You are on page 1of 30

ARTICLE IN PRESS

Prog. Polym. Sci. 33 (2008) 448–477


www.elsevier.com/locate/ppolysci

The development of microgels/nanogels


for drug delivery applications
Jung Kwon Oha,, Ray Drumrighta,
Daniel J. Siegwartb, Krzysztof Matyjaszewskib,
a
The Dow Chemical Co., Dow Coating Solutions R&D Midland, MI 48664, USA
b
Department of Chemistry, Center for Macromolecular Engineering, Carnegie Mellon University, 4400 Fifth Avenue,
Pittsburgh, PA 15213, USA
Received 30 October 2007; received in revised form 22 January 2008; accepted 29 January 2008
Available online 12 February 2008

Abstract

Microgels/nanogels are crosslinked polymeric particles, which can be considered as hydrogels if they are
composed of water soluble/swellable polymer chains. They possess high water content, biocompatibility, and desirable
mechanical properties. They offer unique advantages for polymer-based drug delivery systems (DDS): a tunable size
from nanometers to micrometers, a large surface area for multivalent bioconjugation, and an interior network
for the incorporation of biomolecules. Present and future microgel applications require a high degree of control over
properties. They include stability for prolonged circulation in the blood stream, novel functionality for further
bioconjugation, controlled particle size with uniform diameter, and biodegradability for sustained release of drugs
for a desired period of time and facile removal of empty devices. This review describes the recent developments
of microgel/nanogel particles as drug delivery carriers for biological and biomedical applications. Various synthetic
strategies for the preparation of microgels/nanogels are detailed, including photolithographic and micromolding
methods, continuous microfluidics, modification of biopolymers, and heterogeneous free radical and controlled/living
radical polymerizations.
r 2008 Elsevier Ltd. All rights reserved.

Keywords: Microgels; Nanogels; Hydrogels; Drug delivery; Heterogeneous polymerization; Reverse micelles

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
2. Photolithographic and micromolding methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
2.1. Photolithographic techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
2.2. Micromolding methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 452
3. Microfluidic preparation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453
4. Fabrication of biopolymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453

Corresponding authors.
E-mail addresses: jkoh2@dow.com (J.K. Oh), km3b@andrew.cmu.edu (K. Matyjaszewski).

0079-6700/$ - see front matter r 2008 Elsevier Ltd. All rights reserved.
doi:10.1016/j.progpolymsci.2008.01.002
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 449

4.1. Water-in-oil (W/O) heterogeneous emulsion methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454


4.1.1. Inverse (mini)emulsion method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454
4.1.2. Reverse micellar method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
4.1.3. Membrane emulsification. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
4.2. Aqueous homogeneous gelation method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457
4.3. Spray drying method. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457
4.4. Chemical crosslinking of dextrans. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458
4.4.1. Michael addition reaction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458
4.4.2. Free radical polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
5. Heterogeneous free radical polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
5.1. Dispersion polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
5.2. Precipitation polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
5.3. Inverse (mini)emulsion polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
5.4. Inverse microemulsion polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
6. Heterogeneous controlled/living radical polymerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468

1. Introduction reversibly degrade into the corresponding precur-


sors upon external stimuli.
The design and preparation of hydrogels have Another method involves chemical crosslinking in
attracted a great deal of interest in biomedical the presence of various crosslinkers. Several methods
engineering, pharmaceutical applications, and bio- have been explored for the preparation of effective
materials science because of their tunable chemical hydrogels. The modification of natural biopolymers
and three-dimensional (3D) physical structure, good possessing a high degree of functional groups with
mechanical properties, high water content, and additional functional crosslinkers results in the
biocompatibility. These unique properties offer great formation of biopolymer-based hydrogels [39–42].
potential for the utilization of hydrogels in tissue Free radical polymerization (FRP) of water-soluble
engineering, biomedical implants, drug delivery, and or hydrophilic monomers in the presence of
bionanotechnology [1–5]. Recently, the development either difunctional or multifunctional crosslinkers
of hydrogels hybridized with nano-sized materials allows for the preparation of synthetic hydrogels in
such as inorganic clays [6–9], carbon nanotubes water. Various monomers have been examined,
[10,11], and polyaniline nanosticks [12] has also been including hydroxy-functional methacrylates [43,44],
of increasing interest in the field of materials science. poly((ethylene glycol) fumarate) [45], and (meth)a-
Hydrogels are generally prepared from hydro- crylate derivatives of sugars [46,47], dendrimers [48],
philic polymer matrices that are crosslinked by poly(amino acid) [49], poly(ethylene glycol) (PEG)
several methods. One method is physical cross- [50], and PLA-b-PEG-b-PLA triblock copolymers
linking. Hydrogen bonds, crystallized domains, [51]. In particular, the FRP approach has been
hydrophobic interactions, stereocomplexation, tem- extensively utilized to prepare stimuli-responsive
perature-induced sol–gel transition, host–guest in- hydrogels, mainly based on poly(N-isopropylacryla-
teraction, aggregation, and self-assembly have been mide) (PNIPAM). This is due to their unique
utilized for the synthesis of ‘‘bulk hydrogels’’. They volume phase transition in response to external
include protein-based gels [13,14], polyelectrolyte- stimuli such as temperature and pH [52–57].
based multilayered layer-by-layer (LbL) film hydro- Hydrogels can be in the form of macroscopic
gels [15–17], triblock copolymer-based gelators networks or confined to smaller dimensions such as
[18–21], polylactide (PLA)-based thermogelling microgels, which are crosslinked polymeric parti-
hydrogels [22–25], polyurethane-based hydrogels cles. When the size of microgels is in submicron
[26], poly(acrylic acid-co-acrylamide) (P(AA-co- range, they are known as nanogels [58–60]. Micro-
AAm)) gels [27], hydrophobically modified hydro- gels/nanogels offer unique advantages for polymer-
gels [28,29], organogels [30–33], and supramolecular based drug delivery systems (DDS) over polymer–
gels [34–38]. These physically crosslinked gels could protein conjugates [61], polymer–drug conjugates
ARTICLE IN PRESS
450 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

Nomenclature PSS poly(sodium 4-styrenesulfonate)


PAH poly(allyamine hydrochloride )
PLA polylactide LbL layer-by-layer
DDS drug delivery systems PS polystyrene
MPS mononuclear phagocyte system PMMA poly(methyl methacrylate)
PDMS poly(dimethylsiloxane) PHEA poly(2-hydroxyethyl acrylate)
BSA bovin serum albumin PHPMA poly(2-hydroxypropyl methacrylate)
mTM microtransfer molding EGDMA ethylene glycol dimethacrylate
MIMIC micromolding in capillaries PNIPAM poly(N-isopropyl acrylamide)
PFPE perfluoropolyether LCST lower critical solution temperature
PEGDA poly(ethylene glycol diacrylate) DMIAAm dimethylmaleimidoethyl acrylamide
MeHA methacrylated hyaluronic acid PAA poly(acrylic acid)
HRP horseradish peroxidase DOX doxorubicin
SAMs self-assembled monolayers PAAm polyacrylamide
CS chitosan AETMAC 2-acryloxyethyltrimethylammonium
HA hyaluronan chloride
Dex dextran CMC critical micellar concentration
W/O water-in-oil PEO-b-PPO-b-PEO poly(ethylene glycol)-b-poly
ADH adipic dihydrazide (propylene glycol)-b-poly(ethylene
pDNA plasmid-DNA glycol)
EDCI ethyl-3-[3-dimethylamino]propyl OEGDMA oligo(ethylene glycol) dimethacrylate
carbodiimide PHEAS a,b-poly(N-2-hydroxyethyl)-D,L-aspar-
HAALD aldehyde-modified hyaluronan tamide
HAADH hydrazide-modified hyaluronan mTHPC meta-tetra(hydroxyphenyl)chlorine
SPG Shirasu porous glass AETMAC 2-acryloxyethyltrimethylaminium
DLS dynamic light scattering chloride
TPP tripolyphosphate CRP controlled/living radical polymeriza-
PEG poly(ethylene glycol) tion
PEI polyethyleneimine Mw/Mn molecular weight distribution
EDTAA ethylenediaminetetraacetic dianhydride PDI polydispersity
CTP camptothecin ATRP atom transfer radical polymerization
TEM transmission electron microscopy SFRP stable free radical polymerization
AFM atomic force microscopy RAFT reversible addition-fragmentation
Dex-SH thiol-functionalized dextrans chain transfer
HA-SH thiol-functionalized hyaluronans POEOMA poly(oligo(ethylene glycol)
GMA glycidyl methacrylate monomethyl ether methacrylate)
LA-HEMA hydroxy-terminated HEMA-lactate RES reticuloendothelial system
HEMA 2-hydroxyethyl methacrylate FITC fluorescein isothiocyanate
DMAEMA dimethylaminoethyl methacrylate RITC rhodamine B isothiocyanate

[62,63], micelles [64–66], and vesicles [67,68] based network, as well as their in vitro release behavior,
on amphiphilic and doubly hydrophilic block have been extensively investigated as targeted drug
copolymers, dendrimers [69], and submicron-sized delivery carriers for biomedical applications. In
particulates [70,71]. Microgels/nanogels have tun- addition, the incorporation of inorganic nanocrys-
able size from nanometers to several micrometers tals has been also reported, including quantum dots
and a large surface area for multivalent bioconjuga- [73,74] and magnetic nanoparticles [75,76] for
tion [72]. They also have an interior network for the optical and magnetic imaging of living cells, and
incorporation of biorelated molecules. Physical gold nanorods for photodynamic therapy [77].
entrapment of bioactive molecules such as drugs, However, the present and future microgel appli-
proteins, carbohydrates, and DNA in the polymeric cations still require a high degree of control over
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 451

properties. Several criteria are required to design (mini)emulsion, and inverse microemulsion. In
and develop effective microgel-based DDS for addition, recent advance on controlled/living radical
in vivo applications. One criterion is the stability polymerization (CRP) in heterogeneous media to
of microgels/nanogels for prolonged circulation in prepare well-defined microgels/nanogels with un-
the blood stream. This is important because the ique properties for drug delivery will also be
instability of DDS may result in premature release discussed.
of therapeutics, causing adverse side effects. An-
other one is their novel functionality for further 2. Photolithographic and micromolding methods
bioconjugation of microgel surfaces with specific
ligands that can recognize receptors on diseased 2.1. Photolithographic techniques
cells. The effective ligands for cancer cells include
folic acid derivatives, peptides, proteins, and anti- Soft- and photolithography [102–104] has been
bodies [71,78,79]. Effective bioconjugation allows explored to fabricate 3D hydrogel particles
for active targeting of microgels/nanogels to specific [105,106] and microgel rings [107] for drug delivery
cells, especially cancercells. Another criterion is and tissue engineering. The photolithographic
control over the dimension to be less than 200 nm method requires the development of techniques for
in diameter. This can facilitate cellular uptake of the surface treatment of stamps or new materials for
nanogels through a receptor-mediated endocytosis replica molds to permit the release of molded gels
to cross cell membranes as well as reduce nanopar- from stamps or replica molds. Poly(dimethylsilox-
ticle uptake by mononuclear phagocyte system ane) (PDMS) stamps have generally been used to
(MPS); consequently, this will significantly increase mold, release, and stack gels into 3D structures.
their circulation time in blood [80]. The last criterion Surface modification of PDMS stamps with hex-
is the biodegradability of microgels/nanogels. Bio- a(ethylene glycol)-terminated self-assembled mono-
degradation should not only modulate the release of layers (SAMs) or adsorbed monolayers of bovin
drugs for a desired period of time, but also enable serum albumin (BSA) promoted the release of
removal of the empty device after drug release. A molded gels to a substrate without distortion.
significant amount of work has been directed Replica molding, microtransfer molding (mTM),
toward the development of crosslinkers functiona- micromolding in capillaries (MIMIC), and other
lized with (bio)degradable linkages. These include standard lithographic techniques were applied to
peptides [39,81–83], anhydrides [84], oligo(lactate) fabricate gels with various microstructures including
esters [85–89], polyperoxides [90], disulfides [91,92], monolithic gels, isolated microstructures, and inter-
acetals [93–95], poly(3-hydroxybutyrate) [96], and connected networks. In addition, various gel pre-
polyphosphoesters [97,98]. The resulting hydrogels cursors were examined, such as protein-based
crosslinked with these linkages were degraded to collagen, gelatin, and tumor extract Matrigel as
water-soluble polymers in external environments. well as sugar-based agarose gel [105].
For example, oligo(lactate) ester linkages were A top-down method called ‘‘Particle Replication
hydrolyzed in physiological conditions at pH ¼ 7.0 In Nonwetting Templates (PRINT)’’ [106] was
at 37 1C. Disulfides were degraded into the corre- developed to fabricate submicron-sized microgels
sponding thiols in presence of water-soluble redu- with control over particle size, shape, and composi-
cing agents, including dithiothreitol [99,100] and a tion. A photocurable perfluoropolyether (PFPE)
tripeptide glutathione [101]. Acetals were degraded replica was used as the molding material. This
under acidic conditions (pHo6.5), while they are eliminated the formation of a residual interconnect-
stable at physiological pH ¼ 7.4 [93]. ing film between molded objects, thus allowing for
This review will describe the recent developments the preparation of isolated objects (Fig. 1). In the
of microgel/nanogel particles as drug delivery approach, silicon master templates were mainly
carriers for biological and biomedical applications. made by using electron beam lithography. Dimeth-
Various synthetic strategies for the preparation of acrylate-functionalized PFPE oligomers containing
microgels/nanogels will be presented. They include a photoinitiator were poured onto various patterned
photolithographic and micromolding methods, mi- silicon master templates, and then photochemically
crofluidics, modification of biopolymers with var- cured to form patterned elastomeric PFPE replica
ious approaches, and free radical heterogeneous mold. PFPE-based materials have many advantage-
polymerization in dispersion, precipitation, inverse ous properties including solvent resistance, chemical
ARTICLE IN PRESS
452 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

Fig. 1. Illustration of the PRINT process (upper) and TEM images of microgels of PEGDA with various shapes prepared using PRINT
(lower): trapezoidal (a), bar (b), conical (c), and arrow shape (d). Reprinted with permission from Ref. [106]. Copyright 2005 American
Chemical Society.

robustness, and durability. They have also been size and shape of microgels, which are important for
used for different applications to fabricate organic biomedical applications.
solvent resistant microfluidic devices with features The preparation of shape-controlled, harvestable
on the order of hundreds of microns [108,109]. hydrogel particles encapsulated with mammalian
Using PRINT, monodisperse microgels of poly cells was demonstrated using micromolding method
(ethylene glycol diacrylate) (PEGDA), triacrylate [110]. In the process, cells were suspended in a
resin, PLA, and polypyrrole were prepared. Their hydrogel precursor solution consisting of either
sizes were ranged from 200 nm to micron-scale methacrylated hyaluronic acid (MeHA) or PEGDA
in diameter with various shapes such as trapezoidal, and a photoinitiator in water. The resulting mixture
bar, conical, and arrow. In addition, DNA, was deposited onto plasma-cleaned hydrophilic
proteins, and small-molecule therapeutics were PDMS patterns and then photocrosslinked via
incorporated into 200 nm PEG-based microgels exposure to UV light. The resulting cell-laden
using a simple encapsulation technique to demon- microgels were removed, hydrated, and then har-
strate the compatibility of PRINT with (bio) vested. They were also molded into various shapes
molecules. including square prisms, disks, and strings.
A similar method was reported for the prepara-
2.2. Micromolding methods tion of cell-laden chemically and physically cross-
linked hydrogel particles for tissue engineering
Micromolding of hydrogels has been examined as [111]. This approach utilized the controlled release
a potential method to fabricate submicron-sized of gelling agents from the mold to enable rapid
microgels. The methods are similar to photolitho- gelling upon contact of hydrogel-forming precur-
graphic techniques. However, they can minimize the sors with the gelling agent in the mold. Ionically
need to use costly lithographic equipment and clean crosslinked hydrogels of alginate, a naturally
room facilities. Moreover, by controlling the fea- occurring carbohydrate-based polymer, were pre-
tures on a mold stamp, they enable control over the pared from crosslinking of alginates molded
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 453

between PDMS and calcium-containing agarose internal and external gelation. For the former,
slab upon the release of calcium ions from the slab. droplets consist of a gelling polymer and a cross-
The obtained microstructured hydrogel particles linking agent precursor in inactive form. Contin-
had lateral dimensions between 5 and 2000 mm and uous phase contains an activator that can diffuse
vertical dimensions between 10 and 200 mm. into droplets and react with crosslinking agent
precursors, triggering the release of crosslinking
3. Microfluidic preparation agents. The released crosslinking agents can cause
gelation. For the latter, droplets are emulsified in
Microfluidic methods have been recently explored continuous phase that contains a crosslinking agent.
for the preparation of monodisperse micron-sized Diffusion of crosslinking agents to emulsified
microgels. The methods require the fabrication of droplets leads to gelation. As a consequence, the
microfluidic devices by soft lithography using elasto- crosslinking agent should be soluble in aqueous
meric materials, particularly PDMS or polyurethane droplet phase.
elastomers as building blocks. The devices generally Recently, physical gelation by ionic crosslinking
consist of inlets for monomers (or oligomers) and using internal and external crosslinking approaches
continuous liquids, and microchannels with a tapered was explored for the microfluidic preparation of
junction where two immiscible phases are merged. microgels of alginate crosslinked with calcium ions
Emulsification of monomers by breaking up liquid (Ca2+) (Fig. 2) [112]. For internal gelation, an
threads to droplets and in-situ crosslinking of the aqueous sodium alginate dispersion containing
resulting droplets by photopolymerization or poly- CaCO3 as a crosslinking agent precursor was
condensation are the two general steps involved in the supplied from the central channel and soybean oil
continuous microfluidic preparation of microgels. containing acetic acid as an activator was supplied
Confinement of droplets, variation of flow rates of from the side channels. Two liquid streams passed
liquids, and precise control of reaction time are key through a tapered junction to form emulsified
parameters to generate monodisperse particles with a droplets in soybean oil continuous phase. Acetic
variety of shapes and morphologies [112]. More acid that diffused to the aqueous droplets reacted
recently, the microfluidic preparation of complicated with CaCO3. The generated Ca2+ ions as cross-
nanostructured particles such as Janus and ternary linking agents involved in crosslinking reactions
particles has been reported [113]. with carboxylic acid residues of sodium alginate,
Several approaches were examined for the micro- resulting in the formation of alginate microgel
fluidic preparation of micron-sized microgels of particles. However, weak gelation was identified as
both synthetic and biological polymers. They are the most important shortcoming, along with poor
based on gelation methods in microchannels; control over microgel morphology for the method.
chemical gelation, physical gelation by temperature For external gelation, aqueous sodium alginate
change [114], reversible shear thinning [115], and solution was emulsified in soybean oil containing
ionic crosslinking [116,117], and coalescence-in- sodium acetate. Downstream of the channel, Ca2+
duced gelation [118]. In this review, chemical ions diffused to aqueous droplets to crosslink
gelation and ionic crosslinking will be discussed. alginate. Contrary to internal crosslinking, this
Details on other gelation methods have been method resulted in the successful preparation of
summarized in a mini-review [112]. stable monodisperse spherical alginate microgels.
Chemical gelation utilizes photoinduced polymer- Their sizes varied from 50 to 70 mm with the flow
ization to crosslink low-viscosity monomer droplets rates of aqueous and organic phases. In addition,
stably dispersed in a continuous phase with an aid control over the diffusion time of crosslinking agent
of surfactants. This method was mostly examined in aqueous phase and the concentration of cross-
for the preparation of microgels of synthetic linker in organic phase allowed for the preparation
polymers. Examples include the preparation of of various alginate microgels with capsular, gradi-
10-mm-sized monodisperse dextran (Dex)-modified ent, and uniform structures [121].
microgels [119] and photocrosslinked microgels
immobilized with biocatalysts such as glucose 4. Fabrication of biopolymers
oxidase (GOx) and horseradish peroxidase (HRP)
[120]. Physical gelation by ionic crosslinking can be Chitosan (CS), hyaluronan (HA), and Dex are
achieved in a microchannel using two approaches; naturally occurring carbohydrate-based biopolymers.
ARTICLE IN PRESS
454 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

Fig. 2. Illustration of the microfluidic production of alginate microgels by internal gelation (a) and external gelation (b). Reprinted with
permission from Ref. [112]. Copyright 2007 Wiley InterScience.

Fig. 3 shows their chemical structures. CS is a also be utilized for further bioconjugation with cell-
polysaccharide composed of 2-amino-deoxy-D-glucan targeting agents.
combined with glycosidic b(1-4) linkages. It is Many methods have been developed for the
commercially obtained by hydrolysis of aminoacetyl preparation of microgels of these biopolymers.
groups of naturally occurring chitin, which is the They can be classified into four categories: water-
main component of shells, crabs, shrimp, and krill in-oil (W/O) heterogeneous emulsion, aqueous
[122,123]. HA is a naturally occurring mucopoly- homogeneous gelation, spray drying method, and
saccharide composing of N-acetyl-D-glucosamine chemical crosslinking of Dex. For CS, the prepara-
and D-glucuronic acid, found in the extracellular tion of micro/nanoparticles as drug delivery carriers
tissue matrix of vertebrates. Dex is a bacterial- has recently been reviewed [125].
derived polysaccharide consisting of a-1,6-linked
D-glucopyranose residues, and generally produced 4.1. Water-in-oil (W/O) heterogeneous
by enzymes from certain strains [124]. These emulsion methods
carbohydrate-based biopolymers are non-toxic and
offer high water solubility, biocompatibility, and W/O emulsion methods involve generally two
biodegradability. Due to their unique properties, steps: emulsification of aqueous droplets of water-
these biopolymers have been used for various soluble biopolymers in continuous oil phase with an
applications in biomedicine, pharmaceutics, materi- aid of oil-soluble surfactants and crosslinking of
als science, cosmetics, and food industry. Another biopolymers with water-soluble crosslinkers. The
feature of these biopolymers is a high content of general approaches include inverse (mini)emulsion
functional groups, including amino groups in CS (often called emulsion crosslinking technique),
and carboxylic acid groups in HA. These functional reverse micelle, and membrane emulsification.
groups can be utilized in crosslinking with addi-
tional functional crosslinkers, resulting in the 4.1.1. Inverse (mini)emulsion method
formation of functional microgels of biopolymers. A W/O emulsion is formed from a mixture
Furthermore, the residual functional groups can consisting of aqueous biopolymer droplets and a
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 455

continuous oil phase using either a homogenizer or stability of the resulting inverse emulsion. Various
a high-speed mechanical stirrer. Mineral oil and drugs, DNA, and cells are physically incorporated
hexane as hydrophobic organic solvents and Span into aqueous droplets. The resulting aqueous
80 (sorbitan monooleate) and Aerosol OT (sodium droplets of biopolymers are then crosslinked with
bis(2-ethylhexyl) sulfosuccinate) as oil-soluble sur- appropriate crosslinking agents. The resulting cross-
factants were often used to implement colloidal linked microgel particles are prepared as dispersion
in organic solvents, and thus purified by precipita-
tion, centrifugation, washing with organic solvents
such as isopropanol, and lyophilization. By this
method, the size of the prepared microgel particles
can be controlled by amount of surfactants and
crosslinking agents as well as stirring speed during
the formation of inverse emulsion.
For preparation of HA-based microgels, car-
boxylic acids of HA were crosslinked with adipic
dihydrazide (ADH) as a crosslinker in the presence
of ethyl-3-[3-dimethylamino]propyl carbodiimide
(EDCI) in aqueous droplets (Fig. 4). Plasmid-
DNA (pDNA) was then incorporated into the
robust microspheres with a size distribution of
5–20 mm for sustained gene delivery and site-specific
targeting application [126]. In another case, HA was
chemically modified with aldehyde functionalities
generated from vicinal secondary alcohols on HA
by oxidation with sodium periodate, resulting
in the formation of aldehyde-modified hyaluronan
(HAALD). HA was also modified with dihydrazide
functionalities generated from carboxylic acids on
HA by coupling with glutahydrazide, forming
hydrazide-modified hyaluronan (HAADH). The
resultant HAALD and HAADH were homogenized
in mineral oil with Span 80 to form microspheres
with a size of 10–15 mm for vocal fold regeneration
Fig. 3. Chemical structures of repeating unit of chitosan, [127]. The microgels were crosslinked with other
hyaluronan, and dextran. reactive polymers to form doubly crosslinked

Fig. 4. Illustration of crosslinking reaction of hyaluronan with adipic dihydrazide [126].


ARTICLE IN PRESS
456 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

networks that exhibited elastic moduli similar to through a carbodiimide coupling reaction with
those of vocal fold lamina propria at frequencies cystamine at pH ¼ 7.0 and then thiol-disulfide
close to the range of human phonation. exchange in the presence of dithiothreitol at
For preparation of CS-based microgels, glutar- pH ¼ 8.0. The resulting thiol-HA was crosslinked
aldehyde has been mainly used as an effective in reverse micelles containing small interfering RNA
crosslinker that reacts with amino groups on CS in (siRNA) by reduction of thiols to disulfides to form
aqueous droplets. Various drugs including diclofe- nanogels with a diameter of 198 nm size for target-
nac sodium, 5-fluorouracil, phenobarbitone, theo- specific intracellular delivery of siRNA [138].
phylline, pentazocine, and progesterone were added
into aqueous droplets [128–133]. The resulting drug- 4.1.3. Membrane emulsification
loaded CS-based microspheres had a relatively large Membrane emulsification is a relatively new
size ranging from 40 to 300 mm. A similar approach method for the preparation of spherical particles
has been applied to prepare complex microgels by with a highly uniform size distribution [139]. The
coacervation of CS glutamates and HAs without method involves the use of a membrane, initially
external crosslinkers in mineral oil containing Span Shirasu porous glass (SPG) membrane with a highly
80. Gentamicin sulfate as model drug was incorpo- uniform pore size ranging from 0.1 to 18 mm, through
rated into microgels to examine its in vitro release which a liquid is allowed to permeate under adequate
profile [134]. pressure [140]. The resulting uniform-sized droplets
are dispersed in the continuous phase, resulting in the
4.1.2. Reverse micellar method formation of simple W/O and O/W emulsions,
Similar to the inverse (mini)emulsion method, the multiple emulsions such as O/W/O, O/W/O, and
reverse micellar method also involves a W/O solid/O/W dispersions, and the corresponding parti-
dispersion; however, a relatively large amount of cles with various shapes including microspheres,
oil-soluble surfactants is used to form a thermo- hollow spheres, core–shell microcapsules, and orga-
dynamically stable micellar solution consisting of nic–inorganic hybrid materials. Details on such
aqueous droplets dispersed in the continuous oil methods using membrane emulsification are sum-
phase [135,136]. The resulting micellar droplets have marized in several review papers [141–144].
a submicron size ranged from tens to hundreds of This technique combined with a step-wise cross-
nanometers in diameter (Fig. 5). linking method was recently developed to prepare
Tumor targeted CS-based nanogels were pre- microgel particles with a uniform size distribution
pared in inverse microemulsion of hexane contain- [145]. Fig. 6 illustrates the preparation process of CS
ing Aerosol OT as a stabilizer in the presence of microspheres containing insulin. First, a stable W/O
doxorubicin (Dox)-modified Dex. Aqueous glutar- emulsion with a uniform droplet size was formed by
aldehyde was used to crosslink CS. The resulting passing an aqueous CS/acetic acid solution contain-
Dox-encapsulating CS-based nanogels have a dia- ing insulin through the uniform pores of a porous
meter of around 100 nm [137]. HA-based nanogels glass membrane into a paraffin/petroleum ether
were also prepared in hexane containing a surfac- mixture containing hexaglycerin pentaester (PO-500)
tant mixture of Span 65, Span 80, and Tween 80. emulsifier. Next, the uniform droplets of CS were
This approach involves the modification of HA to hardened by both ionic crosslinking with tripoly-
corresponding thiol-functionalized HA (HA-SH) phosphate (TPP) and chemical crosslinking with

Fig. 5. Illustration of the reverse micellar method for the preparation of nanogels.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 457

Fig. 6. Illustration of a process for the preparation of chitosan microspheres containing insulin by a membrane emulsification technique
combined with a step-wise crosslinking method. Reprinted with permission from Ref. [145]. Copyright 2006 Elsevier.

glutaraldehyde. The solidification condition was opti- possible toxicity of crosslinkers and reagents that
mized based on microsphere morphology, encapsula- are involved in chemical crosslinking method. For
tion efficiency, drug activity, and release profile the development of drug delivery carriers to target
in vitro. The resulting uniformly sized insulin-loaded tumors, CS was recently modified with glycidyltri-
microgels had a size range from 4 to 15 mm in methylammonium chloride to form N-[(2-hydroxy-
diameter. A similar method was applied to prepare 3-trimethylammonium)propyl]chitosan chloride,
CS-based microgels incorporated with BSA [146] and, which was in turn crosslinked with TPP in the
more recently, agarose beads with different particle presence of methotrexate, an anticancer drug.
size ranging from 15 to 60 mm [147]. The resulting CS-nanogels underwent a change in
volume upon decreasing pH, varying the diameter
4.2. Aqueous homogeneous gelation method from 180 nm at pH ¼ 7.4 to 450 nm at pH ¼ 5.0.
This volume transition is effective for targeted drug
Covalent chemical crosslinking was utilized for delivery of cancer therapeutics in that methotrexates
the preparation of biopolymer-based nanoparticles are physically entrapped in CS-based nanogels at
in water. An example includes the utilization of a physiological pH ¼ 7.4, while they are released
facile carbodiimide coupling reaction of CS with from the nanogels in tumor cells under acidic
a PEG dicarboxylic acid as a water-soluble cross- conditions. In addition, these nanogels were con-
linker, resulting in the formation of CS-based jugated with transferrin, a cell-targeting protein, in
nanogels with a diameter of 4–24 nm by transmission aqueous media, which facilitates receptor-medicated
electron microscopy (TEM) and 50–120 nm endocytosis. The resulting methotrexate-loaded CSs
by dynamic light scattering (DLS) [148]. In another nanogel bioconjugates could be used for targeted
example, ethylenediaminetetraacetic dianhydride pH-mediated intracellular release of cancer thera-
(EDTAA) was used to react with CS for the peutics (Fig. 7) [154,155].
preparation of pH-sensitive nanogels with a dia-
meter of 70–80 nm. They enabled reversible switch of 4.3. Spray drying method
their surface upon pH change to be positive
at pHo4.8 and negative at pH45.2, and were Spray drying is widely used in pharmaceutical and
stable in the entire pH range. These characteristics materials science to prepare capsules, granules, fine
suggest that they are an effective candidate for powders, and agglomerates. This method involves
encapsulation of pH-sensitive anticancer agents such the use of a spray dryer, mainly consisting of
as highly water-insoluble camptothecin (CPT) [149]. atomizer and drying chamber. Solutions and suspen-
Reversible physical crosslinking has also been sions of drugs, polymers, and particles are atomized
explored to prepare fine nanoparticles. In particu- to fine droplets. A stream of hot air induces quick
lar, ionic gelation involves electrostatic interaction evaporation of solvent from the droplets in drying
of cationic CS with polyanion, polyethyleneimine chamber, resulting in the formation of microspheres
(PEI) [150], and typically TPP [151–153] in water. or microgels. The obtained particles settle into a
The method is advantageous in that it avoids the bottom collector, which are further dried in a
ARTICLE IN PRESS
458 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

Fig. 7. A schematic illustration of the preparation of chitosan-based microgels using ionic gelation (a), bioconjugation (b), loading (c), and
release of drugs (d) for drug delivery targeted to tumors. Reprinted with permission from Ref. [155]. Copyright 2006 American Chemical
Society.

vacuum chamber or modified in separated experi- chemical crosslinking, including carbodiimide cou-
ments. The size of the resulting microspheres is pling [42], Michael addition, and free radical
determined by nozzle size, spray flow rate, atomiza- polymerization. The next section will detail Michael
tion speed, and extent of crosslinking. addition and free radical polymerization. Physical
This method has been explored to prepare drug- crosslinking based on stereocomplexation and self-
loaded microspheres of CS [156–160] and HA [161] assembly was also reported for the preparation of
as biodegradable drug delivery carriers. An aqueous nanogels [164,165].
solution of these biopolymers containing cross-
linkers was spray-dried with various drugs, such as 4.4.1. Michael addition reaction
cimetidine, sodium dichofenac, vitamin D2, ampi- A Michael addition-type reaction has been
cillin, betamethasone disodium phosphate, and explored to prepare hydrogels with thiol-acrylate
cromolyn sodium salt. The resulting spheres had a networks. This method requires the modification of
size of 1–10 mm in diameter. Further encapsulation hydroxy groups of Dex with either thiols or
of the resultant biomicrospheres was achieved. As acrylates [166]. Thiol-functionalized Dex (Dex-SH)
an example, CS-based microspheres with a size of was prepared by a two-step reaction: activation of
1.8–2.9 mm were prepared by spray drying method, hydroxy groups of Dex with 4-nitrophenylchloro-
and then encapsulated into Eudragits microspheres formate and then reaction of the resultant nitro-
with a size ranging from 152 to 223 mm using an phenyl-substituted Dex (Dex-NP) with cysteamine
oil-in-oil solvent evaporation method [162]. (Fig. 8). The thiol-Dex was then added into a PEG
The spray drying technique has also been used tetra-acrylate, resulting in the formation of Dex-
to prepare submicron-sized nanoparticles of silica/ based hydrogels [167]. In another case, vinylsulfone
poly(L-lysine)/alginate [163]. In-situ reduction of functionalized Dex (Dex-VS) was prepared by
Co2+ ions resulted in the formation of magnetic reaction of Dex with vinyl sulfone alkanionic acid,
cobalt silicate nanoparticles. The resulting nano- which was prepared by the reaction of a mercapto-
particles could enter cells through endocytosis and alkanoic acid with divinyl sulfone (Fig. 8). They
degrade in fibroblast cells, suggesting that they may were crosslinked with linear and four-arm star
be suitable for targeted drug delivery applications. mercaptopoly(ethylene glycol), which was prepared
by multi-step organic synthesis, to form Dex-
4.4. Chemical crosslinking of dextrans hydrogels [168].
Michael addition reaction was also utilized for
Biodegradable Dex-based microgels and hydro- the preparation of HA-based hydrogels. In this case,
gels were prepared by various methods based on HA-SH was prepared by reaction of carboxylic
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 459

Fig. 8. Schematic representation for the synthesis of thiol-functionalized dextran (Dex-SH) [167] and vinylsulfone-functionalized dextran
(Dex-VS) [168].

Fig. 9. Illustration of crosslinking of hyaluronan with PEGDA using a Michael addition reaction [169].

acids of HA with cysteamine, and then added into


PEGDA (Fig. 9) [169]. However, none of papers
reported the preparation of Dex-based microgel
particles by Michael addition-type reactions to our
best knowledge.

4.4.2. Free radical polymerization


Free radical polymerization of methacrylate-
functionalized Dexs allows for the preparation of
Dex-based hydrogels, as well as nanogels. Various
methacrylated dextrans were synthesized by utiliz-
ing the hydroxy groups of Dex that react with Fig. 10. Chemical structures of various methacrylated dextrans.
different methacrylate precursors. They include
methacrylated Dex derivatives of glycidyl metha- oxy-terminated HEMA-lactate (Dex-LA-HEMA)
crylate (Dex-GMA) [170,171], 2-hydroxyethyl (Fig. 10) [87]. These monomers are homopoly-
methacrylate (dex-HEMA) [172], and hydr- merized or copolymerized with other monomers.
ARTICLE IN PRESS
460 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

For example, dex-GMA [171] was copolymeri- as a continuous phase. At the onset, polymerization
zed with amidoethyl methacrylate to prepare occurs in a homogeneous reaction mixture; how-
macroporous, cell-adhesive and cell-penetrable ever, the formed polymers become insoluble in
Dex-hydrogels. Dex-LA-HEMA [87] was also co- the continuous medium, ultimately leading to the
polymerized with NIPAM for the preparation of formation of stable dispersion of polymeric particles
thermoresponsive hydrogels. with an aid of colloidal stabilizers [177,178]. The
An interesting approach utilized photoinitiated method has been mainly applied to prepare uniform
free radical polymerization in liposomes as reactors microspheres of hydrophobic polymers including
to prepare lipid coated and naked biodegradable polystyrene (PS) [179,180] and poly(methyl metha-
nanogels with tunable degradation properties crylate) (PMMA) [181].
[173,174]. In this method, the liposomes were Hydrophilic monodisperse micron-sized particles
prepared by multiple extrusions of lipid bilayers of PHEMA were also prepared by dispersion
hydrated with dex-HEMA through a polycarbonate polymerization in the presence of PEO-b-poly
membrane with a well-defined pore size. Photoini- (1,1,2,2-tetrahydroperfluorodecyl acrylate) diblock
tiated polymerization was followed in liposomes of copolymer as a stabilizer in supercritical carbon
dex-HEMA to form lipid-coated Dex-nanogels with dioxide [182], and methacryloyl-terminated PMMA
a diameter of around 400 nm. The surrounding lipids in a 55/45 (wt/wt) mixture of 2-butanol/tolune [183].
were removed from lipid-coated nanoparticles by Recently, crosslinked microgel particles based on
the addition of a nonionic surfactant, resulting in PHEMA, PGMA, poly(2-hydroxyethyl acrylate)
the formation of lipid naked nanoparticles [172]. (PHEA), and poly(2-hydroxypropyl methacrylate)
This approach was further extended to prepare (PHPMA) were prepared in the presence of difunc-
self-rupturing microcapsules, in which nanogels tional crosslinkers such as ethylene glycol dimetha-
consisting of copolymers of Dex-HEMA with crylate (EGDMA) and 4-[(E)-[(3Z)-3-(4-(acryloyloxy)
dimethylaminoethyl methacrylate (DMAEMA) were benzylidene)-2-hexylidene]methyl]phenyl acrylate.
first prepared in liposomes. The resulting nanogels Drugs and magnetic nanoparticles were either
were then coated with poly(sodium 4-styrenesulfonate) physically incorporated or chemically attached to
(PSS) as poly anions and poly(allylamine hydro- microgels. The resulting microgels were effective as
chloride ) (PAH) as polycations using LbL deposi- drug delivery carriers and for DNA applications
tion to form microcapsules consisting of Dex-based [44,184–186].
nanogel core with polyelectrolyte shell. The poly-
electrolyte shell was impermeable to dextrans produced 5.2. Precipitation polymerization
from degradation of nanogel core upon hydrolysis,
leading to rupturing of microcapsules [175,176]. Similar to dispersion polymerization, precipita-
tion polymerization involves the formation of
5. Heterogeneous free radical polymerization homogeneous mixture at its initial stage and the
occurrence of initiation and polymerization in
Various heterogeneous polymerization reactions the homogeneous solution. As the formed polymers
of hydrophilic or water-soluble monomers in the are not swellable but soluble in the medium, the use
presence of either difunctional or multifunctional of crosslinker is necessary to crosslink polymer
crosslinkers have been mostly utilized to prepare chains for the isolation of particles. As a conse-
well-defined synthetic microgels. They include dis- quence, the resulting crosslinked particles often
persion, precipitation, inverse (mini)emulsion, and have an irregular shape with high polydispersity
inverse microemulsion polymerization utilizing an (PDI) [178,187–190].
uncontrolled free radical polymerization process. Preparation of hydrogels and microgels based on
PNIPAM and its derivatives by precipitation
5.1. Dispersion polymerization polymerization in water has been extensively
explored for biomedical applications [191–193]. This
Dispersion polymerization is a technique that is due to the thermosensitive properties of PNI-
allows for the preparation of micron-sized particles PAM-microgels that undergo volume change at a
with narrow size distribution. In the process, most lower critical solution temperature (LCST) in water,
ingredients including monomers, polymeric stabili- at around 32 1C. Above the LCST, PNIPAM
zers, and initiators are soluble in an organic solvent hydrogels and microgels are hydrophobic and expel
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 461

water; below LCST, they are hydrophilic and served as reactive sites for post-modification of
swollen in water. These unique properties facilitate microgels. Examples include the bioconjugation of
the loading of drugs into microgels as well as P(NIPAM-co-AA) microgels with cell-targeting
enhance the controllable release of drugs encapsu- biomolecules such as folic acids, proteins, and
lated in microgels [87]. However, the use of antibodies for targeted drug delivery [198–200],
PNIPAM-based microgels shows a certain limita- and the reaction of P(NIPAM-co-AA) microgels
tion to biomedical applications due to relatively with boronic acid derivatives to make a glucose
narrow range of physical and chemical properties of sensor [201,202]. Second, carboxylic acids in micro-
PNIPAM. gels immobilized inorganic precursors through ionic
An introduction of functional groups is a interactions. Microgels were utilized as polymeric
promising way to overcome such limitation and reactors for semiconductor, metal, and magnetic
broaden various applications of PNIPAM-based nanoparticles [203–205]. In particular, gold-loaded
microgels. Towards that end, various functional nanogels were used for site-specific drug delivery
monomers were copolymerized with NIPAM under and photodynamic therapy through photothermally
precipitation polymerization conditions, resulting in triggered volume transitions under physical condi-
the preparation of functional microgels [194]. tions [77]. Third, acid-functionalized microgels were
Copolymerization with more hydrophilic or hydro- doubly responsive to two external stimuli (tempera-
phobic monomers enabled to tune the volume phase ture and pH) [206–208]. Fourth, core–shell P
transition of the microgels [195,196]. Copolymeriza- (NIPAM-co-AA) microgels were prepared [209] as
tion with a tetrazole-containing AAm could bind to effective delivery carriers of insulin [210], Dox [211],
various protonated amines in aqueous medium at and anionic surfactant [212]. They were effectively
millimolar concentration [197]. Dimethylmaleimi- assembled on an aminopropyltrimethoxysilane-
doethyl acrylamide (DMIAAm) was synthesized functionalized glass substrate via electrostatic inter-
and copolymerized with NIPAM to prepare photo- actions, resulting in the fabrication of microlens
crosslinkable poly(NIPAM-co-DMIAAm). These arrays for biological processes (Fig. 11) [213–216].
copolymers aggregated to form particles in water Recently, enzyme-responsive hydrogel particles
above of LCST, and in turn the resulting aggregates were prepared by copolymerization of AAm with
were crosslinked by UV irradiation, resulting in the an amine-containing PEG-functionalized AAm
preparation of microgels [60]. in water. The amine groups on microgels were
Acid-containing monomers such as AA, utilized for attachment of enzyme cleavable peptide
methacrylic acid, and undecanoic acid were copo- linkers that can be cleaved by a protease
lymerized to prepare PNIPAM-based microgels enzyme. The volume change of PAAm-based
with pendent carboxylic acid groups. These car- microgels upon responsiveness to pH enhanced the
boxylic acids provided many advantages. First, they incorporation of drugs and the volume change upon

Fig. 11. Illustration of hydrogel microlens assay, where P(NIPAM-co-AA) microgels were prepared by free-radical precipitation
polymerization method. Reprinted with permission from Ref. [216]. Copyright 2005 American Chemical Society.
ARTICLE IN PRESS
462 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

responsiveness to enzymes facilitated the release of organic solvents. Temperature- and pH-sensitive
drugs [217,218]. P(NIPAM-co-AA) minigels [227] and PAAm/PAA
interpenetrating polymer network microgels [228]
5.3. Inverse (mini)emulsion polymerization were prepared in the presence of N,N0 -methylenebi-
sacrylamide. Their swelling properties were studied
Inverse (mini)emulsion polymerization is a W/O in water by measuring particle diameter upon
polymerization process that contains aqueous dro- change in temperature and pH. Stable, crosslinked,
plets (including water-soluble monomers) stably amphiphilic nanoparticles based on acrylated tri-
dispersed with the aid of oil-soluble surfactants in block copolymer of poly(ethylene glycol)-b-poly
a continuous organic medium. Stable dispersions (propylene glycol)-b-poly(ethylene glycol) (PEO-b-
are formed by mechanical stirring for inverse PPO-b-PEO) were prepared by inverse emulsion
emulsion process and by sonification for inverse photopolymerization (Fig. 12). The hydrophobic
miniemulsion polymerization. Upon addition of PPO-rich domains enhanced incorporation of Dox,
radical initiators, polymerization occurs within the an amphiphilic anticancer drug, up to 9.8 wt%. The
aqueous droplets producing colloidal particles. resulting microgels had the diameter of 50 and
Several reports have demonstrated the synthesis of 500 nm [229]. In addition, poly(magnesium acrylate)
hydrophilic or water-soluble particles of PHEMA microgels with the diameter of 40 mm were prepared
[219], PAA [220], and PAAm [219], temperature- for immobilizing enzymes such as Glucose oxidase
sensitive hollow microspheres of PNIPAM [221], (Gox) as bioreactors [230].
core–shell nanocapsules with hydrophobic shell and Biodegradable microgels were prepared via poly-
hydrophilic interior [132], and polyaniline nanopar- merization of a macromonomer of diacrylated
ticles [222]. In addition, this method has also been Pluronic–oligo(lactic acid) copolymer, which was
utilized to prepare stable organic–inorganic hybrid synthesized by a ring opening polymerization of
particles containing magnetic iron oxide nanoparti- D,L-lactide initiated with PEO-b-PPO-b-PEO, fol-
cles [223,224] and clays [225] in cyclohexane based lowed by reaction with acryloyl chloride [231]. The
inverse miniemulsions. Details on such systems are size of the resulting microgels varied from 200 to
also summarized in a review paper [226]. 600 mm as a stirring speed decreased during poly-
Recently, this method has been explored to merization; however their size distributions were
prepare crosslinked microgels in the presence of relatively monodisperse. BSA as a model protein
difunctional crosslinkers for effective drug delivery. was encapsulated and its release behavior was
This is due to a facile confinement of water-soluble examined as a function of temperature [232]. Acid-
drugs in aqueous droplets dispersed in continuous degradable microgels based on PAAm were also

Fig. 12. Illustration of preparation for microgels of PEO-b-PPO-b-PEO via inverse emulsion polymerization. Reprinted with permission
from Ref. [229]. Copyright 2005 American Chemical Society.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 463

prepared for delivery of protein and pDNA [235,236]. This method has been explored for the
[233,234]. A water-soluble diacrylamide crosslinker preparation of well-defined nanoparticles based on
containing an acid-labile acetal group was synthe- PNIPAM [237,238], PAAm [239–241], and poly
sized [93] and introduced into inverse emulsion poly- (dimethylacrylamide) [242] as well as magnetic
merization. The resulting microgels were degraded polymeric particles of PAAm and PEO [75] contain-
under acidic condition (pH ¼ 5.0) to the correspond- ing iron oxide nanoparticles [243].
ing linear polymers, resulting in the release of Inverse microemulsion polymerization was ex-
encapsulated molecules. In addition, acid-degradable plored for the synthesis of well-defined nanogels.
cationic microgels were prepared by introducing Poly(vinylpyrrolidone)-based nanogels incorporated
2-acryloxyethyltrimethylammonium chloride (AET- with Dex as a water-soluble macromolecular
MAC), a cationic monomer, for antigen delivery. carbohydrate drug were prepared [244,245]. Catio-
nic nanogels of poly(HEA-co-AETMAC) were
5.4. Inverse microemulsion polymerization prepared in the presence of oligo(ethylene glycol)
dimethacrylate (OEGDMA) as a crosslinker. The
While inverse (mini)emulsion polymerization diameter of the resulting nanogels decreased from
forms kinetically stable macroemulsions at, below, 150 to 40 nm as the amount of crosslinker increased.
or around the critical micellar concentration The resulting nanogels had potential for gene
(CMC), inverse microemulsion polymerization pro- delivery, since the presence of quaternary ammo-
duces thermodynamically stable microemulsions nium ion side groups appeared to enhance incor-
upon further addition of emulsifier above the poration of DNA into nanogels via electrostatic
critical threshold. This process also involves aqu- association with the phosphate groups [246]. Poly
eous droplets, stably dispersed with the aid of a (amino acid)-based nanoparticles with different
large amount of oil-soluble surfactants in a con- surface PEGylation were prepared. a,b-Poly
tinuous organic medium; polymerization occurs (N-2-hydroxyethyl)-D,L-aspartamide (PHEAS) and
within the aqueous droplets, producing stable PEG-modified PHEA (PHEAS-PEG) were functio-
hydrophilic and water-soluble colloidal nanoparti- nalized with a methacrylate group and then poly-
cles having a diameter of less than 50–100 nm merized by UV irradiation in inverse microemulsion.

Fig. 13. Fluorescence images of rat C6 glioma cells after incubation with free meta-tetra(hydroxyphenyl)chlorine as a photosensitizer
(upper) and ultrafine hydrophilic PAAm-based nanogels incorporated with meta-tetra(hydroxyphenyl)chlorine (lower): (a, d) before light
exposure, (b, e) after light exposure, and (c, d) zoom out of (b) and (e). Scale bar ¼ 30 mm for (a), (b), (d), and (e), and 40 mm for (c) and (f).
Reprinted with permission from Ref. [248]. Copyright 2006 Wiley InterScience.
ARTICLE IN PRESS
464 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

The resulting nanoparticles had a size of around (ATRP) [254–260], stable free radical polymeriza-
250 nm in diameter by TEM. The fluorescein-loaded tion (SFRP) [261,262], and reversible addition-
PHEA-based nanoparticles were prepared in the fragmentation chain transfer (RAFT) polymeriza-
presence of fluorescein sodium salts, and examined tion [263–265]. Peptide sequences [266,267], biotin
for cellular uptake using macrophage cells [247]. [268], and streptavidin [269] have been successfully
Recently, ultrafine hydrophilic PAAm-based na- modified to become ATRP initiators. Many CRP
nogels incorporated with meta-tetra(hydroxyphe- reactions of hydrophobic monomers have been
nyl)chlorine (mTHPC) as a photosensitizer were examined in heterogeneous aqueous dispersions,
prepared for photodynamic therapy. In the process, including conventional emulsion, microemulsion,
both AAm and N,N-methylene(bis acrylamide) as a miniemulsion, dispersion, and suspension media.
crosslinker were directly emulsified into hexane/ They resulted in the preparation of stable latex
Aerosol OT, without water, which allowed for the particles consisting of well-defined hydrophobic
preparation of tiny PAAm nanoparticles with a polymers. Several reviews [270–273] and papers
diameter of 2–3 nm [248]. The presence of water concerning CRP in aqueous dispersion have been
in hexane/Aerosol OT produced 20 nm diameter published using ATRP [274–281], SFRP [282–288],
PAAm particles with a relatively broad size and RAFT [289–295].
distribution [249]. The resulting PAAm-nanogels However, only a few reports on CRP of water-
incorporated with mTHPC were effective to kill rat soluble or hydrophilic monomers in heterogeneous
C6 glioma cells upon exposure to 650 nm visible media have been published. The first successful
light (Fig. 13). CRP using ATRP in inverse miniemulsion described
the synthesis of stable nanoparticles of well-con-
6. Heterogeneous controlled/living radical trolled water-soluble poly(oligo(ethylene glycol)
polymerization monomethyl ether methacrylate) (POEOMA) with
Mw/Mno1.3. The resulting nanoparticles had uni-
CRP provides a versatile route for preparation of form size distribution (Fig. 14) [296,297]. This
(co)polymers with controlled molecular weight, method has also been applied for the synthesis of
narrow molecular weight distribution (i.e., Mw/Mn, stable nanoparticles of well-controlled hydrophilic
or PDIo1.5), designed architectures, and useful PEO-b-PHEMA block copolymers with Mw/Mno
end-functionalites [250–252]. Recently, CRP has 1.3. Interestingly, the resulting doubly hydrophilic
been explored as a tool to preparation of well- block copolymers self-assembled to form well-
controlled polymer–protein/peptide bioconjugates defined micelles with a diameter of 10–20 nm in
[253]. Various methods for CRP have been devel- water [298]. In addition, a RAFT inverse miniemul-
oped; however, the most successful techniques sion polymerization was also reported for the
include atom transfer radical polymerization preparation of hydrophilic latex of PAAm [299].

Fig. 14. Preparation of stable nanoparticles of well-controlled poly(oligo (ethylene glycol) monomethyl ether methacrylate) with
Mw/Mno1.3. Evolution of molecular weight and molecular weight distribution (a) and CONTIN plot of particles dispersed in cyclohexane
inverse miniemulsion (b). Inset is their atomic force microscopy (AFM) image. Reprinted with permission from Ref. [297]. Copyright 2006
American Chemical Society.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 465

CRP techniques have been explored for the bioconjugated with fluorescein isothiocyanate
synthesis of gels [300–305] and crosslinked nano- (FITC)-labeled avidin. The number of biotin
particles of well-controlled polymers in the presence molecules in each nanogel was determined to be
of crosslinkers. Hydrogel nanoparticles of PNIPAM 142,000 and the formation of bioconjugates of
was prepared by precipitation polymerization via nanogels with avidin was confirmed using fluores-
ATRP in water [306]. Acid cleavable poly(butyl cence microscopy (Fig. 16) [308].
acrylate) microgel core surrounded with linear Second, they enable incorporation of a high
POEOMA shell for delivery of hydrophobic drugs loading level of drugs such as the anticancer
were prepared by combination of acid-labile micro- drug Dox after polymerization [308] and water-
gel approach with RAFT-mediated seeded disper- soluble biomacromolecules such as rhodamine B
sion polymerization [307].
ATRP in inverse miniemulsion for the synthesis
and functionalization of stable (bio)degradable
crosslinked nanogels of well-controlled water-solu-
ble polymers in the presence of a disulfide-functio-
nalized dimethacrylate (DMA) was reported
[297,308]. This approach allowed for the prepara-
tion of biomaterials with many useful features
(Fig. 15).
First, the resulting particles preserve a high
degree of halide end-functionality to enable further
chain extension to form functional block copoly-
mers and facile functionalization with biorelated
molecules, such as by utilizing click reactions
[309–311]. Further, several other ways are available
for the preparation of novel functional nanogels
using the method involving ATRP in inverse
miniemulsion. They include the use of functional
ATRP initiators and the facile copolymerization
with functional monomers. The latter approach
was successfully demonstrated with the preparation
Fig. 16. Fluorescence microscopy image of FITC–avidin–
of OH-functionalized nanogels by copolymerizing biotin–nanogel conjugates. Scale bar ¼ 5 mm. Reprinted with
with 2-hydroxyethyl acrylate (HEA). The nanogels permission from Ref. [308]. Copyright 2006 American Chemical
were then conjugated with biotin, and further Society.

Fig. 15. Illustration of unique features of stable biodegradable nanogels of well-controlled water-soluble polymers in the presence of a
disulfide-functionalized dimethacrylate for effective targeted drug delivery.
ARTICLE IN PRESS
466 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

isothiocyanate (RITC)-labeled Dex during polymer- (Mw/Mno1.5), indicating the formation of a uni-
ization [312]. Furthermore, the RITC-dextran- formly crosslinked network in the individual
loaded nanogels could enter cells through an particles. This uniform structure is anticipated to
endocytosis. Fig. 17 shows laser confocal fluores- improve control over the release of encapsulated
cence microscope image of RITC-Dex loaded agents (Fig. 18). More importantly, nanogels are
nanogels (green) within MC3TC cells stained for biodegraded in the presence of a biocompatible
nucleus (blue) and actin (red). glutathione tripeptide, which is commonly found in
Third, they are degradable in a reducing en- cells at mM concentrations [92,313–315]. The
vironment to individual polymeric chains with a biodegradation of nanogels can trigger the release
relatively narrow molecular weight distribution of encapsulated molecules as well as facilitate the
removal of empty vehicles.
Fourth, properties including swelling ratio, de-
gradation behavior, and colloidal stability of
nanogels prepared by ATRP are superior to those
prepared by conventional free radical inverse
miniemulsion polymerization. Fifth, nanogels will
enhance circulation time in the blood, since they
consist of POEOMA, an analog of linear PEO,
which can prevent nanoparticle uptake by reticu-
loendothelial system (RES) [80].
Well-defined functional nanogels prepared by this
newly developed method hold great potential as
drug delivery carriers to target-specific cells for
biomedical applications. Future nanogels will con-
tain targeting residues such as peptides, proteins,
and antibodies for delivery to specific regions and
cells.

7. Conclusion

Recent developments of microgel/nanogel parti-


Fig. 17. Laser confocal fluorescence microscopy image of green
cles as drug delivery carriers for biological and
RITC-Dex loaded nanogels (abs. 570 nm (red), but the color was
changed to green to distinguish it from the actin), blue nucleus biomedical applications were reviewed in this paper.
(abs. 350 nm, Hoechst blue) and red actin (abs. 649 nm cy5- Major synthetic strategies for the preparation of
conjugated phalloidin), demonstrating endocytosis of the nanogels. microgels/nanogels were described in detail. Photo-

Fig. 18. Nanogels containing RITC-labeled dextran in water before and after the addition of dithiothreitol (DTT) (a); and fluorescence
microscopy images of nanogels loaded with RITC-dextran before (b) and after (c) degradation in water. The scale bars in b and c are
50 mm. Reprinted with permission from Ref. [312]. Copyright 2007 American Chemical Society.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 467

lithographic methods have been explored for pre- various applications of PNIPAM-based microgels.
paration of well-defined 3D hydrogel particles and Inverse (mini)emulsion polymerization utilizes a
microgel rings. In particular, the PRINT method W/O dispersion that provides a facile confinement
allowed for the preparation of monodisperse sub- of water-soluble drugs in aqueous droplets dis-
micron-sized microgels with control over particle persed in continuous organic solvents. Stable
size and composition, and various shapes including microgels based on amphiphilic PEO-b-PPO-b-
trapezoidal, bar, conical, and arrow. However, the PEO and Pluronic–oligo(lactic acid) copolymer,
methods require the development of surface treat- and acid-degradable microgels based on PAAm
ment of stamps or new materials for replica molds were developed for effective delivery of drugs,
to enhance a facile release of a molded gel from proteins, and DNA. Inverse microemulsion poly-
stamps or replica molds. Micromolding methods of merization involves thermodynamically stable
hydrogels allowed for fabrication of submicron- microemulsions, allowing for the preparation of
sized microgels. Furthermore, they can minimize the well-defined nanogels. Poly(vinylpyrrolidone)-based
need for the use of costly lithographic equipment nanogels, cationic nanogels of poly(HEA-co-AET-
and clean room facilities. Continuous microfluidic MAC), and poly(amino acid)-based nanogels were
preparation of microgels involves two steps: emul- prepared. In particular, ultrafine PAAm-based
sification of monomers by breaking up the liquid nanogels incorporated with a photosensitizer were
threads to droplets and in-site crosslinking of the developed for photodynamic therapy.
droplets by photopolymerization or polycondensa- CRP techniques have been recently explored to
tion. Chemical gelation and physical gelation by prepare stable microgels/nanogels of well-controlled
temperature change, reversible shear thinning, and polymers. A new method utilizing ATRP, inverse
ionic crosslinking, and coalescence-induced gelation miniemulsion polymerization, and thiol-disulfide
methods have been developed. In particular, ionic exchange reaction allowed for the development
crosslinking using external gelation resulted in and functionalization of stable biodegradable
microfluidic preparation of stable monodisperse nanogels of well-controlled water-soluble polymers
spherical alginate microgels, while internal cross- in the presence of a disulfide-functionalized di-
linking led to weak gelation with poor control over methacrylate (DMA). This approach allowed for
microgel morphology. Modification of naturally the preparation of biomaterials with many useful
occurring carbohydrate-based biopolymers such as features; preservation of a high degree of halide end-
CS, HA, and Dex has been explored for the functionality for facile functional block copolymer-
preparation of functional microgels for targeted ization and bioconjugation, biodegradation in the
drug delivery. This is due to unique properties of presence of a biocompatible glutathione tripeptide,
these biopolymers, including non-toxicity, high formation of uniform networks, larger and better
water solubility, biocompatibility, biodegradability swelling ratio, degradation behavior, and colloidal
as well as high degree of functionality. Inverse stability compared to conventional counterparts,
(mini)emulsion, reverse micelle, membrane emulsi- high loading level of anticancer drugs, and en-
fication, aqueous homogeneous gelation, spray hanced circulation time in the blood.
drying, Michael addition, and free radical polymer- Future design and development of effective micro-
ization have been developed. gel-based DDSs for in vivo applications require a high
The most general methods for the preparation of degree of control over properties. These include
well-defined synthetic microgels are heterogeneous excellent stability for prolonged circulation in the
polymerization of hydrophilic or water-soluble blood stream, novel functionality for further biocon-
monomers in the presence of either difunctional or jugation; dimension less than 200 nm in diameter, and
multifunctional crosslinkers. Free radical disper- biodegradability for facile removal of the empty
sion, precipitation, inverse (mini)emulsion, and device and sustained release of drugs.
inverse microemulsion polymerization have been One future goal of research in this area should be
explored. Dispersion polymerization generally pro- the improved design of microgels/nanogels with
duced micron-sized particles. Precipitation polymer- specific targeting residues to enable highly selective
ization has been extensively explored to prepare uptake into particular cells. This will be especially
microgels based on PNIPAM and its derivatives, important for the targeting of cancer cells, thereby
due to their volume change at LCST. Copolyme- reducing non-specific uptake into healthy cells.
rization with functional monomers broadened Furthermore, it will increase the efficiency of the
ARTICLE IN PRESS
468 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

DDS. As the bridge between chemistry and biology single-walled carbon nanotubes (SWNTs) hydrogel made
continues to become shorter, biologists will need to by hybrids of swnts and cyclodextrins. J Am Chem Soc
elucidate the specific interactions between biomole- 2007;129:4878–9.
[12] Meng L, Lu Y, Wang X, Zhang J, Duan Y, Li C. Facile
cules and cellular integral receptors. Chemists can synthesis of straight polyaniline nanostick in hydrogel.
then design microgels/nanogels to present these Macromolecules 2007;40:2981–3.
proteins or antibodies on the surface of the [13] Mi L, Fischer S, Chung B, Sundelacruz S, Harden JL. Self-
microgels/nanogels as a way to precisely target- assembling protein hydrogels with modular integrin bind-
specific cells. In this way, careful control over ing domains. Biomacromolecules 2006;7:38–47.
[14] Reddy TT, Lavenant L, Lefebvre J, Renard D. Swelling
stability, size, biodegradability, and functionality behavior and controlled release of theophylline and
for bioconjugation will guide the development of sulfamethoxazole drugs in b-lactoglobulin protein gels
the next generation of microgels/nanogels. obtained by phase separation in water/ethanol mixture.
Biomacromolecules 2006;7:323–30.
[15] Wood KC, Boedicker JQ, Lynn DM, Hammond PT.
Acknowledgments
Tunable drug release from hydrolytically degradable layer-
by-layer thin films. Langmuir 2005;21:1603–9.
The Dow Chemical Co. is greatly acknowledged [16] Kozlovskaya V, Kharlampieva E, Mansfield ML,
for the support of the work and the permission to Sukhishvili SA. Poly(methacrylic acid) hydrogel films
publish this work. J.K.O thanks the NSERC and capsules: response to pH and ionic strength, and
encapsulation of macromolecules. Chem Mater 2006;18:
Postdoctoral Fellowship of Canada for financial
328–36.
support during his stay at Carnegie Mellon Uni- [17] Berg MC, Zhai L, Cohen RE, Rubner MF. Controlled
versity. drug release from porous polyelectrolyte multilayers.
Biomacromolecules 2006;7:357–64.
[18] Li CTY, Armes SP. Synthesis and characterization
References of biocompatible thermo-responsive gelators based on
ABA triblock copolymers. Biomacromolecules 2005;6:
[1] Hoffman AS. Applications of thermally reversible poly- 994–9.
mers and hydrogels in therapeutics and diagnostics. [19] Madsen J, Armes SP, Lewis AL. Preparation and aqueous
J Control Release 1987;6:297–305. solution properties of new thermoresponsive biocompatible
[2] Langer R. Drug delivery: drugs on target. Science 2001;293: ABA triblock copolymer gelators. Macromolecules 2006;
58–9. 39:7455–7.
[3] Langer R, Vacanti JP. Tissue engineering. Science 1993; [20] Nykaenen A, Nuopponen M, Laukkanen A, Hirvonen
260:920–6. S-P, Rytelae M, Turunen O, et al. Phase behavior
[4] Peppas NA, Hilt JZ, Khademhosseini A, Langer R. and temperature-responsive molecular filters based on
Hydrogels in biology and medicine: from molecular self-assembly of polystyrene-block-poly(N-isopropyla-
principles to bionanotechnology. Adv Mater 2006;18: crylamide)-block-polystyrene. Macromolecules 2007;40:
1345–60. 5827–34.
[5] Brandl F, Sommer F, Goepferich A. Rational design of [21] Guvendiren M, Messersmith PB, Shull KR. Self-assembly
hydrogels for tissue engineering: impact of physical factors and adhesion of DOPA-modified methacrylic triblock
on cell behavior. Biomaterials 2006;28:134–46. hydrogels. Biomacromolecules 2008;9:122–8.
[6] Haraguchi K, Takehisa T, Ebato M. Control of cell [22] Li S, El Ghzaoui A, Dewinck E. Rheology and drug release
cultivation and cell sheet detachment on the surface of properties of bioresorbable hydrogels prepared from
polymer/clay nanocomposite hydrogels. Biomacromole- polylactide/poly(ethylene glycol) block copolymers. Macro-
cules 2006;7:3267–75. mol Symp 2005;222:23–35.
[7] Miyazaki S, Endo H, Karino T, Haraguchi K, Shibayama [23] Lee J, Bae YH, Sohn YS, Jeong B. Thermogelling aqueous
M. Gelation mechanism of poly(N-isopropylacrylamide)– solutions of alternating multiblock copolymers of poly
clay nanocomposite gels. Macromolecules 2007;40: (L-lactic acid) and poly(ethylene glycol). Biomacromole-
4287–95. cules 2006;7:1729–34.
[8] Okay O, Oppermann W. Polyacrylamide–clay nanocom- [24] Shim WS, Kim SW, Lee DS. Sulfonamide-based pH- and
posite hydrogels: rheological and light scattering character- temperature-sensitive biodegradable block copolymer hy-
ization. Macromolecules 2007;40:3378–87. drogels. Biomacromolecules 2006;7:1935–41.
[9] Haraguchi K, Song L. Microstructures formed in co-cross- [25] Park SY, Lee Y, Bae KH, Ahn C-H, Park TG.
linked networks and their relationships to the optical and Temperature/pH-sensitive hydrogels prepared from pluro-
mechanical properties of PNIPA/clay nanocomposite gels. nic copolymers end-capped with carboxylic acid groups via
Macromolecules 2007;40:5526–36. an oligolactide spacer. Macromol Rapid Commun 2007;28:
[10] Wang Z, Chen Y. Supramolecular hydrogels hybridized 1172–6.
with single-walled carbon nanotubes. Macromolecules [26] Mequanint K, Patel A, Bezuidenhout D. Synthesis,
2007;40:3402–7. swelling behavior, and biocompatibility of novel physically
[11] Ogoshi T, Takashima Y, Yamaguchi H, Harada A. cross-linked polyurethane-block-poly(glycerol methacry-
Chemically-responsive sol–gel transition of supramolecular late) hydrogels. Biomacromolecules 2006;7:883–91.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 469

[27] Dai H, Chen Q, Qin H, Guan Y, Shen D, Hua Y, dressing containing ciprofloxacin and its drug release
et al. A temperature-responsive copolymer hydrogel in studies. J Mater Sci Mater Med 2005;16:95–100.
controlled drug delivery. Macromolecules 2006;39: [44] Arun A, Reddy BSR. In vitro drug release studies from the
6584–9. polymeric hydrogels based on HEA and HPMA using 4-
[28] Miquelard-Garnier G, Demoures S, Creton C, Hourdet D. {(E)-[(3Z)-3-(4-(acryloyloxy)benzylidene)-2-hexylidene]-
Synthesis and rheological behavior of new hydrophobically methyl}phenyl acrylate as a crosslinker. Biomaterials 2005;
modified hydrogels with tunable properties. Macromole- 26:1185–93.
cules 2006;39:8128–39. [45] Dadsetan M, Szatkowski JP, Yaszemski MJ, Lu L.
[29] Kretschmann O, Schmitz S, Ritter H. Microwave-assisted Characterization of photo-cross-linked oligo[poly(ethylene
synthesis of associative hydrogels. Macromol Rapid Com- glycol) fumarate] hydrogels for cartilage tissue engineering.
mun 2007;28:1265–9. Biomacromolecules 2007;8:1702–9.
[30] Ajayaghosh A, Varghese R, Praveen VK, Mahesh S. [46] Okada M. Molecular design and syntheses of glycopoly-
Evolution of nano- to microsized spherical assemblies of mers. Prog Polym Sci 2001;26:67–104.
a short oligo(p-phenyleneethynylene) into superstructured [47] Park DW, Haam S, Lee TG, Kim H-S, Kim W-S.
organogels. Angew Chem Int Ed 2006;45:3261–4. Chemoenzymic synthesis of sugar-containing biocompati-
[31] Wang C, Zhang D, Xiang J, Zhu D. New organogels based ble hydrogels: crosslinked poly(b-methylglucoside acrylate)
on an anthracene derivative with one urea group and its and poly(b-methylglucoside methacrylate). J Biomed Mater
photodimer: fluorescence enhancement after gelation. Res A 2004;71A:497–507.
Langmuir 2007;23:9195–200. [48] Soentjens SHM, Nettles DL, Carnahan MA, Setton LA,
[32] Maeda H, Haketa Y, Nakanishi T. Aryl-substituted C3- Grinstaff MW. Biodendrimer-based hydrogel scaffolds for
bridged oligopyrroles as anion receptors for formation of cartilage tissue repair. Biomacromolecules 2006;7:310–6.
supramolecular organogels. J Am Chem Soc 2007;129: [49] Pitarresi G, Pierro P, Palumbo FS, Tripodo G, Giammona
13661–74. G. Photo-cross-linked hydrogels with polysaccharide-
[33] Kang SH, Jung BM, Chang JY. Polymerization of an poly(amino acid) structure: new biomaterials for pharma-
organogel formed by a hetero-bifunctional gelator in a ceutical applications. Biomacromolecules 2006;7:1302–10.
monomeric solvent: preparation of nanofibers embedded in [50] Lin-Gibson S, Bencherif S, Antonucci JM, Jones RL,
a polymer matrix. Adv Mater 2007;19:2780–4. Horkay F. Synthesis and characterization of poly(ethylene
[34] Li JL, Liu XY, Strom CS, Xiong JY. Engineering of small glycol) dimethacrylate hydrogels. Macromol Symp 2005;
molecule organogels by design of the nanometer structure 227:243–54.
of fiber networks. Adv Mater 2006;18:2574–8. [51] Shah NM, Pool MD, Metters AT. Influence of network
[35] Miyauchi M, Takashima Y, Yamaguchi H, Harada A. structure on the degradation of photo-cross-linked PLA-b-
Chiral supramolecular polymers formed by host–guest PEG-b-PLA hydrogels. Biomacromolecules 2006;7:3171–7.
interactions. J Am Chem Soc 2005;127:2984–9. [52] Tanaka T, Nishio I, Sun ST, Ueno-Nishio S. Collapse of
[36] Miyauchi M, Hoshino T, Yamaguchi H, Kamitori S, gels in an electric field. Science 1982;218:467–9.
Harada A. A [2]rotaxane capped by a cyclodextrin and a [53] Okano T, Bae YH, Jacobs H, Kim SW. Thermally on–off
guest: formation of supramolecular [2]rotaxane polymer. switching polymers for drug permeation and release.
J Am Chem Soc 2005;127:2034–5. J Control Release 1990;11:255–65.
[37] Weng W, Beck JB, Jamieson AM, Rowan SJ. Under- [54] Feil H, Bae YH, Feijen J, Kim SW. Mutual influence
standing the mechanism of gelation and stimuli-responsive of pH and temperature on the swelling of ionizable
nature of a class of metallo-supramolecular gels. J Am and thermosensitive hydrogels. Macromolecules 1992;25:
Chem Soc 2006;128:11663–72. 5528–30.
[38] Deng W, Yamguchi H, Takashima Y, Harada A. [55] Matsukuma D, Yamamoto K, Aoyagi T. Stimuli-respon-
A chemical-responsive supramolecular hydrogel from sive properties of N-isopropylacrylamide-based ultrathin
modified cyclodextrins. Angew Chem Int Ed 2007;46: hydrogel films prepared by photo-cross-linking. Langmuir
5144–7. 2006;22:5911–5.
[39] Levesque SG, Shoichet MS. Synthesis of enzyme-degrad- [56] Ho E, Lowman A, Marcolongo M. Synthesis and
able, peptide-cross-linked dextran hydrogels. Bioconjugate characterization of an injectable hydrogel with tunable
Chem 2007;18:874–85. mechanical properties for soft tissue repair. Biomacromo-
[40] Ramamurthi A, Vesely I. Evaluation of the matrix- lecules 2006;7:3223–8.
synthesis potential of crosslinked hyaluronan gels for tissue [57] Robb SA, Lee BH, McLemore R, Vernon BL. Simulta-
engineering of aortic heart valves. Biomaterials 2005;26: neously physically and chemically gelling polymer system
999–1010. utilizing a poly(NIPAAm-co-cysteamine)-based copolymer.
[41] Crescenzi V, Cornelio L, Di Meo C, Nardecchia S, Biomacromolecules 2007;8:2294–300.
Lamanna R. Novel hydrogels via click chemistry: synthesis [58] Graham NB, Cameron A. Nanogels and microgels: the new
and potential biomedical applications. Biomacromolecules polymeric materials playground. Pure Appl Chem 1998;70:
2007;8:1844–50. 1271–5.
[42] Murakami S, Aoki N. Bio-based hydrogels prepared [59] Sahiner N, Godbey WT, McPherson GL, John VT.
by cross-linking of microbial poly(g-glutamic acid) Microgel, nanogel and hydrogel–hydrogel semi-IPN com-
with various saccharides. Biomacromolecules 2006;7: posites for biomedical applications: synthesis and char-
2122–7. acterization. Colloid Polym Sci 2006;284:1121–9.
[43] Tsou T-L, Tang S-T, Huang Y-C, Wu J-R, Young J-J, [60] Kuckling D, Vo CD, Adler HJP, Voelkel A, Coelfen H.
Wang H-J. Poly(2-hydroxyethyl methacrylate) wound Preparation and characterization of photo-cross-linked
ARTICLE IN PRESS
470 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

thermosensitive PNIPAAm nanogels. Macromolecules volume transitions under physiological conditions. Lang-
2006;39:1585–91. muir 2007;23:196–201.
[61] Harris JM, Chess RB. Effect of PEGylation on pharma- [78] Kohler N, Sun C, Wang J, Zhang M. Methotrexate-
ceuticals. Nat Rev Drug Discovery 2003;2:214–21. modified superparamagnetic nanoparticles and their in-
[62] Vicent MJ, Greco F, Nicholson RI, Paul A, Griffiths PC, tracellular uptake into human cancer cells. Langmuir
Duncan R. Polymer therapeutics designed for a combina- 2005;21:8858–64.
tion therapy of hormone-dependent cancer. Angew Chem [79] Bellocq NC, Pun SH, Jensen GS, Davis ME. Transferrin-
Int Ed 2005;44:4061–6. containing, cyclodextrin polymer-based particles for tu-
[63] Khandare J, Minko T. Polymer–drug conjugates: mor-targeted gene delivery. Bioconjugate Chem 2003;14:
progress in polymeric prodrugs. Prog Polym Sci 2006;31: 1122–32.
359–97. [80] Seymour LW, Duncan R, Strohalm J, Kopecek J. Effect of
[64] Nishiyama N, Kataoka K. Nanostructured devices based molecular weight (Mw) of N-(2-hydroxypropyl)methacry-
on block copolymer assemblies for drug delivery: designing lamide copolymers on body distribution and rate of
structures for enhanced drug function. Adv Polym Sci excretion after subcutaneous, intraperitoneal, and intrave-
2006;193:67–101. nous administration to rats. J Biomed Mater Res 1987;
[65] Harada A, Kataoka K. Supramolecular assemblies of block 21:1341–58.
copolymers in aqueous media as nanocontainers relevant to [81] Plunkett KN, Berkowski KL, Moore JS. Chymotrypsin
biological applications. Prog Polym Sci 2006;31:949–82. responsive hydrogel: application of a disulfide exchange
[66] Bae Y, Fukushima S, Harada A, Kataoka K. Design protocol for the preparation of methacrylamide containing
of environment-sensitive supramolecular assemblies for peptides. Biomacromolecules 2005;6:632–7.
intracellular drug delivery: polymeric micelles that are [82] Kim S, Healy KE. Synthesis and characterization
responsive to intracellular pH change. Angew Chem Int Ed of injectable poly(N-isopropylacrylamide-co-acrylic acid)
2003;42:4640–3. hydrogels with proteolytically degradable cross-links.
[67] Buetuen V, Liu S, Weaver JVM, Bories-Azeau X, Cai Y, Biomacromolecules 2003;4:1214–23.
Armes SP. A brief review of ‘schizophrenic’ block [83] Perez P, Plieva F, Gallardo A, San Roman J, Aguilar MR,
copolymers. React Funct Polym 2006;66:157–65. Morfin I, et al. Bioresorbable and nonresorbable macro-
[68] Wu J, Eisenberg A. Proton diffusion across membranes of porous thermosensitive hydrogels prepared by cryopoly-
vesicles of poly(styrene-b-acrylic acid) diblock copolymers. merization. Role of the cross-linking agent. Biomacro-
J Am Chem Soc 2006;128:2880–4. molecules 2008;9:66–74.
[69] Tomalia DA. Birth of a new macromolecular architecture: [84] Muggli DS, Burkoth AK, Keyser SA, Lee HR, Anseth KS.
dendrimers as quantized building blocks for nanoscale Reaction behavior of biodegradable, photo-cross-linkable
synthetic polymer chemistry. Prog Polym Sci 2005;30: polyanhydrides. Macromolecules 1998;31:4120–5.
294–324. [85] Martens PJ, Bryant SJ, Anseth KS. Tailoring the de-
[70] Farokhzad OC, Cheng J, Teply BA, Sherifi I, Jon S, gradation of hydrogels formed from multivinyl poly
Kantoff PW, et al. Targeted nanoparticle-aptamer biocon- (ethylene glycol) and poly(vinyl alcohol) macromers for
jugates for cancer chemotherapy in vivo. Proc Natl Acad cartilage tissue engineering. Biomacromolecules 2003;4:
Sci USA 2006;103:6315–20. 283–92.
[71] Kim SH, Jeong JH, Chun KW, Park TG. Target-specific [86] Eichenbaum KD, Thomas AA, Eichenbaum GM,
cellular uptake of PLGA nanoparticles coated with poly Gibney BR, Needham D, Kiser PF. Oligo-a-hydroxy ester
(L-lysine)–poly(ethylene glycol)–folate conjugate. Langmuir cross-linkers: impact of cross-linker structure on biode-
2005;21:8852–7. gradable hydrogel networks. Macromolecules 2005;38:
[72] Jung T, Kamm W, Breitenbach A, Kaiserling E, 10757–62.
Xiao JX, Kissel T. Biodegradable nanoparticles for [87] Huang X, Lowe TL. Biodegradable thermoresponsive
oral delivery of peptides: is there a role for polymers to hydrogels for aqueous encapsulation and controlled release
affect mucosal uptake? Eur J Pharm Biopha 2000;50: of hydrophilic model drugs. Biomacromolecules 2005;6:
147–60. 2131–9.
[73] Hasegawa U, Nomura S-iM, Kaul SC, Hirano T, Akiyoshi [88] Rice MA, Sanchez-Adams J, Anseth KS. Exogenously
K. Nanogel-quantum dot hybrid nanoparticles for live cell triggered, enzymatic degradation of photopolymerized
imaging. Biochem Biophys Res Commun 2005;331:917–21. hydrogels with polycaprolactone subunits: experimental
[74] Fukui T, Kobayashi H, Hasegawa U, Nagasawa T, observation and modeling of mass loss behavior. Bioma-
Akiyoshi K, Ishikawa I. Intracellular delivery of nanogel- cromolecules 2006;7:1968–75.
quantum dot hybrid nanoparticles into human periodontal [89] Leemhuis M, Kruijtzer JAW, Van Nostrum CF, Hennink
ligament cells. Drug Metab Lett 2007;1:131–5. WE. In vitro hydrolytic degradation of hydroxyl-functio-
[75] Gupta Ajay K, Wells S. Surface-modified superparamag- nalized poly(a-hydroxy acid)s. Biomacromolecules 2007;8:
netic nanoparticles for drug delivery: preparation, char- 2943–9.
acterization, and cytotoxicity studies. IEEE Trans [90] Kitamura T, Matsumoto A. Facile synthesis of degradable
Nanobiosci 2004;3:66–73. gels by oxygen cross-linking of polymers including a dienyl
[76] Chatterjee J, Haik Y, Chen CJ. Biodegradable magnetic group on their side chain or at chain ends. Macromolecules
gel: synthesis and characterization. Colloid Polym Sci 2007;40:6143–9.
2003;281:892–6. [91] Aliyar HA, Hamilton PD, Ravi N. Refilling of ocular lens
[77] Das M, Sanson N, Fava D, Kumacheva E. Microgels capsule with copolymeric hydrogel containing reversible
loaded with gold nanorods: photothermally triggered disulfide. Biomacromolecules 2005;6:204–11.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 471

[92] Li C, Madsen J, Armes SP, Lewis AL. A new class of Teflon’’ for micro fluidic device fabrication. J Am Chem
biochemically degradable, stimulus–responsive triblock Soc 2004;126:2322–3.
copolymer gelators. Angew Chem Int Ed 2006;45:3510–3. [110] Yeh J, Ling Y, Karp JM, Gantz J, Chandawarkar A, Eng
[93] Murthy N, Thng YX, Schuck S, Xu MC, Frechet JMJ. G, et al. Micromolding of shape-controlled, harvestable
A novel strategy for encapsulation and release of proteins: cell-laden hydrogels. Biomaterials 2006;27:5391–8.
hydrogels and microgels with acid-labile acetal cross- [111] Franzesi GT, Ni B, Ling Y, Khademhosseini A.
linkers. J Am Chem Soc 2002;124:12398–9. A controlled-release strategy for the generation of cross-
[94] Themistou E, Patrickios CS. Star polymers and polymer linked hydrogel microstructures. J Am Chem Soc 2006;
networks containing a novel, hydrolyzable diacetal-based 128:15064–5.
dimethacrylate cross-linker: synthesis, characterization, [112] Zhang H, Tumarkin E, Sullan RMA, Walker GC,
and hydrolysis kinetics. Macromolecules 2007;40:5231–4. Kumacheva E. Exploring microfluidic routes to microgels
[95] Kaihara S, Matsumura S, Fisher JP. Synthesis and of biological polymers. Macromol Rapid Commun 2007;
properties of poly[poly(ethylene glycol)-co-cyclic acetal] 28:527–38.
based hydrogels. Macromolecules 2007;40:7625–32. [113] Nie Z, Li W, Seo M, Xu S, Kumacheva E. Janus and
[96] Neugebauer D, Rydz J, Goebel I, Dacko P, Kowalczuk M. ternary particles generated by microfluidic synthesis: de-
Synthesis of graft copolymers containing biodegradable sign, synthesis, and self-assembly. J Am Chem Soc 2006;
poly(3-hydroxybutyrate) chains. Macromolecules 2007;40: 128:9408–12.
1767–73. [114] Xu S, Nie Z, Seo M, Lewis P, Kumacheva E, Stone HA, et
[97] Li Q, Wang J, Shahani S, Sun DDN, Sharma B, Elisseeff al. Generation of monodisperse particles by using micro-
JH, et al. Biodegradable and photocrosslinkable polypho- fluidics: control over size, shape, and composition. Angew
sphoester hydrogel. Biomaterials 2006;27:1027–34. Chem Int Ed 2005;44:724–8.
[98] Du J-Z, Sun T-M, Weng S-Q, Chen X-S, Wang J. [115] Berli CLA, Quemada D, Parker A. Gel transition of
Synthesis and characterization of photo-cross-linked hy- depletion flocculated emulsions. Colloids Surf A: Physico-
drogels based on biodegradable polyphosphoesters and chem Eng Asp 2003;215:201–4.
poly(ethylene glycol) copolymers. Biomacromolecules 2007; [116] Liu K, Ding H-J, Liu J, Chen Y, Zhao X-Z. Shape-
8:3375–81. controlled production of biodegradable calcium alginate
[99] Li Y, Armes SP. Synthesis and chemical degradation of gel microparticles using a novel microfluidic device.
branched vinyl polymers prepared via ATRP: use of a Langmuir 2006;22:9468–72.
cleavable disulfide-based branching agent. Macromolecules [117] Tan W-H, Takeuchi S. Monodisperse alginate hydrogel
2005;38:8155–62. microbeads for cell encapsulation. Adv Mater 2007;19:
[100] Tsarevsky NV, Matyjaszewski K. Reversible redox clea- 2696–701.
vage/coupling of polystyrene with disulfide or thiol groups [118] Sugiura S, Oda T, Izumida Y, Aoyagi Y, Satake M, Ochiai
prepared by atom transfer radical polymerization. Macro- A, et al. Size control of calcium alginate beads containing
molecules 2002;35:9009–14. living cells using micro-nozzle array. Biomaterials 2005;26:
[101] Kakizawa Y, Harada A, Kataoka K. Glutathione-sensitive 3327–31.
stabilization of block copolymer micelles composed of [119] De Geest BG, Urbanski JP, Thorsen T, Demeester J, De
antisense DNA and thiolated poly(ethylene glycol)-block- Smedt SC. Synthesis of monodisperse biodegradable
poly(L-lysine): a potential carrier for systemic delivery of microgels in microfluidic devices. Langmuir 2005;21:
antisense DNA. Biomacromolecules 2001;2:491–7. 10275–9.
[102] Unger MA, Chou H-P, Thorsen T, Scherer A, Quake SR. [120] Jeong WJ, Kim JY, Choo J, Lee EK, Han CS, Beebe DJ,
Monolithic microfabricated valves and pumps by multi- et al. Continuous fabrication of biocatalyst immobilized
layer soft lithography. Science 2000;288:113–6. microparticles using photopolymerization and immi-
[103] Xia Y, Whitesides GM. Soft lithography. Annu Rev Mater scible liquids in microfluidic systems. Langmuir 2005;21:
Sci 1998;28:153–84. 3738–41.
[104] Xia Y, Whitesides GM. Soft lithography. Angew Chem Int [121] Zhang H, Tumarkin E, Peerani R, Nie Z, Sullan RMA,
Ed 1998;37:550–75. Walker GC, et al. Microfluidic production of biopolymer
[105] Tang MD, Golden AP, Tien J. Molding of three-dimen- microcapsules with controlled morphology. J Am Chem
sional microstructures of gels. J Am Chem Soc 2003; Soc 2006;128:12205–10.
125:12988–9. [122] Berger J, Reist M, Mayer JM, Felt O, Peppas NA, Gurny
[106] Rolland JP, Maynor BW, Euliss LE, Exner AE, Denison R. Structure and interactions in covalently and ionically
GM, DeSimone JM. Direct fabrication and harvesting crosslinked chitosan hydrogels for biomedical applications.
of monodisperse, shape-specific nanobiomaterials. J Am Eur J Pharm Biopharm 2004;57:19–34.
Chem Soc 2005;127:10096–100. [123] Laurent TC, editor. The chemistry, biology and medical
[107] Wang B, Hong Y, Feng J, Gong Y, Gao C. Rings of applications of hyaluronan and its derivatives. London:
hydrogel fabricated by a micro-transfer technique. Macro- Portland Press; 1998.
mol Rapid Commun 2007;28:567–71. [124] Lindblad MS, Sjoeberg J, Albertsson A-C, Hartman J.
[108] Rolland Jason P, Hagberg Erik C, Denison Ginger M, Hydrogels from polysaccharides for biomedical applica-
Carter Kenneth R, De Simone Joseph M. High-resolution tions. ACS Symp Ser 2007;954:153–67.
soft lithography: enabling materials for nanotechnologies. [125] Agnihotri SA, Mallikarjuna NN, Aminabhavi TM.
Angew Chem Int Ed 2004;43:5796–9. Recent advances on chitosan-based micro- and nano-
[109] Rolland JP, Van Dam RM, Schorzman DA, Quake SR, particles in drug delivery. J Control Release 2004;100:
DeSimone JM. Solvent-resistant photocurable ‘‘Liquid 5–28.
ARTICLE IN PRESS
472 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

[126] Yun Yang H, Goetz Douglas J, Yellen P, Chen W. [143] Vladisavljevic GT, Williams RA. Recent developments in
Hyaluronan microspheres for sustained gene delivery and manufacturing emulsions and particulate products using
site-specific targeting. Biomaterials 2004;25:147–57. membranes. Adv Colloid Interface Sci 2005;113:1–20.
[127] Jia X, Yeo Y, Clifton RJ, Jiao T, Kohane DS, Kobler JB, [144] Charcosset C, Fessi H. Membrane emulsification and micro-
et al. Hyaluronic acid-based microgels and microgel channel emulsification processes. Rev Chem Eng 2005;21:1–32.
networks for vocal fold regeneration. Biomacromolecules [145] Wang L-Y, Gu Y-H, Zhou Q-Z, Ma G-H, Wan Y-H, Su
2006;7:3336–44. Z-G. Preparation and characterization of uniform-sized
[128] Kumbar SG, Kulkarni AR, Aminabhavi TM. Crosslinked chitosan microspheres containing insulin by membrane
chitosan microspheres for encapsulation of diclofenac emulsification and a two-step solidification process. Col-
sodium: effect of crosslinking agent. J Macroencapsulation loids Surf B: Biointerfaces 2006;50:126–35.
2002;19:173–80. [146] Wang L-Y, Ma G-H, Su Z-G. Preparation of uniform sized
[129] Denkbas EB, Seyyal M, Piskin E. 5-Fluorouracil loaded chitosan microspheres by membrane emulsification techni-
chitosan microspheres for chemoembolization. J Macro- que and application as a carrier of protein drug. J Control
encapsulation 1999;16:741–9. Release 2005;106:62–75.
[130] Al-Helw AA, Al-Angary AA, Mahrous GM, Al-Dardari [147] Zhou Q-Z, Wang L-Y, Ma G-H, Su Z-G. Preparation of
MM. Preparation and evaluation of sustained release uniform-sized agarose beads by microporous membrane
crosslinked chitosan microspheres containing phenobarbi- emulsification technique. J Colloid Interface Sci 2007;311:
tone. J Macroencapsulation 1998;15:373–82. 118–27.
[131] Thanoo BC, Sunny MC, Jayakrishnan A. Crosslinked [148] Bodnar M, Hartmann JF, Borbely J. Synthesis and study of
chitosan microspheres: preparation and evaluation as cross-linked chitosan-N-poly(ethylene glycol) nanoparti-
a matrix for the controlled release of pharmaceuticals. cles. Biomacromolecules 2006;7:3030–6.
J Pharm Pharmacol 1992;44:283–6. [149] Shen X, Zhang L, Jiang X, Hu Y, Guo J. Reversible surface
[132] Sankar C, Rani M, Srivastava AK, Mishra B. Chitosan switching of nanogel triggered by external stimuli. Angew
based pentazocine microspheres for intranasal systemic Chem Int Ed 2007;46:7104–7.
delivery: development and biopharmaceutical evaluation. [150] Zhou X, Liu B, Yu X, Zha X, Zhang X, Chen Y, et al.
Pharmazie 2001;56:223–6. Controlled release of PEI/DNA complexes from mannose-
[133] Jameela SR, Kumary TV, Lal AV, Jayakrishnan A. bearing chitosan microspheres as a potent delivery system
Progesterone-loaded chitosan microspheres: a long acting to enhance immune response to HBV DNA vaccine.
biodegradable controlled delivery system. J Control Re- J Control Release 2007;121:200–7.
lease 1998;52:17–24. [151] Ko JA, Park HJ, Hwang SJ, Park JB, Lee JS. Preparation
[134] Lim ST, Martin GP, Berry DJ, Brown MB. Prepa- and characterization of chitosan microparticles intended
ration and evaluation of the in vitro drug release pro- for controlled drug delivery. Int J Pharm 2002;249:165–74.
perties and mucoadhesion of novel microspheres of [152] Shu XZ, Zhu KJ. A novel approach to prepare tripolypho-
hyaluronic acid and chitosan. J Control Release 2000;66: sphate/chitosan complex beads for controlled release drug
281–92. delivery. Int J Pharm 2000;201:51–8.
[135] Maitra A. Determination of size parameters of water– [153] Xu Y, Du Y. Effect of molecular structure of chitosan on
Aerosol OT–oil reverse micelles from their nuclear mag- protein delivery properties of chitosan nanoparticles. Int J
netic resonance data. J Phys Chem 1984;88:5122–5. Pharm 2003;250:215–26.
[136] Leong YS, Candau F. Inverse microemulsion polymeriza- [154] Zhang H, Oh M, Allen C, Kumacheva E. Monodisperse
tion. J Phys Chem 1982;86:2269–71. chitosan nanoparticles for mucosal drug delivery. Bioma-
[137] Mitra S, Gaur U, Ghosh PC, Maitra AN. Tumor targeted cromolecules 2004;5:2461–8.
delivery of encapsulated dextran–doxorubicin conjugate [155] Zhang H, Mardyani S, Chan WCW, Kumacheva E. Design
using chitosan nanoparticles as carrier. J Control Release of biocompatible chitosan microgels for targeted pH-
2001;74:317–23. mediated intracellular release of cancer therapeutics.
[138] Lee H, Mok H, Lee S, Oh Y-K, Park TG. Target- Biomacromolecules 2006;7:1568–72.
specific intracellular delivery of siRNA using degradable [156] He P, Davis SS, Illum L. In vitro evaluation of the
hyaluronic acid nanogels. J Control Release 2007;119: mucoadhesive properties of chitosan microspheres. Int J
245–52. Pharm 1998;166:75–88.
[139] Ma GH, Nagai M, Omi S. Study on preparation and [157] Giunchedi P, Genta I, Conti B, Muzzarelli RAA, Conte U.
morphology of uniform artificial polystyrene–poly(methyl Preparation and characterization of ampicillin loaded
methacrylate) composite microspheres by employing the methylpyrrolidinone chitosan and chitosan microspheres.
SPG (Shirasu porous glass) membrane emulsification Biomaterials 1998;19:157–61.
technique. J Colloid Interface Sci 1999;214:264–82. [158] Shi X-Y, Tan T-W. Preparation of chitosan/ethylcellulose
[140] Omi S, Ma G-H, Nagai M. Membrane emulsification— complex microcapsule and its application in controlled
a versatile tool for the synthesis of polymeric microspheres. release of Vitamin D2. Biomaterials 2002;23:4469–73.
Macromol Symp 2000;151:319–30. [159] Huang YC, Yeh MK, Chiang CH. Formulation factors in
[141] Nakashima T, Shimizu M, Kukizaki M. Particle control of preparing BTM—chitosan microspheres by spray drying
emulsion by membrane emulsification and its applications. method. Int J Pharm 2002;242:239–42.
Adv Drug Delivery Rev 2000;45:47–56. [160] Grenha A, Seijo B, Serra C, Remunan-Lopez C. Chitosan
[142] Drioli E, Curcio E, Di Profio G. State of the art and recent nanoparticle-loaded mannitol microspheres: structure
progresses in membrane contactors. Chem Eng Res Des and surface characterization. Biomacromolecules 2007;8:
2005;83:223–33. 2072–9.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 473

[161] Esposito E, Menegatti E, Cortesi R. Hyaluronan-based [176] De Geest BG, Dejugnat C, Verhoeven E, Sukhorukov GB,
microspheres as tools for drug delivery: a comparative Jonas AM, Plain J, et al. Layer-by-layer coating of
study. Int J Pharm 2005;288:35–49. degradable microgels for pulsed drug delivery. J Control
[162] Lorenzo-Lamosa ML, Remunan-Lopez C, Vila-Jato JL, Release 2006;116:159–69.
Alonso MJ. Design of microencapsulated chitosan micro- [177] Cawse JL. Dispersion polymerization. In: Lovell P,
spheres for colonic drug delivery. J Control Release El-Aasser MS, editors. Emulsion polymerization and emulsion
1998;52:109–18. polymers. West Sussex, England: Wiley; 1997. p. 743–61.
[163] Boissiere M, Meadows PJ, Brayner R, Helary C, Livage J, [178] Arshady R. Suspension, emulsion, and dispersion poly-
Coradin T. Turning biopolymer particles into hybrid merization: a methodological survey. Colloid Polym Sci
capsules: the example of silica/alginate nanocomposites. 1992;270:717–32.
J Mater Chem 2006;16:1178–82. [179] Song J-S, Tronc F, Winnik MA. Two-stage dispersion
[164] Nagahama K, Mori Y, Ohya Y, Ouchi T. Biodegradable polymerization toward monodisperse, controlled micro-
nanogel formation of polylactide-grafted dextran copoly- meter-sized copolymer particles. J Am Chem Soc 2004;
mer in dilute aqueous solution and enhancement of its 126:6562–3.
stability by stereocomplexation. Biomacromolecules [180] Lee H, Lee JM, Shim SE, Lee BH, Choe S. Synthesis of
2007;8:2135–41. carboxylic acid functionalized nanoparticles by reversible
[165] Akiyama E, Morimoto N, Kujawa P, Ozawa Y, Winnik addition-fragmentation chain transfer (RAFT) miniemul-
FM, Akiyoshi K. Self-assembled nanogels of cholesteryl- sion polymerization of styrene. Polymer 2005;46:3661–8.
modified polysaccharides: effect of the polysaccha- [181] Woods HM, Nouvel C, Licence P, Irvine DJ, Howdle SM.
ride structure on their association characteristics in the Dispersion polymerization of methyl methacrylate in
dilute and semidilute regimes. Biomacromolecules 2007;8: supercritical carbon dioxide: an investigation into Stabilizer
2366–73. Anchor Group. Macromolecules 2005;38:3271–82.
[166] Rydholm AE, Held NL, Bowman CN, Anseth KS. Gel [182] Ma Z, Lacroix-Desmazes P. Dispersion polymerization of
permeation chromatography characterization of the chain 2-hydroxyethyl methacrylate stabilized by a hydrophilic/
length distributions in thiol-acrylate photopolymer net- CO2-philic poly(ethylene oxide)-b-poly(1,1,2,2-tetrahy-
works. Macromolecules 2006;39:7882–8. droperfluorodecyl acrylate) (PEO-b-PFDA) diblock copo-
[167] Hiemstra C, van der Aa LJ, Zhong Z, Dijkstra PJ, Feijen J. lymer in supercritical carbon dioxide. Polymer 2004;45:
Rapidly in situ-forming degradable hydrogels from dextran 6789–97.
thiols through Michael addition. Biomacromolecules [183] Takahashi K, Miyamori S, Uyama H, Kobayashi S.
2007;8:1548–56. Preparation of micron-size monodisperse poly(2-hydro-
[168] Hiemstra C, van der Aa LJ, Zhong Z, Dijkstra PJ, Feijen J. xyethyl methacrylate) particles by dispersion polymeriza-
Novel in situ forming, degradable dextran hydrogels by tion. J Polym Sci A: Polym Chem 1996;34:175–82.
Michael addition chemistry: synthesis, rheology, and [184] Horak D, Rittich B, Spanova A, Benes MJ. Magnetic
degradation. Macromolecules 2007;40:1165–73. microparticulate carriers with immobilized selective ligands
[169] Peattie RA, Rieke ER, Hewett EM, Fisher RJ, Shu XZ, in DNA diagnostics. Polymer 2005;46:1245–55.
Prestwich GD. Dual growth factor-induced angiogenesis in [185] Horak D, Svec F, Adamyan A, Titova M, Skuba N,
vivo using hyaluronan hydrogel implants. Biomaterials Voronkova O, et al. Hydrogels in endovascular emboliza-
2006;27:1868–75. tion. V. Antitumor agent methotrexate-containing
[170] Levesque SG, Lim RM, Shoichet MS. Macroporous P(HEMA). Biomaterials 1992;13:361–6.
interconnected dextran scaffolds of controlled porosity [186] Horak D, Lednicky F, Petrovsky E, Kapicka A. Magnetic
for tissue-engineering applications. Biomaterials 2005;26: characteristics of ferrimagnetic microspheres prepared by
7436–46. dispersion polymerization. Macromol Mater Eng 2004;289:
[171] Levesque SG, Shoichet MS. Synthesis of cell-adhesive 341–8.
dextran hydrogels and macroporous scaffolds. Biomaterials [187] Guha S, Ray B, Mandal BM. Anomalous solubility of
2006;27:5277–85. polyacrylamide prepared by dispersion (precipitation)
[172] Van Thienen TG, Lucas B, Flesch FM, Nostrum CF, polymerization in aqueous tert-butyl alcohol. J Polym Sci
Demeester J, De Smedt SC. On the synthesis and A: Polym Chem 2001;39:3434–42.
characterization of biodegradable dextran nanogels with [188] Liu T, Desimone JM, Roberts GW. Continuous precipita-
tunable degradation properties. Macromolecules 2005;38: tion polymerization of acrylic acid in supercritical carbon
8503–11. dioxide: the polymerization rate and the polymer molecular
[173] Kazakov S, Kaholek M, Teraoka I, Levon K. UV-induced weight. J Polym Sci A: Polym Chem 2005;43:2546–55.
gelation on nanometer scale using liposome reactor. [189] Bai F, Yang X, Zhao Y, Huang W. Synthesis of core–shell
Macromolecules 2002;35:1911–20. microspheres with active hydroxyl groups by two-stage
[174] Kazakov S, Kaholek M, Kudasheva D, Teraoka I, Cow- precipitation polymerization. Polym Int 2005;54:168–74.
man MK, Levon K. Poly(N-isopropylacrylamide-co-1- [190] Li W-H, Stover HDH. Mono- or narrow disperse poly
vinylimidazole) hydrogel nanoparticles prepared and (methacrylate-co-divinylbenzene) microspheres by precipi-
hydrophobically modified in liposome reactors: atomic tation polymerization. J Polym Sci A: Polym Chem
force microscopy and dynamic light scattering study. 1999;37:2899–907.
Langmuir 2003;19:8086–93. [191] Duracher D, Elaissari A, Pichot C. Preparation of poly
[175] De Geest BG, Dejugnat C, Sukhorukov GB, Braeckmans (N-isopropylmethacrylamide) latexes kinetic studies and
K, De Smedt SC, Demeester J. Biodegradable dextran characterization. J Polym Sci A: Polym Chem 1999;37:
based microgel. Adv Mater 2005;17:2357–61. 1823–37.
ARTICLE IN PRESS
474 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

[192] Hazot P, Chapel JP, Pichot C, Elaissari A, Delair T. [209] Jones CD, Lyon LA. Synthesis and characterization of
Preparation of poly(N-ethyl methacrylamide) particles multiresponsive core–shell microgels. Macromolecules
via an emulsion/precipitation process: the role of the 2000;33:8301–6.
crosslinker. J Polym Sci A: Polym Chem 2002;40: [210] Nolan CM, Gelbaum LT, Lyon LA. 1H NMR investiga-
1808–17. tion of thermally triggered insulin release from poly
[193] Huang G, Gao J, Hu Z, St John JV, Ponder BC, Moro D. (N-isopropylacrylamide) microgels. Biomacromolecules
Controlled drug release from hydrogel nanoparticle net- 2006;7:2918–22.
works. J Control Release 2004;94:303–11. [211] Serpe MJ, Yarmey KA, Nolan CM, Lyon LA. Doxor-
[194] Hoare T, Pelton R. Functional group distributions in ubicin uptake and release from microgel thin films.
carboxylic acid-containing poly(N-isopropylacrylamide) Biomacromolecules 2005;6:408–13.
microgels. Langmuir 2004;20:2123–33. [212] Bradley M, Vincent B, Burnett G. Uptake and release of
[195] Leobandung W, Ichikawa H, Fukumori Y, Peppas NA. anionic surfactant into and from cationic core–shell
Monodisperse nanoparticles of poly(ethylene glycol) microgel particles. Langmuir 2007;23:9237–41.
macromers and N-isopropylacrylamide for biomedical [213] Kim J, Serpe MJ, Lyon LA. Hydrogel microparticles as
applications. J Appl Polym Sci 2003;87:1678–84. dynamically tunable microlenses. J Am Chem Soc 2004;
[196] Berndt I, Pedersen JS, Lindner P, Richtering W. Influence 126:9512–3.
of shell thickness and cross-link density on the structure of [214] Kim J, Serpe MJ, Lyon LA. Photoswitchable microlens
temperature-sensitive poly-N-isopropylacrylamide-poly-N- arrays. Angew Chem Int Ed 2005;44:1333–6.
isopropylmethacrylamide core–shell microgels investigated [215] Kim J, Singh N, Lyon LA. Influence of ancillary binding
by small-angle neutron scattering. Langmuir 2006;22: and nonspecific adsorption on bioresponsive hydrogel
459–68. microlenses. Biomacromolecules 2007;8:1157–61.
[197] Tominey A, Andrew D, Oliphant L, Rosair GM, Dupre J, [216] Kim J, Nayak S, Lyon LA. Bioresponsive hydrogel
Kraft A. Supramolecular binding of protonated amines to microlenses. J Am Chem Soc 2005;127:9588–92.
a receptor microgel in aqueous medium. Chem Commun [217] Thronton PD, Mart RJ, Ulijn RV. Enzyme-responsive
2006:2492–4. polymer hydrogel particles for controlled release. Adv
[198] Nayak S, Lee H, Chmielewski J, Lyon LA. Folate- Mater 2007;19:1252–6.
mediated cell targeting and cytotoxicity using thermore- [218] Ulijn RV, Bibi N, Jayawarna V, Thornton PD, Todd SJ,
sponsive microgels. J Am Chem Soc 2004;126:10258–9. Mart RJ, et al. Bioresponsive hydrogels. Mater Today
[199] Debord JD, Lyon LA. On the unusual stability of 2007;10:40–8.
succinimidyl esters in pNIPAm-AAc microgels. Bioconju- [219] Landfester K, Willert M, Antonietti M. Preparation of
gate Chem 2007;18:601–4. polymer particles in nonaqueous direct and inverse mini-
[200] Das M, Mardyani S, Chan WCW, Kumacheva E. emulsions. Macromolecules 2000;33:2370–6.
Biofunctionalized pH-responsive microgels for cancer cell [220] Kriwet B, Walter E, Kissel T. Synthesis of bioadhesive
targeting: rational design. Adv Mater 2006;18:80–3. poly(acrylic acid) nano- and microparticles using an inverse
[201] Zhang Y, Guan Y, Zhou S. Synthesis and volume phase emulsion polymerization method for the entrapment of
transitions of glucose-sensitive microgels. Biomacromole- hydrophilic drug candidates. J Control Release 1998;56:
cules 2006;7:3196–201. 149–58.
[202] Hoare T, Pelton R. Engineering glucose swelling responses [221] Sun Q, Deng Y. In situ synthesis of temperature-sensitive
in poly(N-isopropylacrylamide)-based microgels. Macro- hollow microspheres via interfacial polymerization. J Am
molecules 2007;40:670–8. Chem Soc 2005;127:8274–5.
[203] Zhang J, Xu S, Kumacheva E. Polymer microgels: reactors [222] Marie E, Rothe R, Antonietti M, Landfester K. Synthesis
for semiconductor, metal, and magnetic nanoparticles. of polyaniline particles via inverse and direct miniemulsion.
J Am Chem Soc 2004;126:7908–14. Macromolecules 2003;36:3967–73.
[204] Dong Y, Ma Y, Zhai T, Shen F, Zeng Y, Fu H, et al. [223] Wormuth K. Superparamagnetic latex via inverse emul-
Silver nanoparticles stabilized by thermoresponsive microgel sion polymerization. J Colloid Interface Sci 2001;241:
particles: synthesis and evidence of an electron donor– 366–77.
acceptor effect. Macromol Rapid Commun 2007;28: [224] Xu ZZ, Wang CC, Yang WL, Deng YH, Fu SK.
2339–45. Encapsulation of nanosized magnetic iron oxide by
[205] Brugger B, Richtering W. Magnetic, thermosensitive polyacrylamide via inverse miniemulsion polymerization.
microgels as stimuli-responsive emulsifiers allowing for J Magn Magn Mater 2004;277:136–43.
remote control of separability and stability of oil in water- [225] Voorn DJ, Ming W, Van Herk AM. Polymer–clay
emulsions. Adv Mater 2007;19:2973–8. nanocomposite latex particles by inverse pickering emul-
[206] Hu Z, Xia X, Marquez M, Weng H, Tang L. Controlled sion polymerization stabilized with hydrophobic montmor-
release from and tissue response to physically bonded illonite platelets. Macromolecules 2006;39:2137–43.
hydrogel nanoparticle assembly. Macromol Symp 2005; [226] Antonietti M, Landfester K. Polyreactions in miniemul-
227:275–84. sions. Prog Polym Sci 2002;27:689–757.
[207] Lopez VC, Hadgraft J, Snowden MJ. The use of colloidal [227] Dowding PJ, Vincent B, Williams E. Preparation and
microgels as a (trans)dermal drug delivery system. Int J swelling properties of poly(NIPAM) ‘‘Minigel’’ particles
Pharm 2005;292:137–47. prepared by inverse suspension polymerization. J Colloid
[208] Berndt I, Pedersen JS, Richtering W. Temperature-sensitive Interface Sci 2000;221:268–72.
core–shell microgel particles with dense shell. Angew Chem [228] Owens III DE, Jian Y, Fang JE, Slaughter BV, Chen Y-H,
Int Ed 2006;45:1737–41. Peppas NA. Thermally responsive swelling properties of
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 475

polyacrylamide/poly(acrylic acid) interpenetrating polymer nanoparticles evading reticuloendothelial system. Int J


network nanoparticles. Macromolecules 2007;40:7306–10. Pharm 2000;202:1–10.
[229] Missirlis D, Tirelli N, Hubbell JA. Amphiphilic hydrogel [245] Bharali DJ, Sahoo SK, Mozumdar S, Maitra A. Cross-
nanoparticles. Preparation, characterization, and prelimin- linked polyvinylpyrrolidone nanoparticles as a potential
ary assessment as new colloidal drug carriers. Langmuir carrier for hydrophilic drugs. J Colloid Interface Sci
2005;21:2605–13. 2003;258:415–23.
[230] Rubio-Retama J, Tamimi FM, Heinrich M, Lopez- [246] McAllister K, Sazani P, Adam M, Cho MJ, Rubinstein M,
Cabarcos E. Synthesis and characterization of poly(mag- Samulski RJ, et al. Polymeric nanogels produced via
nesium acrylate) microgels. Langmuir 2007;23:8538–43. inverse microemulsion polymerization as potential gene
[231] Zhu W, Wang B, Zhang Y, Ding J. Preparation of a and antisense delivery agents. J Am Chem Soc 2002;
thermosensitive and biodegradable microgel via polymer- 124:15198–207.
ization of macromonomers based on diacrylated Pluronic/ [247] Craparo EF, Cavallaro G, Bondi ML, Mandracchia D,
oligoester copolymers. Eur Polym J 2005;41:2161–70. Giammona GPE. Gylated nanoparticles based on a poly-
[232] Zhang Y, Zhu W, Ding J. Preparation of thermosensitive aspartamide. Preparation, physico-chemical characterization,
microgels via suspension polymerization using different and intracellular uptake. Biomacromolecules 2006;7:3083–92.
temperature protocols. J Biomed Mater Res A 2005;75A: [248] Gao D, Xu H, Philbert MA, Kopelman R. Ultrafine
342–9. hydrogel nanoparticles: synthetic approach and therapeutic
[233] Murthy N, Xu M, Schuck S, Kunisawa J, Shastri N, application in living cells. Angew Chem Int Ed 2007;46:
Frechet JMJ. A macromolecular delivery vehicle for 2224–7.
protein-based vaccines: acid-degradable protein-loaded [249] Gao D, Agayan RR, Xu H, Philbert MA, Kopelman R.
microgels. Proc Natl Acad Sci USA 2003;100:4995–5000. Nanoparticles for two-photon photodynamic therapy in
[234] Goh SL, Murthy N, Xu M, Frechet JMJ. Cross-linked living cells. Nano Lett 2006;6:2383–6.
microparticles as carriers for the delivery of plasmid DNA [250] Matyjaszewski K, Davis TP, editors. Handbook of radical
for vaccine development. Bioconjugate Chem 2004;15: polymerization. New York: Wiley; 2002.
467–74. [251] Davis KA, Matyjaszewski K. Statistical, gradient, block,
[235] Candau F. Inverse emulsion and microemulsion polymer- and graft copolymers by controlled/living radical polymer-
ization. In: Lovell P, El-Aasser MS, editors. Emulsion izations. Adv Polym Sci 2002;159:2–166.
polymerization and emulsion polymers. West Sussex, [252] Coessens V, Pintauer T, Matyjaszewski K. Functional
England: Wiley; 1997. p. 723–41. polymers by atom transfer radical polymerization. Prog
[236] Nagarajan R, Wang C-C. Theory of surfactant aggregation Polym Sci 2001;26:337–77.
in water/ethylene glycol mixed solvents. Langmuir 2000;16: [253] Nicolas J, Mantovani G, Haddleton DM. Living radical
5243–52. polymerization as a tool for the synthesis of polymer–
[237] Braun O, Selb J, Candau F. Synthesis in microemulsion protein/peptide bioconjugates. Macromol Rapid Commun
and characterization of stimuli-responsive polyelectrolytes 2007;28:1083–111.
and polyampholytes based on N-isopropylacrylamide. [254] Wang J-S, Matyjaszewski K. Controlled/living radical
Polymer 2001;42:8499–510. polymerization. Atom transfer radical polymerization in
[238] Fernandez VVA, Tepale N, Sanchez-Diaz JC, Mendizabal the presence of transition-metal complexes. J Am Chem
E, Puig JE, Soltero JFA. Thermoresponsive nanostructured Soc 1995;117:5614–5.
poly(N-isopropylacrylamide) hydrogels made via inverse [255] Patten TE, Xia J, Abernathy T, Matyjaszewski K.
microemulsion polymerization. Colloid Polym Sci 2006; Polymers with very low polydispersities from atom transfer
284:387–95. radical polymerization. Science 1996;272:866–8.
[239] Juranicova V, Kawamoto S, Fujimoto K, Kawaguchi H, [256] Patten TE, Matyjaszewski K. Atom-transfer radical poly-
Barton J. Inverse microemulsion polymerization of acryla- merization and the synthesis of polymeric materials. Adv
mide in the presence of N,N-dimethylacrylamide. Angew Mater 1998;10:901–15.
Makromol Chem 1998;258:27–31. [257] Matyjaszewski K, Xia J. Atom transfer radical polymeriza-
[240] Barton J. Inverse microemulsion polymerization of oil- tion. Chem Rev 2001;101:2921–90.
soluble monomers in the presence of hydrophilic polyacry- [258] Kamigaito M, Ando T, Sawamoto M. Metal-catalyzed
lamide nanoparticles. Macromol Symp 2002;179:189–208. living radical polymerization. Chem Rev 2001;101:
[241] Renteria M, Munoz M, Ochoa JR, Cesteros LC, Katime I. 3689–745.
Acrylamide inverse microemulsion polymerization in a [259] Tsarevsky NV, Matyjaszewski K. ‘‘Green’’ atom transfer
paraffinic solvent: rolling-M-245. J Polym Sci A: Polym radical polymerization: from process design to preparation
Chem 2005;43:2495–503. of well-defined environmentally friendly polymeric materi-
[242] Kaneda I, Sogabe A, Nakajima H. Water-swellable als. Chem Rev 2007;107:2270–99.
polyelectrolyte microgels polymerized in an inverse micro- [260] Braunecker WA, Matyjaszewski K. Controlled/living
emulsion using a nonionic surfactant. J Colloid Interface radical polymerization: features, developments, and per-
Sci 2004;275:450–7. spectives. Prog Polym Sci 2007;32:93–146.
[243] Deng Y, Wang L, Yang W, Fu S, Elaissari A. Preparation [261] Georges MK, Veregin RPN, Kazmaier PM, Hamer GK.
of magnetic polymeric particles via inverse microemulsion Narrow molecular weight resins by a free-radical polymer-
polymerization process. J Magn Magn Mater 2003;257: ization process. Macromolecules 1993;26:2987–8.
69–78. [262] Hawker CJ, Bosman AW, Harth E. New polymer synthesis
[244] Gaur U, Sahoo SK, De TK, Ghosh PC, Maitra A, Ghosh by nitroxide mediated living radical polymerizations. Chem
PK. Biodistribution of fluoresceinated dextran using novel Rev 2001;101:3661–88.
ARTICLE IN PRESS
476 J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477

[263] Perrier S, Takolpuckdee P. Macromolecular design via [281] Min K, Oh JK. Matyjaszewski Kpreparation of gradient
reversible addition-fragmentation chain transfer (RAFT)/ copolymers via ATRP in miniemulsion II. Forced gradient.
xanthates (MADIX) polymerization. J Polym Sci A: Polym J Polym Sci A: Polym Chem 2007;45:1413–23.
Chem 2005;43:5347–93. [282] Szkurhan AR, Georges MK. Stable free-radical emulsion
[264] Chiefari J, Chong YK, Ercole F, Krstina J, Jeffery J, Le polymerization. Macromolecules 2004;37:4776–82.
TPT, et al. Living free-radical polymerization by reversible [283] Cunningham M, Lin M, Buragina C, Milton S, Ng D, Hsu
addition-fragmentation chain transfer: the RAFT process. CC, et al. Maximizing polymer livingness in nitroxide-
Macromolecules 1998;31:5559–62. mediated miniemulsion polymerizations. Polymer 2005;46:
[265] Barner L, Davis TP, Stenzel MH, Barner-Kowollik C. 1025–32.
Complex macromolecular architectures by reversible addi- [284] Nicolas J, Charleux B, Guerret O, Magnet S. Nitroxide-
tion fragmentation chain transfer chemistry: theory and mediated controlled free-radical emulsion polymerization
practice. Macromol Rapid Commun 2007;28:539–59. of styrene and n-butyl acrylate with a water-soluble
[266] Rettig H, Krause E, Borner HG. Atom transfer radical alkoxyamine as initiator. Angew Chem Int Ed 2004;43:
polymerization with polypeptide initiators: a general 6186–9.
approach to block copolymers of sequence-defined poly- [285] Nicolas J, Charleux B, Guerret O, Magnet S. Nitroxide-
peptides and synthetic polymers. Macromol Rapid Com- mediated controlled free-radical emulsion polymerization
mun 2004;25:1251–6. using a difunctional water-soluble alkoxyamine initiator.
[267] Mei Y, Beers KL, Byrd HCM, Vanderhart DL, Washburn Toward the control of particle size, particle size distribu-
NR. Solid-phase ATRP synthesis of peptide–polymer tion, and the synthesis of triblock copolymers. Macro-
hybrids. J Am Chem Soc 2004;126:3472–6. molecules 2005;38:9963–73.
[268] Qi K, Ma Q, Remsen EE, Clark Jr CG, Wooley KL. [286] Nicolas J, Charleux B, Guerret O, Magnet S. Novel SG1-
Determination of the bioavailability of biotin conjugated based water-soluble alkoxyamine for nitroxide-mediated
onto shell crosslinked nanoparticles. J Am Chem Soc controlled free-radical polymerization of styrene and n-
2004;126:6599–607. butyl acrylate in miniemulsion. Macromolecules 2004;37:
[269] Bontempo D, Maynard HD. Streptavidin as a macro- 4453–63.
initiator for polymerization: in situ protein–polymer con- [287] Delaittre G, Nicolas J, Lefay C, Save M, Charleux B.
jugate formation. J Am Chem Soc 2005;127:6508–9. Surfactant-free synthesis of amphiphilic diblock copolymer
[270] Qiu J, Charleux B, Matyjaszewski K. Controlled/living nanoparticles via nitroxide-mediated emulsion polymeriza-
radical polymerization in aqueous media: homogeneous tion. Chem Commun 2005:614–6.
and heterogeneous systems. Prog Polym Sci 2001;26: [288] McHale R, Aldabbagh F, Zetterlund PB, Okubo M.
2083–134. Nitroxide-mediated radical dispersion polymerization of
[271] Cunningham MF. Living/controlled radical polymeriza- styrene in supercritical carbon dioxide using a poly
tions in dispersed phase systems. Prog Polym Sci 2002; (dimethylsiloxane-block-styrene) alkoxy amine as initiator
27:1039–67. and stabilizer. Macromol Rapid Commun 2006;27:
[272] Asua JM. Miniemulsion polymerization. Prog Polym Sci 1465–71.
2002;27:1283–346. [289] Biasutti JD, Davis TP, Lucien FP, Heuts JPA. Reversible
[273] Schork FJ, Luo Y, Smulders W, Russum JP, Butte A, addition-fragmentation chain transfer polymerization of
Fontenot K. Miniemulsion polymerization. Adv Polym Sci methyl methacrylate in suspension. J Polym Sci A: Polym
2005;175:129–255. Chem 2005;43:2001–12.
[274] Li M, Jahed NM, Min K, Matyjaszewski K. Preparation of [290] Bussels R, Bergman-Goettgens C, Meuldijk J, Koning C.
linear and star-shaped block copolymers by atrp using Multiblock copolymers synthesized in aqueous dispersions
simultaneous reverse and normal initiation process in bulk using multifunctional RAFT agents. Polymer 2005;46:
and miniemulsion. Macromolecules 2004;37:2434–41. 8546–54.
[275] Min K, Gao H, Matyjaszewski K. Preparation of homo- [291] Ferguson CJ, Hughes RJ, Nguyen D, Pham BTT, Gilbert
polymers and block copolymers in miniemulsion by ATRP RG, Serelis AK, et al. Ab initio emulsion polymerization
using activators generated by electron transfer (AGET). by RAFT-controlled self-assembly. Macromolecules
J Am Chem Soc 2005;127:3825–30. 2005;38:2191–204.
[276] Min K, Gao H, Matyjaszewski K. Development of an ab [292] Liu S, Hermanson KD, Kaler EW. Reversible addition-
initio emulsion atom transfer radical polymerization: from fragmentation chain transfer polymerization in microemul-
microemulsion to emulsion. J Am Chem Soc 2006;128: sion. Macromolecules 2006;39:4345–50.
10521–6. [293] Huang X, Sudol ED, Dimonie VL, Anderson CD, El-
[277] Min K, Matyjaszewski K. Atom transfer radical polymer- Aasser MS. Stability in styrene/HD miniemulsions
ization in microemulsion. Macromolecules 2005;38:8131–4. containing a RAFT agent. Macromolecules 2006;39:
[278] Okubo M, Minami H, Zhou J. Preparation of block 6944–50.
copolymer by atom transfer radical seeded emulsion [294] Simms RW, Cunningham MF. Reverse atom transfer
polymerization. Colloid Polym Sci 2004;282:747–52. radical polymerization of butyl methacrylate in a mini-
[279] Eslami H, Zhu S. Emulsion atom transfer radical emulsion stabilized with a cationic surfactant. J Polym Sci
polymerization of 2-ethylhexyl methacrylate. Polymer A: Polym Chem 2006;44:1628–34.
2005;46:5484–93. [295] Simms RW, Cunningham MF. High molecular weight
[280] Min K, Jakubowski W, Matyjaszewski K. AGET ATRP in poly(butyl methacrylate) by reverse atom transfer radical
the presence of air in miniemulsion and in bulk. Macromol polymerization in miniemulsion initiated by a redox
Rapid Commun 2006;27:594–8. system. Macromolecules 2007;40:860–6.
ARTICLE IN PRESS
J.K. Oh et al. / Prog. Polym. Sci. 33 (2008) 448–477 477

[296] Oh JK, Perineau F, Matyjaszewski K. Preparation of [306] Kim KH, Kim J, Jo WH. Preparation of hydrogel
nanoparticles of well-controlled water-soluble homopoly- nanoparticles by atom transfer radical polymerization of
mers and block copolymers using an inverse miniemulsion N-isopropylacrylamide in aqueous media using PEG
ATRP. Macromolecules 2006;39:8003–10. macro-initiator. Polymer 2005;46:2836–40.
[297] Oh JK, Tang C, Gao H, Tsarevsky NV, Matyjaszewski K. [307] Chan Y, Bulmus V, Zareie MH, Byrne FL, Barner L,
Inverse miniemulsion ATRP: a new method for synthesis Kavallaris M. Acid-cleavable polymeric core–shell particles
and functionalization of well-defined water-soluble/cross- for delivery of hydrophobic drugs. J Control Release 2006;
linked polymeric particles. J Am Chem Soc 2006;128: 115:197–207.
5578–84. [308] Oh JK, Siegwart DJ, Lee H-i, Sherwood G, Peteanu L,
[298] Oh JK, Dong H, Zhang R, Matyjaszewski K, Schlaad H. Hollinger JO, et al. Biodegradable nanogels prepared by
Preparation of nanoparticles of double-hydrophilic PEO- atom transfer radical polymerization as potential
PHEMA block copolymers by AGET ATRP in inverse drug delivery carriers: synthesis, biodegradation, in vitro
miniemulsion. J Polym Sci A: Polym Chem 2007;45: release, and bioconjugation. J Am Chem Soc 2007;129:
4764–72. 5939–45.
[299] Qi G, Jones CW, Schork FJ. RAFT inverse miniemulsion [309] Hawker CJ, Wooley KL. The convergence of syn-
polymerization of acrylamide. Macromol Rapid Commun thetic organic and polymer chemistries. Science 2005;309:
2007;28:1010–6. 1200–5.
[300] Tsarevsky NV, Matyjaszewski K. Combining atom transfer [310] Lutz J-F, Boerner HG, Weichenhan K. Combining ATRP
radical polymerization and disulfide/thiol redox chemistry: and ‘‘click’’ chemistry: a promising platform toward
a route to well-defined (Bio)degradable polymeric materi- functional biocompatible polymers and polymer bioconju-
als. Macromolecules 2005;38:3087–92. gates. Macromolecules 2006;39:6376–83.
[301] Ide N, Fukuda T. Nitroxide-controlled free-radical copo- [311] Devaraj NK, Miller GP, Ebina W, Kakaradov B, Collman
lymerization of vinyl and divinyl monomers. Evaluation of JP, Kool ET, et al. Chemoselective covalent coupling of
pendant-vinyl reactivity. Macromolecules 1997;30:4268–71. oligonucleotide probes to self-assembled monolayers. J Am
[302] Huang J, Cusick B, Pietrasik J, Wang L, Kowalewski T, Chem Soc 2005;127:8600–1.
Lin Q, et al. Synthesis and in situ atomic force microscopy [312] Oh JK, Siegwart DJ, Matyjaszewski K. Synthesis
characterization of temperature-responsive hydrogels based and biodegradation of nanogels as delivery carriers
on poly(2-(dimethylamino)ethyl methacrylate) prepared by for carbohydrate drugs. Biomacromolecules 2007;8:
atom transfer radical polymerization. Langmuir 2007;23: 3326–31.
241–9. [313] Carelli S, Ceriotti A, Cabibbo A, Fassina G, Ruvo M, Sitia
[303] Xu F-J, Kang E-T, Neoh K-G. pH- and temperature- R. Cysteine and glutathione secretion in response to protein
responsive hydrogels from crosslinked triblock copolymers disulfide bond formation in the ER. Science 1997;277:
prepared via consecutive atom transfer radical polymeriza- 1681–4.
tions. Biomaterials 2006;27:2787–97. [314] Reichelt W, StabelBurow J, Pannicke T, Weichert H,
[304] Gao H, Min K, Matyjaszewski K. Determination of gel Heinemann U. The glutathione level of retinal Muller glial
point during atom transfer radical copolymerization with cells is dependent on the high-affinity sodium-dependent
cross-linker. Macromolecules 2007;40:7763–70. uptake of glutamate. Neuroscience 1997;77:1213–24.
[305] An Z, Shi Q, Tang W, Tsung C-K, Hawker CJ, Stucky GD. [315] Soderdahl T, Enoksson M, Lundberg M, Holmgren A,
Facile RAFT precipitation polymerization for the micro- Ottersen OP, Orrenius S, et al. Visualization of the
wave-assisted synthesis of well-defined, double hydrophilic compartmentalization of glutathione and protein–
block copolymers and nanostructured hydrogels. J Am glutathione mixed disulfides in cultured cells. Faseb J 2002;
Chem Soc 2007;129:14493–9. 16:124–6.

You might also like