You are on page 1of 24

Review

For reprint orders, please contact reprints@future-science.com

Silicon–polymer hybrid materials


for drug delivery

Silicon and its oxides are widely used in biomaterials research, tissue engineering and drug delivery. These materials
are highly biocompatible, easily surface functionalized, degrade into nontoxic silicic acid and can be processed into
various forms such as micro- and nano-particles, monoliths, membranes and micromachined structures. The large
surface area of porous forms of silicon and silica (up to 1200 m2/g) permits high drug loadings. The degradation
kinetics of silicon- and silica-based materials can be tailored by coating or grafting with polymers. Incorporation of
polymers also improves control over drug-release kinetics. The use of stimuli-responsive polymers has enabled
environmental stimuli-triggered drug release. Simultaneously, silicon microfabrication techniques have facilitated
the development of sophisticated implantable drug-delivery microdevices. This paper reviews the synthesis, novel
properties and biomedical applications of silicon–polymer hybrid materials with particular emphasis on drug delivery.
The biocompatible and bioresorptive properties of mesoporous silica and porous silicon make these materials
attractive candidates for use in biomedical applications. The combination of polymers with silicon-based materials
has generated a large range of novel hybrid materials tailored to applications in localized and systemic drug delivery.

Drug-delivery principles Silicon-based materials for Steven JP McInnes &


Enhanced therapeutic effects have been drug delivery Nicolas H Voelcker †
observed using controlled drug-delivery sys- Silicon-based materials are commonly used in †
Author for correspondence
tems in comparison with conventional drug drug delivery for the simple fact that they are The Flinders University of South
delivery. Consequently, improving the delivery relatively bio-inert and degrade into nontoxic Australia, School of Chemistry,
of an existing drug is often considered as a com- silicic acid (Si[OH]4). Silicon-based materials Physics and Earth Sciences,
mercially viable alternative to the development are also easily surface functionalized with con- GPO Box 2100, Adelaide,
of a new drug [1,2] . Controlled drug-delivery ventional methods of synthesis. This allows a SA 5001, Australia
systems enable drug delivery to specific sites wide variety of hybrid materials to be produced Tel.: +61 882 015 338
and allow the rate of delivery to be finely tuned, by the addition of organic or polymeric layers. Fax: +61 882 012 905
thereby minimizing toxicity, side effects and There are three main forms of silicon used in E-mail: nico.voelcker@
increasing therapeutic benefits (Figure 1) [2–9] . drug-delivery applications: silica, porous silicon flinders.edu.au
To sustain the maximum therapeutic benefit (pSi) and micromachined silicon. Within these
of drugs, a constant sustained-release (zero- three broad categories exist many possible mor-
order) profile with little or no burst effect is phologies, including films, membranes, micro- Porous silicon
usually desired [1,10,11] . However, it is impor- and nano-particles and monoliths. A form of the chemical
tant that the release profile is carefully con- element silicon containing
trolled and matched to the application. For Silica
„„ an array or network of
nanoscale pores produced by
example, wound-healing patches need to be Silicon dioxide or silica (SiO2) is composed of electrochemical etching
tailored with a release profile that matches repeating SiO4 tetrahedrons. The Si-O bond
natural wound healing to fully facilitate long- length is 0.162 nm, smaller than the covalent
term healing and minimize scarring [3] . The radii of the constituent Si and O atoms. This
design of new drug-delivery systems should creates a partial ionic character [16] . Synthetic
also focus on extending the therapeutic release silica is generally fabricated by sol–gel chem-
by providing a pulsatile or continuous-release istries using the condensation of Si(OH) 4 and
or, alternatively, exploiting a triggered-release tetraethyl­orthosilicate (TEOS). The freshly
mechanism [10,12–15] . synthesized silica surface contains a mixture
The silicon–polymer hybrid materials of surface siloxane bridges, silanediol groups
reviewed here have useful properties for con- and silanol groups (Figure 2A–C) , which allow
trolled drug release and are, therefore, attrac- for further functionalization via silanization,
tive candidates for the development of systems esterification and other reactions (Figure  2D) .
and devices for drug delivery. Sol–gel-synthesized silica nanoparticles can have

10.4155/FMC.09.90 © 2009 Future Science Ltd Future Med. Chem. (2009) 1(6), 1051–1074 ISSN 1756-8919 1051
Review | McInnes & Voelcker
electrolyte [32] . The chemistry and mechanism of
Maximum desired (toxic) drug level this electrochemical process has been reviewed
elsewhere [14,33] . pSi has four particularly
attractive qualities for in vivo drug delivery:
n High surface area (up to 800 m 2 /g) [34] ;
Drug level

Therapeutic window n Photonic properties (exploitable for bio­sensing


[35–38] and for self-reporting drug delivery
[33,39] );

n Ability to be processed into membranes and


micro- or nano-particles [40–45] (suitable for
Minimum effective applications including implantation
drug level Second dose Third dose or injection);
Time n Degradation into silicic acid in aqueous
environments, such as exist in vivo [46] .
Figure 1. Drug level in body fluid over time from traditional drug-delivery
methods using multiple doses (solid line) and drug levels in a simple The biocompatibility of pSi has been dem-
controlled-release system (dashed line). onstrated by the attachment and proliferation
Adapted with permission from [9] . of mammalian cells on various pSi structures as
well as by in vivo studies [36,47–50] .
a surface area of up to 1200 m2 /g [10] . Sol–gel- Porous silicon can be functionalized by a
Biocompatibility
produced silica can also be processed to create range of chemical reactions including oxidation,
The ability of a material to
perform with an appropriate
xerogels and bioactive glass monoliths. silanization, hydrosilylation and electrografting.
host response in a Silica xerogel solids are formed from the dehy- Hydrosilylation of hydride-terminated freshly
specific application dration of a SiO2 polymer network and they etched pSi can be achieved using thermal [51,52] ,
usually retain high porosity (25%) and large white light [53–55] , Lewis acid [56–58] and micro-
surface area (150–900 m 2 /g), along with very wave-assisted [59] methods. Both cathodic and
small pore size (1–10 nm). They can also be used anodic electrografting methods have also been
as monoliths [17–19] or as particles [20–22] in both performed on freshly etched pSi and are reviewed
wet [23] and dry [24–26] forms. These materials elsewhere [60,61] . The ageing of freshly prepared
are popular for drug delivery [18,21,24–26] because pSi under ambient conditions or treatment with
release kinetics can be tuned by changing the ozone or high temperatures leads to the formation
sol–gel synthesis parameters, such as the silica of silanol (Si-OH) and silicon (di)oxide (SiO2)
precursor:catalyst ratio [18] , the gelation time [26]
or temperature [19] . A B
Another type of silica, bioactive glass, is com-
O O HO OH
monly composed of CaO and SiO2, but can also
Si Si Si Si
contain P2O5. These materials are able to induce
O O
carbonate hydroxyapatite layer formation on the
surface of the material upon contact with physi- C D
ological fluids, facilitating bonding to both hard CH3
and soft tissue [27] . Research has been directed H3C CH3
Si
towards the incorporation of growth factors H H H
and proteins that promote cell recognition and O O O O O
adhesion [28–30] . Bioglasses also feature a range
Si Si Si Si Si
of porosities and hence surface areas, which is of
O O O
interest for controlled drug release. In addition,
exposed hydroxyl groups on the glass surface
are available for chemical functionalization [31] . Figure 2. (A) Stable surface siloxane bridges,
(B) silanediol groups, (C) silanol groups that
can be free (isolated) or hydrogen bonded
Silicon
„„ (vicinal) and (D) silica surface functionalized
The most common form of elemental silicon with a monoalkoxy silane (left) to create
used for drug-delivery applications is pSi. pSi Si-O-Si bonds and esterified (right) to create
is fabricated by anodization of monocrystalline Si-O-C bonds.
Adapted with permission from [16] .
silicon, typically carried out in hydrofluoric acid

1052 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review

Calcination
and template
Surfactant Deposition removal

Template
nanoparticle

Figure 3. Synthesis of porous hollow silica nanoparticles using calcium carbonate


nanoparticles as templates.
Adapted with permission from [87] .

bonds. Other types of oxidations may be per- as CsOH and KOH. The design of these silicon-
formed to stabilize pSi, including chemical [62– based devices can be as simple as an array of wells
65] , anodic [66,67] and photo-induced [68,69] oxida- or needles [75] or as complicated as the production
tions. Nitridization of pSi has also been described of intricate flaps that control the release of drugs
[70,71] . The presence of Si-OH groups on oxidized from polymer-coated reservoirs [76] . During the
pSi surfaces allows for the functionalization with design stage of any MEMS device for drug deliv-
alkoxy- or chloro-silanes. Ambient conditions ery, there are four major components that should
and only moderately elevated temperatures are be considered for inclusion: microreservoirs,
sufficient for common silanization reactions. micropumps, valves and sensors [15] .
Silanizations can be used to introduce a wide The three major forms of silicon discussed
range of functional groups, including amines, above are all easily functionalized with vari-
isocyanates, methacrylate and polyethylene gly- ous polymer chemistries (e.g., surface-initiated
col (PEG) functionalities. The functionalization and surface-tethered polymerization), grafting
of pSi has been previously reviewed [60,72] . techniques and noncovalent depositions (e.g.,
An additional benefit of pSi-based drug deliv- spin- and dip-coating). In this manner, sili-
ery is that the optical properties of the mate- con–polymer hybrid materials with enhanced
rial allow the noninvasive measurement of the properties for drug delivery can be formed. This
drug-release rate. A self-reporting drug-delivery review will focus on the use of these three silicon
system based on pSi particles has been recently forms in three major approaches to drug delivery
reported in living animal eyes [73] . with silicon–polymer hybrid materials, including
Flat (single crystal) silicon lacks the high sur- micro- and nano-particles, monoliths (e.g., films
face area of pSi, but is a popular model system and membranes) and micromachined devices.
for developers of drug-delivery platforms. Flat When designing materials for drug delivery,
Si can be surface-functionalized by silanization issues such as structural parameters, release and
and hydrosilylation in much the same way as pSi. biodegradation kinetics and mechanisms, load-
However, when performing silanization reac- ing capacity and material toxicity must be taken
tions, the silicon wafers must first be cleaned and into account. It is important to note that the
activated with piranha solution, which is known hybrid materials discussed in this paper are novel
to generate Si-OH groups [74] . On the other hand, and require further testing to fully characterize
complete removal of the native oxide layer with their properties. We have endeavored to discuss Microelectromechanical
HF (to create Si–H surfaces) is required before all relevant data where available. systems

functionalization via hydrosilylation reactions. The integration of mechanical


Due to its simple surface chemistry, wide avail- Micro- & nano-particles for structures with
microelectronics.
ability, ease of ana­lysis and flat morphology, drug delivery Microelectromechanical systems
single crystalline silicon is useful as a testing „„Mesoporous silica nanomaterials are custom designed, allowing
platform for many functionalization techniques High surface area silica nanoparticles mechanical actions to be
that can be applied later to pSi. (~700–1200 m 2 /g) have been investigated for controlled by external stimuli
their ability to store and release drugs in a con-
Micromachined
„„ silicon trolled manner [10] . These silica nanoparticles Biodegradation
Microelectromechanical systems (MEMSs) are can be used as drug carriers and release platforms The capability of decomposition
commonly produced by the micromachining of without further surface modification or polym- of a material into innocuous
complex structures into the surface of single crys- erization. However, a lack of functionalization products by natural biological
activity within the body
talline silicon wafers with etching solutions such can lead to uncontrollable payload release [77,78] .

future science group www.future-science.com 1053


Review | McInnes & Voelcker
A drugs has been published by Chen et al. who
studied the controlled-release behavior of
avermectin (a pesticide) from porous hollow
TMOS Calcination
silica nanoparticles (PHSNs) (Figure  3) [87,90] .
The synthesis was performed via a sol–gel route
using inorganic calcium carbonate nanoparti-
Microgel Microgel–silica Porous silica cles as templates. Avermectin was adsorbed on
the external surface, the pore channels in the
B wall and also loaded in the inner core of the
PHSN carriers, thus giving rise to a multistage
Shell cross-linking TMOS sustained-release pattern. It was found that the
shell thickness was one of the most important
factors in controlling the release rate: by increas-
ing the shell thickness a more sustained release at
Diblock Crosslinked Silica-coated an overall decreased rate ensued [90] . The same
polymer micelle core–shell micelle micelle
approach has been adapted to release other com-
pounds including Brilliant Blue® and Cefradine®
Figure 4. (A) Microgel-templated porous silica and (B) core–shell nanoparticles [89,96] . With appropriate washing procedures,
with a silica shell. drugs adsorbed to the outer surface could be
TMOS: Tetramethyl orthosilicate. removed and the molecules stored in the pores
Adapted with permission from [97,98] .
and the internal core of the nanoparticles could
be controllably released without a burst effect.
Mesoporous silica Two of the most widely studied mesoporous These PHSN carriers could allow for the devel-
Common mesoporous silica materials are Mobil composition of matter opment of new drug-delivery technologies with
nanoparticles, such as MCM-41 (MCM)-41 and Santa Barbara Amorphous type variable nanoscale morphologies, which, owing
and SBA-15 contain well-defined material (SBA)-15. MCM-41 was first synthe- to their small size (compared with conventional
hexagonal pores less than 30 nm
diameter. Applications include sized in 1992 by the Mobil Inc. corporation and drug carriers), hold promise for transmucosal
catalysis, drug delivery possesses uniform hexagonal arrays of pores with and systemic drug-delivery applications [89] .
and imaging pore sizes in the range of 1.5–10 nm [79] . SBA-15
was synthesized 6 years later and possesses the Silica–polymer
„„ hybrids
Controlled release same ordered pore structure but with larger pores To improve the release kinetics and inherent
ranging from 5 to 30 nm [80] . These mesoporous properties of silica particles, research has focused
The release of drugs at
predetermined intervals or silica structures are synthesized by condensation on the creation of silica–polymer hybrid mate-
gradually over time, in order to of TEOS around micellar rods used as templates. rials. For example, Zhou et  al. used cationic
maintain therapeutic levels Calcination removes the template rods and leaves microgels to template the deposition of silica
during the treatment period
particles with a regular pore structure. The use from tetramethyl orthosilicate (TMOS) precur-
of mesoporous silica structures in place of solid sors (Figure 4A) [97] . The hybrid nanoparticles
particles is advantageous in terms of their loading described possessed tunable porosities and iso-
capacity, since the drug can attach to the internal electric points that could be varied depending on
pore volume as well as the external surface. The the TMOS concentration. This may allow for
presence of an internal structure also serves to pro- the controlled delivery of biomolecules such as
tect the drug from the external environment and DNA. On a similar theme, Yuan et al. synthesized
to control the release via capping moieties [81,82] . silica nanoparticles with cationic coronas and
Approaches to improve control of drug release hydrophobic polymer cores [98] . They employed
from mesoporous silica have involved varying diblock copolymers of poly(2-[dimethylamino]
synthetic parameters  [83,84] , composition  [85,86] , ethylmethacrylate)-co-(2-[diisopropylamino]
morphology [87–90] , pore size [91] , loading param- ethylmethacrylate) to template the hydrolysis
eters [92] or surface chemistry [77,78,93–95] of the of TMOS (Figure 4B) in the hydrophilic coro-
silica nanoparticles. nas. This produced silica nanoparticles with a
diameter of 35 nm and well-defined core–shell
„„Silica particle morphology & morphology. Silica deposition effectively cross-
pore-size modulation linked the polymeric nanoparticles, leading to
One particularly intriguing example exploiting an increased stability at pH 2 (where the non-
morphological changes in silica nanoparticles crosslinked polymeric nanoparticles would com-
to control the release profile of small molecular pletely dissociate). This technique has currently

1054 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
not been used for drug-delivery tests; however, encapsulating dibutylphtalate with both hydro-
one could envisage the biphasic release of both phobic and hydrophilic silica nanoparticles
hydrophobic and hydrophilic drugs from these [100,101] . The resulting microparticles featured
nanoparticle preparations. silica multilayers, the architecture of which was
A system that has been tested for its drug- controlled by nanoparticle properties and elec-
delivery properties was synthesized by Huo et al. trolyte concentration. The layered microstruc-
The authors used F127 micelles of ethylene ture, its thickness and hydrophobicity deter-
oxide and propylene oxide (EO106 -PO70-EO106) mined the drug-release rate from the PDMS
to template TEOS hydrolysis to form core–shell droplets. Colloidal and spray-drying approaches
nanoparticles [81] . These particles showed signifi- may also be used to coat polymeric micropar-
cantly slower release rates and increased loading ticles [102] . Such preparations have been shown
capacities compared with noncrosslinked F127 to have good gastric resistance (minimal release
surfactant micelles with papaverine (basic), at pH 2) and subsequent release at neutral pH,
nabumetone (neutral) and naproxen (acidic) making them suitable for the oral delivery of
loadings. This system showed almost zero-order drugs requiring release in the lower digestive sys-
release kinetics. In general, silica crosslinked tem. Engineering of the silica nanoparticle layer
nanoparticles retain the biocompatibility of the offers a wide range of possible release profiles and
original micelles [99] , but are much more stable has the potential to help enhance the delivery of
and do not break down upon dilution, mak- relatively insoluble drugs.
ing them excellent candidates for drug-delivery
applications. The particles also show reduced Polymer
„„ coatings on silica nanoparticles
diffusion of drugs due to the presence of the The inverse architecture, polymeric multilay-
silica shell; they have physiochemical proper- ers coated on silica microparticles, has also
ties that can be tuned by changing the synthe- been reported with a view to exploiting con-
sis parameters and a surface chemistry that is trolled drug delivery. These structures can be
tailorable by one-pot and postfunctionalization created via layer-by-layer (LBL) techniques or
techniques. Due to the hydrophobic core, hydro- encapsulation.
philic and hydrophobic drugs can both be loaded The encapsulation of SiO2 xerogel particles
and delivered. has also been performed by Xue et  al. SiO2-
poly(dl-lactide-co-glycolide) (PLGA) single
„„Silica coatings on polymeric nanoparticles and double layered microspheres (Figure 5) were
Silica nanoparticles are able to self-assemble to formed via solvent evaporation [20] . The thus-
form a coating on polymeric microparticles. formed microspheres allowed the controlled
Prestidge et al. coated micron-sized polydimeth- release of the antibiotic gentamicin. The release
ylsiloxane (PDMS) emulsion droplets, pattern from these composites included a 1-day

A 100 B 80
Cumulative drug release (wt%)

Cumulative drug release (wt%)

80
60

60
40
40

20
20
10wt% xerogel loading
50 µm 20wt% xerogel loading 50 µm
0 0
0 10 20 30 40 50 0 10 20 30 40 50 60
Time (days) Time (days)

Figure 5. (A) Gentamicin release from SiO2–poly(dl-lactide-co-glycolide)-encapsulated single- and (B) double-layered silica xerogel
microspheres. Insets show representative scanning electron microscopy images of encapsulated microspheres.
Adapted with permission from [20] .

future science group www.future-science.com 1055


Review | McInnes & Voelcker
The LBL deposition of poly(diallylmethyl­
+ +
+ +
ammonium chloride) and poly(sodium 4-sty-
- renesulfonate) on the surface of negatively
+ + -
- - charged silica microparticles allows stable poly-
+ + electrolyte films to be formed (Figure  7) [104] .
PAA Release
+ + coating at pH 7 Such a coating could obviate the need for the
+ + SiO2 to encapsulate the drug.
Stimuli-responsive LBL coatings have also
been described as a capping layer on drug-loaded
BSA-loaded SBA-15 PPA-coated PAA and BSA
BSA-loaded SBA-15 release from SBA-15 hollow mesoporous silica spheres, incorporating
ibuprofen. These structures allowed loading up
Figure 6. Electrostatic coating of SBA-15 microparticles containing BSA to threefold higher than uncoated controls. The
with PAA. The system is stable at low pH and unstable at high pH, preventing release of the stored ibuprofen could be stimu-
drug release in the stomach, but facilitating release in the intestine. lated by changing the environmental pH or the
BSA: Bovine serum albumin; PAA: Poly(acrylic acid); SBA: Santa Barbara amorphous. salt concentration [105] .
Adapted with permission from [103] .
Other stimuli-responsive drug-delivery sys-
tems typically rely on poly(N-isopropylacryl-
initial burst of less than 10%, followed by three amide) (PNIPAAm), which is thermosensitive
Stimuli responsiveness
sustained-release phases. The first sustained- and switches from a hydrophilic to a hydropho-
A physicochemical change in a release stage was attributed to the erosion of bic state above the lower critical solution temper-
material (e.g., color or
wettability) upon introduction the outer PLGA layer, the second stage was ature (LCST) of approximately 30–35°C [106] .
of environmental stimuli such as attributed to the erosion of the inner PLGA Silica can be functionalized with PNIPAAm via
pH and temperature layer and the last release phase was attributed surface-initiated polymerization from surface-
to the release of the gentamicin from the xero- bound atom transfer radical polymerization
Surface-initiated gel. Release was sustained for approximately (ATRP) initiators, such as 3-trimethoxysilylpro-
polymerization 9 weeks. By contrast, single-layered PLGA com- pyl methacrylate [107] and surface-bound 2-bro-
A polymerization technique that posites showed a 50% burst release in the first mopropionyl bromide [108,109] . Nanoparticles
allows for the direct growth of day followed by 6 weeks of slow release. possessing photoluminescence (from CdTe),
polymer chains from surface-
bound initiators In order to model protein drug delivery, Song thermosensitivity (from PNIPAAm) and mag-
et al. loaded bovine serum albumin (BSA) into netism (from Fe3O4) have been demonstrated
amino-modified SBA-15 and then encapsulated to deliver water-soluble, photosensitizing drugs
the microparticle, via electrostatic assembly, for photodynamic therapy [106] . Nanoparticles
with poly(acrylic acid) (PAA) (Figure  6) [103] . were taken up by cells below the LCST and the
This imparted stability to the system at low pH drug was successfully released into the cyto-
(uncharged carboxylic acid side-chains allow plasm upon switching to 37°C. Bikram et al.
PAA to contract on the SBA) and instability created silica–gold nanoparticles embedded with
at neutral pH (carboxylic acid side-chains are poly(N-isopropylacrylamide-co-acrylamide)
charged, causing the PAA to swell and dissociate hydrogels that could slowly release the drug in a
from the SBA). The authors propose that this manner dependent on its molecular weight or be
technique could be used to enhance the deliv- switched when heated by means of laser irradia-
ery of peptide and protein drugs into the lower tion to induce complete release [110] . The devel-
digestive tract, where the pH is close to neutral. opment of these stimulated-release systems will

Cationic Anionic
polymer polymer Multiple
- - deposition - - deposition - - depositions - -
- - - - - - - -
- - - - - - - -
- - - - - - - -
- - - - - - - -
- - - - - - - -

Silica nanoparticle Polyelectrolyte-coated


silica nanoparticle

Figure 7. Layer-by-layer deposition of polyelectrolytes on silica particles.


Adapted with permission from [104] .

1056 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
prove important for future drug-delivery appli- could be controlled by addition of more P2O5 or
cations. The main advantage of these stimuli- poly(vinyl alcohol), which decreases the surface
responsive systems is the ability to switch from area and porosity, in principle allowing tunabil-
a passive release of drugs to an accelerated one ity of drug release [115] . These silicon–polymer
upon application of stimuli. Release rates can be hybrid systems could be used as nonload-bear-
tuned by variations in the polymer composition, ing bone defect fillers and control the release of
loading technique and the stimulation condi- small-molecule drugs, such as ibuprofen, to the
tions. The use of external stimuli such as heat, surrounding bone tissue, in order to suppress
magnetism or light to induce drug release could tissue inflammation. There is potential to incor-
lead to the creation of drug-delivery devices that porate a wider range of drugs into polymer-mod-
can be surgically implanted subcutaneously and ified bioglass monoliths to stimulate bone regen-
activated by the patient when required, eliminat- eration, including drugs such as bisphosphonates
ing the need for multiple injections or frequent and platinum anti-tumor compounds [24,116] .
medical consultations. The incorporation and release of drugs and
Particle encapsulation is a simple yet powerful biomolecules such as the antibiotic vancomy-
technique to prepare silicon–polymer hybrids cin  [21] , the anticancer drug doxorubicin [25] ,
for drug delivery. The encapsulation can be per- novel platinum compounds [24] and larger
formed by electrostatic or physiosorption tech- molecules such as heparin [18] have already
niques and the drug-release rates can be tuned by been studied from unmodified silica xerogels.
the addition of multiple polymeric layers. However, the drug-release rate from silica xero-
gel monoliths can be tuned by the addition
Monoliths, films & membranes of biocompatible polymers such PEG [17,19,22] ,
„„ Bioglasses & xerogels PDMS [17] or PLGA [20] . Addition of PEG to the
Unmodified bioglasses (or bioactive glasses) have sol–gel mix during preparation of silica xerogels
been investigated for their ability to store and was shown to increase the drug-release rate in
release drugs in a controlled manner [111,112] . One general, owing to the increased hydrophylic-
of the most interesting recent examples of drug ity of the network  [19] . Pegylation of silicon-
delivery from bioglasses is the incorporation of based materials also helps avoid rapid degrada-
mesoporous silica (SBA-15) by precipitation into tion or clearance as well as evade the immune
a bioactive glass–ceramic (SiO2-CaO-K 2O) scaf- response  [22,117–122] . Pegylation and associated
fold. This material combines the biocompatibil- advantages gained for biomedical applications
ity of the bioglass and the drug-release properties have been reviewed extensively and will not be
of the SBA-15 [113] , and could find applications discussed further [122,123] .
as an implantable sustained-release device for the Similar work has indicated that the release
therapy of bone diseases or for bone regeneration rate is controlled by diffusion from the porous
after trauma. structure [22] . Czarnobaj et al. have also used
To further improve the properties of these more hydrophobic SiO2-PDMS xerogels to slow
bioglasses, they can be combined with poly- the release of cisplatin [17] . It was found that, for
mers. Bioglasses, of the general form SiO2- PDMS-containing xerogels, drug release is by
CaO-P2O5, have been combined with polymers diffusion only, while the PEG-containing xero-
to improve their properties. Ladron de Guevara- gel releases drugs via a combination of diffusion,
Fernandez et al. prepared bioactive glass by the erosion and swelling of the composite materi-
hydrolysis and polycondensation of TEOS, als. Silica xerogels require only mild synthesis
triethylphosphate and calcium nitrate tetrahy- conditions, which allows for the presynthesis
drate, followed by the free radical polymeriza- incorporation of drugs, creating homogeneous
tion of a mixture of poly(l-lactic acid) (PLA), drug distributions within the composite mate-
poly(methylmethacrylate) (PMMA) and meth- rial, not only in the pores as per postsynthesis
ylmethacrylate [114] . They found that ibuprofen loading. This direct incorporation of the drug
release (incorporated during the polymerization into the matrix also acts to protect the drug from
stage) lasted for approximately 3 weeks and that premature in vivo degradation, often encoun-
the composites were still bioactive and able to tered with the systemic administration of drugs.
induce the formation of an apatite layer in SBF. Other advantages include the nontoxicity, hard-
Bioactive glasses coated with poly(vinyl alcohol) ness and tunable porosity of the composites.
have also been shown to induce the formation There is certainly also the potential to convert
of apatite layers. Degradation of the composite the monolithic structures into microparticles.

future science group www.future-science.com 1057


Review | McInnes & Voelcker
Polymeric monoliths can also be produced et al. who coated silicon wafers with alginate/
by casting techniques. Casting involves the use chitosan via spin coating [126] . Here, the release
of a preshaped mould into which a polymeric of fluorescent biomolecules was monitored in
solution can be poured and then dried to form both phosphate-buffered saline and simulated
the required monolithic structure; subsequently, body fluid. A substantial burst release of over
smaller monoliths can also be cut from larger 50% of the loaded molecules occurred within
structures if required. One such example was the first 20 min. Release was then sustained over
demonstrated by Lin et  al. who synthesized the next 26 days. The release rate could be tuned
PMMA–silica composites via a sol–gel approach by increasing the thickness of the multilayer
followed by casting with acetylsalicylic acid stack or by variation of the molecular weight
(aspirin) into a Teflon dish [124] . This approach of the release molecule. Peng et al. argue that
resulted in a monolith composed primarily of these thin coatings could prove useful in appli-
polymer with interdispersed silica micropar- cations such as wound healing, which requires a
ticles. This method still allows for the release large burst of the drug in the first few hours fol-
rate to be tuned, in this case by the addition of lowed by a slow, but sustained, release. Likewise,
a higher percentage of 3-(trimethoxysilyl) pro- multilayered imine crosslinked poly(allylamine
pylmethacrylate into the PMMA component, hydrochloride)-dextran and poly(styrene sul-
causing more polymer to be attached to the silica fonate) polyelectrolyte microgels fabricated via
and slowing the release rate of the incorporated alternating dip coating on silicon wafers have
aspirin. The amount of drug released could be been shown to be capable of storing and releas-
controlled by the percentage of silica in the com- ing model drug entities [127] . Furthermore, due
posite. This allowed for the generation of tunable to the crosslinked nature of the structures, they
drug-release materials that have demonstrated were found to be robust enough to be reloaded
aspirin release for up to 90 days. Biopolymer- and reused for multiple drug-delivery cycles.
coated bioactive glass has also been incorporated Another example of a crosslinked LBL multi-
into a collagen monolith and used as a growth layered film for the controlled release of DNA
factor-delivery device. Bergeron et al. cast bio- utilized the formaldehyde-induced crosslink-
active glass microspheres (incorporating bone ing of single-stranded DNA and BSA on sili-
morphogenic proteins) into a collagen gel. The con wafers [128] . The resulting film was stable
authors demonstrated the release of bone mor- and the amount of single-stranded DNA and
phogenic proteins to MC3T3-E1 cells and pro- BSA could be tailored by the simple addition
posed that this system could be used for bone or removal of more layers. The formation of
regeneration [125] . this multilayered stack was not controlled by
The monolith structures described can be electrostatic interactions and, subsequently, the
used to create implantable devices that will layer dissolution could be performed by the
release their payload as they degrade in  vivo. addition of proteinase K, allowing the amount
The ability to tune these monoliths via the addi- of DNA released to be tuned by limiting the
tion of various polymeric components or surface proteinase incubation time. The development
polymerization has led to the development of of systems such as this could lead to films that
structures that can potentially be used as bone are more stable and provide greater control over
defect fillers or implantable devices for the con- the release of biomolecules when compared with
trolled release of incorporated drugs. The abil- electrostatically constructed LBL systems.
ity of the xerogels to incorporate drugs during Vazquez et al. were the first to report a LBL
one-pot synthesis is of great importance to the layer system deposited on silicon that was con-
development of homogeneous matrices that will structed from specially synthesized poly(b-
release drugs at a constant predetermined rate. amino esters) (cationic) and calf thymus DNA
(anionic) layers on silicon surfaces [129] . This sys-
„„LBL deposition on flat silicon tem was able to release DNA via the slow erosion
Commonly, silicon is used as a base material of the LBL assembly under physiological condi-
to deposit multilayered structures for funda- tions. Since then, much work with this multi-
mental studies of drug release. Silicon is one layer approach has been performed [130–132] . It
of the most favored substrates as it is flat, eas- has been found that the released DNA is able
ily functionalized and primed for LBL coating to transfect mammalian cells and promote
and, most importantly, it is amenable for sur- the expression of proteins [130] . This leads to
face ana­lysis. An example is the work of Peng many possible applications in gene transfer. For

1058 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review

Table 1. Polymers with potential application to silicon- and silica-based drug-delivery composites.
Polymer Useful properties Ref.
Poly(7-allyloxycoumarin) Photodimerization (with drugs or interchain) [138]
Poly(vinylbenzylthymine) Photodimerization (with drugs or interchain) [139]
Poly(ethyleneimine)-co-poloxamer diacrylate High transfection efficiency, good DNA binding, low cytotoxicity [140,141]
or
poly(ethyleneimine)-co-glycerol
dimethacrylate
Poly(N-isopropylacrylamide)-co-(4- Temperature and pH sensitive, fluorescent [142]
vinylpyridine)-co-(N-
pyrenemethylacrylamide)
High-molecular-weight polyethylene glycols Easily modified, reductively cleavable S-S bonds, hydrolytically degradable, [143]
modified with cysteine bonding to drugs, proteins and DNA
Poly(glycerol-adipate) Nontoxic, biological monomer, low cytotoxicity, high drug loading [144]
Poly(pyridyldisulfide ethylmethacrylate) Multifunctional (able to bind DNA, proteins and sulfur-containing compounds), [145]
variable physicochemical properties
Crosslinked acid liable poly(acrylamide) pH-stimulated release, protection from nuclease degradation [146]

example, in coatings for biomedical devices such stack size to allow higher loading can lead to
as intravascular stents, the coating could locally the retardation of drug diffusion from the film.
release gene-based therapeutics for cardiovascu- Other issues include the inability to release drugs
lar disease [131] . In these works, the time for com- under physiological conditions and the need to
plete drug release was between 30 and 100 h, modify some substrates before deposition [127] .
however these release times could be tuned by However, the LBL deposition of polymers onto
introducing variations in the layer thickness and surfaces such as silicon has many advantages,
number of bilayers used. Zhang et al. have also including the high number of polymers available
released antisense DNA oligonucleotide strands as well as the composition and stack-size of the
from phosphorylcholine-containing polymer polyelectrolyte films. The ability to coat compli-
films on flat silicon wafers [133] . Oligonucleotide cated structures and to store and release a wide
loading increased with increasing film thick- range of compounds, including nanoparticles,
ness and also when the charge density of the still makes these systems exciting candidates for
cationic side chains increased. The film sus- use in drug-delivery applications.
tained the release of oligonucleotides for 8 days.
Released oligonucleotides were able to enter Polymer
„„ grafting on flat silicon
vascular smooth muscle cells. LBL assemblies The grafting of polymers to surfaces can be
have been tuned to do more than just release performed via ‘grafting-to’ or ‘grafting-from’
functional DNA. These multilayered structures polymerization techniques. The ‘grafting-to’
are able to infer protein resistance to surfaces, or, method uses prepolymerized macromolecules
if coated with a top layer such as PLL-g–PEG/ (commonly biomolecules) with functional
PEG–biotin, they are able to selectively bind end-groups to react with complementary func-
streptavidin-bound proteins [134] . Polyelectrolyte tional groups residing on the surface; whereas
LBL-deposited films on silicon have also been the ‘grafting-from’ approach uses surface-
used to deliver therapeutic polysaccharides [7] bound monomer (surface-tethered polym- Surface-tethered
and gentamicin [135] . erization) or initiator groups (surface-initi- polymerization

Although LBL films on silicon are themselves ated polymerization) as anchor points for the A polymerization technique that
allows the tethering of growing
unlikely to find applications in in vivo drug deliv- polymerization process.
polymer chains to surface-
ery, they are widely studied as model coatings, This type of surface ‘grafting-from’ approach bound monomer groups
with the aim to transfer the optimized coatings has been used by Cole et al. to covalently link
to more relevant substrates such as intravascular PNIPAAm to flat silicon [107] . These surfaces
Ti stents [131] . There are some issues involved were anchored via the use of a methacrylate-
with the use of LBL systems for drug release, terminated silane, followed by the free radi-
the most notable being the inability of the films cal polymerization of N-isopropylacrylamide
to load large quantities of drugs. Poor loading (NIPAAm), tethering the resulting polymer
is usually caused by the ionic nature of the mul- chains to the surface. The resulting coatings
tilayered stack; however, simply increasing the were protein-binding above the LCST and

future science group www.future-science.com 1059


Review | McInnes & Voelcker

Table 2. Polymers that have been covalently attached to silicon, silica and pSi (since 2007).
Polymerization Polymer Surface Switchable? Ref.
technique (stimulus)
ATRP Poly(2-hydroxyethyl methacrylate) Silica and silicon No [147,148]

Poly(styrene) Silica and silicon No [149,150]


Poly(4-[10-methacryloydecyloxy]-4´-pentylazobenzene) Silicon Yes [151,152]
(light)
Poly(methyl methacrylate) Silica No [150]
Poly(methyl methacrylate) pSi No [153]
Poly(N-isopropylacrylamide) (PNIPAAm) SiO2 nanoparticles Yes [109]
(temperature)
Polyacrylamide (PAAm) Silicon No [154]
Poly(ethyleneglycol methacrylate) Silicon No [155–157]
Poly(1-[4-vinylbenzyl]-3-butylimidazolium hexafluorophosphate) Silicon Yes (counter- [158]
anions)
Poly(2-[dimethylamino] ethylmethacrylate) Silicon No [159]
Co-polymer preparations Silicon No [160–162]
Poly(butylmethacrylate)-co-(boc-aminoethyl methacrylate)
Poly(butyl methacrylate)-co-2,2-dimethylaminoethyl methacrylate)
Poly(styrene)-b-(acrylic acid)
‘Grafting-to’ Poly(glycidyl methacrylate) and aromatic aliphatic Silicon No [163,164]
hyperbranched polyester
Poly(glycidyl methacrylate) Silicon No [163,164]
Poly(methyl methacrylate) Silicon No [165,166]
Poly(ethylene glycol) Silicon No [165–168]
Poly(N-isopropylacrylamide) pSi Yes [169]
(temperature)
Poly(acrylamide) and poly(2-vinylpyridine) Silicon Yes [170]
(pH)
Poly(tert-butyl acrylate) Silicon No [171]
RAFT Poly(methyl methacrylate) Silicon No [172,173]
Poly(styrene) and poly(butyl acrylate) Silicon No [174]
Poly(styrene), poly(styrene)-b-acrylic acid), poly(styrene)-b- Silicon Yes [175]
(N-isopropylacrylamide) and poly(methyl methacrylate)-b-poly (solvent)
(N,N-(dimethylamino)ethylacrylate)
Free radical Poly(N-isopropylacrylamide) Silicon Yes [107,176]
(temperature)
Poly(N-isopropylacrylamide)-co-poly(acrylic acid) Silicon Yes [130]
(temperature)
Nitroxide-mediated Poly(styrene) Silicon No [177]
Poly(styrene) and poly(acrylate) SiO2 No [178]
ROMP Poly(spiropyan) Silicon No [179]
ROP Poly(l-lactide) pSi No [32]
Electrografting Poly(acrylic acid) Silicon No [180]
Poly(nitrophenylene) Silicon No [181]
ATRP: Atom-transfer radical polymerization; pSi: Porous silicon; RAFT: Reversible addition–fragmentation chain transfer; ROMP: Ring-opening metathesis
polymerization; ROP: Ring-opening polymerization.

resisted protein adsorption below the LCST. coatings of PNIPAAm and PAAc to reversibly
PNIPAAm surfaces have also been used by bind fluorescein isothiocyanate (FITC)-labeled
Tsukagoshi et al. to load and release aspirin [8] . BSA [136] . In this case, the polymer network was
At temperatures above the LCST, the release crosslinked with N,N´-methylenebisacrylamide
of aspirin was much slower due to the polymer and the maximum switching of the hydrogel,
chains in the collapsed state entrapping the aspi- in terms of wettability, occurred at a monomer
rin molecules. Kurkuri et al. prepared copolymer ratio of 36:1 PNIPAAm:PAAc. LeMieux et al.

1060 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
have incorporated an additional hydrophobic
layer of poly(butyl acrylate) over the PNIPAAm
layer [137] . This poly(butyl acrylate) layer could Temperature 34 nm
be used to control premature drug release from change
the composite until the PNIPAAm layer is PBA
17 nm
PNIPAAm
switched. The authors found that the distance Silicon
of poly(butyl acrylate) from the surface could
50°C PNIPAAm 10°C PNIPAAm
be changed from 17 to 34 nm upon switching compressed state expanded state
of the underlying PNIPAAm layer (Figure 8) . It
is conceivable that the aforementioned proof-of-
Figure 8. PNIPAAm (light region) and hydrophobic PBA (dark region) on a
principle studies will soon lead to commercial flat silicon substrate. The poly(N-isopropylacrylamide) layer can exist in the
temperature-switchable drug-delivery devices. contracted or expanded state depending on the temperature.
Novel monomers are also being constantly PBA: Poly(butyl acrylate); PNIPAAm: Poly(N-isopropylacrylamide).
designed and incorporated into drug-delivery Adapted with permission from [137] .
systems. Table 1 contains examples of polymers
that have interesting properties and have not high biopolymer binding. Popular in this con-
yet been applied to porous scaffolds and should text are NHS-modified monomers, which can
be considered when designing new materials be conjugated to any amine-presenting biomol-
for biomedical devices [138–146] . Table 2 shows a ecule. NHS-containing polymers have been
list of polymers and polymerization techniques, applied to silicon surfaces via spin coating [189] ,
which have been applied to silicon, silica and pSi surface-initiated ATRP [190] and polymeric self-
since 2007 [32,107,109,136,147–181] . assembled monolayers [191] , and to silica via oxi-
dative polymerization [192] . The surface-initiated
Biopolymer
„„ grafting on flat silicon ATRP of PAA on silicon surfaces has also been
Just as synthetic polymers can be grafted onto used to generate a high loading surface for any
silicon, biopolymers (e.g., peptides and oligo- NH2-containing biomolecules (Figure 9) , again
nucleotides) can also be used as the polymer via NHS-mediated amide bond formation [193] .
for surface modification, however they are Recent research in this area has also exploited
commonly attached via ‘grafting-to’ methods, click chemistry [194,195] to immobilize biomol-
for example, the immobilization of DNA onto ecules on silicon functionalized with a polymeric
undecylenic acid-patterned silicon surfaces was self-assembled monolayer-containing terminal
performed by Voicu et al. as early as 2004 [182] . alkyne groups. However, this method requires
This coupling required activation of the surface the biomolecule to be azido-functionalized prior
COOH groups with N-hydroxysuccinimide and to forming the self-assembled monolayer [196] .
then a subsequent reaction with an amine-termi- Burnham et al. have grafted hydrogels on silicon
nated DNA strand. Böcking et al. have also used surfaces containing a disulfide crosslinker, which
epoxide groups to immobilize thiol-terminated can be reduced to generate reactive sulfhydryl
DNA [183] . Similarly, Hone et al. have conju- groups suitable for biomolecule conjugation [197] .
gated proteins to silicon surfaces functionalized This approach also allowed the authors to pat-
by hydrosilylation with undecenylaldehyde [184] . tern multiple biomolecules onto the hydrogel
Click chemistry has also been used to covalently surface via microcontact printing techniques.
bind trypsin into mesoporous silica [185] . To Potentially, biomolecules attached to the
improve the density of the biopolymer chains, a hydrogel via disulfide bonds could be released
few groups have examined the grafting of DNA when exposed to a reducing environment inside
[43,186] and peptides [187,188] from the surface with the body.
conventional solid-phase synthesis protocols. Flat crystalline silicon is not considered a bio-
However, the design of linker chemistry that is degradable or bioresorbable material due to its
cleavable under physiological conditions in order stability in bodily fluids. The surface is passivated
to release bioactive biopolymers has not come and therefore hydrolyzes very slowly, at time-
to fruition. scales not relevant to drug delivery and other
The modification of surfaces with biomol- biomedical applications. However, the behavior
ecules such as peptides and DNA can also of crystalline silicon can be tuned from rela-
be performed by means of polymeric spacers tively bioinert to bioactive and even bioresorb-
between the silicon and the biomolecule, spe- able by the introduction of porosity [34,198] . The
cifically designed for the purpose of attaining introduction of porosity or micro­structuring of

future science group www.future-science.com 1061


Review | McInnes & Voelcker
crystalline silicon increases the low surface area be used for the sustained release of drugs [204] .
and assists in improving drug loadings [10,41,199] . Demonstrating increased control over drug
Devices made from polymer and flat silicon release, Wu and Sailor prepared a pSi scaffold
hybrid materials would need to be surgically containing a crosslinked chitosan–hydrogel cap-
retrieved after drug release. Postsurgical removal ping layer. The insulin release was stimulated by
of the device is not desirable. The development immersion in buffer at pH 6, avoiding the burst
of drug-delivery devices composed of only biore- release observed for uncapped pSi [205] .
sorbable materials would enable the device to Koh et  al. investigated composites of pho-
automatically be eliminated by the body upon tonic pSi films and polymers as a smart patch
degradation in  vivo. For these reasons, there that allows for the delivery of drugs to be moni-
needs to be scope to transfer the polymer coat- tored in real-time [39] . The reflectance peak of the
ings developed on flat silicon to more biodegrad- photonic material (Distributed Bragg Reflector
able scaffolds, such as mesoporous silica and pSi [DBR]) can be tuned to a wavelength spanning
films and microparticles [32,200] . the visible spectrum by varying the layer thick-
ness and the refractive index contrast. In particu-
„„Porous silicon lar, reflection peaks in the region of low optical
There are many examples of the release of drugs absorption for human tissue (500–600 nm in
from polymer-free pSi scaffolds [42,201,202] . this case) will allow the smart-patch DBR to be
However, in order to improve control over the monitored in vivo. The free-standing pSi DBR
release behavior of drugs from pSi scaffolds, was cast in a PMMA solution containing the
polymeric functionalization is currently being drug to be released. The resulting composite
investigated. Some of the most interesting and was approximately 140 µm thick, flexible and
novel developments in recent years include the chemically robust. Release of caffeine from the
work of Batra et  al., who have covered dye- smart patch was monitored in vitro by UV–Vis
loaded pSi substrates via repetitive coating with and changes in the reflection peak. The caffeine
a poly(e-caprolactone) (PCL) layer of varying release could be controlled by varying the num-
thickness [203] . The PCL layer slowed down the ber of etching repeats used to create the initial
release rate of the dyes as well as lowering the DBR. The porous structure of pSi can also be
equilibrium amount of the dye released. The transferred to polymers via the use of templating
release rate could be controlled by tailoring the techniques. Polymer infusion and subsequent pSi
thickness of the PCL layer. Similarly, Coffer dissolution allows for the creation of porous poly-
et al. have formed composites of high-porosity meric materials that retain photonic properties of
pSi particles and PCL to create scaffolds that the pSi and allow for monitoring of drug release
are highly biocompatible and could potentially or scaffold dissolution [206,207] . These polymer

H
Br ONa Br O N Br N
( )n

Br O O O O

O O O O O
O O O O
ONa EDC, NHS NH2

CuBr, CuBr2, H2O,


2,2´-bipyridine, rt
Si Si Si Si

Surface silanization Surface-initiated ATRP NHS activation Binding of NH2-containing


of sodium acrylate biomolecule

Figure 9. Grafting of biomolecules to the activated carboxyl groups of surface-initiated poly(acrylic acid) chains.
ATRP: Atom transfer radical polymerization.
Adapted with permission from [193] .

1062 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
replicas of pSi can also be formed into micropar- earliest examples was demonstrated by Yoon
ticles via the use of microdroplet patterning [208] et al., who impregnated a fresh pSi membrane
and bead patterning [209] . Structures such as with a ring-opening metathesis precursor and
these can be made from biocompatible and bio- subsequently crosslinked the pSi scaffold with
degradable polymers and could allow for high poly(norbornene), generating a composite
drug loading and, at the same time, self-reporting material that was chemically and mechanically
drug release. very stable [212] . Recently, ATRP has also been
The Sailor group produced a thermo­ used for the surface grafting of polymers such
responsive pSi–PNIPAAm composite by polym- as PMMA [153] . The resulting composites have
erizing a NIPAAm hydrogel into the pores of been studied to determine the effect the porosity
pSi [210] . The temperature-dependent swelling exerts on the molecular weight and polydisper-
of the polymer led to refractive index changes sity during the surface-initiated polymerization.
that were monitored by interferometric reflec- These composites have not yet been studied for
tance spectroscopy. It is conceivable that this their ability to store and release drugs; however,
composite material is infused with drugs and we feel that the expansion of these surface-
used for controlled release triggered by a tem- initiated polymerization techniques with new
perature change that is sustained for a certain polymers, such as those listed in Table 1, could
time period. This could potentially give greater lead to the creation of composites with new and
control over the release of drugs, particularly if interesting properties for applications such as
multiple delivery cycles are required. Mukherjee drug delivery and tissue engineering.
et al. prepared drug-loaded blocks of various
shapes and sizes, approximately 3 mm3, from Micromachined silicon
PCL. pSi microparticles were subsequently Silicon microfabrication techniques allow the
embedded in selected faces of the blocks by production of complex structures with a wide
localized melting of the PCL. The blocks could range of advantages, including very well-defined
be self-assembled into networks by exploiting size, shape and patterns. MEMS are commonly
the hydrophilic attraction of the oxide-ter- produced by the micromachining of complex
minated pSi regions and the use of starch to structures into the surface of single crystalline
help drive the coupling of the enrichment sites.
The composites released the widely studied
model compound Ru(bpy)3Cl 2 at varying rates
depending on the physical structure, spatial
organization, pSi enrichment site and compo-
sition of the polymer blocks (Figure  10) [211] .
It was found that the blocks of Si-containing
cubes are nontoxic to HEK 293 cells.
In the above examples, there is no covalent
bond between the polymer phase and the pSi
scaffold. However, our group recently reported
on a new type of hybrid biodegradable material
composed of a pSi substrate covalently grafted
with PLLA chains [32] . The grafting of PLLA
from the pSi-tethered hydroxyl initiator groups
was studied by various surface analyses, includ-
ing atomic force microscopy, that revealed PLLA
nanobrushes. While no drug-release studies have
been carried out with this material, we anticipate
that these hybrid materials will widen the scope
of applications for biodegradable materials in
medicine and, in particular, will be useful in sit-
uations where drug delivery devices, implants
and scaffolds for tissue engineering with complex
biodegradation behavior are desired. There are a Figure 10. Hexagonal poly(e-caprolactone) blocks enriched with porous
few other examples of ‘grafting-from’ approaches silicon (dark interfacial regions) for the controlled release of Ru(bpy) 3Cl2.
Reproduced with permission from [211] .
that use pSi as the scaffold material. One of the

future science group www.future-science.com 1063


Review | McInnes & Voelcker
silicon wafers to create microscale mechanical
Drug-loaded hyrogel
structures. There have been many recent exam-
Stratum corneum
ples of polymer-free micromachined devices
used for the delivery of drugs [213–217] . These Viable epidermis
devices feature unit operations at the nano- and
micro-scale:
n Nanochannel arrays can be tailored with chan-

nel sizes ranging from 7 to 100 nm to control Diffusion of drug


into epidermis
the release rates of molecules flown across them
in a zero-order manner [213,215,216] . The size of
the channel can be tuned depending on the
drug to be delivered; small channel widths Figure 12. An array of microscale
(7 nm) are useful for controling the release of channels generated by the application of
small organic drugs (~3–5 nm), while larger a microneedle array to the skin. A drug-
channels (13  nm) are suitable for larger loaded hydrogel is then applied to fill the
channels.
molecules such as BSA [213] ; Adapted with permission from [240] .
n Microscale reservoirs can be used to store drugs
prior to delivery and can be opened via the efficient valve systems adapted for drug-delivery
dissolution of a biocompatible capping material applications is currently being explored. Many
such as gold [217,218] , gelatin or starch [15] . Drug researchers have turned to hydrogels  [222] or
release can be stimulated via the electrochem- PDMS [223,224] to generate such valves;
ically triggered dissolution of the gold capping n Chemo- or bio-sensors may be incorporated into
layer [217] ; an implanted micromachined delivery device to
n Micromachined pumps are able to move and detect certain environmental conditions, such
deliver nanolitre amounts of fluids in a con- as pH, blood pressure and the presence of cer-
trolled fashion. These devices have been tain analytes including glucose [15,225,226] . Bio-
reviewed recently [219] and some examples of sensors and bio-MEMS systems and device
pioneering works on implantable micro-pumps fabrication have been extensively reviewed
Microneedle were demonstrated by Ikemoto and Sharpe [220] elsewhere [226–230] .
Micromachined needle that
and Havlik et al. [221] ;
operates by mechanically
When loaded and surgically implanted into
n Valves in conventional MEMS systems can be patients, such devices maximize the local effec-
perforating the epidermis and
allowing for transdermal drug controlled pneumatically or piezoelectrically [15]; tiveness of drug delivery and minimize the toxic
absorption or fluid sampling however, the development of more reliable, effects of fully systemic therapies [3,5,6] .
Micromachined devices, which have featured
prominently in the recent literature, are silicon
microneedles [75,231,232] for transdermal drug
delivery. Microneedle arrays (Figure  11) are a
viable alternative for the delivery of drugs that
are unable to be effectively delivered via oral
ingestion or hypodermic injection. The array of
microneedles disrupts the outer layer of the skin
at multiple sites and increases drug permeabil-
ity through the epidermis into the bloodstream,
thereby increasing the drug-delivery efficiency.
Micro­needle injection is painless as the needles are
not long enough to reach the nerves and induce a
20 kV ×230 100 µm pain response. The therapeutic agent to be deliv-
ered can be coated onto the microneedles before
Figure 11. Fabrication procedure and insertion, applied to the skin as a gel after the
scanning electron microscropy application of the microneedle array (Figure 12)
micrograph of a silicon microneedle array or delivered from an external reservoir in the case
with porous tips. of hollow microneedles [231,233,234] . Microneedles
Reproduced with permission from [232] .
have been reviewed elsewhere [235,236] .

1064 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review

A B
Photoresist
PMMA
Silicon
First mask and
reactive ion etch

Second photoresist
150 µm

C
Second mask and
reactive ion etch

Photoresist and
device removal

PMMA drug-
150 µm
delivery devices

Figure 13. (A) Fabrication process for poly(methylmethacrylate) drug-delivery devices. (B) 150 ×
150 × 3 µm poly(methylmethacrylate) devices with four reservoirs. (C) 150 × 150 × 5 µm PMMA
devices with single reservoirs.
PMMA: Poly(methylmethacrylate).
Adapted from [241] .

Microneedles have also been fabricated with examples act as actuators to control the release
porous tips (Figure 11) that could potentially be of drugs from the device. Tsai et al. have used
used to store increased amounts of drugs and a polypyrrole film to induce a conformational
to safeguard against any biocompatibility issues change on a gold membrane upon the applica-
caused by broken tips [231–233] . New fabrication tion of a voltage. This gold membrane is laid
processes have been used to generate arrays fea- across a micromachined silicon aperture inside
turing both short (400-µm) and long-shanked a PDMS drug reservoir. This system is actuated
(900-µm) microneedles that can simultaneously based on the volume change in the polypyrrole
deliver drugs (short needles) and withdraw blood layer as cations move in and out of the polymer
samples (long needles) [237,238] . layer. When the bias is applied, the gold flap
Microneedles have proven difficult to coat opens and releases the drug to the surrounding
uniformly [239] . As an alternative, drug-loaded area [76] .
polymer gels have been used after application of
the microneedle array. To this end, the hydrogel Micromachined
„„ polymeric
formulation containing therapeutic DNA was drug‑delivery devices
placed into the microscale channels created in As early as 2000, microfabrication processes
the skin after the insertion and removal of the were being applied to biodegradable polymers in
microneedles to the epidermis (Figure 12) . The order to create implantable biomedical micro-
DNA can now diffuse from this reservoir into devices. Armani et  al. used micromachined
the surrounding epidermis [240] . silicon moulds to create 3D microstructures in
PCL, as well as demonstrating that both dry-
„„Polymer-enhanced MEMS and liquid-filled microcavities could be covered
The use of polymers in MEMS is not limited with thin gold films, which could potentially
to biocompatible coatings. On the contrary, be removed via anodic dissolution in biological
the polymers in some of the most interesting tissue [218] .

future science group www.future-science.com 1065


Review | McInnes & Voelcker
A more recent example of a micromachined their final form, they have been created via the use
drug-delivery device comes from Tao et al., who of common silicon micromachining techniques,
used micromachined PMMA layers on silicon such as photolithography and etching, or from
substrates to create microdevices that can poten- micromachined moulds. These polymeric drug
tially be used for oral drug delivery to the GI devices could prove to be advantageous as they
tract. The devices were manufactured to be 150 × will degrade naturally in vivo and avoid any need
150 µm2 and 3-µm thick with multiple reservoirs for surgical retrieval [218] .
(Figure 13) [241] . The devices were also functional-
ized with lectin to facilitate binding to the cells in Conclusion
the GI tract and ensure that the drug is released The preparation of silicon-based monolithic mate-
in close proximity to the intended delivery site. rials for implant applications such as bone regen-
Very recently, biopolymer delivery has been eration can be tailored with various polymers to
attempted with micromachined systems. One ensure that they release therapeutic agents such as
of the most versatile is the particle replication in anti-inflammatories. The incorporation of poly-
the nonwetting templates method pioneered by mers into these silicon-based materials can help to
DeSimone [242,243] . This technique allows for the tune the drug release by modifying the structural
creation of ‘smart particles’ as small as 160 nm properties, such as hydrophobicity and porosity,
with controlled and monodisperse sizes, shapes, which in turn can influence the degradation and
compositions, cargo encapsulation and surface drug-release rate. Polymeric layers can also be used
functionality [242] . Such nanomolded particles as coatings for silicon-based particles to control
can be varied in size and shape via the creation the drug release from the particle; these coatings
of a suitable silicon master template. The pro- can be deposited via LBL techniques, electrostatic
cess allows the encapsulation of biological drugs assembly, encapsulation or covalent polymeriza-
including DNA, proteins and RNA into polymers tion. In some cases, inverse architectures have been
such as PEG, poly(d-lactic acid) and poly(pyrrole). described where silica has been used to coat PDMS
Most importantly, they can be easily released and microparticles and slow down the release of drugs.
dissolved under physiological conditions and used Single crystalline silicon has been modified by
for therapeutic drug-delivery systems [242] . LBL techniques and grafting approaches in a range
Glangchai et  al. have also fabricated drug- of fundamental studies with drug-delivery appli-
loaded nanoparticles with controlled size, shape cations in mind. The in vivo use of these devices
and composition using an imprint lithography is unlikely, as silicon is not biodegradable and its
technique [244] . They have demonstrated a high- low surface area limits drug-loading capabilities.
throughput nanofabrication technique using pSi, however, shares common functionalization
synthetic and biological macromonomers (PEG techniques with single crystalline silicon and ben-
diacrylates, PEG dimethylacrylates and a synthe- efits from a much larger drug-loading capacity.
sized enzymatically degradable peptide [Gly-Phe- The use of pSi in composite production is also
Leu-Gly-Lys-diacrylate]) to produce highly mono- advantageous as it is biodegradable, so no fur-
disperse, enzymatically triggered nanoparticles. ther surgical intervention is required to remove
Biodegradable and nonbiodegradable particles as implantable devices. pSi can be processed into
small as 50 nm were created via imprinting with micro- and nano-particles, which increases the
a quartz template and were harvested directly into range of delivery options.
aqueous buffers using a biocompatible, one-step- Stimuli-responsive polymers integrated into
release technique. The authors also demonstrate silicon-based materials allow environmental
successful encapsulation (via mixing with the stimulus-triggered drug release. Alternatively,
monomer precursor) and enzyme-triggered con- micromachining techniques offer drug release
trolled release of antibodies and nucleic acids from from reservoirs upon electronic actuations.
these nanoparticles. In summary, this review has shown a range of
Micromachining techniques are being improved silicon–polymer hybrid architectures resulting in
constantly, mostly driven by a drive within the materials with properties well suited for controlled
electronics industry to increase feature density drug delivery.
and functionality in order to develop higher-
performing devices. Some of these advancements Future perspective
will no doubt be translated to micromachined The composite structures and devices described
drug-delivery devices. Although the microfabri- above have demonstrated many potential uses
cated devices presented here are fully polymeric in of silicon–polymer hybrid materials for the

1066 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
controlled delivery of small-molecule and macro­ accessible [245] . Another recent example relates to
molecular drugs. In the near future, improvements the demonstration of nanotapes of silica nano-
in surface modification with polymers, proteins, composites combining self-assembly and pep-
antibodies and other site-directing agents could tide-directed silicification of Si(OH) 4 [246,247] .
allow for site-specific targeting and stimuli- Although not yet demonstrated as drug-delivery
responsive drug release from nanoparticle com- devices, the future incorporation of drugs into
positions. Further research into new monomers these new materials could lead to the creation of
and polymers could lead to improved composite new structures or devices that display promising
materials that are capable of better protecting drug-release properties.
therapeutic agents in vivo, while also improving
the release properties. We envision the future use Acknowledgements
of fully biodegradable silicon–polymer composite Our thanks go to Emily Anglin and Frances Harding for
implants that have tailored release characteristics. critical reading of the manuscript.
The progression of silicon- and polymer-based
MEMS devices could lead to the development of Financial & competing interests disclosure
environmentally responsive, pulsatile drug-release Financial support from Flinders University and the Australian
systems that are able to sense the need for a drug Research Council is duly acknowledged. The authors have no
and release the required amount of a therapeutic other relevant affiliations or financial involvement with any
molecule accordingly. organization or entity with a financial interest in or financial
Some very recent newly emerging nanostruc- conflict with the subject matter or materials discussed in the
tured silicon–polymer morphologies have the manuscript. This includes employment, consultancies, hono-
potential to be used for drug-delivery applica- raria, stock ownership or options, expert t­estimony, grants or
tions. For example, polymer brush-coated silica patents received or pending, or royalties.
nanowires have been recently synthesized and No writing assistance was utilized in the production of this
vesicular and tubular topography may also be manuscript.

Executive summary
„„ Single crystalline silicon is a popular platform for the initial design and testing of new silicon–polymer hybrid materials. However, the
inability of the silicon to degrade in vivo renders these devices unsuitable for in vivo applications.
„„ Mesoporous and porous silica are attractive candidates for biomedical applications as they are highly tunable for pore size and

morphology, and they are also easily modifiable with a wide variety of chemical moieties.
„„ The optical properties of porous silicon (pSi) or its polymer replicas are sensitive to the release of molecules from the porous matrices.

This allows for self-reporting of drug release.


„„ Encapsulation of silica and pSi with polymers can help improve biocompatibility and tune the biodegradation and drug-release kinetics,

as well as facilitate the site-targeted release of drugs.


„„ The use of composite materials could help improve drug resistance to first-pass metabolism and degradation in vivo and increase the

bioavailability at the site of delivery.


„„ Microneedles can penetrate the epidermis and allow the painless delivery of therapeutic agents from porous or hollow reservoir or

polymeric coatings.
„„ Microelectromechanical system devices can control the release of drugs via electrically stimulated flaps or the dissolution of

capping layers.
„„ The application of microelectromechanical system technology to drug-delivery devices including features such as reservoirs,

sensors, pumps and valves is producing smart devices with improved control over drug delivery and the possibility of sensor-release
feedback loops.
„„ The development of polymeric devices based on micromachining principles could lead to implantable biodegradable devices that will not

require further surgical removal after their payload is exhausted.

2 Castro G, Panilaitis B, Kaplan D. Emulsan, a 4 Elvira C, Gallardo A, San Roman J,


Bibliography
tailorable biopolymer for controlled release. Cifuentes A. Covalent polymer–drug
Papers of special note have been highlighted as:
n of interest
Bioresour. Technol. 99(11), 4566–4571 conjugates. Molecules 10, 114–125 (2005).
nn of considerable interest
(2008). 5 Sharma S, Jasper Nijdam A, Sinha P et al.
1 Hughes G. Nanostructure-mediated drug 3 Andreadis S, Geer D. Biomimetic approaches Controlled-release microchips. Expert Opin.
delivery. Nanomed. Nanotech. Biol. Med. 1(1), to protein and gene delivery for tissue Drug Deliv. 3(3), 379–394 (2006).
22–30 (2005). regeneration. Trends Biotechnol. 24(7),
331–337 (2006).

future science group www.future-science.com 1067


Review | McInnes & Voelcker
6 Soppimath K, Aminabhavi T, Kulkarni A, 20 Xue J, Tan C, Lukito D. Biodegradable 33 Anglin E, Cheng L, Freeman W, Sailor M.
Rudzinski W. Biodegradable polymeric polymer–silica xerogel composite Porous silicon in drug delivery devices and
nanoparticles as drug delivery devices. microspheres for controlled release of materials. Adv. Drug Delivery Rev. 60,
J. Control. Release. 70(1–2), 1–20 (2001). gentamicin. J. Biomed. Mater. Res. 78B(2), 1266–1277 (2008).
7 Wood K, Boedicker J, Lynn D, Hammond P. 417–422 (2006). 34 Canham L. Properties of Porous Silicon. Short
Tunable drug release from hydrolytically 21 Radin S, El-Bassyouni G, Vresilovic E, Run Press, London, UK (1997).
degradable layer-by-layer thin films. Langmuir Schepers E, Ducheyne P. In vivo tissue 35 Worsfold O, Voelcker N, Nishiya T.
21(4), 1603–1609 (2005). response to resorbable silica xerogels as Biosensing using lipid bilayers suspended on
8 Tsukagoshi T, Kondo Y, Yoshino N. controlled-release materials. Biomaterials porous silicon. Langmuir 22(16), 7078–7083
Preparation of thin polymer films with 26(9), 1043–1052 (2005). (2006).
controlled drug release. Colloids Surf. B 57(2), 22 Prokopowicz M. Silica–polyethylene glycol 36 Alvarez S, Derfus A, Schwartz M, Bhatia S,
219–225 (2007). matrix synthesis by sol–gel method and Sailor M. The compatibility of hepatocytes
9 Brannon-Peppas L. Polymers in controlled evaluation for Diclofenac diethyloammonium with chemically modified porous silicon with
drug delivery. Med. Plastics Biomat. Mag. 1(1), release. Drug Delivery 14, 129–138 (2007). reference to in vitro biosensors. Biomaterials
39–90 (1997). 23 Teoli D, Parisi L, Realdon N et al. Wet 30(1), 26–34 (2009).
10 Vallet-Regi M, Balas F, Arcos D. Mesoporous sol–gel derived silica for controlled release of 37 Coffer J, Montchamp J-L, Aimone J, Weis R.
materials for drug delivery. Angew. Chem. Int. proteins. J. Control. Release 116(3), 295–303 Routes to calcified porous silicon:
Ed. 46(40), 7548–7558 (2007). (2006). implications for drug delivery and biosensing.
11 Narasimhan B. Handbook of Pharmaceutical 24 Margiotta N, Ostuni R, Teoli D et al. Phys. Status Solidi A 197(2), 336–339 (2003).
Controlled Release Technology. Marcel Dekker, Bisphosphonate complexation and calcium 38 Palestino G, Agarwal V, Aulombard R, Perez
NY, USA (2000). doping in silica xerogels as a combined E, Gergely C. Biosensing and protein
strategy for local and controlled release of fluorescence enhancement by functionalized
12 Stubbe B, De Smedt S, Demeester
active platinum antitumor compounds. porous silicon devices. Langmuir 24(23),
J. ‘Programmed polymeric devices’ for pulsed
Dalton Trans. 3131–3139 (2007). 13765–13771 (2008).
drug delivery. Pharm. Res. 21(10), 1732–1740
(2004). 25 Prokopowicz M. In vitro controlled release of 39 Koh Y, Jang S, Kim J et al. DBR PSi/PMMA
doxorubicin from silica xerogels. J. Pharm. composite materials for smart patch
13 Gardner P. Microfabricated nanochannel
Pharmacol. 59, 1365–1373 (2007). application. Colloids Surf. A 313–314,
implantable drug delivery devices. trends,
limitations and possibilities. Expert Opin. 26 Prokopowicz M, Przyjazny A. Synthesis of 328–331 (2008).
Drug Deliv. 3(4), 479–487 (2006). sol–gel mesoporous silica materials providing nn Composites of photonic pSi films and
a slow release of doxorubicin.
14 Salonen J, Kaukonen A, Hirvonen J, Lehto polymers were used as a smart-patch,
J. Microencapsulation 24(7), 694–713 (2007).
V-P. Mesoporous silicon in drug delivery which could allow for the delivery of
applications. J. Pharm. Sci. 97(2), 632–653 27 Lenza R, Jones J, Vasconcelos W, Hench L. drugs to be monitored in real-time for
(2008). In vitro release kinetics of proteins from in vivo applications.
bioactive foams. J. Biomed. Mater. Res.
15 Richards Grayson A, Scheidt Shawgo R, Li Y, 40 Salonen J, Laitinen L, Kaukonen A et al.
67A(1), 121–129 (2003).
Cima M. Electronic MEMS for triggered Mesoporous silicon microparticles for oral
delivery. Adv. Drug Delivery Rev. 56(2), 28 Ducheyne P, Qiu Q. Bioactive ceramics: the drug delivery. Loading and release of five
173–184 (2004). effect of surface reactivity on bone formation model drugs. J. Control. Release 108(2–3),
and bone cell function. Biomaterials 362–374 (2005).
16 Bergna H. Colloidal Silica. Fundamentals and
20(23–24), 2287–2303 (1999).
Applications. Taylor and Francis, London, UK 41 Salonen J, Paski J, Vaha-Heikkila K et al.
(2006). 29 Nicoll S, Radin S, Santos E, Tuan R, Determination of drug load in porous silicon
Ducheyne P. In vitro release kinetics of microparticles by calorimetry. Phys. Status
17 Czarnobaj K, Lukasiak J. In vitro release of
biologically active transforming growth Solidi A 202(8), 1629–1633 (2005).
cisplatin from sol–gel processed organically
factor-b1 from a novel porous glass carrier.
modified silica xerogels. J. Mater. Sci. Mater. 42 Lehto V-P, Vähä-Heikkilä K, Paski J, Salonen
Biomaterials 18(12), 853–859 (1997).
Med. 18(10), 2041–2044 (2007). J. Use of thermoanalytical methods in
30 Sheridan M, Shea L, Peters M, Mooney D. quantification of drug load in mesoporous
18 Roveri N, Morpurgo M, Palazzo B, Parma B, Bioabsorbable polymer scaffolds for tissue
Vivi L. Silica xerogels as a delivery system for silicon microparticles. J. Therm. Anal.
engineering capable of sustained growth Calorim. 80(2), 393–397 (2005).
the controlled release of different molecular factor delivery. J. Control. Release 64(1–3),
weight heparins. Anal. Bioanal. Chem. 381(3), 43 McInnes S, Graney S, Khung Y-L, Voelcker
91–102 (2000).
601–606 (2005). N. Porous silicon microparticles as an
31 Lin S, Claudia I, Pike KJ, Smith ME, Jones alternative support for solid phase DNA
19 Czarnobaj K, Prokopowicz M, Lukasiak JR. Nanostructure evolution and calcium
J. The effect of PEG concentration on the synthesis. Proc. SPIE 6036, 445–454 (2006).
distribution in sol–gel derived bioactive glass.
release rate of cisplatin from PEG-modified 44 Meade S, Sailor M. Microfabrication of
J. Mater. Chem. 19, 1276–1282 (2009).
silica xerogels. Drug Delivery 13, 339–344 freestanding porous silicon particles
32 McInnes S, Thissen H, Choudhury N, containing spectral barcodes. Phys. Status
(2006).
Voelcker N. New biodegradable materials Solidi RRL 1(2), R71–R73 (2007).
n The inclusion of biocompatible polymers produced by ring opening polymerisation of
such as poly(ethyleneglycol) (PEG) has 45 Wu E, Park J-H, Park J et al. Oxidation-
poly(l-lactide) on porous silicon substrates.
allowed for the release rate of cisplatin from triggered release of fluorescent molecules or
J. Colloid Interface Sci. 332(2), 336–344
silica xerogel monoliths to be tuned. drugs from mesoporous Si microparticles.
(2009).
ACS Nano 2(11), 2401–2409 (2008).

1068 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
46 Canham L, Reeves C, King D et al. Bioactive 61 Buriak J. Organometallic chemistry on silicon 76 Tsai H, Ma K, Wang C et al. Development of
polycrystalline silicon. Adv. Mater. 8(10), and germanium surfaces. Chem. Rev. 102(5), integrated protection system for a miniturised
850–852 (1996). 1271–1308 (2002). drug delivery system. Smart Mater. Struct. 16,
47 Low S, Williams K, Canham L, Voelcker N. 62 Frotscher U, Rossow U, Ebert M et al. S295–S299 (2007).
Evaluation of mammalian cell adhesion on Investigation of different oxidation processes 77 Yang J, Lee J, Kang J et al. Hollow silica
surface-modified porous silicon. Biomaterials for porous silicon studied by spectroscopic nanocontainers as drug delivery vehicles.
27(26), 4538–4546 (2006). ellipsometry. Thin Solid Films 276(1–2), Langmuir 24(7), 3417–3421 (2008).
48 Bayliss S, Buckberry L, Harris P, Tobin M. 36–39 (1996). 78 Tang Q, Yao X, Dong W et al. Studies on a
Nature of the silicon–animal cell interface. 63 Salonen J, Lehto V, Laine E. The room new carrier of trimethylsilyl-modified
J. Porous Mat. 7(1), 191–195 (2000). temperature oxidation of porous silicon. Appl. mesoporous material for controlled drug
49 Khung YL, Barritt G, Voelcker NH. Using Surf. Sci. 120(3–4), 191–198 (1997). delivery. J. Control. Release 114(1), 41–46
continuous porous silicon gradients to study 64 Song J, Sailor M. Dimethyl sulfoxide as a mild (2006).
the influence of surface topography on the oxidising agent for porous silicon and its effect 79 Beck J, Vartuli J, Roth W et al. A new family
behaviour of neuroblastoma cells. Exp. Cell on photoluminescence. Inorg. Chem. 37, of mesoporous molecular sieves prepared with
Res. 314, 789–800 (2008). 3355–3360 (1998). liquid crystal templates. J. Am. Chem. Soc. 114,
50 Low SP, Voelcker NH, Canham LT, Williams 65 Mattei G, Alieva E, Petrov J, Yakovlev V. 10834–10843 (1992).
KA. The biocompatibility of porous silicon in Quick oxidation of porous silicon in presence 80 Zhao D, Feng J, Huo Q et al. Triblock
tissues of the eye. Biomaterials 30, 2873–2880 of pyridine vapor. Phys. Status Solidi A 182(1), copolymer syntheses of mesoporous silica with
(2009). 139–143 (2000). periodic 50 to 300 angstrom pores. Science
51 Boukherroub R, Morin S, Wayner D, 66 Halimaoui A, Oules C, Bomchil G et al. 279, 548–552 (1998).
Lockwood D. Thermal route for chemical Electroluminescence in the visible range 81 Huo Q, Liu J, Wang L-Q et al. A new class of
modification and photoluminescence during anodic oxidation of porous silicon silica crosslinked micellar core-shell
stabilization of porous silicon. Phys. Status films. Appl. Phys. Lett. 59(3), 304–306 (1991). nanoparticles. J. Am. Chem. Soc. 128(19),
Solidi A 182(1), 117–121 (2000). 67 Vial J, Bsiesy A, Gaspard F et al. Mechanisms 6447–6453 (2006).
52 Boukherroub R, Morin S, Wayner D et al. of visible-light emission from electro-oxidised nn F127 micelles of ethylene oxide and propylene
Ideal passivation of luminescent porous porous silicon. Phys. Rev. B Condens. Matter oxide (EO106-PO70-EO106) were used to
silicon by thermal, noncatalytic reaction with Mater. Phys. 45(24), 14171–14176 (1992). template TEOS hydrolysis to form core-shell
alkenes and aldehydes. Chem. Mater. 13(6), 68 Tischler M, Collins R, Stathis J, Tsang nanoparticles. These particles showed almost
2002–2011 (2001). J. Luminescence degradation in porous silicon. zero-order release kinetics.
53 Buriak J. Silicon–carbon bonds on porous Appl. Phys. Lett. 60(5), 639–641 (1992). 82 Giri S, Trewyn B, Lin V. Mesoporous silica
silicon surfaces. Adv. Mater. 11(3), 265–267 69 Salonen J, Lehto V-P, Laine E. Photo- nanomaterial-based biotechnological and
(1999). oxidation studies of porous silicon using biomedical delivery systems. Nanomedicine
54 Stewart M, Buriak J. Photopatterned microcalorimetric method. J. Appl. Phys. 2(1), 99–111 (2007).
hydrosilylation on porous silicon. Angew. 86(10), 5888–5893 (1999). 83 Viitala R, Jokinen M, Maunu S, Jalonen H,
Chem. Int. Ed. 37(23), 3257–3260 (1998). 70 Anderson R, Muller R, Tobias C. Chemical Rosenholm J. Chemical characterization of
55 Stewart M, Buriak J. Exciton-mediated surface modification of porous silicon. bioresorbable sol–gel derived SiO2 matrices
hydrosilylation on photoluminescent J. Electrochem. Soc. 140, 1393 (1993). prepared at protein-compatible pH. J. Non-
nanocrystalline silicon. J. Am. Chem. Soc. 71 Kimoto K, Arai T. Photoluminescence of rapid Cryst. Solids 351(40–42), 3225–3234 (2005).
123(32), 7821–7830 (2001). thermal treated porous silicon in nitrogen 84 Barbe C, Kong L, Finnie K et al. Sol–gel
56 Buriak J, Allen M. Photoluminescence of atmosphere. Phys. Status Solidi A 182, 133 matricies for controlled release: from macro to
porous silicon surfaces stabilized through (2000). nano using emulsion polymerisation. J. Sol–gel
Lewis acid mediated hydrosilylation. 72 Schmeltzer J, Buriak J. The Chemistry of Sci. Technol. 46, 393–409 (2008).
J. Lumin. 80(1–4), 29–35 (1998). Nanomaterials. Synthesis, Properties and 85 Hwang Y-J, Oh C, Oh S-G. Controlled release
57 Buriak J, Stewart M, Geders T et al. Lewis Applications (Volume 2). Wiley-VCH, of retinol from silica particles prepared in
acid mediated hydrosilylation on porous Weinheim, Germany (2004). O/W/O emulsion: the effects of surfactants
silicon surfaces. J. Am. Chem. Soc. 121(49), 73 Cheng L, Anglin E, Cunin F et al. Intravitreal and polymers. J. Control. Release 106(3),
11491–11502 (1999). properties of porous silicon photonic crystals. a 339–349 (2005).
58 Holland J, Stewart M, Allen M, Buriak J. potential self-reporting intraocular drug 86 Shen S, Chow P, Chen F, Tan R. Submicron
Metal mediated reactions on porous silicon delivery vehicle. Brit. J. Opthalmol. 92, particles of SBA-15 modified with MgO as
surfaces. J. Solid State Chem. 147(1), 251–258 705–711 (2008). carriers for controlled drug delivery. Chem.
(1999). 74 Veiseh M, Wickes B, Castner D, Zhang M. Pharm. Bull. 55(7), 985–991 (2007).
59 Boukherroub R, Petit A, Loupy A, Chazalviel Guided cell patterning on gold–silicon dioxide 87 Wen L-X, Li Z-Z, Zou H-K, Liu A-Q,
J-N, Ozanam F. Microwave-assisted chemical substrates by surface molecular engineering. Chen J-F. Controlled release of avermectin
functionalization of hydrogen-terminated Biomaterials. 25(16), 3315–3324 (2004). from porous hollow silica nanoparticles. Pest
porous silicon surfaces. J. Phys. Chem. B 75 Ji J, Tay F, Miao J, Iliescu C. Microfabricated Manage. Sci. 61(6), 583–590 (2005).
107(48), 13459–13462 (2003). microneedle with porous tip for drug delivery. 88 Begu S, Pouessel A, Lerner D, Tourne-Peteilh
60 Stewart M, Buriak J. Chemical and biological J. Micromech. Microeng. 16(5), 958–964 C, Devoisselle J. Liposil, a promising
applications of porous silicon technology. (2006). composite material for drug storage and
Adv. Mater. 12(12), 859–869 (2000). release. J. Control. Release 118(1), 1–6 (2007).

future science group www.future-science.com 1069


Review | McInnes & Voelcker
89 Li Z-Z, Wen L-X, Shao L, Chen J-F. 101 Simovic S, Prestidge C. Nanoparticle layers 111 Xia W, Chang J. Well-ordered mesoporous
Fabrication of porous hollow silica controlling drug release from emulsions. bioactive glasses (MBG): A promising
nanoparticles and their applications in drug Eur. J. Pharmaceutics Biopharmaceutics bioactive drug delivery system. J. Control.
release control. J. Control. Release 98(2), 67(1), 39–47 (2007). Release 110(3), 522–530 (2006).
245–254 (2004). 102 Beck R, Pohlmann A, Guterres S. 112 Zhao L, Yan X, Zhou X et al. Mesoporous
90 Li Z-Z, Xu S-A, Wen L-X et al. Nanoparticle-coated microparticles: bioactive glasses for controlled drug release.
Controlled release of avermectin from preparation and characterisation. Microporous Mesoporous Mat. 109(1–3),
porous hollow silica nanoparticles: influence J. Microencapsulation 21(5), 499–512 210–215 (2008).
of shell thickness on loading efficiency, (2004). 113 Cauda V, Fiorilli S, Onida B et al. SBA-15
UV-shielding property and release. 103 Song S-W, Hidajat K, Kawi S. pH- ordered mesoporous silca inside a bioactive
J. Control. Release 111(1–2), 81–88 (2006). controllable drug release using hydrogel glass–ceramic scaffold for local drug delivery.
91 Carino I, Pasqua L, Testa F et al. Silica- encapsulated mesoporous silica. Chem. J. Mater. Sci. Mater. Med. 19, 3303–3310
based mesoporous materials as drug delivery Commun. 4396–4398 (2007). (2008).
system for Methotrexate release. Drug nn Bovine serum albumin (BSA) was loaded n Mesoporous silica (SBA-15) was precipitated
Delivery 14, 491–495 (2007).
into amino-modified SBA-15 and the into bioactive glass-ceramic (SiO2-CaO-K2O)
92 Charnay C, Begu S, Tourne-Peteilh C et al. particles were encapsulated via electrostatic scaffolds. This material could find application
Inclusion of ibuprofen in mesoporous assembly with poly(acrylic acid) (PAA). in implantable sustained release devices.
templated silica: drug loading and release This technique could enhance the delivery 114 Ladron de Guevara-Fernandez S, Ragel CV,
property. Eur. J. Pharmaceutics of peptide and protein drugs. Vallet-Regi M. Bioactive glass–polymer
Biopharmaceutics 57, 533–540 (2004).
104 Trotter H, Zaman A, Partch R. Preparation materials for controlled release of ibuprofen.
93 Wang G-H, Zhang L-M. Manipulating Biomaterials 24(22), 4037–4043 (2003).
and characterization of polymer composite
formation and drug-release behavior of new multilayers on SiO2 . J. Colloid Interface Sci. 115 Martin A, Salinas A, Vallet-Regi M. Bioactive
sol–gel silica matrix by hydroxypropyl guar 286(1), 233–238 (2005). and degradable organic–inorganic hybrids.
gum. J. Phys. Chem. B 111(36), 10665–
105 Zhu Y, Shi J, Shen W et al. Stimuli- J. Eur. Ceram. Soc. 25(16), 3533–3538
10670 (2007).
responsive controlled drug release from a (2005).
94 Balas F, Manzano M, Colilla M,
hollow mesoporous silica sphere/ n Bioactive glasses coated with poly(vinyl
Vallet‑Regi M. l-Trp adsorption into silica polyelectrolyte multilayer core-shell
mesoporous materials to promote bone alcohol) (PVA) were shown to induce the
structure. Angew. Chem. Int. Ed. 44(32), formation of apatite layers. Composite
formation. Acta Biomaterialia 4(3), 514–522 5083–5087 (2005).
(2008). degradation and drug release could be
106 He J, Chen J-Y, Wang P et al. Poly(N- controlled by addition of more P 2O5
95 Roy I, Ohulchanskyy TY, Bharali DJ et al.
isopropylacrylamide)-coated thermo- or PVA.
Optical tracking of organically modified responsive nanoparticles for controlled
silica nanoparticles as DNA carriers: a 116 Balas F, Manzano M, Horcajada P, Vallet-
delivery of sulfonated Zn-phthalocyanine in
nonviral, nanomedicine approach for gene Regi M. Confinement and controlled release
Chinese hamster ovary cells in vitro and
delivery. PNAS 102(2), 279–284 (2005). of bisphosphonates on ordered mesoporous
zebra fish in vivo. Nanotechnology 18(41),
silica-based materials. J. Am. Chem. Soc.
96 Chen J-F, Ding H-M, Wang J-X, Shao L. 415101 (2007).
Preparation and characterization of porous 128(25), 8116–8117 (2006).
n Nanoparticles composed of CdTe,
hollow silica nanoparticles for drug delivery 117 Britcher L, Barnes TJ, Griesser HJ, Prestidge
poly(N-isopropylacrylamide) and Fe3O4
application. Biomaterials 25(4), 723–727 CA. PEGylation of porous silicon using click
have been used to deliver water-soluble,
(2004). chemistry. Langmuir 24(15), 7625–7627
photosensitizing drugs for (2008).
97 Zhou F, Li S, Vo C et al. Biomimetic
photodynamic therapy.
deposition of silica templated by a cationic 118 Boulmedais F, Frisch B, Etienne O et al.
107 Cole M, Jasieniak M, Voelcker N et al. Polyelectrolyte multilayer films with
polyamine-containing microgel. Langmuir
23(19), 9737–9744 (2007). Switchable surface coatings for control over pegylated polypeptides as a new type of
protein adsorption. Biomed. App. Micro. anti-microbial protection for biomaterials.
98 Yuan J-J, Mykhaylyk O, Ryan A, Armes S.
Nanoeng. III Proc SPIE 6416, 6–10 (2006). Biomaterials 25(11), 2003–2011 (2004).
Crosslinking of cationic block copolymer
108 Lei Z, Bi S. Preparation of polymer 119 Prakash S, Long TM, Selby JC, Moore JS,
micelles by silica deposition. J. Am. Chem.
Soc. 129(6), 1717–1723 (2007). nanocomposites of core–shell structure via Shannon MA. “Click” modification of silica
surface-initiated atom transfer radical surfaces and glass microfluidic channels.
99 Lee KE, Kim BK, Yuk SH. Biodegradable
polymerizations. Mater. Lett. 61(16), Anal. Chem. 79(4), 1661–1667 (2006).
Polymeric Nanospheres Formed by 3531–3534 (2007).
Temperature-Induced Phase Transition in a 120 Ostaci R-V, Damiron D, Capponi S et al.
109 Bi S, Wei X, Li N, Lei Z. In situ formation Polymer brushes grafted to “passivated”
Mixture of Poly(lactide-co-glycolide) and
Poly(ethylene oxide)-Poly(propylene of Fe3O 4 nanoparticles within the silicon substrates using click chemistry.
oxide)-Poly(ethylene oxide) Triblock thermosensitive hairy hybrid particles. Langmuir 24(6), 2732–2739 (2008).
Copolymer. Biomacromolecules 3(5), Mater. Lett. 62(17–18), 2963–2966 (2008).
121 Yague C, Moros M, Grazu V, Arruebo M,
1115–1119 (2002). 110 Bikram M, Gobin A, Whitmire R, West Santamaria J. Synthesis and stealthing study
100 Prestidge C, Simovic S. Nanoparticle
J. Temperature-sensitive hydrogels with of bare and PEGylated silica micro- and
encapsulation of emulsion droplets. Int. SiO2 –Au nanoshells for controlled drug nanoparticles as potential drug-delivery
J. Pharm. 324(1), 92–100 (2006). delivery. J. Control. Release 123, 219–227 vectors. Chem. Eng. J. 137(1), 45–53 (2008).
(2007).

1070 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
122 Wattendorf U, Merkle HP. PEGylation as a 134 Heuberger R, Sukhorukov G, Voros J, 146 Goh S, Murthy N, Xu M, Frechet
tool for the biomedical engineering of Textor M, Mohwald H. Biofunctional J. Crosslinked microparticles as carriers for
surface modified microparticles. J. Pharm. polyelectrolyte multilayers and the delivery of plasmid DNA for vaccine
Sci. 97(11), 4655–4669 (2008). microcapsules: control of non-specific and development. Bioconjugate Chem. 15(3),
123 Jain A, Jain SK. PEGylation: an approach for bio-specific protein adsorption. Adv. Funct. 467–474 (2004).
drug delivery. A review. Crit. Rev. Ther. Mater. 15(3), 357–366 (2005). 147 Yoshikawa C, Goto A, Ishizuka N et al.
Drug Carrier Sys. 25(5), 403–447 (2008). 135 Chuang H, Smith R, Hammond P. Size-exclusion effect and protein repellency of
124 Lin M, Wang H, Meng S et al. Structure and Polyelectrolyte multilayers for tunable concentrated polymer brushes prepared by
release behavior of PMMA/silica composite release of antibiotics. Biomacromolecules surface-initiated living radical polymerization.
drug delivery system. J. Pharm. Sci. 96(6), 9(6), 1660–1668 (2008). Macromol. Symposia 248(1), 189–198 (2007).
1518–1526 (2007). 136 Kurkuri M, Nussio M, Deslandes A, 148 Edmondson S, Vo C-D, Armes SP, Unali G-F,
125 Bereron E, Marquis M, Chretien I, Faucheux Voelcker N. Thermosensitive copolymer Weir MP. Layer-by-layer deposition of
N. Differentiation of preosteoblasts using a coatings with enhanced wettability polyelectrolyte macroinitiators for enhanced
delivery system with BMPs and bioactive switching. Langmuir 24(8), 4238–4244 initiator density in surface-initiated ATRP.
glass microspheres. J. Mater. Sci. Mater. Med. (2008). Langmuir 24(14), 7208–7215 (2008).
18, 255–263 (2007). 137 LeMieux M, Peleshanko S, Anderson K, 149 Remzi-Becer C, Haensch C, Hoeppener S,
126 Peng P, Voelcker N, Kumar S, Griesser H. Tsukruk V. Adaptive nanomechanical Schubert U. Patterned polymer brushes
Nanoscale eluting coatings based on response of stratified polymer brush grafted from bromine-functionalized,
alginate/chitosan hydrogels. Biointerphases structures. Langmuir 23(1), 265–273 chemically active surface templates. Small
2(2), 95–104 (2007). (2007). 3(2), 220–225 (2007).

127 Wang L, Wang X, Xu M, Chen D, 138 Mal N, Fujiwara M, Tanaka Y. 150 Wang T-L, Ou C-C, Yang C-H. Synthesis and
Sun J. Layer-by-layer assembled microgel Photocontrolled reversible release of guest properties of organic/inorganic hybrid
films with high loading capacity: reversible molecules from coumarin-modified nanoparticles prepared using atom transfer
loading and release of dyes and mesoporous silica. Nature 421, 350–353 radical polymerization. J. Appl. Polym. Sci.
nanoparticles. Langmuir 24(5), 1902–1909 (2003). 109(5), 3421–3430 (2008).
(2008). 139 Saito K, Ingalls L, Lee J, Warner J. Core- 151 Uekusa T, Nagano S, Seki T. Unique
128 Lu Z, Li C, Zhou Q, Bao Q-L, Cui X. bound polymeric micellar system based on molecular orientation in a smectic liquid
Covalently linked DNA/protein multilayered photocrosslinking of thymine. Chem. crystalline polymer film attained by
film for controlled DNA release. J. Colloid Commun. 24, 2503–2505 (2007). surface-initiated graft polymerization.
Interface Sci. 314(1), 80–88 (2007). 140 Kim T, Cook S, Arote R et al. A degradable Langmuir 23(8), 4642–4645 (2007).

n Crosslinked LBL multilayered films of hyperbranched poly(ester amine) based on 152 Uekusa T, Nagano S, Seki T. Highly ordered
poloxamer diacrylate and polyethylenimine in-plane photoalignment attained by the brush
ssDNA and BSA on silicon wafers were
as a gene carrier. Macromol. Biosci. 7(5), architecture of liquid crystalline azobenzene
used for the controlled release of DNA. The
611–619 (2007). polymer. Macromol. 42(1), 312–318 (2009).
dissolution could be activated and
controlled by proteinase K. 141 Arote R, Hwang S-K, Yoo M-K et al. 153 Gorman CB, Petrie RJ, Genzer J. Effect of
Biodegradable poly(ester amine) based on substrate geometry on polymer molecular
129 Vazquez E, Dewitt D, Hammond P, Lynn D.
glycerol dimethacrylate and weight and polydispersity during surface-
Construction of hydrolytically-degradable
polyethylenimine as a gene carrier. J. Gene initiated polymerization. Macromol. 41(13),
thin films via layer-by-layer deposition of
Med. 10(11), 1223–1235 (2008). 4856–4865 (2008).
degradable polyelectrolytes. J. Am. Chem.
Soc. 124(47), 13992–13993 (2002). 142 Ozen K, Elmas B, Ozsar O, Senel S, Tuncel 154 Cringus-Fundeanu I, Luijten J, van der Mei
A. Thermoresponsive-fluorescent HC, Busscher HJ, Schouten AJ. Synthesis and
130 Zhang J, Chua L, Lynn D. Multilayered thin
isopropylacrylamide–vinylpyridine– characterization of surface-grafted
films that sustain the release of functional
pyrenemethylacrylamide terpolymer and its polyacrylamide brushes and their inhibition of
DNA under physiological conditions.
complexation behaviour with DNA. React. microbial adhesion. Langmuir 23(9),
Langmuir 20(19), 8015–8021 (2004).
Funct. Polym. 68(2), 623–633 (2008). 5120–5126 (2007).
131 Jewell C, Zhang J, Fredin N et al. Release of
143 Braunova A, Pechar M, Laga R, Ulbrich K. 155 Feng W, Zhu S, Ishihara K, Brash JL. Protein
plasmid DNA from intravascular stents
Hydrolytically and reductively degradable resistant surfaces: comparison of acrylate graft
coated with ultrathin multilayered
high-molecular-weight poly(ethylene glycol) polymers bearing oligo-ethylene oxide and
polyelectrolyte films. Biomacromolecules 7(9),
s. Macromol. Chem. Phys. 208(24), phosphorylcholine side chains. Biointerphases
2483–2491 (2006).
2642–2653 (2007). 1(1), 50–60 (2006).
132 Zhang J, Lynn D. Multilayered films
144 Kallinteri P, Higgins S, Hutcheon G, 156 Stadler V, Beyer M, Konig K et al.
fabricated from combinations of degradable
St Pourcain C, Garnett M. Novel Multifunctional CMOS microchip coatings
polyamines: tunable erosion and release of
functionalized biodegradable polymers for for protein and peptide arrays. J. Proteome Res.
anionic polyelectrolytes. Macromolecules
nanoparticle drug delivery systems. 6(8), 3197–3202 (2007).
39(26), 8928–8935 (2006).
Biomacromol. 6(4), 1885–1894 (2005). 157 Gao X, Feng W, Zhu S, Sheardown H, Brash
133 Zhang Z, Cao X, Zhao X et al. Controlled
145 Wong L, Boyer C, Jia Z et al. Synthesis of JL. A facile method of forming nanoscale
delivery of antisense oligodeoxynucleotide
versatile thiol-reactive polymer scaffolds via patterns on poly(ethylene glycol)-based
from cationically modified
R AFT polymerization. Biomacromol. 9(7), surfaces by self-assembly of randomly grafted
phosphorylcholine polymer films.
1934–1944 (2008). block copolymer brushes. Langmuir 24(15),
Biomacromolecules 7(3), 784–791 (2006).
8303–8308 (2008).

future science group www.future-science.com 1071


Review | McInnes & Voelcker
158 He X, Yang W, Pei X. Preparation, 171 Walters KB, Hirt DE. Synthesis and 182 Voicu R, Boukherroub R, Bartzoka V et al.
characterization, and tunable wettability of characterization of a tertiary amine polymer Formation, characterization, and chemistry
poly(ionic liquid) brushes via surface- series from surface-grafted poly(tert-butyl of undecanoic acid-terminated silicon
initiated atom transfer radical acrylate) via diamine reactions. Macromol. surfaces: patterning and immobilization of
polymerization. Macromol. 41(13), 40(14), 4829–4838 (2007). DNA. Langmuir 20(26), 11713–11720
4615–4621 (2008). 172 Yuan K, Li Z-F, Li L-L, Shi X-N. Synthesis (2004).
159 Slim C, Tran Y, Chehimi MM et al. and characterization of well-defined polymer 183 Bocking T, Kilian K, Gaus K,
Microelectrochemical patterning of surfaces brushes grafted from silicon surface via Gooding J. Single-step DNA immobilization
with polymer brushes. Chem. Mat. 20(21), surface reversible addition-fragmentation on antifouling self-assembled monolayers
6677–6685 (2008). chain transfer (RAFT) polymerization. Mat. covalently bound to silicon(111). Langmuir
160 Madkour AE, Dabkowski JM, Nusslein K, Lett. 61(10), 2033–2036 (2007). 22(8), 3494–3496 (2006).
Tew GN. Fast disinfecting antimicrobial 173 Wang L-P, Wang Y-P, Yuan K, Lei Z-Q. 184 Hong Q, Rogero C, Lakey J et al.
surfaces. Langmuir 25(2), 1060–1067 Synthesis and characterization of surface- Immobilisation of proteins at silicon surfaces
(2009). initiated polymer brushes on silicon substrates using undecenylaldehyde: demonstration of
161 Rakhmatullina E, Braun T, Kaufmann T by reversible addition fragmentation chain the retention of protein functionality and
et al. Functionalization of gold and silicon transfer polymerization. Polymers Adv. Tech. detection strategies. Analyst 134, 593–601
surfaces by copolymer brushes using 19(4), 285–290 (2008). (2009).
surface-initiated ATRP. Macromol. Chem. 174 Li D, Luo Y, Li B-G, Zhu S. Effect of rate 185 Schlossbauer A, Schaffert D, Kecht J, Wagner
Phys. 208(12), 1283–1293 (2007). retardation in RAFT grafting polymerization E, Bein T. Click chemistry for high-density
162 Zhu Y, Shi M, Wu X, Yang S. Amphiphilic from silicon wafer surface. J. Polymer Sci. A biofunctionalization of mesoporous silica.
copolymer grafted “smart surface” enhanced Polymer Chem. 46(3), 970–978 (2008). J. Am. Chem. Soc. 130(38), 12558–12559
by surface roughness. J. Colloid Interface Sci. 175 Rowe MD, Hammer BAG, Boyes SG. (2008).
315(2), 580–587 (2007). Synthesis of surface-initiated stimuli- 186 Lie L, Patole S, Pike A et al. Immobilisation
163 Reichelt S, Gohs U, Simon F et al. responsive diblock copolymer brushes and synthesis of DNA on Si(111),
Immobilization of a hyperbranched polyester utilizing a combination of ATRP and RAFT nanocrystalline porous silicon and silicon
via grafting-to and electron beam irradiation. polymerization techniques. Macromol. 41(12), nanoparticles. Faraday Disc. 125, 235–249
Langmuir 24(17), 9392–9400 (2008). 4147–4157 (2008). (2004).

164 Reichelt S, Jakisch L, Simon F et al. Two 176 Ishida N, Biggs S. Salt-induced structural 187 Furbert P, Lu C, Winograd N, DeLouise L.
routes for immobilization of a hyperbranched behavior for poly(N-isopropylacryamide) Label-free optical detection of peptide
OH-terminated polyester on a silicon surface. grafted onto solid surface observed directly by synthesis on a porous silicon scaffold/sensor.
Macromol. Symposia 254(1), 240–247 (2007). AFM and QCM-D. Macromol. 40(25), Langmuir 24(6), 2908–2915 (2008).
9045–9052 (2007). 188 Mosse W, Koppens M, Gengenbach T et al.
165 Feng L, Gu G, Chen M, Wu L. Synthesis and
surface properties of poly(methyl 177 Lewis GT, Cohen Y. Controlled nitroxide- Peptides rafted from solids for the control of
methacrylate)/poly(ethylene glycol) binary mediated styrene surface graft polymerization interfacial properties. Langmuir 25(3),
brushes. Macromol. Mat. Eng. 292(6), with atmospheric plasma surface activation. 1488–1494 (2009).
754–761 (2007). Langmuir 24(22), 13102–13112 (2008). 189 Feng C, Zhang Z, Forch R et al. Reactive
166 Ostaci R-V, Damiron D, Capponi S et al. 178 Brinks M, Hirtz M, Chi L, Fuchs H, Studer thin polymer films as platforms for the
Polymer brushes grafted to passivated silicon A. Site-selective surface-initiated immobilization of biomolecules.
substrates using click chemistry. Langmuir polymerization by Langmuir-Blodgett Biomacromolecules 6(6), 3243–3251 (2005).
24(6), 2732–2739 (2008). lithography. Angew. Chem. Int. Ed. 46(27), 190 Yao Y, Ma Y-Z, Qin M et al. NHS-ester
5231–5233 (2007). functionalized poly(PEGMA) brushes on
167 Dyer MA, Ainslie KM, Pishko MV. Protein
adhesion on silicon-supported hyperbranched 179 Samanta S, Locklin J. Formation of silicon surface for covalent protein
poly(ethylene glycol) and poly(allylamine) photochromic spiropyran polymer brushes via immobilization. Colloids Surf. B 66(2),
thin films. Langmuir 23(13), 7018–7023 surface-initiated, ring-opening metathesis 233–239 (2008).
(2007). polymerization: reversible photocontrol of 191 Park S, Lee K-B, Choi I, Langer R, Jon S.
wetting behavior and solvent dependent Dual functional, polymeric self-assembled
168 Howarter J, Youngblood J. Self-cleaning and
morphology changes. Langmuir 24(17), monolayers as a facile platform for
anti-fog surfaces via stimuli-responsive
9558–9565 (2008). construction of patterns of biomolecules.
polymer brushes. Adv. Mater. 19(22),
3838–3843 (2007). 180 Charlot A, Gabriel S, Detrembleur C, Jerome Langmuir 23(22), 10902–10905 (2007).
R, Jerome C. Combination of electrografting 192 Azioune A, Ben Slimane A, Ait Hamou L
169 Segal E, Perelman LA, Moore T, Kesselman
and layer-by-layer deposition: an efficient way et al. Synthesis and characterization of active
E, Sailor M. Grafting stimuli-responsive
to tailor polymer coatings of (semi)- ester-functionalized polypyrrole-silica
polymer brushes to freshly etched porous
conductors. Chem. Commun. 4656–4658 nanoparticles: application to the covalent
silicon. Phys. Status Solidi C 6(7), 1717–1720
(2007). attachment of proteins. Langmuir 20(8),
(2009).
181 Ghorbal A, Grisotto F, Laude M, Charlier J, 3350–3356 (2004).
170 Synytska A, Stamm M, Diez S, Ionov L.
Palacin S. The in situ characterization and 193 Dong R, Krishnan S, Baird B, Lindau M,
Simple and fast method for the fabrication of
structuring of electrografted polyphenylene Ober C. Patterned biofunctional poly(acrylic
switchable bicomponent micropatterned
films on silicon surfaces. An AFM and XPS acid) brushes on silicon surfaces.
polymer surfaces. Langmuir 23(9),
study. J. Colloid Interface Sci. 328(2), Biomacromol. 8(10), 3082–3092 (2007).
5205–5209 (2007).
308–313 (2008).

1072 Future Med. Chem. (2009) 1(6) future science group


Silicon–polymer hybrid materials for drug delivery | Review
194 Ciampi S, Bocking T, Kilian K, Harper J, 208 Li YY, Kollengode VS, Sailor MJ. Porous- 222 Eddington D, Beebe D. Flow control with
Gooding J. Click chemistry in mesoporous silicon/polymer nanocomposite photonic hydrogels. Adv. Drug Delivery Rev. 56(2),
materials: functionalization of porous crystals formed by microdroplet patterning. 199–210 (2004).
silicon rugate filters. Langmuir 24(11), Adv. Mater. 17(10), 1249–1251 (2005). 223 Unger M, Chou H-P, Thorsen T, Scherer A,
5888–5892 (2008). 209 Park J, Meade S, Segal E, Sailor M. Porous Quake S. Monolithic microfabricated valves
195 Kolb HC, Finn MG, Sharpless KB. Click silicon-based polymer replicas formed by bead and pumps by multilayer soft lithography.
chemistry: diverse chemical function from a patterning. Phys. Status Solidi A 204(5), Science 288, 113–116 (2000).
few good reactions. Angew. Chem. Int. Ed 40, 1383–1387 (2007). 224 Jeon N, Chiu D, Wargo C et al. Design and
2004–2021 (2001). 210 Segal E, Perelman L, Cunin F et al. fabrication of intergrated passive valve
196 Chen L, Rengifo H, Grigoras C et al. Confinement of thermoresponsive hydrogels in pumps for flexible polymer 3-dimensional
Spin-on end-functional diblock copolymers nanostructured porous silicon dioxide microfluidic systems. Biomed. Microdevices
for quantitative DNA immobilization. templates. Adv. Funct. Mater. 17(7), 4, 117–121 (2002).
Biomacromol. 9(9), 2345–2352 (2008). 1153–1162 (2007). 225 Adrega T, Prazeres D, Chu V,
197 Burnham M, Turner J, Szarowski D, Martin 211 Mukherjee P, Whitehead M, Senter R et al. Conde J. Thin‑film silicon MEMS DNA
D. Biological functionalization and surface Biorelevant mesoporous silicon/polymer sensors. J. Non-Cryst. Solids 352(9–20),
micropatterning of polyacrylamide composites: directed assembly, dissassembly, 1999–2003 (2006).
hydrogels. Biomat. 27(35), 5883–5891 and controlled release. Biomed. Microdevices 8, 226 Moser I. Multi-parameter BioMEMS for
(2006). 9–15 (2006). clinical monitoring. Microsystems 16, 15–39
198 Canham LT. Bioactive silicon structure 212 Yoon M, Ahn K, Cheung R et al. Covalent (2007).
fabrication through nanoetching techniques. crosslinking of 1-D photonic crystals of 227 Koschinsky T, Heinemann L. Sensors for
Adv. Mat. 7(12), 1033–1037 (1995). microporous Si by hydrosilylation and glucose monitoring: technical and clinical
199 Anglin E, Schwartz M, Ng V, Perelman L, ring-opening metathesis polymerization. aspects. Diabetes Metab. Res. Rev. 17,
Sailor M. Engineering the chemistry and Chem. Commun. 6, 680–681 (2003). 113–123 (2001).
nanostructure of porous silicon Fabry-Perot 213 Martin F, Walczak R, Boiarski A et al. 228 McNichols R, Cote G. Optical glucose
films for loading and release of a steroid. Tailoring width of microfabricated sensing in biological fluids: an overview.
Langmuir 20(25), 11264–11269 (2004). nanochannels to solute size can be used to J. Biomed. Opt. 5, 5–16 (2000).
200 Commandeur S, Van Beusekom H, Van Der control diffusion kinetics. J. Control. Release
229 Chaniotakis N, Sofikiti N. Novel
Giessen W. Polymers, drug release, and 102(1), 123–133 (2005).
semiconductor materials for the
drug-eluting stents. J. Interventional Cardiol. 214 Neeves K, Lo C, Foley C, Saltzman W, development of chemical sensors and
19(6), 500–506 (2006). Olbricht W. Fabrication and characterization biosensors: a review. Anal. Chim. Acta
201 Fujii M, Minobe S, Usui M et al. Generation of microfluidic probes for convection enhanced 615(1), 1–9 (2008).
of singlet oxygen at room temperature drug delivery. J. Control. Release 111(3),
230 Chilcott T, Coster H, Boecking T.
mediated by energy transfer from 252–262 (2006).
Organic–silicon interface. Biophys. Rev.
photoexcited porous Si. Phys. Rev. B Condens. 215 Lesinski G, Sharma S, Varker K et al. Release Lett. 2(2), 191–205 (2007).
Matter Mater. Phys. 70(8), 85311 (2004). of biologically functional interferon-a from a
231 Rajaraman S, Henderson H. A unique
202 Kleps I, Danila M, Angelescu A et al. Gold nanochannel delivery system. Biomed.
fabrication approach for microneedles using
and silver/Si nanocomposite layers. Mater. Microdevices 7(1), 71–79 (2005).
coherent porous silicon technology. Sens.
Sci. Eng. C 27(5–8), 1439–1443 (2007). 216 Prasad S, Quijano J. Nanoporous noninvasive Actuators. B 105(2), 443–448 (2005).
203 Batra I, Coffer J, Canham L. Electronically- cellular electrical activity-based analysis
232 Ji J, Tay F, Miao J, Iliescu C.
responsive delivery from a calcified devices. J. Ass. Lab. Automation 11(2), 65–74
Microfabricated silicon microneedle array
mesoporous silicon structure. Biomed. (2006).
for transdermal drug delivery. J. Physics
Microdevices 8, 93–97 (2006). 217 Li Y, Shawgo R, Tyler B et al. In vivo release Conf. Series 34, 1127–1131 (2006).
204 Coffer J, Whitehead M, Nagesha D et al. from a drug delivery MEMS device. J. Control.
233 Rodriguez A, Molinero D, Valera E et al.
Porous silicon-based scaffolds for tissue Release 100(2), 211–219 (2004).
Fabrication of silicon oxide microneedles
engineering and other biomedical 218 Armani D, Liu C. Microfabrication technology from macroporous silicon. Sens. Actuators B
applications. Phys. Status Solidi A 202(8), for polycaprolactone, a biodegradable polymer. 109(1), 135–140 (2005).
1451–1455 (2005). J. Micromech. Microeng. 10, 80–84 (2000).
234 Ma B, Liu S, Gan Z et al. A PZT insulin
205 Wu J, Sailor MJ. Chitosan hydrogel-capped 219 Nisar A, Afzulpurkar N, Mahaisavariya B, pump integrated with a silicon microneedle
porous SiO2 as a pH responsive nano-valve Tuantranont A. MEMS-based micropumps in array for transdermal drug delivery.
for triggered release ofinsulin. Adv. Funct. drug delivery and biomedical applications. Microfluid Nanofluid 2, 417–423 (2006).
Mater. 19(5), 733–741 (2009). Sens. Actuators. B 130(2), 917–942 (2008).
235 McAllister DV, Allen MG, Prausnitz MR.
206 Li Y, Cunin F, Link J et al. Polymer replicas 220 Ikemoto S, Sharpe L. A head-attachable device Microfabricated microneedles for gene and
of photonic porous silicon for sensing and for injecting nanoliter volumes of drug drug delivery. Annu. Rev. Biomed. Eng. 2,
drug delivery applications. Science 299, solutions into brain sites of freely moving rats. 289–313 (2000).
2045–2047 (2003). J. Neurosci. Meth. 110(1–2), 135–140 (2001).
236 Chabri F, Bouris K, Jones T et al.
207 Kim J, Park C, Kim SJ et al. Fabrication and 221 Havlik R, Kerkhoffs W, Jiao L et al. Microfabricated silicon microneedles for
optical characterisation of rugate-structured Intravascular micropump for augmented liver nonviral cutaneous drug delivery. Brit.
polymer replicas. Bull. Korean Chem. Soc. perfusion: first in vivo experience. Artificial J. Derm. 150, 869–877 (2004).
28(11), 2079–2082 (2007). Organs 25(5), 392–394 (2001).

future science group www.future-science.com 1073


Review | McInnes & Voelcker
237 Moon S, Lee S, Lee H, Kwon T. Fabrication 242 Kelly J, DeSimone J. Shape-specific, nn Drug-loaded biodegradable and non-
of microneedle array using LIGA and hot monodisperse nano-molding of protein biodegradable nanoparticles as small as 50 nm
embossing process. Microsys. Technol. 11(4), particles. J. Am. Chem. Soc. 130, 5438–5439 were created via imprinting with a quartz
311–318 (2005). (2008). template. The authors demonstrate
238 Moon S, Lee S. A novel fabrication method of 243 Rolland JP, Maynor BW, Euliss LE et al. Direct enzyme-triggered controlled release of
a microneedle array using inclined deep X-ray fabrication and harvesting of monodisperse, antibodies and nucleic acids from
exposure. J. Micromech. Microeng. 15, shape-specific nanobiomaterials. J. Am. Chem. these nanoparticles.
903–911 (2005). Soc. 127(28), 10096–10100 (2005). 245 Yuan J, Xu Y, Walther A et al .Water-soluble
239 Gill H, Prausnitz M. Coating formulations nn ‘Smart particles’ as small as 160 nm with organo–silica hybrid nanowires. Nat. Mater. 7,
for microneedles. Pharm. Res. 24(7), controlled and monodisperse size, shape, 718–722 (2008).
1369–1380 (2007). composition, cargo encapsulation and surface 246 Kessel S, Borner HG. High rate silicification of
240 Pearton M, Allender C, Brain K et al. Gene functionality were created. These nanomolded peptide–polymer assemblies toward composite
delivery to the epidermal cells of human skin particles could be easily released and dissolved nanotapes. Macromol. Rapid Comm. 29(5),
explants using microfabricated microneedles under physiological conditions and used for 419–424 (2008).
and hydrogel formulations. Pharm. Res. 25(2), therapeutic drug-delivery systems. 247 Kessel S, Borner HG. Self-assembled
407–416 (2008). PEO-peptide nanotapes as ink for plotting
244 Glangchai LC, Caldorera-Moore M, Shi L, Roy
241 Tao S, Lubeley M, Desai T. Bioadhesive K. Nanoimprint lithography based fabrication nonwoven silica nanocomposites and
poly(methyl methacrylate) microdevices for of shape-specific, enzymatically-triggered smart mesoporous silica fiber networks. Macromol.
controlled drug delivery. J. Control. Release nanoparticles. J. Control. Release 125(3), Rapid Comm. 29(4), 316–320 (2008).
88(2), 215–228 (2003). 263–272 (2008).

1074 Future Med. Chem. (2009) 1(6) future science group

You might also like