function of active targeting is to facilitate the interaction
4.4.2. Fighting Multidrug Resistance with cancer cells, retain the vehicle into the tumor site, and
Multidrug resistance is a major obstacle in cancer facilitate the cellular uptake [15]. MSNs have been con-
chemotherapy and severely limits the efficacy of cancer jugated with various targeting ligands including sugar
therapeutics. Both cell intrinsic and extrinsic mechanisms moieties [121, 162, 163], folic acid [164, 165], DNA
can lay behind this property. Cellular mechanisms that affect aptamers [167], as well as monoclonal antibodies, e.g.,
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1181

Drug
siRNA
Targeting moiety
Targeted receptor

Endosome

Cytotoxic action

Nucleus

Antiapoptotic mechanisms
Drug resistance mechanisms

Cell specific apoptosis


Fig. (4). MSNs can be designed to overcome multidrug resistance mechanisms by increasing the intracellular concentration of chemotherapy
agents as well as silencing the synthesis of proteins involved in drug resistance to enhance the effect of the loaded cytotoxic drug. Adapted
from ref. [122].

against the human epidermal growth factor receptor 2 168]. PDT involves the administration of photosensitizers
(HER2) [166]. Successful cell specific drug delivery to (PS) with subsequent irradiation of appropriate wavelengths
cancer cells have been demonstrated by Zink and co- to induce cell death. The overall reaction generates free
workers. They showed elevated cytotoxicity in pancreatic radicals and reactive oxygen species, such as singlet oxygen
PANC-1 cancer cells (60 % cell death based on a viability (1O2), which are toxic [162]. Light acts like a trigger to
assay) by MSNs surface functionalized with folate and induce toxicity, causing subsequent cell damage or even
loaded with the anticancer drug camptothecin, as compared death. The large surface area of MSNs thus facilitates a high
to the HFF cell line (30 % cell death) used as reference cell loading of PS while the outer particle surface can be tailored
line [135]. Similar numbers were reported by Zhu et al. for efficient cellular uptake or even targeting [168]. More-
[114], who investigated the effect of DOX-loaded MSNs over, the ability of PS to enter cells is critical, since the
surface functionalized with aptamer conjugates on cell short-lived 1O2 can then directly interact with intracellular
viability in cancerous HeLa cells and healthy QGY7703 machinery for maximal cytotoxicity and thus when
cells. About 40 % and 60 % viability was reported after 24 h, conjugated inside the pores of MSNs, the PS remain well-
respectively. In a recent study, Rosenholm et al. [83] con- protected from its local environment within a rigid porous
jugated the anticancer drug MTX, structurally similar to structure. Mou and co-workers thus reported a surface
folate, mainly onto the surface of MSNs. MTX acted dually modification process conjugating a PS, protoporphyrin IX
as a targeting ligand and a cytostatic drug. The MTX-loaded (PpIX) [167] as well as a Pd-porphyrin (PdTTP) [168], with
MSNs induced an apoptotic cell death of about 33 % after 72 MSNs through covalent bonding. In vitro evaluation of
h in the cancerous HeLa cell line, compared to non-can- PdTTP-MSNs revealed significant photo-induced cyto-
cerous HEK293 cells where no apoptosis over the control toxicity as studied in a MDA-MB-231 breast cancer cell line,
was observed. and furthermore combined the ability of PdTPP to serve as
an in vivo contrast agent for oxygen sensing and imaging.
We are still awaiting demonstrations on the ability of
The team concluded that PdTTP-MSNs holds great promise
MSNs for targeted drug-delivery in vivo, but the above
mentioned studies and the fairly straightforward functiona- as a useful nanoplatform for cancer theragnostics [168]. In
vitro tests of PpIX-MSN performed with HeLa cells also
lization of MSNs for targeting purposes make them pro-
revealed high cellular-uptake efficiency, and the
mising candidates for targeted drug delivery.
phototoxicity was found to be both irradiation time- and
4.4.4. Photodynamic Therapy dose-dependent, while cytotoxicity would occur only in the
irradiated area. They thus concluded that small amounts of
A recent development in the therapeutic application of
PpIX-MSNs accumulated inside healthy cells are fairly
MSNs is the attachment of cell-death-inducing photosensi- tolerable and would not cause severe cell damages to unra-
tizes in the pores for photodynamic therapy (PDT) [162, 167,
1182 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

diated regions. This would be beneficial in cancer treatment, DOX = Doxorobucin


as the resulting photodynamic effect can then be limited to
EDC = 1-Ethyl-3-(3-dimethylaminopropyl)-
the area of interest, leaving the surrounding healthy tissues
carbodiimide
and cells undamaged. Another strategy to accomplish this is,
naturally, active cellular targeting of the cancer cells. In this EPR effect = Enhanced permeability and retention effect
way, a recent report by Brevet et al. designed mannose- Gd = Gadolinium
targeted MSNs for PDT and showed that targeting of also
MDA-MB-231 cells with mannose was essential (as GFP = Green fluorescent protein
compared to non-functionalized MSNs) to get a high PDT HER2 = Human epidermal growth factor receptor 2
efficiency in this case [162].
hMSCs = Human mesenchymal stem cells
MRI = Magnetic resonance imaging
OUTLOOK
Fe3O4 = Magnetite
MSNs have been shown during the last ten years to
exhibit numerous attractive properties that could be syner- MIMs = Mechanically interlocked molecules
gistically exploited in the development of theragnostic MSNs = Mesoporous silica nanoparticles
devices for targeted medicine. On the therapeutic side,
MSNs have proven to function as delivery vehicles for a MTX = Methotrexate
range of different drugs, proteins, and nucleic acids, and MCM-41 = Mobil composition of matter No. 41
have also recently demonstrated successful multidrug deli-
very potential. Different cargo release strategies can be built NIR = Near infra-red
into the system, which can be based on either simple diffu- NMR = Nuclear magnetic resonance
sion that can be tuned by the material characteristics, or more
sophisticated mechanisms based on either intrinsic (release PdTTP = Pd-porphyrin
triggered by intracellular changes in pH, redox potential, or PBS = Phosphate buffered saline
enzymes) or external (irradiation, magnetization, ultrasound,
PDT = Photodynamic therapy
temperature) triggers. Hereby caps consisting of other
nanomaterials, mechanically interlocked molecules (MIMs), PS = Photosensitizers
proteins (even antibodies) or supramolecular assemblies can
PAH/PSS = Poly(allylamine) hydrochloride/
function as gate-keepers locking the drug molecules prior to
poly(sodium-4-styrene sulfonate)
exposure to a trigger. Also stimuli-responsive polymeric
chains can be utilized for this purpose, all of the former exhi- PEG = Polyethylene glycol
biting some true synergistic capabilities of hybrid inorganic- PEI = Polyethyleneimine
organic materials. On the imaging side, MSNs have shown
to be suitable for multimodal imaging, to date demonstrated PET = Positron emission tomography
under both in vitro and in vivo conditions. Due to their PLGA = Poly(D,L-lactide-co-glycolide)
multifunctionality, these imaging properties can be readily
combined with the drug delivery ability to yield interesting PNIPAAm = Poly(N-isopropylacrylamide)
theragnostic agents. Furthermore, as targetability has re- PpIX = Protoporphyrin IX
cently been successfully demonstrated for these systems,
they contain all elements for the design of smart theragnostic RES = Reticuloendothelial system
nanoplatforms. Even though the therapeutic benefits of G2-PAMAM = Second generation polyamidoamine
MSNs in vivo remains to be demonstrated, emerging bio-
compatibility and biodistribution studies together with the SBF = Simulated body fluid
promising in vitro outcomes suggest in the proximate future 1O2 = Singlet oxygen
the expansion of MSNs into such in vivo applications.
SPIO = Superparamagnetic iron oxide
TEM = Transmission electron micrographs
ABBREVIATIONS
XRD = X-ray diffraction
AMP = Adenosine monophosphate
ADP = Adenosine diphosphate REFERENCES
ATP = Adenosine triphosphate [1] Liversidge G. Merisko-Liversidge E. Ruddy SB. Callanan, F. Will
CTAB = Cetyltrimethylammonium bromide nanoparticles deliver? Drug Discov Develop 2009; 12: 30-4.
[2] Sahoo SK, Parveen S, Panda JJ. The present and future of
DCC = N,N'-dicyclohexylcarbodiimide nanotechnology in human health care. Nanomed Nanotech Biol
Med 2007; 3: 20-31.
DiI = 1,1'-Dioctadecyl-3,3,3',3'-tetramethyl- [3] Hartmann M. Ordered mesoporous materials for bioadsorption and
indocarbocyanine perchlorate biocatalysis. Chem Mater 2005; 17: 4577-93.
[4] Vallet-Regi M, Balas F, Arcos D. Mesoporous materials for drug
DiO = 3,3'-Dioctadecyloxacarbocyanine delivery. Angew Chem Int Ed 2007; 46: 7548-58.
perchlorate [5] Avnir D, Coradin T, Lev O, Livage, J. Recent bio-applications of
sol-gel materials. J Mater Chem 2006; 16: 1013-30.
DTTA = Diethylenetriaminetetraacetic acid
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1183

[6] Trewyn BG, Giri S, Slowing II, Lin, VSY. Mesoporous silica [30] Jiang X, Brinker CJ. Aerosol-assisted self-assembly of single-
nanoparticle based controlled release, drug delivery, and biosensor crystal core/nanoporous shell particles as model controlled release
systems. Chem Commun 2007; 3236-45. capsules. J Am Chem Soc 2006; 128: 4512-3.
[7] Giri S, Trewyn BG, Lin VSY. Mesoporous silica nanomaterial- [31] Kim J, Lee JE, Lee J, et al. Magnetic fluorescent delivery vehicle
based biotechnological and biomedical delivery systems. using uniform mesoporous silica spheres embedded with
Nanomedicine 2007; 2: 99-111. monodisperse magnetic and semiconductor nanocrystals. J Am
[8] Slowing, II, Trewyn, BG.; Giri, S, Lin VSY. Mesoporous silica Chem Soc 2006; 128: 688-9.
nanoparticles for drug delivery and biosensing applications. Adv [32] Liong M, France B, Bradley KA, Zink JI. Antimicrobial Activity of
Funct Mater 2007; 17: 1225-36. Silver Nanocrystals Encapsulated in Mesoporous Silica
[9] Trewyn, BG, Slowing, II, Giri S, Chen HT, Lin VSY. Synthesis and Nanoparticles. Adv Mater 2009; 21: 1684-9.
functionalization of a mesoporous silica nanoparticle based on the [33] Wang Y, Ren J, Liu X, et al. Facile synthesis of ordered magnetic
sol-gel process and applications in controlled release. Acc Chem mesoporous γ-Fe2O3/SiO2 nanocomposites with diverse mesostruc-
Res 2007; 40: 846-53. tures. J Colloid Interface Sci 2008; 326: 158-65.
[10] Slowing II, Vivero-Escoto L, Wu CW, Lin VSY. Mesoporous silica [34] Kim J, Kim HS, Lee N, et al. Multifunctional uniform nanoparticles
nanoparticles as controlled release drug delivery and gene composed of a magnetite nanocrystal core and a mesoporous silica
transfection carriers. Adv Drug Deliv Rev 2008; 60: 1278-88. shell for magnetic resonance and fluorescence imaging and for drug
[11] Angelos S, Liong M, Choi E, Zink, JI. Mesoporous silicate delivery. Angew Chem Int Ed 2008; 47: 8438-41.
materials as substrates for molecular machines and drug delivery. [35] Lin YS, Haynes CL. Synthesis and characterization of biocom-
Chem Eng J 2008; 137: 4-13. patible and size-tunable multifunctional porous silica nanoparticles.
[12] Liong M, Angelos S, Choi E, Patel K, Stoddart JF, Zink JI. Chem Mater 2009; 21: 3979-86.
Mesostructured multifunctional nanoparticles for imaging and drug [36] Wu P, Zhu J, Xu Z. Template-assisted synthesis of mesoporous
delivery. J Mater Chem 2009; 19: 6251-7. magnetic nanocomposite particles. Adv Funct Mater 2004; 14: 345-
[13] Pasqua L, Cundari S, Ceresa C, Cavaletti G. Recent development, 51.
applications, and perspectives of mesoporous silica particles in [37] Zhao W, Gu J, Zhang L, Chen H, Shi J. Fabrication of uniform
medicine and biotechnology. Curr Med Chem 2009; 16: 3054-63. magnetic nanocomposite spheres with a magnetic core/mesoporous
[14] Wang SB. Ordered mesoporous materials for drug delivery. silica shell structure. J Am Chem Soc 2005; 127: 8916-7.
Microporous Mesoporous Mater 2009; 117: 1-7. [38] Deng Y, Qi D, Deng C, Zhang X, Zhao D. Superparamagnetic high-
[15] Rosenholm J, Sahlgren C, Lindén M. Cancer-cell targeting and cell- magnetization microspheres with an Fe3O4@SiO 2 core and
specific delivery by mesoporous silica nanoparticles. J Mater Chem perpendicularly aligned mesoporous SiO2 shell for removal of
2010; 20: 2707-13. microcystins. J Am Chem Soc 2008; 130: 28-9.
[16] Kresge CT, Leonowicz ME, Roth WJ, Vartuli JC. Ordered [39] Zhao W, Shi J, Chen H, Zhang L. Particle size, uniformity, and
mesoporous molecular-sieves synthesized by a liquid-crystal mesostructure control of magnetic core/mesoporous silica shell
template mechanism. Nature 1992; 359: 710-2. nanocomposite spheres. J Mater Res 2006; 21: 3080-9.
[17] Beck JS, Vartuli JC, Roth WJ, et al. A new family of mesoporous [40] Stöber W, Fink A, Bohn E. Controlled growth of monodisperse
molecular-sieves prepared with liquid-crystal templates. J Am silica spheres in micron size range. J Colloid Interface Sci 1968; 26:
Chem Soc 1992; 114: 10834-43. 62-9.
[18] Yanagisawa T, Shimizu T, Kuroda K, Kato C. The preparation of [41] Graf C, van Blaaderen A. Metallodielectric colloidal core-shell
alkyltrimethylammonium-kanemite complexes and their conversion particles for photonic applications. Langmuir 2002; 18: 524-32.
to microporous materials. Bull Chem Soc Jpn 1990; 63: 988-92. [42] Yoon SB, Kim JY, Kim JH, et al. Synthesis of monodisperse
[19] For examples of MSN synthesis, see for instance: Urata C, Aoyama spherical silica particles with solid core and mesoporous shell:
Y, Tonegawa A, Yamauchia Y, Kuroda K. Chem Commun 2009; mesopore channels perpendicular to the surface. J Mater Chem
5094-6, and references therein. 2007; 17: 1758-61.
[20] Berggren A, Holmberg K, Palmqvist AEC. Synthesis of stable [43] Kim JH, Yoon SB, Kim JY, Chae YB, Yu JS. Synthesis of
colloidal suspensions of ordered mesostructured silica from sodium monodisperse silica spheres with solid core and mesoporous shell:
metasilicate using pluronic P123 and mildly acidic conditions. Stud Morphological control of mesopores. Colloids Surf A Physicochem
Surf Sci Catal 2007; 165: 53-6. Eng Aspects 2008; 313-314: 77-81.
[21] He Q, Cui X, Cui F, Guo L, Shi J. Size-controlled synthesis of [44] Nakamura T, Mizutani M, Nozaki H, Suzuki N, Yano K. Formation
monodispersed mesoporous silica nano-spheres under a neutral mechanism for monodispersed mesoporous silica spheres and its
condition Microporous Mesoporous Mater 2009; 117: 609-16. application to the synthesis of core/shell particles. J Phys Chem C
[22] Kapoor MP, Fujii W, Yanagi M, et al. Environmental friendly rapid 2007; 111: 1093-100.
mass production synthetic process of highly ordered nanometer [45] For a recent review, see for instance: Athens GL, Shayib RM,
sized mesoporous silica using a combination of acid-base and Chmelka BF. Functionalization of mesostructured inorganic-
evaporation approach. Microporous Mesoporous Mater 2008; 116: organic and porous inorganic materials. Curr Opin Colloids
370-7. Interface Sci 2009; 14: 281-92.
[23] Lin KJ, Chen LJ, Prasad MR, Cheng CY. Core-shell synthesis of a [46] Che S, Garcia-Bennett AE, Yokoi T, et al. A novel anionic
novel, spherical, mesoporous silica/platinum nanocomposite: surfactant templating route for synthesizing mesoporous silica with
Pt/PVP@MCM-41. Adv Mater 2004; 16: 1845-9. unique structure. Nat Mater 2003; 2: 801-5.
[24] Joo SH, Park JY, Tsung CK, Yamada Y, Yang P, Somorjai GA. [47] de Juan F, Ruiz-Hitzky E. Selective functionalization of
Thermally stable Pt/mesoporous silica core-shell nanocatalysts for mesoporous silica. Adv Mater 2000; 12: 430-2.
high-temperature reactions. Nature Mater 2009; 8: 126-31. [48] Rosenholm JM, Duchanoy A, Lindén M. Hyperbranching surface
[25] Nooney RI, Dhanasekaran, T, Chen Y, Josephs R, Ostafin AE. Self- polymerization as a tool for preferential functionalization of the
assembled highly ordered spherical mesoporous silica/gold outer surface of mesoporous silica. Chem Mater 2008; 20: 1126-33.
nanocomposites. Adv Mater 2002; 14: 529-32. [49] Kecht J, Schlossbauer A, Bein T. Selective Functionalization of the
[26] Nooney RI, Thirunavukkarasu D, Chen Y, Josephs R, Ostafin AE. Outer and Inner Surfaces in Mesoporous Silica Nanoparticles.
Self-assembly of mesoporous nanoscale silica/gold composites. Chem Mater 2008; 20: 7207-14.
Langmuir 2003; 19: 7628-37. [50] He Q, Zhang J, Shi J, et al. The effect of PEGylation of mesoporous
[27] Gorelikov I, Matsuura N. Single-step coating of mesoporous silica silica nanoparticles on nonspecific binding of serum proteins and
on cetyltrimethyl ammonium bromide-capped nanoparticles. Nano cellular responses. Biomaterials 2010; 31:1085-92.
Lett 2008; 8: 369-73. [51] Huang S, Fan Y, Cheng Z, et al. Magnetic mesoporous silica
[28] Fan H, Gabaldon J, Brinker CJ, Jiang YB. Ordered nanocrystal/ spheres for drug targeting and controlled release. J Phys Chem C
silica particles self-assembled from nanocrystal micelles and 2009; 113: 1775-84.
silicate. Chem Commun 2006; 2323-5. [52] Zhu Y, Shi J, Shen W, et al. Stimuli-responsive controlled drug
[29] Botella P, Corma A, Navarro MT. Single gold nanoparticles release from a hollow mesoporous silica sphere/polyelectrolyte
encapsulated in monodispersed regular spheres of mesostructured multilayer core-shell structure. Angew Chem Int Ed 2005; 44:
silica produced by pseudomorphic transformation. Chem Mater 5083-7.
2007; 19: 1979-83.
1184 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

[53] Zhu Y, Shi J. A mesoporous core-shell structure for pH-controlled using supercritical carbon dioxide. J Supercritical Fluids 2009; 51:
storage and release of water-soluble drug. Microporous Mesoporous 278-86.
Mater 2007; 103: 243-9. [79] Hillerström A, van Stam J, Andersson M. Ibuprofen loading into
[54] You YZ, Kalebaila KK, Brock SL, Oupický D. Temperature- mesostructured silica using liquid carbon dioxide as a solvent.
controlled uptake and release in PNIPAM-modified porous silica Green Chem 2009; 11: 662-7.
nanoparticles. Chem Mater 2008; 20: 3354-9. [80] Ritter H, Brühwiler D. Accessibility of amino groups in
[55] Zhu S, Zhou Z, Zhang D. Control of drug release through the in situ postsynthetically modified mesoporous silica. J Phys Chem C 2009;
assembly of stimuli-responsive ordered mesoporous silica with 113: 10667-74.
magnetic particles. ChemPhysChem 2007; 8: 2478-83. [81] Tourné-Péteilh C, Brunel D, Bégu S, et al. Synthesis and
[56] Lim MH, Stein A. Comparative studies of grafting and direct characterisation of ibuprofen-anchored MCM-41 silica and silica
syntheses of inorganic-organic hybrid mesoporous materials. Chem gel. New J Chem 2003; 27: 1415-8.
Mater 1999; 11: 3285-95. [82] Mortera R, Vivero-Escoto J, Slowing II, Garrone E, Onida B, Lin
[57] Zhuravlev LT. Surface characterization of amorphous silica - a VS-Y. Cell-induced intracellular controlled release of membrane
review of work from the former USSR. Colloids Surf A impermeable cysteine from a mesoporous silica nanoparticle-based
Physicochem Eng Aspects 1993; 74: 71-90. drug delivery system. Chem Commun 2009; 3219-21.
[58] Iler RK. The Chemistry of Silica, Wiley, New York 1979. [83] Rosenholm JM, Peuhu E, Bate-Eya LT, Eriksson J-E, Sahlgren C,
[59] Rosenholm JM, Lindén M. Towards establishing structure-activity Lindén M. Cancer-cell-specific induction of apoptosis using
relationships for mesoporous silica in drug delivery applications. J mesoporous silica nanoparticles as drug-delivery vectors. Small
Control Release 2008; 128: 157-64. 2010; 6: 1234-41.
[60] Tang Q, Xua Y, Wua D, Sun Y. A study of carboxylic-modified [84] Carino IS, Pasqua L, Testa F, et al. Silica-based mesoporous
mesoporous silica in controlled delivery for drug famotidine. J materials as drug delivery system for methotrexate release. Drug
Solid State Chem 2006; 179: 1513-20. Deliv 2007; 14: 491-5.
[61] Hartmann M. Ordered mesoporous materials for bioadsorption and [85] Mellaerts R, Jammaer JAG, van Speybroeck M, et al. physical state
biocatalysis. Chem Mater 2005; 17: 4577-93. of poorly water soluble therapeutic molecules loaded into sba-15
[62] Hudson S, Magner E, Cooney J. Proteins in mesoporous silicates. ordered mesoporous silica carriers: a case study with itraconazole
Angew Chem Int Ed 2008; 47: 8582-94. and ibuprofen. Langmuir 2008; 24: 8651-9.
[63] Diaz JF, Balkus KF Jr. Enzyme immobilization in MCM-41 [86] Rosenholm JM, Peuhu E, Eriksson JE, Sahlgren C, Lindén M.
molecular sieve. J Mol Catal B 1996; 2: 115-26. Targeted intracellular delivery of hydrophobic agents using
[64] Deere J, Magner E, Wall JG, Hodnett BK. Adsorption and activity mesoporous hybrid silica nanoparticles as carrier systems. Nano
of cytochrome c on mesoporous silicates. Chem Commun 2001; Lett 2009; 9: 3308-11.
465-6. [87] Chen AM, Zhang M, Wei D, et al. Co-delivery of doxorubicin and
[65] Deere J, Magner E, Wall JG, Hodnett BK. Adsorption of Bcl-2 sirna by mesoporous silica nanoparticles enhances the
cytochrome c onto ordered mesoporous silicates. Stud Surf Sci efficacy of chemotherapy in multidrug-resistant cancer cells. Small
Catal 2001; 135: 233. 2009; 5: 2673-7.
[66] Vinu A, Murugesan V, Tangermann O, Hartmann M. Adsorption of [88] Zhao Y, Trewyn BG, Slowing II, Lin VS-Y. Mesoporous silica
cytochrome c on mesoporous molecular sieves: influence of ph, nanoparticle-based double drug delivery system for glucose-
pore diameter, and aluminum incorporation. Chem Mater 2004; 16: responsive controlled release of insulin and cyclic AMP. J Am
3056-65. Chem Soc 2009; 131: 8398-400.
[67] Vinu, A, Murugesan V, Tangermann O, Hartmann M. Adsorption [89] Ng JBS, Kamali-Zare P, Brismar H, Bergström L. Release and
of lysozyme over mesoporous molecular sieves mcm-41 and sba- molecular transport of cationic and anionic fluorescent molecules in
15: influence of ph and aluminum incorporation. J Phys Chem B mesoporous silica spheres. Langmuir 2008; 24: 11096-102.
2004; 108: 7323-30. [90] Cheng K, Landry CC. Diffusion-based deprotection in mesoporous
[68] Lu S, He J, Guo X. Architecture and performance of mesoporous materials: a strategy for differential functionalization of porous
silica-lipase hybrids via non-covalent interfacial adsorption. AICHE silica particles. J Am Chem Soc 2007; 129: 9674-85.
J 2010; 56: 506-14. [91] Lebold T, Jung,C, Michaelis J, Bräuchle C. Nanostructured silica
[69] Han Y-J, Stucky GD, Butler A. Mesoporous silicate sequestration materials as drug-delivery systems for doxorubicin: single molecule
and release of proteins. J Am Chem Soc 1999; 121: 9897-8. and cellular studies. ACS Nano 2009; 9: 2877-83.
[70] Slowing II, Trewyn BG, Lin VS-Y. Mesoporous silica [92] Cauda V, Muhlstein L, Onida B, Bein T. Tuning drug uptake and
nanoparticles for intracellular delivery of membrane-impermeable release rates through different morphologies and pore diameters of
proteins. J Am Chem Soc 2007; 129: 8845-9. confined mesoporous silica. Microporous Mesoporous Mater 2009;
[71] Solberg SM, Landry CC. Adsorption of DNA into mesoporous 118: 435-42.
silica. J Phys Chem B 2006; 110: 15261-8. [93] Cauda C, Schlossbauer A, Bein T. Bio-degradation study of
[72] Xia T, Kovochich M, Liong M, et al. Polyethyleneimine coating colloidal mesoporous silica nanoparticles: Effect of surface
enhances the cellular uptake of mesoporous silica nanoparticles and functionalization with organo-silanes and poly(ethylene glycol).
allows safe delivery of siRNA and DNA constructs. ACS Nano Microporous Mesoporous Mater 2010; 132: 60-71.
2009; 3: 3273-86. [94] Mortera R, Fiorilli S, Garrone E, Verné E, Onida B. Pores occlusion
[73] Vallet-Regi M, Ramila A, del Real RP, Perez-Pariente J. A new in MCM-41 spheres immersed in SBF and the effect on ibuprofen
property of mcm-41: drug delivery system. Chem Mater 2001; 13: delivery kinetics: A quantitative model. Chem Eng J 2010; 156:
308-11. 184-92.
[74] Andersson J, Rosenholm J, Areva S, Lindén M. Influences of [95] Ng JBS, Kamali-Zare P, Sörensen M, Brismar H, Hedin N,
material characteristics on ibuprofen drug loading and release Bergström L. Intraparticle transport and release of dextran in silica
profiles from ordered micro- and mesoporous silica matrices. Chem spheres with cylindrical mesopores. Langmuir 2010; 26: 466-70.
Mater 2004; 16: 4160-7. [96] Cauda V, Onida B, Platschek B, Mühlstein L, Bein T. Large
[75] Aiello R, Cavallaro G, Giammona G, Pasqua L, Pierro P, Testa F. antibiotic molecule diffusion in confined mesoporous silica with
Mesoporous silicate as matrix for drug delivery systems of non- controlled morphology. J Mater Chem 2008; 18: 5888-99.
steroidal antinflammatory drugs. Stud Surf Sci Catal 2002; 142: [97] Cotí KK, Belowich ME, Liong M, et al. Mechanised nanoparticles
1165-72. for drug delivery. Nanoscale 2009; 1: 16-39.
[76] Hata H, Saeki S, Kimura T, Sugahara Y, Kuroda K. Adsorption of [98] Aznar E, Martínez-Mañez R, Sancenón F. Controlled release using
taxol into ordered mesoporous silicas with various pore diameters. mesoporous materials containing gate-like scaffoldings. Exp Op
Chem Mater 1999; 11: 1110-9. Drug Deliv 2009; 6: 643-55.
[77] Domingo C, García-Carmona J, Fanovich MA, Saurina J. Study of [99] Casasús R, Marcos MD, Martínez-Máñez R, et al. Toward the
adsorption processes of model drugs at supercritical conditions development of ionically controlled nanoscopic molecular gates. J
using partial least squares regression. Anal Chim Acta 2002; 452: Am Chem Soc 2004; 126: 8612-3.
311-9. [100] Bernardos A, Aznar E, Coll C, et al. Controlled release of vitamin
[78] Belhadj-Ahmed F, Badens E, Llewellyn P, Denoyel R, Charbit G. B-2 using mesoporous materials functionalized with amine-bearing
Impregnation of vitamin E acetate on silica mesoporous phases gate-like scaffoldings. J Control Release 2008; 131: 181-9.
Multifunctional Mesoporous Silica Nanoparticles Current Drug Targets, 2011, Vol. 12, No. 8 1185

[101] Song S-W, Hidajat K, Kawi S. pH-controllable drug release using [124] Yu J, Zhao H, Ye L, et al. Effect of surface functionality of
hydrogel encapsulated mesoporous silica. Chem Commun 2007; magnetic silica nanoparticles on the cellular uptake by glioma cells
4396-8. in vitro. J Mater Chem 2009; 19: 1265-70.
[102] Gao Q, Xu Y, Wu D, Sun Y, Li X. pH-Responsive drug release [125] Lin MM, Kim H-H, Kim H, Dobson J, Kim DK. Surface activation
from polymer-coated mesoporous silica spheres. J Phys Chem C and targeting strategies of superparamagnetic iron oxide
2009; 113: 12753-8. nanoparticles in cancer-oriented diagnosis and therapy.
[103] Park HS, Kim CW, Lee HJ, et al. A mesoporous silica nanoparticle Nanomedicine 2010; 5: 109-33.
with charge-convertible pore walls for efficient intracellular protein [126] Lu C-W, Hung Y, Hsiao J-K, et al. Bifunctional magnetic silica
delivery. Nanotechnology 2010; 21: 225101 (9 pp). nanoparticles for highly efficient human stem cell labeling. Nano
[104] Yang Q, Wang S, Fan P, et al. pH-responsive carrier system based Lett 2007; 7: 149.
on carboxylic acid modified mesoporous silica and polyelectrolyte [127] Lin Y-S, Wu S-H, Hung Y, et al. Multifunctional composite
for drug delivery. Chem Mater 2005; 17: 5999-6003. nanoparticles: magnetic, luminescent, and mesoporous. Chem
[105] Wu J, Sailor M. Chitosan hydrogel-capped porous sio2 as a ph Mater 2006; 18: 5170-2.
responsive nano-valve for triggered release of insulin. Adv Funct [128] Liu H-M, Wu S-H, Lu C-W, et al. Mesoporous silica nanoparticles
Mater 2009; 19: 733-41. improve magnetic labeling efficiency in human stem cells. Small
[106] Khashab NM, Belowich ME, Trabolsi A, et al. pH-responsive 2008; 4: 619-26.
mechanised nanoparticles gated by semirotaxanes. Chem Commun [129] Wu S-H, Lin Y-S, Hung Y, et al. Multifunctional mesoporous silica
2009; 5371-3. nanoparticles for intracellular labeling and animal magnetic
[107] Liu R, Zhang Y, Zhao X, Agarwal A, Mueller LJ, Feng P. pH- resonance imaging studies. Chem Bio Chem 2008; 9: 53-7.
Responsive nanogated ensemble based on gold-capped mesoporous [130] Lin Y-S, Hung Y, Su J-K, et al. Gadolinium(III)-incorporated
silica through an acid-labile acetal linker. J Am Chem Soc 2010; nanosized mesoporous silica as potential magnetic resonance
132: 1500-1. imaging contrast agents. J Phys Chem B 2004; 108: 15608-11.
[108] Lai C-Y, Trewyn BG, Jeftinija DM, et al. A mesoporous silica [131] Taylor K-M, Kim J-S, Reiter W-J, An H, Lin W, Lin W.
nanosphere-based carrier system with chemically removable cds Mesoporous silica nanospheres as highly efficient MRI contrast
nanoparticle caps for stimuli-responsive controlled release of agents. J Am Chem Soc 2008; 130: 2154-5.
neurotransmitters and drug molecules. J Am Chem Soc 2003; 125: [132] Hsiao J-K, Tsai C-P, Chung T-H, et al. Mesoporous silica
4451-9. nanoparticles as a delivery system of gadolinium for effective
[109] Torney F, Trewyn BG, Lin VS-Y, Wang K. Mesoporous silica human stem cell tracking. Small 2008; 4: 1445-52.
nanoparticles deliver DNA and chemicals into plants. Nature [133] Carniato F, Tei L, Dastrù W, Marchese L, Botta M. Relaxivity
Nanotechnol 2007; 2: 295-300. modulation in Gd-functionalised mesoporous silicas. Chem
[110] Aznar E, Marcos MD, Martinez-Máñez R, et al. pH- and photo- Commun 2009; 1246.
switched release of guest molecules from mesoporous silica [134] Lin Y-S, Tsai C-P, Huang H-Y, et al. Well-ordered mesoporous
supports. J Am Chem Soc 2009; 131: 6833-43. silica nanoparticles as cell markers. Chem Mater 2005; 17: 4570-3.
[111] Giri S, Trewyn BG, Stellmaker MP, Lin VS-Y. Stimuli-responsive [135] Huang D-M, Hung Y, Ko B-S, et al. Highly efficient cellular
controlled-release delivery system based on mesoporous silica labeling of mesoporous nanoparticles in human mesenchymal stem
nanorods capped with magnetic nanoparticles. Angew Chem Int Ed cells: implication for stem cell tracking. FASEB J 2005; 19: 2014-6.
2005; 44: 5038-44. [136] Lee C-H, Cheng S-H, Wang Y-J, et al. Near-infrared mesoporous
[112] Fujiwara M, Terashima S, Endo Y, Shiokawa K, Ohue H. silica nanoparticles for optical imaging: characterization and in vivo
Switching catalytic reaction conducted in pore void of mesoporous biodistribution. Adv Funct Mater 2009; 19: 215-22.
material by redox gate control. Chem Commun 2006; 4635-7. [137] Liong M, Lu J, Kovochich M, et al. Multifunctional Inorganic
[113] Liu R, Zhao X, Wu T, Feng P. Tunable redox-responsive hybrid Nanoparticles for Imaging, Targeting, and Drug Delivery. ACS
nanogated ensembles. J Am Chem Soc 2008; 130: 14418-9. Nano 2008; 2: 889-96.
[114] Zhu C-L, Song X-Y, Zhou W-H, Yang H-H, Wen Y-H, Wang X-R. [138] Lee J-E, Lee N, Kim H, et al. Uniform mesoporous dye-doped
An efficient cell-targeting and intracellular controlled-release drug silica nanoparticles decorated with multiple magnetite nanocrystals
delivery system based on MSN-PEM-aptamer conjugates. J Mater for simultaneous enhanced magnetic resonance imaging,
Chem 2009; 19; 7765-70. fluorescence imaging, and drug delivery. J Am Chem Soc 2010;
[115] Patel K, Angelos S, Dichtel WR, et al. Enzyme-responsive snap-top 132: 552-7.
covered silica nanocontainers. J Am Chem Soc 2008; 130: 2382-3. [139] Chung T-H, Wu S-H, Yao M, et al. The effect of surface charge on
[116] Park C, Kim H, Kim S, Kim C. Enzyme responsive nanocontainers the uptake and biological function of mesoporous silica
with cyclodextrin gatekeepers and synergistic effects in release of nanoparticles in 3T3-L1 cells and human mesenchymal stem cells.
guests. J Am Chem Soc 2009; 131: 16614-5. Biomaterials 2007; 28: 2959-66.
[117] Schlossbauer A, Kecht J, Bein T. Biotin-avidin as a protease- [140] Lu F, Wu S-H, Hung Y, Mou C-Y. Size effect on cell uptake in
responsive cap system for controlled guest release from colloidal well-suspended, uniform mesoporous silica nanoparticles. Small
mesoporous silica. Angew Chem Int Ed 2009; 48: 3092-5. 2009; 5: 1408-13.
[118] Climent E, Bernardos A, Martinez-Manez R, et al. controlled [141] Vallhov H, Gabrielsson S, Strømme M, Scheynius A, Garcia-
delivery systems using antibody-capped mesoporous nano- Bennett A-E. Mesoporous silica particles induce size dependent
containers. J Am Chem Soc 2009; 131: 14075-80. effects on human dendritic cells. Nano Lett 2007; 7: 3576-82.
[119] Liu J, Jiang X, Ashley C, Brinker CJ. Electrostatically mediated [142] Fisichella M, Dabboue H, Bhattacharyya S, et al. Mesoporous silica
liposome fusion and lipid exchange with a nanoparticle-supported nanoparticles enhance MTT formazan exocytosis in HeLa cells and
bilayer for control of surface charge, drug containment, and astrocytes. Toxicology in vitro 2009; 23: 697.
delivery. J Am Chem Soc 2009; 131: 7567-9. [143] Trewyn B-G, Slowing I-I, Giri S, Chen H-T, Lin V S-Y. synthesis
[120] Oyewumi MO, Rice K.G. In: Nanoparticle Technology for Drug and functionalization of a mesoporous silica nanoparticle based on
Delivery; Gupta RB, Kompella UB, Eds. New York: Taylor & the sol-gel process and applications in controlled release. Acc Chem
Francis Group 2006; pp. 361. Res 2007; 40: 846-53.
[121] Radu DR, Lai CY, Jeftinija K, Rowe EW, Jeftinija S, Lin VS-Y. A [144] Di Pasqua A-J, Sharma K-K, Shi Y-L, et al. Cytotoxicity of
polyamidoamine dendrimer-capped mesoporous silica nanosphere- mesoporous silica nanomaterials. J Inorg Biochem 2008; 102:
based gene transfection reagent. J Am Chem Soc 2004; 126: 13216- 1416-23.
7. [145] Tao Z, Toms BB, Goodisman J, Asefa T. Mesoporosity and
[122] Park IY, Kim IY, Yoo MK, Choi YJ, Cho M-H, Cho CS. Functional Group Dependent Endocytosis and Cytotoxicity of
Mannosylated polyethylenimine coupled mesoporous silica Silica Nanomaterials. Chem Res Toxicol 2009; 22: 1869-80.
nanoparticles for receptor-mediated gene delivery. Int J Pharm [146] Rosenholm JM, Sahlgren C, Lindén M. Towards multifunctional,
2008; 359: 280-7. targeted drug delivery systems using mesoporous silica
[123] Hom C, Lu J, Liong M, et al. Mesoporous silica nanoparticles nanoparticles – opportunities & challenges. Nanoscale 2010; 2:
facilitate delivery of siRNA to shutdown signaling pathways in 1870-83.
mammalian cells. Small 2010; 6: 1185-90.
1186 Current Drug Targets, 2011, Vol. 12, No. 8 Rosenholm et al.

[147] Hudson SP, Padera RF, Langer R, Kohane DS. The [158] Lu J, Liong M, Sherman S, et al. Mesoporous silica nanoparticles
biocompatibility of mesoporous silicates. Biomaterials 2008; 29: for cancer therapy: energy-dependent cellular uptake and delivery
4045-55. of paclitaxel to cancer cells. Nanobiotechnol 2007; 3: 89-95.
[148] Cho M, Cho WS, Choi M, et al. The impact of size on tissue [159] De Smedt SC, Demeester J, Hennink WE. Cationic polymer based
distribution and elimination by single intravenous injection of silica gene delivery systems. Pharm Res 2000; 17: 113-26.
nanoparticles. Toxicol Lett 2009; 189: 177-83. [160] Dai C, Yuan Y, Liu C, et al. Degradable, antibacterial silver
[149] Ferrari M. Cancer nanotechnology: opportunities and challenges. exchanged mesoporous silica spheres for hemorrhage control.
Nat Rev Cancer 2005; 5: 161-71. Biomaterials 2009: 30; 5364-74.
[150] Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an [161] Mellaerts R, Mols R, Jammaer JAG, et al. Increasing the oral
emerging treatment modality for cancer. Nat Rev Drug Discov bioavailability of the poorly water soluble drug itraconazole with
2008; 7: 771-82. ordered mesoporous silica. Eur J Pharm Biopharm 2008; 69: 223.
[151] Wei Y, Dong H, Xu J, Feng Q. Simultaneous Immobilization of [162] Brevet D, Gary-Bobo M, Raehm L, et al. Mannose-targeted
Horseradish Peroxidase and Glucose Oxidase in Mesoporous Sol- mesoporous silica nanoparticles for photodynamic therapy. Chem
Gel Host Materials. ChemPhysChem 2002; 3: 802-8. Commun 2009; 1475-7.
[152] Dai Z, Liu S, Ju H, Chen H. Direct electron transfer and enzymatic [163] Gu J, Fan W, Shimojima A, Okubo T. Organic-inorganic
activity of hemoglobin in a hexagonal mesoporous silica matrix. mesoporous nanocarriers integrated with biogenic ligands. Small
Biosens Bioelectron 2004; 19: 861-7. 2007; 3: 1740-4.
[153] Dai ZH, Xu XX, Ju HX. Direct electrochemistry and electrocata- [164] Slowing I, Trewyn BG, Lin VS-Y. Effect of Surface Functionaliza-
lysis of myoglobin immobilized on a hexagonal mesoporous silica tion of MCM-41-type mesoporous silica nanoparticles on the
matrix. Anal Biochem 2004; 332: 23-31. endocytosis by human cancer cells. J Am Chem Soc 2006; 128:
[154] Radu DR, Lai CY, Wiench JW, Pruski M, Lin VS. Gatekeeping 14792-3.
layer effect: a poly(lactic acid)-coated mesoporous silica nano- [165] Rosenholm JM, Meinander A, Peuhu E, et al. Targeting of porous
sphere-based fluorescence probe for detection of amino-containing hybrid silica nanoparticles to cancer cells. ACS Nano, 2009; 3: 197-
neurotransmitters. J Am Chem Soc 2004; 126: 1640-1. 206.
[155] Descalzo AB, Marcos MD, Monte C, Martínez-Máñez R, Rurack [166] Tsai C-P, Chen C-Y, Hung Y, Chang F-H, Mou C-Y. Monoclonal
K. Mesoporous silica materials with covalently anchored antibody-functionalized mesoporous silica nanoparticles (MSN) for
phenoxazinone dyes as fluorescent hybrid materials for vapour selective targeting breast cancer cells. J Mater Chem 2009; 19:
sensing. J Mater Chem 2007; 44: 4716-23. 5737-43.
[156] Lu J, Liong M, Zink JI, Tamanoi F. Mesoporous silica [167] Tu H-L, Lin Y-S, Lin H-Y, et al. In vitro studies of functionalized
nanoparticles as a delivery system for hydrophobic anticancer mesoporous silica nanoparticles for photodynamic therapy. Adv
drugs. Small 2007; 3: 1341-6. Mater 2009; 21: 172-4.
[157] Vivero-Escoto JL, Slowing II, Wu C-W, Lin VS-Y. Photoinduced [168] Cheng SH, Lee C-H, Yang C-S, Tseng F-G, Mou C-Y, Lo L-W.
intracellular controlled release drug delivery in human cells by Mesoporous silica nanoparticles functionalized with an oxygen-
gold-capped mesoporous silica nanosphere. J Am Chem Soc 2009; sensing probe for cell photodynamic therapy: potential cancer
131: 3462-3. theranostics J Mater Chem 2009; 19: 1252-7.

Received: April 20, 2010 Revised: July 14, 2010 Accepted: August 30, 2010

PMID: 21443474

Você também pode gostar