Sei sulla pagina 1di 26

Advances in Polymeric Systems for Tissue

Engineering and Biomedical Applications


Rajeswari Ravichandran, Subramanian Sundarrajan,*
Jayarama Reddy Venugopal, Shayanti Mukherjee, Seeram Ramakrishna*
1. Introduction
The prerequisites of polymer scaffold materials for tissue
engineering applications are manifold and highly challen-
ging. These include (i) biocompatibility of the polymer
material (ii) the material must not elicit unnecessary
inammatory response (iii) the material should not
demonstrate any adverse immune response or cytotoxicity
(iv) similar to all materials incontact withhumanbody, the
scaffolds must be easily sterilizable to prevent infection.
In addition, the mechanical properties of the polymeric
scaffoldmust be compatible andshouldnot collapse during
surgical implantation or during the patients regular
activities. There is an extensive list of criteria a polymer
has to satisfy in order to be applied safely as polymer
therapeutics or as an agent in tissue regeneration. A
polymeric biomaterial intended for tissue engineering
application can be characterized as a material intended to
Review
R. Ravichandran, S. Sundarrajan, J. R. Venugopal,
S. Mukherjee, S. Ramakrishna
Healthcare and Energy Materials Laboratory, Nanoscience and
Nanotechnology Initiative, Faculty of Engineering, National
University of Singapore, Singapore
E-mail: sundar1@nus.edu.sg; seeram@nus.edu.sg
R. Ravichandran, S. Sundarrajan, S. Ramakrishna
Department of Mechanical Engineering, National University of
Singapore, Singapore 117576
The characteristics of tissue engineered scaffolds are major concerns in the quest to fabricate
ideal scaffolds for tissue engineering applications. The polymer scaffolds employed for tissue
engineering applications should possess multifunctional properties such as biocompatibility,
biodegradability and favorable mechanical properties as it comes in direct contact with the
body uids in vivo. Additionally, the polymer system should also possess biomimetic archi-
tecture and should support stem cell adhesion, proliferation and differentiation. As the
progress in polymer technology continues, polymeric biomaterials have taken characteristics
more closely related to that desired for tissue engineering and clinical needs. Stimuli
responsive polymers also termed as smart biomaterials respond to stimuli such as pH,
temperature, enzyme, antigen, glucose and electrical stimuli that are inherently present in
living systems. This review highlights the exciting advancements in these polymeric systems
that relate to biological and tissue engineering appli-
cations. Additionally, several aspects of technology
namely scaffold fabrication methods and surface
modications to confer biological functionality to
the polymers have also been discussed. The ultimate
objective is to emphasize on these underutilized adap-
tive behaviors of the polymers so that novel appli-
cations and new generations of smart polymeric
materials can be realized for biomedical and tissue
engineering applications.
286
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com DOI: 10.1002/mabi.201100325
interface with biological systems to evaluate, treat,
augment, repair or replace any tissue, organ or function
of the body.
[1]
In tissue engineering applications the
polymeric scaffold material serves as a biomimetic
template for cell adhesion, proliferation, differentiation,
extracellular matrix (ECM) formation and mineralization
thereby providing a favorable environment for the
regeneration of damaged tissue. Similarly in the case of
drug delivery applications, if the polymer is not a drug
itself, it often provides a passive function as a drug
carrier, reducing immunogenicity, toxicity or degradation,
while improving circulation time and potentially a passive
targeting function. Inthis case the polymer has to be water-
soluble, non-toxic, non-immunogenic and it needs to be
safe before and after the drug has been released, including
a safe excretion. If the polymer is non-degradable (e.g.,
polymethacrylates), the size needs to be below the renal
threshold ensuring that it is not accumulated in the body.
If the polymer is degradable (e.g., polyesters), the toxicity
and/or immune response of the byproducts have to be
taken into consideration. As the line of development
continues, polymeric biomaterials will take on character-
istics more closely related to that of pharmaceutical and
clinical needs, where the polymer material is designed
to function in a biospecic manner by interacting with
specic biological and biochemical pathways in vivo. The
objective of this review is to put in evidence the evolution
and potentiality of the emerging polymer approaches and
the advancements made to the existing polymer systems
for tissue engineering applications. This review highlights
the exciting developments in polymer technology that
relate to biomedical applications. Toward these ends,
several aspects of technology have been highlighted,
namelyscaffoldfabricationmethods, surfacemodications
to confer biological functionality to the polymers and the
advanced polymeric systems that can be employed in
combination for several tissue engineering and biomedical
applications.
2. Scaffold Fabrication Techniques for
Biomedical Engineering
Current efforts in polymer scaffold fabrication techniques
have been focused on custom designing and synthesizing
polymers with tailored properties for specic biomedical
applications; for instance (i) developing novel polymeric
materials with unique chemistries to increase the
diversity of polymer structure, (ii) developing biosynthetic
processes for fabricating biomimetic polymer structures
and (iii) adopting combinatorial and computational
approaches for scaffold design. Other highly desirable
features concerning scaffold fabrication include near-net-
shape fabrication and scalability for cost-effective produc-
tion. Polymers are the primary materials for scaffold
fabrication and the requirements for following certain
fabrication techniques has ranged from their potential
ability to create scaffolds with controlled porosities,
Rajeswari Ravichandran received her B.Tech in
Biotechnology from Anna University, India. She
then did her M.Eng in Bioengineering from
National University of Singapore (NUS) and is
currently pursuing her PhDfromNUS. Her research
interests include cardiac tissue engineering, bio-
materials and stem cell biology.
Subramanian Sundarrajan graduated with an
M.Sc. in Inorganic Chemistry from the University
of Madras, India in 1996. Later, he worked in a
project at Indian Institute of Science, India till
1999. He his PhD from the University of Madras
in 2003 and joined the NUS in 2003 and currently
working as Senior Research Fellow and he is
working on electrospinning of nanobers, metal
oxide nanoparticles and nanobers for tissue
regeneration applications.
Shayanti Mukherjee completed her B.Tech in
Industrial Biotechnology fromDr. MGRUniversity,
Chennai, India and is now pursuing her Ph.D in
National University of Singapore, Singapore in
myocardial tissue engineering. Her research inter-
est lays in translational regenerative medicine.
Jayarama Reddy Venugopal received his PhD in
neuroendocrinology from University of Madras,
India. At present, he holds an appointment with
National university of Singapore as senior research
fellow. His research experience spans over fabrica-
tion of biocompatible nanobrous scaffolds for
skin, nerve, bone, vascular and cardiac tissue
engineering. His research interests include cell
and molecular biology and nanomedicine.
SeeramRamakrishna received his PhDinMaterials
Science & Engineering from the University of
Cambridge and General Management training
from the Harvard University. He is a Professor
at the department of Mechanical Engineering in
NUS. He is a former Dean of NUS Faculty of
Engineering from 2003 to 2008, and founding
Co-Director of NUS Nanoscience & Nanotechnol-
ogy Initiative from 2003 to 2010. He is a Fellow of
major professional societies in Singapore, UK and
USA including the American Society for Materials
International (ASM); American Society for Mech-
anical Engineers (ASME); American Institute for
Medical & Biological Engineering (AIMBE); Institu-
tion of Mechanical Engineers (IMechE) UK; Insti-
tute of Materials, Minerals & Mining (IOM3) UK;
and Institution of Engineers Singapore (IES).
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
287
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
encapsulation of growth factors, for controlled delivery of
pharmaceutical agents, the removal of residual solvents
following other fabrication methods, and to produce
biomimetic scaffolds for tissue regeneration.
[2]
Materials
fabrication and design will be more closely related to
and actuated by specic clinical needs and biological
phenomena. Some of the widely employed scaffold
fabrication techniques for tissue engineering applications
are discussed in the following section.
2.1. Thermally Induced Phase Separation (TIPS)
3D resorbable polymer scaffolds with very high porosities
can be produced using TIPS technique to develop scaffolds
with controlled macro- and microstructure architectures
suitable for nerve, muscle, tendon, ligament, intestine,
bone, and teeth applications.
[35]
This procedure requires
the use of a solvent with a low boiling point that is easy to
evaporate. For instance, dioxane could be used to dissolve
aliphatic polyesters and subsequently phase separation is
induced through the addition of a small quantity of water.
This leads totheformationof apolymer-richandapolymer-
poor phase, which upon cooling leads to the formation of a
porous scaffold. The polymeric scaffolds obtained via TIPS
arehighlyporous withanisotropic tubular morphologyand
extensive pore interconnectivity. Using TIPS technique the
pore morphology, mechanical properties, bioactivity and
degradationrates can be controlled by varying the polymer
concentrationinsolution, volume fractionof the secondary
phase, quenching temperature and the polymer and
solvent used.
[3]
Injectable scaffolds for tissue engineering
applications to ll voids in damaged tissue are fascinating
due to their ability to conformto the implant site, whereas
preformed scaffolds require prior cognition of the defect to
be lled and those with irregular size and shape can prove
problematic. Amicrosphere networkwithsufcientlylarge
interstices allows tissue to inltrate the network to bear
the mechanical loads as the scaffold degrades. A novel
application of TIPS in tissue engineering is in the rapid
formation of porous microspheres, wherein the pore
morphology and the pore size can be tailored to facilitate
surface modications like the incorporation of bio-
molecules. Such porous biodegradable microspheres are
desirable for tissue engineering and drug delivery applica-
tions becausetheconstituent amount of polymer is reduced
compared with solid microspheres, yet the scaffold volume
is kept constant and the degradation mechanism is more
predictable.
[6]
A study suggested that TIPS microspheres
produced from liquidliquid phase separation of PLGA/
dimethyl carbonate(DMC) andbyanemulsionroute, PLGA/
silver-dopedphosphate-basedglasses bysolidliquidphase
separation, and dispersion of protein particles by phase
separation of PLGA/DMC in the presence of uorescently
labeled antibody; as a suitable candidate for localized drug
delivery, tissue regeneration/augmentation and tissue
engineering.
[6]
Chitosan and collagen are other proteins
that have beenused to fabricate microspheres using TIPS.
[7]
The microspheres developed by TIPS enable control over
both the open pore structure (determined by microsphere
size) and the internal structure, which could be matched to
the desired tissue by adjusting the processing parameters.
A network of biologically active microspheres fabricated
using TIPS could be applied as a tissue engineering scaffold,
or could act as a ller material for inaccessible soft and hard
tissue repair/augmentation.
2.2. Solvent Casting and Particulate Leaching
Solvent casting of polymeric scaffolds involves the
dissolution of the polymer in an organic solvent, mixing
with porogen granules like sugar, inorganic salt, parafn
spheres, and casting the solution into a predened 3D
mould. The size of the porogen particles used will affect the
scaffold porosity, while the polymer to porogen ratio is
directly correlated to the amount of porosity of the nal
structure. The solvent is later allowed to evaporate and the
porogen particles are removed by leaching, following
the main processing step. The main advantage of this
fabrication technique is the ease of fabrication without the
need for any specialized equipment; however organic
solvents must be fully removed in order to avoid any
possible damage to the cells seeded on the scaffold. A
study reported an enhanced solvent casting/particulate
leaching (SCPL) method developed for preparing three-
dimensional porous polyurethane (PU) scaffolds for
cardiac tissue engineering applications.
[8]
It involved a
combination of SCPL with centrifugation; with the
aim to enhance the pore uniformity and the pore
interconnectivity. These scaffolds showed uniform distri-
bution of the human aortic endothelial cells, useful for
cardiac tissue engineering applications.
2.3. Solid Freeform Fabrication Techniques (SFFT)
This technique is employedto fabricate highlyreproducible
scaffolds with completely interconnected porous net-
works.
[9,10]
Using digital data produced by imaging soft-
ware suchas computer tomography or magnetic resonance
imaging enables appropriate design of the polymeric
scaffold structure.
[10]
Solid freeform (SFF) manufacturing
coupled with conventional foam scaffold fabrication
procedures (phase separation, emulsion-solvent diffusion
or porogen leaching) may be used to fabricate materials
with controlled micro- and macroporous architectures.
Suchbiomimetic internal architectures mayprove valuable
for multi-tissue and structural tissue interface engineering.
Unlike conventional computerized machining techniques
which involve the removal of materials from a stock,
288
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
SFF process employs the underlying concept of layered
manufacturing,
[11]
whereby three-dimensional objects
are fabricated with layer-by-layer building via the proces-
sing of solid sheet, liquid or powder material stocks. For
instance, Xiong et al.
[8]
fabricated composites of PLLA/TCP
with porosities of up to 90% as shown in Figure 1, with
mechanical properties close to human cancellous bone by
using low-temperature depositionbased ona layer-by-layer
manufacturingmethodof SFF fabrication. Theexibilityand
manufacturing advantages of SFF have been employed for
biological applications ranging from the production of
scale replicas of human bones
[12]
and body organs
[13]
to
the advancedcustomized drug deliverydevices
[14]
andother
areas of medical sciences including medical forensics.
[15]
Three-dimensional printing (3D-P) is the most widely
investigated SFF technique for scaffold fabrication.
Kim et al.
[16]
employed 3D-P with particulate leaching
technique for creating porous scaffolds using polylactide-
co-glycolide (PLGA) powder mixed with salt particles and a
suitable organic solvent. Electron microscopy results
performed 2 days after the in vitro cell culture with
hepatocytes (HCs) revealed the successful attachment of
largenumbers of HCs ontothescaffolds. Similarly, Zeltinger
et al.
[17]
employed3D-P-fabricatedporous poly(L-lactic acid)
(L-PLA) disc shaped scaffolds measuring 10 mm (diameter)
by 2 mm (height) to investigate the inuence of pore size
and porosity on cell adhesion, proliferation and matrix
deposition. The scaffolds were constructed with two
different porosities (75% and 90%) and four different pore
size distributions (<38, 3863, 63106 and 106150mm)
andwereassessedwithcell cultureusingthreedifferent cell
types, canine dermal broblasts, vascular smooth muscle
cells and microvascular epithelial cells. Cell culture results
demonstrated the suitability of 3D-P fabricated scaffolds in
supporting tissue development for different cell types.
AlthoughtheSFFtechniques havedisadvantages attributed
to limited material range and small pore sizes, most of the
limitations can be overcome through slight modications
carried out on the original hardware and software set-up
of commercialized systems and also the processing
procedures and material stocks employed. For instance,
on the 3D-P systems, water-based binder systems can be
formulated to replace the use of toxic solvents, which are
harmful for cell culture.
[18]
The poor mechanical strength
of 3D-P scaffolds can be improved by inltrating the
fabricated structure with a polymer. The application of
CAD strategies in connection with SFF fabrication will
allow scaffolds with highly uniform pore morphologies,
unlimited range of pore sizes, porosity and complete pore
interconnectivity to be realized with unprecedented
accuracy and consistency, desired for biological applica-
tions. Dropondemandprinting(DDP) is alsoaSFFtechnique
capable of generating microscale physical features
desired for tissue engineering scaffolds. A study reported
a reproducible manufacturing process for tissue engineer-
ing scaffolds based on injectable porogens fabricated by
DDP technique. They designed scaffolds composed of either
pure polycaprolactone (PCL) or homogeneous composites
of PCL and calcium phosphate (CaP, 10% or 20% w/w) by
injection molding of molten polymer-ceramic composites,
followed by porogen dissolution with ethanol. Human
embryonic palatal mesenchymal (HEPM) cells attached
and proliferated on all these scaffolds, as evidenced by
uorescent nuclear staining with Hoechst 33258 and
the Alamar Blue assay, with increased proliferation
observed on 80:20 PCLCaP scaffolds, proving that SFF-
based injection molding process for the fabrication of
PCL and PCLCaP scaffolds has suitable in vitro cyto-
compatibility and mechanical properties for hard tissue
repair applications.
[19]
2.4. Phase Separation
Phase separation has been employed for several years as a
manufacturing technique to create porous polymer
membranes. Ma and Zhang
[20]
pioneered the use of this
technique to produce nanobrous three-
dimensional scaffolds whose macropor-
ous structure can be tailored to suit the
desired applications. Employing phase
separation, scaffolds can be created
froma variety of biodegradable aliphatic
polyesters, composed of bers with
diameters that range from 50
to 500nm.
[2125]
Phase separation is a
thermodynamic separation of a polymer
solution into a polymer-rich component
and a polymer-poor/solvent-rich compo-
nent. Fundamentally, a polymer is
dissolved in solution and the phase
separation is induced, either thermally
or throughtheadditionof anon-solvent to
Figure 1. SEM images of the cross-section of the PLLA/TCP composite scaffold: (a) low
magnication and (b) high magnication.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
289
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
the polymer solution to create a gel. Water is then used to
extract the solvent from the gel; the gel is cooled to a
temperature below the glass transition temperature of the
polymer and freeze dried under vacuum to produce a
nanobrous scaffold.
[2022]
The desired architecture can be
obtained through the addition of various porogens to
polymer solution during the phase separation process. This
provides engineers with a signicant amount of control in
tailoring both the pore sizes and interconnectivity of the
resultant polymeric material by altering the concentration,
size, and geometry of the porogens used.
[21]
Unlike other
techniques, phase separation is a simple process that does
not require much specialized equipment. Besides, it is also
easy to achieve batch-to-batch consistency; and tailoring of
scaffold mechanical properties and architecture is easily
achieved by varying polymer/porogen concentrations.
However, thisfabricationprocess islimitedtobeingeffective
with only a selective number of polymers and is strictly a
laboratory scale technique.
[20]
2.5. Electrospinning
Inthelast decade, electrospinningtechniquehas attracteda
great interest as it allows the fabrication of brous non-
woven micro/nano fabrics for tissue engineering applica-
tions, mainly due to the structural similarity to the tissue
extracellular matrix (ECM). Electrospinning technique
involves the application of high voltage to a polymeric
solution, in order to create an electrically charged jet
randomly collected onto a grounded target.
[26,27]
Electro-
spinning is a simple and versatile method to prepare ultra
thinbers frompolymer solutions or melts. It utilizes ahigh
voltage source to inject charge of a certain polarity into
a polymer solution or melt, which is then accelerated
toward a collector of opposite polarity. As the electrostatic
attraction between the oppositely charged liquid and
collector and the electrostatic repulsions between like
charges in the liquid become stronger, the leading edge of
the solution changes from a rounded
meniscus to a cone (the Taylor cone). A
ber jet is eventually ejected from the
Taylor cone as the electric eld strength
exceeds the surface tension of the liquid.
The ber jet travels through the atmo-
sphere allowing the solvent to evaporate,
thus leading to the deposition of solid
polymer bers on the collector.
By modifying variables such as the
distance to collector, magnitude of
applied voltage, or solution ow rate,
researchers can dramatically change the
overall scaffold architecture depending
on the desired application. The bres
produced by this technique usually have
a diameter fromseveral nanometers to a fewmicrometers.
Electrospun polymer nanobres possess many extraordin-
ary properties including small diameters, favorable biomi-
metic architecture, concomitant largespecic surfaceareas,
large surface to volume ratio which favors enhanced
protein adsorption, a high degree of structural perfection
andgoodmechanical properties. Inorder tomoreaccurately
mimic the natural ECM, research has also examined the
electrospinning of natural materials such as: collagen,
[28]
chitosan
[29]
and gelatin.
[30]
However, these materials often
lack the desired physical properties or are difcult to
electrospinontheir own, whichhas led to the development
of hybrid materials, which consist of a blend of synthetic
andnatural polymers.
[3032]
Stitzel et al. studied the use of a
hybrid blend of type I collagen (45%), PLGA (40%), and
elastin (15%) to form a vascular prosthesis using electro-
spinning.
[31]
The addition of PLGA was shown to improve
mechanical properties such as burst strength and com-
pliance of the prosthesis in comparison to scaffolds
composed solely of type I collagen and elastin alone.
Electrospun bers can be oriented or arranged randomly,
givingcontrol over boththebulkmechanical properties and
the biological response to the scaffold. For example, in
designing scaffolds meant to replace highly oriented tissue
such as the medial layer of a native artery it is desirable to
generate aligned nanobers. In the medial layer both
smooth muscle cells and ECM brils are aligned circumfer-
entially, which allows for vasoconstriction and vasodila-
tion in response to corresponding stimuli. Xu et al.
developed an aligned nanobrous scaffold using electro-
spinning of a poly (L-lactide-co-e-caprolactone) (P(LLA-CL))
(75:25) copolymer for vascular tissue engineering.
[32]
Smooth muscle cells attached and migrated along the axis
of the aligned bers. Furthermore, the proteins comprising
the cytoskeleton of the smooth muscle cells were aligned
parallel to the aligned bers, demonstrating the cells
proclivity to organize along oriented ber topography.
Figure 2 shows a SEM image of electrospun gelatin
Figure 2. SEM image of electrospun gelatin nanobers at a) 500x and b) 5000x
magnication.
290
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
nanobers at 500x and 5000x magnication. A detailed
review concerning the performance of nanobrous poly-
meric materials in guiding cells to initially adhere
and spread over the material, as well as further triggering
them to differentiate and secrete the appropriate ECM
biomolecules targeted to skin, blood vessel, cartilage,
muscle, adipose, nerve and bone tissue engineering
applications is dealt elsewhere.
[33]
The various advantages
and disadvantages of the scaffold fabrication techniques
aresummarizedinTable1. Whilethesizescale, porosityand
orientation of scaffolds fabricated by these techniques can
be modied to inuence cell functions such as adhesion,
proliferation, and migration, even greater enhancement
over the control of cellular function can be achieved by
attachingbioactive molecules tothe surface of the scaffolds
as discussed in the following section.
3. Surface Functionalization of Polymeric
Materials for Tissue Engineering
Over the years, several advancements in polymeric
materials have addressed biological aspects of increased
complexity, starting on the level of ion interactions
and moving to growth factor and stem cell incorporation.
The polymeric materials were extended from purely
Table 1. Various scaffold fabrication techniques, their advantages and disadvantages and their potential applications.
Fabrication technique Advantages Disadvantages Applications
Thermally induced
phase separation
Can control the porosity
and pore morphology
Achievable sizes range
from only 10 to 2000 mm
in diameter
Proteins and drug delivery
and higher drug encapsulation
efciency
Solvent casting
and particulate
leaching
Simple operation, control
of the pore size and
porosity by selecting the
particle size and the
amount of salt particles
Distribution of salt particles
is often not uniform within
the polymer solution, and
the degree of direct contact
between the salt particles is
not well controlled,
interconnectivity of pores
in a nal scaffold cannot be
well controlled, limited
membrane thickness, lack
of mechanical strength,
problems with residual
solvent, residual porogens.
Cardiac and vascular tissue
engineering applications
Solid freeform
fabrication
techniques
Customized design,
computer-controlled
fabrication, anisotrophic
scaffold microstructures,
processing conditions
Lack of mechanical strength,
limited to small pore sizes
Production of scale replicas
of human bones and body
organs to advanced
customized drug delivery
devices
Phase separation Allows incorporation of
bioactive agents,
highly porous structures
Lack of control over
micro-architecture,
problems with residual
solvent, limited range
of pore sizes
Drug release and protein
delivery applications
Electrospinning Easy process, High porosity,
High surface
area to volume ratio
Limit range of polymers,
lack of mechanical strength,
problems with residual
solvent, lack of control over
micro-architecture
Bone, skin, nerve and cardiac
tissue engineering
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
291
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
synthetic materials to material/biologic hybrids, engi-
neering at the same time bioactivity and biodegradability
by imparting biological cues into the polymer. For
instance, in order to apply biodegradable polyester based
nanocomposite in tissue engineering, their surfaces have
to be chemically and physically modied with the
incorporation of bioactive molecules and cell binding
proteins. This provides the desired biomimetic micro-
environments for cell adhesion, proliferation and differ-
entiation. Therefore, many approaches to functionalize
the surface of biodegradable polymer scaffolds have been
undertaken in order to introduce useful surface character-
istics to the polymer for tissue engineering applications.
The scaffold environment should be able to present
and deliver combinations of biomolecules such as cell
adhesion motifs, protein molecules, growth factors,
angiogenic factors, differentiation factors and immuno-
suppressive or anti-inammatory agents.
3.1. Stem Cells
Stem cell incorporation into polymeric scaffolds is of
immense potential for creating next-generation syn-
thetic/living composite biomaterials that feature
highadaptability to the biological environment. Scaffolds
seeded with stem cells allow the cells to undergo
differentiation to adapt the desired tissue engineering
approach. This approach enables the polymeric scaffold
surface to mimic complex biological functions leading to
in vitro and in vivo growth of tissues and organs. A
combination of mesenchymal stem cells, growth factors,
and bioresorbable polymers can provide a solution for the
treatment of difcult tendon injuries. A knitted PLGA
matrix populated with allogeneic bone-marrow-derived
MSCs was used to bridge Achilles tendon defects in adult
rabbits.
[34]
In this study the specimens treated with MSCs
showed a higher rate of tissue regeneration and remodel-
ing at 2 and 4 weeks after surgery compared with the
group treated withPLGAalone. Similarly ina recent study
byour group, acombinationof MSCs withcardiomyocytes
cultured on an elastomeric poly(glycerol sebacate)/
gelatin nanobrous scaffolds was shown to be favorable
for the regeneration of the infarcted myocardium. In the
study the incorporation of stem cells into the polymeric
material induces paracrine signaling effects, reducing
the cell death of cardiomyocytes.
[35]
Figure 3 shows the
double immunostaining image for both MSC marker
protein and cardiac marker proteins expressed by stem
cells which have undergone cardiogenic differentiation.
Thus the incorporation of stem cells into polymeric
materials lead to the production of next generation
biomimetic scaffolds for various tissue engineering
applications.
3.2. Biomolecules
There is a signicant scope in the application of surface
modications, through the use of protein biomolecules to
provide more cues for cell adhesion, proliferation and
differentiation. Integrins, laminin and Arg-Gly-Asp (RGD)
proteins and also several natural proteins like collagen,
gelatin and brinogen were shown to be essential for cell
attachment to polymeric material surfaces devoid of any
cell recognition sites.
[36,37]
The immobilization of these
proteins to polymers not only promotes cell adhesion and
proliferation but also increases hydrophilicity of hydro-
phobic polymers such as aliphatic polyesters. One such
surface functionalizationfor biopolymer substrate surfaces
is attachment of RGDpeptides that is the most effectiveand
often employed peptide sequence for stimulating cell
adhesion on synthetic polymer surfaces. This peptide
sequence can interact with integrin receptors at the focal
adhesion points. Once the RGD sequence is recognized by
the integrins, it will initiate an integrin-mediated cell
attachment pathway and activate signal transduction
between the cell and ECM, thus inuencing various cell
behaviors on the substrate including proliferation, differ-
entiation, survival and migration.
[38]
In another study,
Jianget al. usedacoaxial electrospinningset-uptofabricate
biodegradable core/shell bers with PCL as the shell and
BSAcontainingdextranas thecore.
[39]
BothBSAloadingand
its release prole could be controlled by varying the feed
rate of the core solution, with higher feed rates giving
higher BSAloading andacceleratedrelease. Additionof PEG
to the shell was used to further control the release prole,
and was shown to increase release of BSA. By varying the
inner solution feed rate as well as the PEG content of
the shell the authors were able to vary the release period
from 1 week to approximately 1 month, for drug delivery
applications.
3.3. Growth Factors
Scaffolds functionalized with immobilized growth factors
is of utmost importance in several tissue engineering
applications as the embedded growth factors (i) could
be released in response to several cell-mediated stimuli,
(ii) could create a highly regulated network of signaling
molecules for mediating several biological pathways,
(iii) able to orchestrate cell attachment, migration, organi-
zation and proliferation nally giving rise to functional
tissue. The focus of studies during the past decade has been
on numerous growth factors that promote soft-tissue
regenerationsuchas platelet-derived growthfactor (PDGF),
epidermal growth factor, basic broblast growth factor,
insulin-like growth factor-I, bone morphogenetic proteins
(BMPs) and transforming growth factors.
[4045]
Polymeric
systems that provide a gradual and controlled release of
292
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
growthfactors to the site of injuryis of extreme importance
for tissue repair and regeneration. Growth factors like
BMPs were shown in in vivo studies to be osteoinductive.
Immobilizing these growth factors on the scaffold
surface might signicantly shorten the bone healing
process, enhance osseointegration and reduce patient
recovery time. Chew et al. examined the release of b-nerve
growth factor (NGF) stabilized in BSA from a copolymer
consisting of e-caprolactone and ethyl ethylene phosphate
(PCLEEP).
[46]
Due to its relatively hydrophobic backbone,
PCLEEP has a slow degradation rate demonstrating a mass
loss of approximately 7%over a 3-month period. Using this
system, the authors observed a sustained release of NGF
over a period of 3 months. Due to the relatively small
amount of mass loss over this period it was inferred
that NGF release was occurring primarily via diffusion,
demonstrating that a biodegradable systemcan be used to
obtain a desirable release prole while still eliminating the
need for a second surgery for implant removal. A detailed
review of various growth factors and their signicance for
tissue engineering has already been discussed.
[47,48]
3.4. Surface Modication Techniques
Surface modicationtechniques suchas plasma treatment,
ion sputtering, oxidation and corona discharge affect the
chemical and physical properties of the polymer surface
without signicantly changing the inherent bulk material
properties. For example, using plasma processes, it is
possibletochangethechemical compositionandproperties
of the polymer system such as wettability, surface energy,
metal adhesion, refractive index, hardness, chemical
inertness and biocompatibility.
[49]
Plasma techniques can
easily be used to induce the desired groups or chains onto
Figure 3. Dual immunocytochemical analysis for the expression of MSC marker protein CD 105 (a, d, g) and cardiac marker protein actinin
(b, e, h) in the coculture samples and the merged image showing the dual expression of both CD 105 and actinin (c, f, i); on the TCP (a, b, c),
gelatin nanobers (d, e, f), and PGS/gelatin core/shell bers (g, h, i) at 60x magnication. Nucleus stained with DAPI.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
293
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
the surface of a polymer.
[5052]
Appropriate selection of
the plasma source provides the introduction of diverse
functional groups on the polymer surface to improve
biocompatibility or to allow subsequent covalent immo-
bilization of various bioactive cues. For instance, plasma
treatments with oxygen, ammonia, or air can generate
carboxyl groups or amine groups on the polymer sur-
face.
[53,54]
Plasma treatment affects the chemistry of the
biodegradable polymer surface, but at the same time it
also introduces signicant changes in topography.
[55,56]
A
variety of ECM protein components such as gelatin,
collagen, laminin, and bronectin could be immobilized
onto the plasma treated surface to enhance cellular
functions.
[57,58]
In a recent study, it was noticed that
broblasts proliferation, morphology, CMFDA dye expres-
sion and secretion of collagen were signicantly improved
on plasma-treated PLACL/gelatin scaffolds compared to
PLACL nanobrous scaffolds, proving that the plasma-
treated PLACL/gelatin nanobrous scaffold is a potential
biocomposite material for skin tissue regeneration.
[59]
4. Advancements in Polymer Systems for
Cell Culture
Living systems respond to external stimuli adapting
themselves to changing environmental conditions. Poly-
mer scientists recently have been trying to mimic this
behavior for creating smart polymeric systems for tissue
engineeringapplications. Thesesmart polymersaredened
as polymers that undergo reversible large, physical or
chemical changes in response to small external changes in
the environmental conditions, such as temperature, pH,
light, magnetic or electric eld, ionic factors, biological
molecules, etc. Smart polymers have very promising
applications in the biomedical eld as delivery systems
of therapeutic agents, tissue engineering scaffolds, cell
culture supports, bioseparation devices, sensors or actua-
tors systems. Hoffman et al.
[60]
demonstrated, in a very
elegant design, that the actionof anenzymatic receptor can
bemodulatedwhenthis kindof polymer is conjugatedclose
to its active place. The authors were able to switch on-off
the receptor using the transition between extended and
coiled form of the molecule.
[61]
Two different kinds of
bioconjugates including stimuli-responsive polymers can
be prepared for biological applications by: a). Random
polymer conjugationtolysine aminogroups of aprotein. b).
Site-specic conjugation of the polymer to genetically
engineered specic amino acid sites. The placement of
stimuli-sensitive polymers near the active place of a
recognition protein can provide a highly environmental-
sensitive system. Some of the widely employed smart
polymeric systems have been dealt with in detail in the
following sections.
4.1. Nanocomposites Embedded Polymer Systems
Nanocomposites are an efcient strategy to upgrade
the structural and functional properties of synthetic
polymers. Aliphatic polyesters such as polylactide (PLA),
poly(glycolides) (PGA), poly(caprolactone) (PCL) have
attracted wide area for their biodegradability and bio-
compatibility in the human body. However all these
desired properties cannot be achieved from a single
polymer system. In fact, although several polymeric
materials are available and have been investigated for
tissue engineering, no single biodegradable polymer can
meet all the requirements for biomedical applications.
Therefore, the design of multi-component polymer
systems represents a viable strategy in order to
develop innovative multifunctional polymeric materials.
A consequence has been the introduction of organic
and inorganic nanollers into biodegradable polymers
to produce nanocomposites based on hydroxyapatite,
metal nanoparticles or carbon nanotructures, to prepare
advanced polymeric systems with enhanced properties.
This combination of bioresorbable polymers and
nanostructures opens new perspectives in the
application of nanomaterials for biomedical
applications with desirable mechanical, thermal and elec-
trical properties.
4.1.1. Polymer/HA Nanocomposites
Natural bone matrix is an organic/inorganic composite
material of collagen and apatites. Composite material
systems based on inorganic nanoparticles, showed a
strongly enhanced polymer degradation rate when com-
pared to pure polymer systems. Studies have shown that
tricalciumphosphate lled polymers showed deposition of
small, 10 nm sized hydroxyapatite crystals on the surface
of the PLGA polymer composite, while for pure PLGA/
nanohydroxyapatite formation was observed during
degradation, indicative of enhanced osteoconductive
properties of PLGA nanocomposites. The fast degradation
and the superior osteoconductivity make these nanocom-
posites a promising material for application in ortho-
pedics.
[62,63]
These polymer/HA nanocomposites can also
be surface modied for the release of biomolecules. For
instance, Nie and Wang examined the release of DNA from
electrospun scaffolds consisting of a blend of PLGA and
hydroxyapatite (HAp) with various HAp contents (0%, 5%,
and 10%) for bone tissue engineering applications.
[64]
DNA
was incorporatedinto the scaffolds inthree ways: (1) naked
DNA, (2) adsorption of DNA/chitosan nanoparticles onto
scaffolds after scaffold fabricationby dripping, or (3) blend-
ing DNA/chitosan nanoparticles with the PLGA/HAp
solution prior to electrospinning. They noticed that higher
HAp contents led to faster DNA release for both free and
encapsulated DNA. This may be due to the hydrophilic
294
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
nature of HAp, which caused the DNA/chitosan bind to
HAp particles in the presence of dichloromethane
during the emulsion procedure. These techniques not only
increase encapsulation efciency, as was noted by the
authors, but it would also increase the release rate.
As the HAp nanoparticles diffuse out of the PLGA bers
they leave pores through which the DNA/chitosan
particles can easily diffuse through the scaffolds.
The authors noted that encapsulated DNA/chitosan nano-
particles enhanced transfection efciency leading to
higher cell attachment and viability in an in vitro study.
Thus, it was demonstrated that the encapsulation of
DNA/chitosan nanoparticles in PLGA/HAp electrospun
scaffolds has the potential to augment bone tissue
regeneration. In a recent study by our group n-HA was
precipitated by calcium-phosphate dipping method on
PLLA/PBLG/Col scaffolds. The incorporation of n-HA drives
the adipose derived stem cells to osteogenic lineage on
these scaffolds, in the absence of any induction medium,
for bone tissue engineering.
[65]
4.1.2. Polymer Embedded Nanoparticles
Silver (Ag) has been known to have a disinfecting effect
and has been applied in traditional medicines. Several
salts of Ag and their derivatives are commercially
employed as antimicrobial agents. Hence, Ag/polymer
nanocomposites have been investigated for their anti-
bacterial property.
[66,67]
The use of nanoparticles has been
limited by difculties associated with handling and
processing of nanoparticles. Embedding of nanoparticles
into biodegradable polymer matrices represents a valid
solution to these stabilization problems and permits a
controlled effect.
[68]
For instance, Liang et al. examined
the incorporation of DNA into PLGA scaffolds fabricated
by electrospinning.
[69]
Plasmid DNA was condensed in a
poor solvent mixture, and was then encapsulated in
micelles composed of a triblock copolymer (PLA-PEG-PLA)
givingencapsulatedDNAnanoparticles.
The micelles were then dissolved in a
solution of DMF with PLGA and electro-
spun, resulting in the formation of PLGA
bers containing encapsulated DNA
nanoparticles. The DNA was encapsu-
lated in the PLA-PEG-PLA triblock copo-
lymer in order to protect it from
degradation during electrospinning
with the PLGA copolymer. An in vitro
release study demonstrated that
approximately 20% of the encapsulated
DNA was released after a period of 7 d.
Such systems are very useful for
gene delivery and drug delivery applica-
tions.
4.1.3. Polymers Incorporating Carbon Nanostructures
Polymers incorporating carbon nanostructures have been
investigated by several groups for a variety of biomedical
applications.
[7072]
Carbon nanotubes (CNT) have the
potential to provide the needed structural reinforcement
for biomedical polymer scaffold. By dispersing a small
fraction of carbon nanotubes into a polymer, signicant
improvements inthe polymer/CNTcomposites mechanical
strength has been observed. The role of the interface
between the CNT and polymer matrix is essential in
transferring the load from the polymer to the nanotubes,
therebyenhancingthe mechanical andelectrical properties
of the composite. The electrical conductivity of CNT based
nanocomposites is auseful tool inorder todirect cell growth
and cell differentiation since they can conduct electrical
stimulus into the desired tissue, thereby stimulating the
healing process in nerve, bone and cardiac tissue engineer-
ing applications. For example when an alternating current
is applied to the nanocomposites of poly(lactic acid) and
MWCNTs, it showed increased osteoblast proliferation and
calcium production, favorable for bone regeneration as
shown in Figures 4a and b respectively.
[73]
4.2. Polymer Blended Hybrid Systems
Whenasingle polymer does not have the properties desired
for a tissue engineering application, a copolymer or blend
(simple mixture) of polymers may be employed to achieve
the desired properties. Studies have shown that PLA:PCL
blends and copolymers possess great solvent exibility and
also exhibit a percent increase in elasticity while main-
taining an ultimate tensile strength that is analogous to
that of pure PLA scaffolds. It was shown that by adding a
small amount of PCL (as little as 5 wt%) to PLA, the strain to
failure of a scaffold increased from less than 25% to
more than 200%.
[26]
Similarly, Kwon et al. successfully
Figure 4. A) Osteoblast proliferation under electrical stimulation: & without electrical
stimulation; & with electrical stimulation. Values are meanSEM; n4;

p <0.05
(compared to controls; Students t test). B) Calcium content in osteoblast cultures
exposed to electrical stimulation: & without electrical stimulation; & with electrical
stimulation. Values are meanSEM; n3;

p <0.05 (compared to controls; Students t
test).
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
295
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
electrospun poly(L-lactide-co-e-caprolactone) (PLCL) at
various weight percents from methylene chloride in ratios
of 70:30 (9wt%), 50:50 (7 wt%), and 30:70 (11 wt%).
It was found that at 25 8C the 70:30 ratio was a hard solid,
the 50:50 ratio was an elastomer, while the 30:70 ratio
was a gummy solid. The two ratios that resulted in brous
scaffolds, 70:30 and 50:50, were determined to have a
Youngs modulus of 14.2 and 0.8 MPa, respectively,
[74]
showing a great deal of promise for use as an arterial graft.
This is because an engineered vascular graft must be
strong enough to accommodate a large pressure increase
while having enough elasticity to passively expand to
allow blood ow downstream. Similarly in another study
Xu et al. electrospun a nanobrous scaffold of P(LLA-CL)
(75:25). This scaffold exhibited a tensile modulus of
156MPa, tensile strength of 5 MPa, and strain at break of
127%. This compares favorably to the mechanical proper-
ties of native coronary artery having a tensile strength of
1.4011.14 MPa and a strain at break of 4599%.
[75]
As is
true with copolymers, blending of natural polymers and/or
synthetic polymers serves to enable further capability of
tuning a material to attain the desired properties for
tissue engineering applications. For instance, Huang et al.
electrospun collagen type I and poly(ethylene oxide) to
tailor ber morphology and mechanical properties of
scaffolds.
[76]
Additionally collagenGAG scaffold has been
utilized extensively as dermal regenerationtemplates with
unprecedented biological activity to achieve enhanced
healing response.
[77,78]
4.3. Stimuli-responsive Polymers
Stimuli-responsive polymers are also termed as smart
polymers. Interest in stimuli-responsive polymers has
persisted over many decades, and a great deal of effort
has been crafted to fabricate newsmart materials. Stimuli-
responsive polymers show an acute change in properties
upon a slight change in environmental condition. This
property can be utilized for the preparation of smart
polymeric systems, which mimic biological response
behavior to a certain extent. Stimuli-responsive polymers
mimic biological systems in a primitive way where an
external stimulus results in a change in properties. This
can be a change in conformation, change in solubility,
alteration of the hydrophilic/hydrophobic balance or
release of biomolecules or combination of two or more
responses. Many advances in stimuli response polymers
are advantageous in the biomedical elds due to their
specicity and their ability to respond to stimuli that
are inherently present in vivo. The physical or chemical
stimulus that triggers specic responses is limited to the
formation or destruction of secondary forces (hydrogen
bonding, hydrophobic effects, electrostatic interactions,
etc.), simple reactions (e.g., acidbase reactions) of moieties
on the polymer backbone, and/or osmotic pressure
differentials that result from such phenomena.
[79]
Besides,
theresponsecanalsobeexpandedtoincludemoredramatic
alterations to the polymeric structure. For instance,
degradation of polymeric hydrogels upon specic stimulus
can occur by reversible or irreversible bond breakage of the
polymeric backbone or cross-linking groups. Such systems
may facilitate the application of smart polymers in drug
delivery, diagnostics, separations and other clinical appli-
cations. The major advantage of these polymer systems is
their ability to apply these stimuli in a non-invasive
manner in living body. The stimuli may occur internally
(such as change in pH or temperature) or externally
(external stimuli such as magnetic or electric eld, light
andultrasound). Figure5shows aschematic representation
of the various stimuli and their responses.
[79]
4.4. Conducting Polymers
Conducting polymers (CPs) have attracted much interest as
suitable matrices for biomolecules owing to their high
electronic and ionic conductivities, which has been used to
enhance the stability, speed and sensitivity of various
biomedical devices. The electrically active tissues like the
brain, heart and skeletal muscle provide opportunities to
couple electronic devices and computers with human or
animal tissues to create therapeutic bodymachine inter-
faces. The conductive and semiconducting properties of CPs
make them an important class of materials related to such
applications. The origin of electrical conduction in CPs has
been ascribed to the formation of nonlinear defects such
as solitons, polarons or bipolarons formed during either
doping or polymerization of a monomer.
[80]
For example,
Abidian et al. has demonstrated the use of the CPs
polypyrrole (PPy) and poly(3,4-ethylenedioxythiophene)
(PEDOT) for nerve tissue engineering applications, by
culturing neuronal cells using an in vitro dorsal root
ganglion model.
[81]
Electrical stimulation with neural
electrodes is used clinically to improve conditions such as
hearingloss (cochlear implants). It was foundthat, whenPPy
CP was coated onto cultured substrates on which cochlear
explants were cultured, the neurite growth improved upon
the incorporation of neurotrophins like NT-3 and brain-
Figure 5. Potential stimuli and responses of synthetic polymers.
296
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
derived neurotrophic factor (BDNF). Such CP-based smart
biomaterials provide a biocompatible substrate to help
protect auditory neurons from degradation after sensor
neural hearing loss and encourage neurite outgrowth
towards the electrodes.
[82]
Conductive neural interfaces
tailoredfor cell interactionbysurfacefunctionalizationwith
incorporation of bioactive factors produce superior neuro-
prosthetic devices with improved charge transfer capabil-
ities. Astudy examined the effect of entrapping NGF within
the CP PEDOT during electrodeposition to create a polymer
capable of stimulating neurite outgrowth from proximal
neural tissue.
[83]
The incorporation of NGF can modify the
biological interactions of the electrode without compromis-
ing on the conductive properties. Another study demon-
strated the use of PEDOT nanotubes polymerized on top of
electrospun PLGA nanobres for the potential release of the
drug dexamethasone. Here, dexamethasone was incorpo-
rated within the PLGA nanobers and then PEDOT was
polymerized around the dexamethasone-loaded PLGA
nanobers. As the PLGA bres degraded, dexamethasone
molecules remained inside the PEDOT nanotubes. These
PEDOT nanotubes favored controlled release upon electrical
stimulation. This was becauseof thechangeinvolumeof the
PEDOT nanotube upon electrical stimulation owing to the
expulsionof anions. Figure6demonstratestheincorporation
and release mechanism of dexamethasone from PEDOT
nanotubes due to electrical stimulation.
[84]
For cardiovas-
cular applications, Li et al. hasdemonstratedthepotential for
usingPANI asanelectroactivepolymer. Theystudiedvarious
advancements in CPs by covalently attaching oligopeptides
to PANI and electrospinning PANIgelatin blend nanober
scaffold. These scaffolds were analyzed as potential candi-
dates for cardiac tissue engineering applications using H9c2
myoblast cells.
[85]
A detailed review on the application of
Figure 6. Schematic illustration of the controlled release of dexamethasone: A) dexamethasone-loaded electrospun PLGA, B) hydrolytic
degradation of PLGA bers leading to release of the drug, and C) electrochemical deposition of PEDOT around the dexamethasone-loaded
electrospun PLGA ber slows down the release of dexamethasone (D). E) PEDOT nanotubes in a neutral electrical condition. F) External
electrical stimulation controls the release of dexamethasone from the PEDOT nanotubes due to contraction or expansion of the PEDOT. By
applying a positive voltage, electrons are injected into the chains and positive charges in the polymer chains are compensated. To maintain
overall charge neutrality, counterions are expelled towards the solution and the nanotubes contract. This shrinkage causes the drugs to
come out of the ends of tubes. G) Cumulative mass release of dexamethasone from: PLGA nanoscale bers (a), PEDOT-coated PLGA
nanoscale bers (b) without electrical stimulation, and PEDOT-coated PLGA nanoscale bers with electrical stimulation of 1 V applied at the
ve specic times indicated by the circled data points (c). H) UV absorption of dexamethasone-loaded PEDOT nanotubes after 16h (a), 87 h
(b), 160h (c), and 730h (d). The UV spectra of dexamethasone have peaks at a wavelength of 237 nm. Data are shown with a standard
deviation (n15 for each case).
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
297
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
conducting polymer for biomedical applications has already
been discussed elsewhere.
[86]
CPs thus represents a class of
smart polymeric materials that provides an excellent
opportunity for fabricating highly selective, biocompatible,
specic, economic and handy biomedical devices. However,
challenges facing CPs include poor stability and mechanical
properties as well as poor control of the mobility,
concentration and presentation of bioactive molecules.
4.5. Glucose-responsive Polymers
While a variety of polymer systems have been reported for
diagnostic applications, polymers that respond to glucose
have received considerable recognition because of their
potential application in both glucose sensing and insulin
delivery applications. Diabetes mellitus, commonly
referred to as diabetes, is a chronic disease characterized
by decient production of insulin. Treatment for diabetic
patients generally involves regular monitoring of blood
sugar concentrations and subcutaneously administering
insulin every day. This need for continuous patient
vigilance often leads to poor compliance with the
prescribed treatment. One potential route proposed taking
advantage of the advancements made in polymer systems,
is the development of smart delivery of polymer systems in
whichinsulindeliveryis automaticallytriggeredbyarisein
blood glucose levels in vivo. While a variety of approaches
can be visualized to achieve this objective, considerable
research by material scientists has been dedicated to
developing self-regulated insulin delivery systems based
on glucose-responsive polymers.
[87]
The majority of reports
detailing glucose-responsive polymers are based on the
GOx-catalyzed reaction of glucose with oxygen. Typically,
glucose-sensitivityis not duetodirect interactionof glucose
with the glucose responsive polymer, but rather by the
response of polymer to the byproducts that result from
the enzymatic oxidationof glucose. The enzymatic reaction
of GOx on glucose is highly specic and leads to byproducts
suchas gluconic acidandH
2
O
2
. Therefore, theincorporation
of a polymer that responds to either of these byproducts
can indirectly trigger a glucose-responsive system. Another
type of glucose-responsive system as reported by Brownlee
and Cerami utilizes competitive binding of glucose with
glycopolymerlectin complexes.
[88]
Such glucose responsive
polymers are of potential importance for diabetic patients
owing to their specic insulin delivery applications.
4.6. pH responsive Polymers
pH-sensitive polymers are polyelectrolytes that bear intheir
structure weak acidic or basic groups that either accept or
release protons inresponse tochanges inenvironmental pH.
Different organs, tissues and cellular compartments may
have large differences in pH, which makes the pHa suitable
stimulus for biological applications. When weak acids
(carboxylic acids, phosphoric acid) and bases (amines) are
linked to the polymer structure, they exhibit a change inthe
ionisation state upon variation of the pH. This leads to a
conformational changeinthe case of soluble polymers anda
change in the swelling behavior is observed in the case of
hydrogels. The pH range that a reversible phase transition
occurs can be generally modulated by two strategies:
1. Selecting the ionizable moiety with a pKa matching the
desired pH range. Therefore, the proper selection
between polyacid or polybase should be considered
for the desired application.
2. Incorporating hydrophobic moieties into the polymer
backbone and controlling their nature, amount and
distribution. When ionizable groups become neutral
non-ionized- and electrostatic repulsion forces disappear
within the polymer network, hydrophobic interactions
dominate. The introduction of a more hydrophobic
moiety can offer a more compact conformation in the
uncharged state and a more accused phase transition.
Ionisable polymers with a pKa value between 3 and 10
are ideal candidates for pH-responsive systems. Some of
the most widely studied pH responsive polymers are
poly(acrylamide), polyacrylic acid, poly(methacrylic acid),
poly(dimethylaminoethyl methacrylate) (PDEAEMA),
poly(dimethylaminoethyl methacrylate) (PDMAEMA). For
example, the change in pH along the gastro-intestinal
tract
[89]
from acidic in the stomach (pH2) to basic in the
intestine (pH58) has to be considered for oral delivery
of any kind of biomolocule, but there are also other, more
subtle changes within different tissues in the body.
Polymers are usually taken up into cells by uid-phase
pinocytosis or receptor-mediated endocytosis. Within the
early endosome towards the lysosomes the pH drops from
6.2 to 5.0 giving a large change in proton concentration
inside these compartments. This drop in pH has been
utilizedinorder toreleasebiomolecules fromthelysosomes
tothecytosol.
[90]
Intracellular deliveryof oligo/poly(nucleic
acids) usually uses cationic polymers, which complex the
negatively charged nucleic acids. These cationic polymers
are then deprotonated within the endosomes, which
triggers endosome membrane disruption and release into
the cytosol before reaching lysosome with its hydrolytic
enzymes as shown in Figure 7.
[91]
Thus, tailoring the
protonation/deprotonation by altering the polymer struc-
ture can largely allow ne-tuning of the response in a
specic compartment with respect to the change in pH.
Similar to glucose responsive polymers, a pH responsive
polymer can also be applied for insulin delivery applica-
tions. For example a pH responsive polymer loaded or
conjugated with GOx, triggers the GOx-catalyzed reaction.
The gluconic acid byproduct that results from the reaction
298
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
with glucose induces a response in the pH-responsive
macromolecule, thereby triggering the release of insulin
biomolecule. For applications specically intended for
diabetes therapy, the pH-response generally causes swel-
ling or collapse of the polymer matrix, releasing insulin.
Imanishi and co-workers have reported the covalent
modication of a cellulose lm with GOx-conjugated
poly(acrylic acid) (PAA) for insulin delivery applications.
[92]
Duncan et al. designed poly(amidoamine)s by combining
both positive and negative charges within the polymer
backbone.
[93]
On one side of which a very unique prole in
size changes upon protonation/deprotonation was found
with neutron scattering and NMR experiments.
[94]
The
amphoteric backbone yields an expanded shape at lowpH,
which slowly collapsed when neutral pH is approached.
This seems to be the reason that these polymers exhibit
endosomolytic properties, which makes them very inter-
esting candidates in cancer therapy, e.g. delivery of non-
permeant toxins like gelonin. The pH-responsive swelling
and collapsing behavior has been used to induce controlled
release of model compounds like caffeine,
[95]
drugs like
indomethacin,
[96]
or cationic proteins like lysozome.
[95,97]
For instance, Poly(L-histidine)-b-PEG in combination with
PLLA-b-PEG and adriamycin as drug was studied for an
extracellular tumor targeting. The system shows a very
sharp transition from non-protonated and hydrophobic
at pH 7.4, where the mixed micelles are stable, to ionized
and micelle-destabilizing at pH 6.6.
Adriamycin is rapidly released from the
micelles at this pHvalue.
[98]
Several other
groups have developed polymeric pro-
drugs (polymers in which the drug is
covalently attached to the macromole-
cular chain) susceptible to hydrolytic
cleavage dependent upon the pH and
hence suitable for colon drug delivery.
This is the case of poly(N-metha-
cryloylaminoethyl 5-aminosalycilamide)
or poly(methacryloylethoxyethyl 5-amino-
salycilic acid)
[99]
or the copolymeric
system developed based on 2-acryl-
amido-2-methylpropane sulfonic acid
(AMPS) and a methacrylic derivative of
an antiaggregant drug called Triu-
sal.
[100]
The system depends on colonic
microora for liberation of entrapped
drug, which seems most suitable, i.e.,
glycosidase activity of the colonic micro-
ora is responsible for liberation of drugs
from glycosidic prodrugs and the pre-
sence of azoreductase fromthe anaerobic
bacteria of the colon plays a main role
in the release of drug from azo bound
prodrugs.
[101,102]
Researchers have
designed more sophisticated pH-sensitive polymers in
order to take advantage of the pH changes that occur in
nature. These materials are inspired by living organisms
trying to mimic their response mechanisms. For instance,
Sauer et al.
[103]
reported the synthesis of pH-sensitive
hollow nanocontainers inspired in virion particles. The
poly(acrylic acid) vehicles were synthesized by vesicular
polymerization and emulsion polymerization. These nano-
capsules combined the protective ability of the nanocon-
tainers in combination with controlled permeability and
therefore can be used to trigger the release of encapsulated
materials from the inner core. Kataoka
[104]
recently
communicated the development of polymeric micelles
as nanocarriers for gene and drug delivery based on
doxorubicin-conjugated block copolymer poly(ethylene
glycol)-poly(aspartame hydrazinedoxorubicin) [(PEG-
p(AspHid-dox)]. The polymer retained drugs and genes at
physiological pH and released the drugs as pH decrease
below 6.0.
Another most promising application of pH-sensitive
polymers is as nonviral gene carriers. Naked DNA is very
difcult to incorporate into the cells because it is negatively
charged and it has a very large size at physiological
conditions. Godbey and Mikos reviewed some of the
advances innon-viral gene deliveryresearch
[105]
describing
the use of poly- (ethylenimine) (PEI) and poly(L-lysine)
(PLL) as two of the most successful candidates for this
Figure 7. Schematic of DNA condensation and encapsulation into polymeric depot
systems. A) DNA complexation with cationic polymers leads to the formation of
nanometer sized polyplexes. B) Condensed or uncondensed DNA can be encapsulated
into polymeric scaffolds for sustained delivery.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
299
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
application. PEI is a highly polycationic
synthetic polymer that condense DNA in
solution, forming complexes that are
directly endocytosed by several cell
types. Chitosan, a biocompatible and
resorbable cationic aminopolysacchar-
ide, has also extensively been used as
DNA carrier.
[106,107]
Lim et al.
[108]
pre-
pared a self-destroying, biodegradable,
polycationic polyester, poly(trans-4-
hydroxy-L-proline ester) (PHP ester), with
hydroxyproline, a major constituent of
collagen, gelatin, and other proteins,
as a repeating unit. PHP ester formed
soluble polymer/DNA complexes with
average diameters of less than 200nm.
These complexes could transfect the
mammalian cells, being comparable to
the transfection obtained with PLL, the
most commonpolymer for gene delivery,
at suitable pH conditions.
4.7. Enzyme-responsive Polymers
Arelativelynewareaof researchinpolymeric systems is the
design of materials that undergo macroscopic property
changes when triggered by selective enzymatic reac-
tions.
[109,110]
These polymeric systems consists of the
following characteristics (i) sensitivity of this system is
unique because enzymes are highly target specic (ii) they
can operate even under mild conditions in vivo, and (iii) are
vital components in many biological pathways. Enzyme-
responsivepolymeric materials are composedof anenzyme-
sensitive substrate and another component that directs or
controls interactions that lead to macroscopic property
variations.
[110]
Catalytic reaction of an enzyme on a
substrate can lead to changes in geometry, supramolecular
architectures, swelling/collapse of gels, or variations of
surface characteristics.
[110]
Xu and co-workers reported the
use of enzymatic dephosphorylation to induce a solgel
transition. In the reaction sequence the small molecule
uorenylmethyloxycarbonyl (FMOC)-tyrosine phosphate
was exposed to a phosphatase, and the resulting removal
of phosphate groups led to a reduction in electrostatic
repulsions, supramolecular assembly and eventual gelation
of the polymer system.
[111,112]
Figure 8 illustrates the design
of a visual assay, wherein the precursor, which acts as the
substrate of an enzyme, transforms into a hydrogelator
when the enzyme catalyzes its conversion. Then, the self-
assembly of the hydrogelators in water induces the
formation of hydrogel. When inhibitors competitively bind
with the enzyme active site and block the conversion of the
precursor catalyzed by the enzyme, no hydrogel forms,
asshowninFigure8.
[112]
Inasimilarexample, Kaplanandco-
workers reported the modication of a genetically engi-
neered variant of spider dragline silk via enzymatic
phosphorylation and dephosphorylation.
[113]
Enzyme-
responsive polymers upon exposure to a specic enzyme,
undergoes changes intheir macroscopic properties owing to
the creation of new covalent linkages. For instance, Ulijin
and co-workers used proteases to cause self-assembly of
polymeric hydrogels via reversed hydrolysis of peptides.
[114]
Transglutaminase, abloodclottingenzyme, has theabilityto
cross-link the side chains of lysine (Lys) residues with
glutamine(Gln) residues withinor across peptidechains.
[110]
This behavior was exploited by Grifth and Sperinde for the
synthesis of hydrogels of cross-linked functionalized PEG
and lysine-containing polypeptides.
[115,116]
4.8. Temperature-responsive Polymers
Temperature-responsive polymers and hydrogels exhibit a
volume phase transition at a certain temperature, which
causes a sudden change in the solvation state. When
hydrogels are prepared by cross-linking temperature-
sensitive polymers, the temperature sensitivity in water
results in changes in the polymer hydration degree. Below
the transition temperature the polymer swells up to
equilibrium hydration degree, being in an expanded state.
Byincreasingthetemperatureabovethetransitionhydrogel
deswells to a collapsed state. This transition is usually
reversible and can be applied in a pulsatile manner making
the polymer to behave as an on-off system when the
stimulus is applied or removed. Polymers, which become
insoluble upon heating, have a lower critical solution
temperature (LCST). Systems, which become soluble upon
Figure 8. The illustration of the design for identifying inhibitors of an enzyme by
hydrogelation.
300
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
heating, have an upper critical solution temperature (UCST).
The change in the hydration state, within the aqueous
environment as the human body, causes the volume phase
transition, where intra- and intermolecular hydrogen
bonding of the polymer molecules are favored compared
toasolubilisationbywater. Typical LCSTpolymers arebased
on N-isopropylacrylamide (NIPAM),
[117,118]
N,N-diethylacry-
lamide (DEAM),
[119]
methylvinylether (MVE),
[120,121]
and N-
vinylcaprolactam (NVCl)
[122,123]
as monomers. A typical
UCSTsystemis basedonacombinationof acrylamide(AAm)
and acrylic acid (AAc).
[124]
Most biomedical applications use
the change from room temperature to body temperature in
order to induce a change in the physical properties for e.g.
gelation, especially in applications using injectable biode-
gradable scaffolds. Polymers with LCST have been tested in
controlled drug delivery matrices and in on-off release
prolesinresponsetoastepwisetemperaturechange. Inthis
sense, polyNIPAAm hydrogels form a thick skin on their
surface above the LCST in the collapsed state, which reduces
the transport of bioactive molecules out of the hydrogels.
NIPAAmhas also been copolymerized with alkyl methacry-
lates in order to increase the hydrogels mechanical proper-
ties, without compromising on the temperature sensitivity.
Poly(NIPAAm-co-butyl methacrylate) (poly(NIPAAm-co-
BM)) was studiedfor the deliveryof insulinina temperature
on-off prole, belowand above the LCST that in this system
was loweredtoabout 258C.
[125]
The temperature-responsive
polymers have the transition temperature in the region
20408C, which is interesting for biomedical applications.
However the transition temperature can be strongly
dependent on factors such as solvent quality, salt concen-
tration and molecular weight. PNIPAM is a very interesting
temperature-responsive material, having good biocompat-
ibilityandthe positionof the LCSTat 32338C, whichis ideal
for biological applications. The LCST of PNIPAM is indepen-
dent of the molecular weight and the concentration,
[126]
but
it can be changed upon shifting the hydrophilic/hydro-
phobic balance, by copolymerization with a second mono-
mer. Hydrophobic co-monomers increase the LCST, whereas
hydrophilic co-monomers have the opposite effect.
[127,128]
PNIPAM copolymers have been mainly studied for cardiac
tissue engineering and oral delivery of biomolecules like
calcitonin and insulin. In the latter case, the peptide or
hormoneis immobilizedinPNIPAMbeads, whichstaystable
while passing through the stomach. Then in the alkaline
intestine the beads disintegrate and the drug is released.
Serres et al.
[129]
and Ramkisson-Ganorkar et al.
[130]
synthe-
sized P(NIPAM-co-BMA-co-AAc) for the intestinal delivery of
human calcitonin. Meanwhile, Kim et al. investigated the
delivery of insulin.
[131]
In both cases the combination of
the hydrophobic BMA moiety (butylmethacrylate) and
acrylic acid (AAc), which is non-ionized at low pH, prevents
disintegration of the beads in the acidic environment of the
stomach. At elevated pH the beads disintegrated due to the
solubilisation by the now ionized AAc. Thus in this case
besides temperature, the pHalso plays an important role in
stimulating the drug release. Besides PNIPAM, Poly(methyl
vinyl ether) also has a transition temperature exactly at
378C, which makes it very interesting for biomedical
application. Poly(N-vinyl caprolactam) (PVCa) has not been
studied as intensively as PNIPAM, but it also possesses very
interesting properties for medical and biotechnological
applications, e.g. solubility in water and organic solvents,
biocompatibility, high absorption ability and a transition
temperature within the settings of these applications
(338C).
[123]
Several approaches have been performed in the
tissueengineeringeldas temperaturesensitivescaffolds or
surface modications for the manipulation of cell sheets.
Poly(NIPAAm-co-acrylic acid) (poly(NIPAAm-co-AA)) gels
have been applied as extracellular matrix for pancreatic
islets in biohydrid pancreas.
[132]
4.9. Antigen-responsive Polymers
Similar to enzymatic reactions, antigenantibody interac-
tions are also highly specic and are associated with
complex immune responses that help to recognize any
foreign bodies in the blood stream. Binding reaction
between antigens and antibodies can rely on a variety of
non-covalent interactions, such as hydrogen bonding, van-
der-Waals forces, and electrostatic and hydrophobic
interactions. Antibodies are employed in a number of
immunological assays for the detection and measurement
of biological and non-biological substances,
[133]
and the
highafnityandtarget specicityof their interactions with
antigens have been harnessed to yield a variety of antigen-
responsive synthetic polymeric systems for biological
applications. In most cases, antigenantibody binding
has been used to induce responses in polymeric systems
prepared by either physically entrapping antibodies or
antigens innetworks, chemical conjugationof theantibody
or antigen to the network, or using antigenantibody pairs
as reversible cross-linkers within networks.
[134]
Miyata
et al. prepared antigen-sensitive hydrogels by coupling
rabbit immunoglobulin G (Rabbit IgG) to N-succinimidy-
lacrylate (NSA). The modiedmonomer was polymerizedin
the presence of goat anti-rabbit IgG as an antibody and
acrylamide, which results in the formation of a hydrogel
cross-linked both covalently and by antigenantibody
interactions. Upon the addition of rabbit IgG as a free
antigen, competitive binding of the goat anti-rabbit IgG
antibodies resulted in the breakage of antigenantibody
cross-linkers and a change in the morphology of the
hydrogel characterized by swelling of the hydrogel.
[135]
4.10. Redox/thiol-responsive Polymers
Redox/thiol sensitive polymers are another class of
advanced polymers that are of immense importance in
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
301
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
bioengineering applications especially in the eld of
controlled drug delivery.
[136,137]
The interconversion of
thiols and disuldes is a key step in many biological
processes as it plays a signicant role in the stability and
rigidity of native proteins in living cells.
[138]
Since disulde
bonds canbereversiblyconvertedtothiolswhenexposedto
various reducingagents and/or undergodisulde exchange
in the presence of other thiols, polymers containing
disulde linkages can be considered for both redox- and
thiol-responsive applications.
[139,140]
For instance, Glu-
tathione (GSH), is the most abundant reducing agent in
living cells. It has an intracellular concentration of about
10 mM, whereas an extracellular concentration of about
0.002 mM in the cell exterior.
[141]
This signicant variation
in concentration has been utilized to design thiol/redox-
responsive polymer drug delivery systems that specically
release therapeutics into cells. For example, Lee and co-
workers synthesized polymer micelles with shells cross-
linked via thiol-reducible disulde bonds as shown in
Figure 9. These served as carriers that preferentially release
anticancer drugs under reducing conditions typical of
cancer tissues.
[142]
4.11. Shape Memory Polymers
Shape memory polymers (SMPs) can rapidly change their
shapes from a temporary shape to their permanent shapes
under appropriate stimulus such as temperature, light,
electric eld, magnetic eld, pH, specic ions or enzyme.
Schematic representation of thermally induced shape
memory effect is given in Figure 10, where heating a
sample above the switching transition temperature
(T Trans) induces the recovery of the permanent shape of
polymer. SMPs have the advantages of light weight, low
cost, good processability, high shape deformability, high
shape recoverability and tailorable switching tempera-
ture.
[143145]
For example, poly(D,L-lactide) is a good shape
memory biomaterial having good biodegradability and
biocompatibility; and have been utilized for several tissue
engineering applications. Zheng et al.
[146]
reinforced
poly(D,L-lactide) (PDLLA) with hydroxylapatite for hard
tissue engineering applications. The composite showed
good biodegradation, biocompatibility and shape memory
properties. The study showed that PDLLA/hydroxylapatite
composites at a suitable fraction ratio of between 2.0:1 and
2.5:1 had much higher shape recovery ratios and recover
speed than pure PDLLA. By using a crystalline polymer as
the xing phase and a second crystalline polymer as the
reversible phase, SMP blends were created for bioengineer-
ing eld. In that context, Behl et al.
[147]
reported binary
biodegradable polymer blends from two crystalline poly-
mers poly(p-dioxanone) (PPDO) and poly(caprolactone)
with poly(alkylene adipate) mediator as a compatibilizer.
The crystalline PPDO provides the hard segment to
determinethepermanent shapeandthepoly(caprolactone)
provides the soft segment to determine the switching
temperature. Besides, Zhang et al.
[148]
introduced poly-
(ethyleneglycol) dimethacrylate (PEGDMA) to PLGA/iso-
phorone diisocyanate (IPDI). This system showed good
shape memory and hydrophilic properties useful for
biomedical applications. Further, Zhu et al.
[149]
improved
the radiation efciency of polycaprolactone by blending it
with polymethylvinylsiloxane (PMVS)
before radiation cross-linking, for biome-
dical applications.
4.12. Electro-responsive Polymers
Electro-responsive polymers (ERPs) are
very benecial for biological applications
because of their potential to being direc-
tional, which can give rise to anisotropic
deformation. ERPs can be used to prepare
materials that swell, shrink, or bend in
response to an electric eld.
[150,151]
Since
ERP can transform electrical energy into
mechanical energy, they have promising
applications in biomechanics, articial
muscle actuation, sensing, energy trans-
duction, sound dampening, chemical
Figure 9. Illustration of shell cross-linking in drug -loaded polymer micelles and facili-
tated drug release in response to cellular GSH.
Figure 10. Schematic representation of the thermally induced
one-way shape-memory effect.
302
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
separations, andcontrolleddrugdeliveryapplications.
[150153]
Gel deformation, whichinvolves bendinginanelectric eld
is inuenced by a number of factors like variable osmotic
pressure based on the voltage-induced motions of ions in
the solution, pH or salt concentration of the surrounding
medium, position of the gel relative to the electrodes,
thickness or shapeof thegel, andtheappliedvoltage.
[153,154]
Transforming the application of an electric eld into a
physical response by a polymer generally results in certain
changes in the properties of the polymer matrix like,
collapse of a gel in an electric eld, electrochemical
reactions, electrically activated complex formation, ionic
polymermetal interactions, electrorheological effects, or
changes in electrophoretic mobility.
[150]
Typically, ERPs
have been investigated in the form of polyelectrolyte
hydrogels
[150,151]
which are capable of deformation under
anelectric eldduetoanisotropic swellingor deswelling, as
charged ions are directed toward the anode or cathode
sideof thegel.
[150]
For example, under anelectric eld, inthe
case of hydrolyzed polyacrylamide gels, mobile H

ions
migrate toward the cathode while the negatively charged
immobile acrylate groups in the polymer networks are
attracted toward the anode, creating a uniaxial stress
withinthegel. Theregionsurroundingtheanodeundergoes
the greatest stress while the area in the vicinity of the
cathode exhibits the smallest stress. This stress difference
contributes to the anisotropic gel deformation under an
electric eld.
[155]
The natural polymers used to prepare
electro-responsive materials include chitosan,
[156]
chon-
droitin sulfate,
[157]
hyaluronic acid,
[158]
and alginate.
[159]
The monomers of synthetic polymers used include
allylamine,
[160]
2-acrylamido-2-methylpropane sulfonic
acid,
[161]
aniline,
[162]
methacrylic
acid,
[159]
acrylic acid
[162]
and vinyl sulfo-
nic acids.
[162]
These polymers are of
increasing importance as a class of smart
materials for biological applications.
4.13. Magneto-responsive Polymers
Polymers that respond to either the
presence or absence of magnetic elds
are called magneto-responsive polymers.
These can exist as free chains in solution,
be immobilized to surfaces, or be cross-
linked within networks.
[163]
Generally,
inorganic magnetic particles are physi-
cally entrapped within or covalently
immobilized to a three-dimensional
cross-linked network,
[164,165]
leading to
materials with shape and size distortion
that occurs reversibly and instanta-
neously in the presence of a non-uniform
magnetic eld.
[166,167]
In this case, the
magnetophoretic force conferred to the polymeric material
as a result of the magnetic susceptibilityof the particles has
led to such smart polymers receiving signicant attention
for useas soft biomimetic actuators, sensors, cancer therapy
agents, articial muscles, switches, separation media,
membranes, and bioengineering systems.
[168,169]
Super-
paramagnetic iron oxide nanoparticles (SPION, g-Fe
2
O
3
or
Fe
3
O
4
) are widely used as core, coated with organic or
polymeric materials as shell were used for various drug
targeting applications. Figure 11 demonstrates a procedure
for preparing iron oxide (g-Fe
2
O
3
) nanoparticles encapsu-
lated with Arg peptide (RRRRR CK-FITC) conjugated with
PLGA. Such encapsulation of magnetic particles with
preformed natural or synthetic polymers is the simple
and classical method to prepare magnetic polymeric
materials which can be used in biomedical applications
like cell separation and gene and protein delivery.
[155]
4.14. Ultrasound-responsive Polymers
The concept of using ultrasound-responsive polymers for
biological applications is attractive because the method is
essentially noninvasive and has been successfully used in
other areas of medical treatment and diagnostics.
[170]
The
success of ultrasonic mediationof drugdeliveryis generally
ascribed to cavitation, which is the alternating growth
and shrinkage of gas-lled microbubbles that results from
high and low pressure waves generated by ultrasound
energy.
[171]
Subsequently, these cavitating microbubbles
implode, generating local shock waves that can disrupt
polymer assemblies in their vicinity as shown in
Figure 12.
[172]
The ultrasound-induced release rate of
Figure 11. A) The schematic diagram for preparing the g-Fe
2
O
3
-PLGA-Arg-FITC nanopar-
ticles. B) The sequence of arginine-peptide.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
303
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
immobilized biomolecules from biodegradable polymers,
polyglycolides, polylactides, and poly[bis(p-carboxy-
phenoxy)alkane] anhydrides with sebacic acid and non-
biodegradable ethylene-vinyl acetate copolymers have
been extensively studied by Langer and co-workers for
biological applications.
[173,174]
4.15. Photo-responsive Polymers
Photo-responsive polymers are macromolecules that
change their properties when irradiated with light of
appropriate wavelength.
[175,176]
Typically these variations
are the result of light-inducedstructural transformations of
specic functional groups along the polymer backbone or
side chains.
[177]
These photoresponsive polymers can be
employed in photomechanical transduction and actuation,
bioactivity switching of proteins, tissue engineering, and
drug delivery applications.
[178,179]
Figure 13 shows a
schematic representation of the photo-responsive enzyme
switch.
[179]
An important aspect of photo-sensitive poly-
mer systemis usingirradiationas astimulus, inwhichnon-
invasive mechanism is involved to induce responsive
behavior. The well-studied examples of photo-responsive
polymers are those that contain azobenzene groups.
[175]
Azobenzene is a chromophore with an irradiation-induced
cis-to-trans isomerizationthat is accompaniedbyafast and
complete change in electronic structure, geometric shape,
and polarity of the polymer.
[178]
For example, Wang and co-
workers described photo-induced deformation of epoxy-
based azobenzene-containing polymer colloids.
[180]
This
concept has been employed to prepare a variety of
spiropyran-containing photo-responsive polymers, for
example, PAA,
[181]
PHPMA,
[182]
and PNIPAM
[183]
which
have been widely used for tissue engineering applications.
4.16. Photoluminescent Polymers
Polymers such as polyesters, polyamides, polyimides, and
polyurethanes canhaveemissiveproperties whenaphoton
absorbing chromophore is attached with these polymers.
Multiblockcopolymers inwhichdistyryl blocks werelinked
bydifferent functional groups withnon-conjugatedspacers
can be synthesized. By altering these spacers, different
classes of photoluminescent polycondensates with
chromophoric groups can be obtained. The reaction of
dihydroxy functionalized, substituted phenylene vinylene
chromophores with a variety of dicarboxylic acid chlorides
can yield high molecular weight electroluminescent
polyesters emitting light in the blue-green region.
[184]
Similarly, copolyesters with varying concentrations of
isolated 1,2-naphthylene chromophore units can emit in
the blue to blue-green region.
[185]
Distyryl naphthalene,
distyryl anthracene, and terthiophene units in the chain
of copolyesters also emit blue and green light,
[186]
and
copolyesters with isolated dimethoxy quarterthiophene
emitted in the orange region.
[187]
Additionally, polyur-
ethanes with a diphenylamino-substituted-1,4-bistyryl
benzene acted as an emissive and charge transport
chromophore and showed luminance of 35 cd/m
2
at
26 V.
[188]
Such photoluminescent polymers can be applied
for label-free cellular imaging and bioimaging applica-
tions.
[189]
Table 2 illustrates the various stimuli-responsive
polymers that have been dealt with in the present review
along with their properties and applications.
4.17. Dual responsive Polymers
It is possible to obtain polymeric structures sensitive to
both temperature and pH, by the simple combination of
ionisable and hydrophobic (inverse thermosensitive) func-
tional groups. It has mainly been achieved by the
copolymerization of monomers bearing these functional
groups,
[190,191]
combining thermosensitive polymers with
polyelectrolytes (SIPN, IPN)
[192]
or by the development of
new monomers that respond simultaneously to both
stimuli.
[193]
Leung et al.
[194]
prepared smart core/shell
microgels based on PNIPAAm, MBAAm and chitosan or
poly(ethyleneimine) in the absence of surfactants. The
materials were obtained by graft copolymerization and
presented a well dened core/shell structure consisting of
Figure 12. A) Layout of the experiment. The piezoelectric trans-
ducer generates a standing ultrasound eld. B) High-speed time
series (bottom view through inverted microscope) of vesicle
motion (interframe time 10ms). The bright object is a bubble
of radius a 15 mm at the cuvette wall, whose oscillation ampli-
tude is too small to be seen here. The dark object on the right is a
lipid vesicle, whose shape far fromthe bubble would be spherical.
Here it is severely deformed as it approaches the bubble, collides
with it, and is then expelled away from the observer (upwards in
the cuvette), blurring as it leaves the focal plane. C) Experimen-
tally observed trajectories of vesicles in a side view (z is the axis
perpendicular to the cuvette wall to which the bubble is
attached).
304
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
temperature-sensitive cores (based on PNIPAAm) with pH-
sensitive shells (based oncationic water-soluble polymers).
Kurata and Dobashi
[195]
published the preparation of
potential intelligent drug carriers based on N-acryloyl-N

-
alkylamide derivatives of both L-glutamic acid and L-
aspartic acid. New copolymeric systems derived from
N,N-dimethylaminoethylmethacrylate (DMAEM) and
acrylic acid (AAc) or itaconic acid (IAc) were obtained by
UVirradiation. TheyrespondedtobothpHandtemperature
as a polyampholyte according to the monomeric composi-
tions and combination of temperature and pH condi-
tions.
[195]
Alginate has been modied using PNIPAAm
forming dual stimuli responsive SIPNs that could be useful
in biomedical elds for stimuli-responsive drug delivery
systems.
[196]
Ning et al. demonstrated
[197]
the synthesis by
g-irradiation and characterization of poly(N,N-dimethyla-
minoethyl methacrylate) (PDMAEMA), a temperature-
sensitive material in a temperature range of 3840 8C
and pH-sensitive at pH2.5. Electricity-responsive beha-
vior at a eld voltage of approximately 3.0V was shown to
take place. As the synthesized materials
were transparent, elastic, and had a good
swellingcapacitytheir utilizationas drug
delivery systems was proposed.
5. Future Perspectives
Surface modication with stimuli-
responsive polymers leads to the pre-
paration of responsive interfaces which
may show a very different behavior in
response to small changes in the envir-
onmental parameters. Surface may
change from hydrophobic to hydrophi-
lic
[198,199]
or if a membrane is chemically
modied, it may vary the pore size
itself.
[200,201]
These surface functionaliza-
tion processes can produce smart sur-
faces or smart conjugates, useful for
enhanced cell-biomaterial interactions.
Rodr guez-Cabellos group
[202]
demon-
strated the potential in developing well
dened smart polymers with tailored
surfaces covering a wide range of proper-
ties. They developed elastin-like poly-
mers (ELPs) by genetic engineering,
which showed clear environmental
advantages. The ELPs presented a modu-
lated pH- and temperature-sensitivity
covering the most interesting range of
biomedical applications. ELPs have also
been modied with photoresponsive
molecules as azobenzenes
[202]
and spir-
opyranes
[203]
getting photosensitive macromolecules. Sur-
face modications incorporating temperature responsive
polymers have been carried out in order to immobilize
specic molecules or to manipulate cell sheets in tissue
engineering applications. In this sense, the application of
temperature responsive polymers to modied surfaces
exploits the fact that most proteins show signicantly
greater adsorption on hydrophobic surfaces than in
hydrophilic ones. AbovetheLCSTthetemperature-sensitive
polymer will adsorb peptides and proteins from a solution
and these biomolecules can be desorbed by decreasing the
temperature, which has been employed in chromato-
graphic supports incorporating PNIPAAm and using water
as eluent.
[204]
Another approachhas shownthat the surface
of tissue culture polystyrene grafted with PNIPAAmallows
cells toadhereandproliferateabovetheLCSTof thepolymer
whereas a cell detachment was detected at temperatures
below LCST.
[199]
This type of surface modication has also
been performed on reversible 3D-matrix for the culture of
articular chondrocytes, cells that proliferate vastly on the
Figure 13. Schematic model for the photoresponsive enzyme switch. The photo-
responsive copolymer compositions are shown with the end-modied vinyl sulfone
terminus for thiol-specic conjugations. The 3D model of EG 12A displays the relative
locations of position 55 (with schematic polymer coil attached) and the catalytic
active site residues D99, E116, and E200. The ONPC substrate is shown schematically
to show the orientation of the active site groove. The polymer coil is shown as a
10-kDa chain with a distribution of nine DMA monomers to one azobenzene
monomer.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
305
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
Table 2. Various stimuli responsive polymers and their properties and applications.
Functional polymer Examples Properties Applications
Conducting polymer PEDOT, PPy, PANI Possess high electronic
and ionic conductivities
Nerve and cardiac
tissue engineering
Glucose responsive
polymer
Phenylboronic acid (PBA) groups Responses to the
byproducts that result
from the enzymatic
oxidation of glucose
Glucose sensing
and insulin
delivery
pH responsive
polymer
Poly(acrylamide), polyacrylic acid,
poly(methacrylic acid),
poly(dimethylaminoethyl
methacrylate) (PDEAEMA),
poly(dimethylaminoethyl
methacrylate) (PDMAEMA).
Exhibits a change in
the ionisation state
upon variation of
the pH leading to
conformational
change
Insulin delivery,
drug delivery
Enzyme responsive
polymer
Genetically engineered variant
of spider dragline silk
Undergoes macroscopic
property changes when
triggered by selective
enzymatic reactions
Drug delivery
Temperature-responsive
polymer
PNIPAM Undergoes a
conformational
change upon the
change in
temperature
Cardiac tissue
engineering,
drug release
Antigen responsive
polymer
N-succinimidylacrylate
(NSA, Poly(acrylamide)
Antigenantibody
interactions
Biosensors and
drug release
Redox/thiol responsive
polymer
Polymers containing
disulde-functional
dimethacrylate
Responds to the
interconversion of thiols
and disuldes
Bioengineering
and drug delivery
Shape memory
polymer
Poly(D,L-lactide) Can rapidly change their
shapes from a temporary
shape to their permanent
shapes under appropriate
stimulus
Biomedical
applications
Electro responsive
polymer
Polyacrylamide gels,
chondroitin sulfate,
hyaluronic acid
They can swell, shrink,
or bend in response to
an electric eld
Biomechanics,
articial muscle
actuation, sensing,
controlled drug
delivery
Magneto responsive
polymer
Superparamagnetic iron
oxide nanoparticles
(SPION, g-Fe
2
O
3
or Fe
3
O
4
)
are widely used as core,
coated with organic or
polymeric materials as shell.
They respond to either
the presence or absence
of magnetic elds
Soft biomimetic
actuators, sensors,
cancer therapy
agents, articial
muscles
306
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
matrixthat arethenremovedbytemperaturelowering.
[205]
Modications can also be induced to the polymer by
modifyingthepolymer composition. For instance, Pluronics
and Tetronics are FDA approved thermo-reversible gels
used for tissue engineering processes.
[206]
Their gelation
temperaturedepends onpolymer compositionandsolution
concentration. For example Pluronic F127 gels at 37 8C in
solutions that contain about 20 wt% of polymer. These
systems have been used in treatment of burns and other
wound healing applications.
[207]
More recent PEO-PPO-PEO
triblock copolymers and Poloxamer 407 (Poloxamers are
also known by the trade name Pluronics), were mixed with
isolated chondrocytes when applied with a brush on an
osseous surface resulted inthe formationof a sticky gel ina
short period of time. A new cartilage was formed on the
osseous substrate in the bone cartilage interface.
[208]
Poloxamer 407 in solution with isolated chondrocytes
was also applied as an injectable cartilage formulation,
testing the formation of tissue after subcutaneous injec-
tions in mice. Histological examination of all samples
demonstrated the presence of new cartilage formation
indicating that the polymer/cell suspension is very
promising for orthopaedic and reconstructive surgery.
[208]
Poloxamer is again the most commonly used in this kind of
application and it has been tested in the delivery of protein
and peptides (insulin, urease, bone morphogenic protein
and growth factors) showing inmost of the cases sustained
release proles over several hours.
[209]
Such smart poly-
mers with smart surfaces or smart conjugates, fabricated
depending on the desired application, are emerging as
promising next generation of engineered tissues and is
relying on producing scaffolds with an informational
function, e.g., material fabricated to produce the exact
physical parameters similar to native tissue and material
containing peptide molecules and growth factors sequence
which facilitates cell attachment, proliferation and differ-
entiation that is far better than non-informational smart
polymeric systems.
6. Conclusion
Advances in synthetic organic chemistry and novel
bioprocesses enable the development of a wide range of
smart polymeric materials as candidates for developing
next generationbiomedical devices, transient implants and
drug delivery vehicles. Ultimately, engineers must match
applications, materials, and fabrication processes to best
meet the needs of the tissue they wish to engineer. Stimuli-
responsive polymers offer great advantages for biomedical
applications and drug delivery. Instead of acting passively
as pure drug carriers, they will interact and respond to the
environmental setting. This allows us to aim further for
tailor-made drug delivery devices with superior pharma-
cokinetics while having all safety questions addressed.
Stimuli-responsive polymers that respond to two or
more stimulus can be synthesized and studied for tissue
engineering and biomedical applications, which will open
new frontiers not only in basic research for understanding
the concept but also exploitation of their potential for real
clinical applications. This review shows how polymers can
be used in a smart fashion potentially leading to multiple
responses and functionalities at the desired point of action.
Thus the ultimate goal is to produce advanced polymeric
systems in a controlled, reliable and reproducible approach
inthe eld of biomedical applications. It is also necessary to
Table 2. (Continued)
Functional polymer Examples Properties Applications
Ultrasound responsive
polymer
Polyglycolides, polylactides, and
poly[bis(p-carboxyphenoxy)alkane
anhydrides with sebacic acid
and ethylene-vinyl acetate
copolymers
Responds to cavitation
that results from high
and low pressure waves
generated by ultrasound
energy
Medical treatment
and diagnostics
Photo responsive
polymer
Azobenzene and spiropyran
containing polymer
Macromolecules that change
their properties when
irradiated with light of
appropriate wavelength
Biological
applications
Photoluminescent
polymer
Chromophores attached to
polyesters, polyamides,
polyimides, polyurethanes
Polymers with emissive
properties
Biosensors and
medical diagnostics
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
307
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
emphasize these underutilized adaptive behaviors of these
polymers, so that novel applications and new generations
of smart materials can be realized for stemcell therapy and
tissue engineering applications.
Acknowledgements: This study was supported by the NRF-
Technion (R-398-001-065-592), Ministry of Education (R-265-000-
318-112), and NUSNNI, National University of Singapore, Singapore.
Received: August 17, 2011; Revised: November 16, 2011;
Published online: January 25, 2012; DOI: 10.1002/mabi.201100325
Keywords: pH and temperature responsive polymers; smart
polymers; stimuli-responsive polymers; surface modication;
tissue engineering
[1] D. F. Williams, The Williams dictionary of biomaterials,
Liverpool University Press, Liverpool 1999.
[2] R. A. Quirk, R. M. France, K. M. Shakesheff, S. M. Howdle, Curr.
Opin. Solid St. M. 2004, 8, 313.
[3] R. Boccaccini, V. Maquet, Compos. Sci. Technol. 2003, 63,
2417.
[4] P. X. Ma, R. Zhang, J. Biomed. Mater. Res. 2001, 56, 469.
[5] V. Maquet, D. Martin, F. Scholtes, R. Franzen, J. Schoenen,
G. Moonen, Biomaterials 2001, 22, 1137.
[6] J. J. Blaker, J. C. Knowles, R. M. Day, Acta Biomaterialia. 2008,
4, 264.
[7] J. Roh, I. C. Kwon, J. Biomater. Sci. Polym. Ed. 2002, 13, 769.
[8] D. C. Sin, X. Miao, G. Liu, F. Wei, G. Chadwick, C. Yan, T. Friis,
Mater. Sci. Eng. C 2010, 30, 78.
[9] M. Taboas, R. D. Maddox, P. H. Krebsbach, S. J. Hollister,
Biomaterials 2003, 24, 181.
[10] D. W. Hutmacher, Biomaterials 2001, 21, 2529.
[11] P. Kruth, Ann. CIRP 1991, 40, 603.
[12] P. S. DUrso, W. J. Earwaker, T. M. Barker, M. J. Redmond, R. G.
Thompson, D. J. Effeney, F. H. Tomlinson, Br. J. Plast. Surg.
2000, 53, 200.
[13] B. Sanghera, S. Naique, Y. Papaharilaou, A. Amis, Rapid
Prototyping J. 2001, 7, 275.
[14] F. Leong, K. K. Phua, C. K. Chua, Z. H. Du, K. O. Teo, Proc. Inst.
Mech. Eng. H 2001, 215, 191.
[15] P. Vanezi, M. Vanezis, G. McCombe, T. Niblett, Forensic Sci.
Int. 2000, 108, 81.
[16] S. S. Kim, H. Utsunomiya, J. A. Koski, B. M. Wu, M. J. Cima,
J. Sohn, K. Mukai, L. G. Grifth, J. P. Vacanti, Ann. Surg. 1998,
228, 8.
[17] J. Zeltinger, J. K. Sheerwood, D. M. Graham, R. Mueller, L. G.
Grifth, Tissue Eng. 2001, 7, 557.
[18] C. X. F. Lam, X. M. Mo, S. H. Teoh, D. W. Hutmacher, Mater. Sci.
Eng. C 2002, 20, 49.
[19] J. Mondrinos, R. Dembzynski, L. Lu, V. K. C. Byrapogu, D. M.
Wootton, P. I. Lelkes, J. Zhou, Biomaterials 2006, 27, 4399.
[20] P. X. Ma, R. Zhang, J. Biomed. Mater. Res. 1999, 46, 60.
[21] Z. Xiong, Y. N. Yan, S. G. Wang, R. J. Zhang, C. Zhang, Scr.
Mater. 2002, 46, 771.
[22] K. Jayaraman, M. Kotaki, Y. Zhang, X. Mo, S. Ramakrishna,
J. Nanosci. Nanotech. 2004, 4, 52.
[23] L. A. Smith, P. X. Ma, Colloids Surf. B Biointerfaces 2004,
39, 125.
[24] Z. Ma, M. Kotaki, R. Inai, S. Ramakrishna, Tissue Eng. 2005,
1, 101.
[25] V. J. Chen, P. X. Ma, Biomaterials 2004, 25, 2065.
[26] C. P. Barnes, S. A. Sell, E. D. Boland, D. G. Simpson, G. L.
Bowlin, Adv. Drug Deliv. Rev. 2007, 59, 1413.
[27] R. Zhang, P. X. Ma, J. Biomed. Mater. Res. B 2000, 52, 430.
[28] J. A. Matthews, G. E. Wnek, D. G. Simpson, G. L. Bowlin,
Biomacromolecules 2002, 3, 232.
[29] A. Matsuda, G. Kagata, R. Kino, J. Tanaka, J. Nanosci. Nano-
technol. 2007, 7, 852.
[30] J. H. Song, H. E. Kim, H. W. Kim, J. Mater. Sci. Mater. Med.
2007, 19, 95.
[31] J. Stitzel, J. Liu, S. J. Lee, M. Komura, J. Berry, S. Soker, G. Lim,
M. VanDyke, R. Xzerw, J. Yoo, A. Atala, Biomaterials 2006, 27,
1088.
[32] C. Y. Xu, R. Inai, M. Kotaki, S. Ramakrishna, Biomaterials
2004, 25, 877.
[33] R. Ravichandran, S. Liao, C. C. H. Ng, C. K. Chan, M. Raghunath,
S. Ramakrishna, World J. Stem Cells 2009, 1, 55.
[34] H. W. Ouyang, J. C. Goh, A. Thambyah, S. H. Teoh, E. H. Lee,
Tissue Eng. 2003, 9, 431.
[35] R. Ravichandran, J. R. Venugopal, S. Sundarrajan, S. Mukherjee,
S. Ramakrishna, Tissue Eng. Part A 2011, 17, 1363.
[36] K. Anselme, Biomaterials 2000, 21, 667.
[37] M. Mrksich, Curr. Opin. Chem. Biol. 2002, 6, 794.
[38] U. Hersel, C. Dahmen, H. Kessler, Biomaterials 2003, 24, 4385.
[39] H. Jiang, Y. Hu, P. Zhao, Y. Li, K. Zhu, J. Biomed. Mater. Res. B
Appl. Biomater. 2006, 79, 50.
[40] J. Lou, Y. Tu, M. Burns, M. J. Silva, P. Manske, J. Orthop. Res.
2001, 19, 1199.
[41] S. O. Abrahamsson, G. Lundborg, L. S. Lohmander, J. Orthop.
Res. 1991, 9, 495.
[42] M. B. Klein, N. Yalamanchi, H. Pham, M. T. Longaker, J. Chang,
J. Hand Surg. 2002, 27, 615.
[43] M. Rickert, M. Jung, M. Adiyaman, W. Richter, H. G. Simank,
Growth Factors 2001, 19, 115.
[44] C. Forslund, Acta Orthop. Scand. Suppl. 2003, 74, 1.
[45] X. T. Wang, P. Y. Liu, J. B. Tang, J. Hand Surg. 2004, 29, 884.
[46] S. Y. Chew, J. Wen, E. K. Yim, K. W. Leong, Biomacromolecules
2005, 6, 2017.
[47] J. A. Jansen, J. W. M. Vehof, P. Q. Ruhe, H. Kroeze-Deutman,
Y. Kuboki, H. Takita, J. Control. Rel. 2005, 101, 127.
[48] J. E. Babensee, L. V. McIntire, A. G. Mikos, Pharm. Res. 2000,
17, 497.
[49] Y. Wan, X. Qu, J. Lu, C. Zhu, L. Wan, J. Yang, Biomaterials 2004,
25, 4777.
[50] G. H. Ryu, W. S. Yang, H. W. Roh, I. S. Lee, J. K. Kim, G. H. Lee,
Surf. Coat. Technol. 2005, 193, 60.
[51] P. Favia, R. dAgostino, Surf. Coat. Technol. 1998, 98, 1102.
[52] S. H. Hsu, W. C. Chen, Biomaterials 2000, 21, 359.
[53] D. Hegemann, H. Brunner, C. Oehr, Nucl. Instrum. Methods
Phys. Res. B Beam Interact. Mater. Atoms 2003, 208, 281.
[54] H. Park, K. Y. Lee, S. J. Lee, K. E. Park, W. H. Park, Macromol.
Res. 2007, 15, 238.
[55] H. Shen, X. Hu, J. Bei, S. Wang, Biomaterials 2008, 29,
2388.
[56] X. Qu, W. Cui, F. Yang, C. Min, H. Shen, J. Bei, Biomaterials
2007, 28, 9.
[57] H. S. Baek, Y. H. Park, C. S. Ki, J. C. Park, D. K. Rah, Surf. Coat.
Technol. 2008, 202, 5794.
308
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
[58] W. He, T. Yong, Z. W. Ma, R. Inai, W. E. Teo, S. Ramakrishna,
Tissue Eng. 2006, 12, 2457.
[59] R. Chandrasekaran, J. Venugopal, S. Sundarrajan,
S. Ramakrishna, Biomed. Mater. 2011, 6, 015001.
[60] Z. Ding, G. Chen, A. S. Hoffman, J. Biomed. Mater. Res. 1998,
39, 498.
[61] P. S. Stayton, T. Shimoboji, C. Long, A. Chilkoti, G. Chen, J. M.
Harris, A. S. Hoffman, Nature 1995, 378, 472.
[62] S. Dunn, P. G. Campbell, K. G. Marra, J. Mater. Sci. Mater. Med.
2001, 12, 673.
[63] S. Loher, V. Reboul, T. J. Brunner, M. Simonet, C. Dora,
P. Neuenschwander, Nanotechnology 2006, 17, 2054.
[64] H. Nie, C. H. Wang, J. Controlled Release 2007, 120, 111.
[65] R. Ravichandran, J. R. Venugopal, S. Sundarrajan, S. Mukherjee,
S. Ramakrishna, Biomaterials 2012, 33, 846.
[66] J. R. Morones, J. L. Elechiguerra, A. Camacho, K. Holt, J. B.
Kouri, J. T. Ramirez, Nanotechnology 2005, 16, 2346.
[67] C. Baker, A. Pradhan, L. Pakstis, D. J. Pochan, S. I. Shah,
J. Nanosci. Technol. 2005, 5, 244.
[68] J. Y. Lee, J. L. R. Nagahata, S. Horiuchi, Polymer 2006, 47, 7970.
[69] D. Liang, Y. K. Luu, K. Kim, B. S. Hsiao, M. Hadjiargyrou,
B. Chu, Nucleic Acids Res. 2005, 33, e170.
[70] E. T. Thostenson, Z. Ren, T. Chou, Compos. Sci. Technol. 2001,
61, 899.
[71] I. Armentano, M. Dottori, D. Puglia, J. M. Kenny, J. Mater. Sci.
Mater. Med. 2008, 19, 2377.
[72] I. Armentano, C. Del Gaudio, A. Bianco, M. Dottori, F. Nanni,
E. Fortunati, J. Mater. Sci. 2009, 44, 4789.
[73] P. R. Supronowicz, P. M. Ajayan, K. R. Ullmann, B. P. Arula-
nandam, D. W. Metzger, R. Bizios, J. Biomed. Mater. Res. 2002,
59, 499.
[74] K. Kwon, S. Kidoaki, T. Matsuda, Biomaterials 2005, 26, 3929.
[75] C. Xu, R. Inai, M. Kotaki, S. Ramakrishna, Tissue Eng. 2004, 10,
1160.
[76] L. Huang, K. Nagapudi, R. P. Apkarian, E. L. Chaikof,
J. Biomater. Sci. Polym. Ed. 2001, 12, 979.
[77] V. Yannas, J. F. Burke, P. L. Gordon, C. Huang, R. H. Ruben-
stein, J. Biomed. Mater. Res. 1980, 14, 107.
[78] R. Brau, I. V. Yannas, Tissue engineering skin, in: Encyclo-
pedia of Biomaterials and Biomedical Engineering, (Eds. G. E.
Wnek, G. L. Bowlin), Marcel Dekker, Inc., New York, USA
2004, pp. 16521660.
[79] B. Schmajljohann, Adv. Drug. Deliv. Rev. 2006, 58, 1655.
[80] V. Saxena, B. D. Malhotra, Handbook of polymers in
electronics, Rapra Technology, Shrewsbury, UK 2002, p. 3.
[81] M. R. Abidian, J. M. Corey, D. R. Kipke, D. C. Martin, Small
2010, 6, 421.
[82] B. C. Thompson, R. T. Richardson, S. E. Moulton, A. J. Evans,
O. Stephen, G. M. Clark, G. C. Wallace, J. Controlled Release
2010, 141, 161.
[83] A. Green, N. H. Lovell, L. A. Poole-Warren, Acta Biomater.
2009, 6, 63.
[84] M. R. Abidian, D. H. Kim, D. C. Martin, Adv. Mater. 2006, 18,
405.
[85] M. Li, Y. Guo, Y. Wei, A. G. MacDiarmid, P. I. Lelkes, Bioma-
terials 2006, 27, 2705.
[86] R. Ravichandran, J. R. Venugopal, S. Sundarrajan, S. Mukherjee,
S. Ramakrishna, J. R. Soc. Interface 2010, 7, 559.
[87] E. S. Gil, S. M. Hudson, Prog. Polym. Sci. 2004, 29, 1173.
[88] M. Brownlee, A. Cerami, Science 1979, 206, 1190.
[89] A. T. Florence, D. Attwood, Physicochemical Principles of
Pharmacy, 3rd ed., Macmillan Press, London 1998, 380.
[90] R. Duncan, Nat. Rev. Drug Discov. 2003, 2, 347.
[91] H. Storrie, D. J. Mooney, Adv. Drug Deliv. Rev. 2006, 58, 500.
[92] Y. Ito, M. Casolaro, K. Kono, Y. Imanishi, J. Controlled Release
1989, 10, 195.
[93] N. Lavignac, M. Lazenby, P. Foka, B. Malgesini, I. Verpilio,
P. Ferruti, R. Duncan, Macromol. Biosci. 2004, 4, 922.
[94] Z. Khayat, P. C. Grifths, I. Grillo, R. K. Heenan, S. M. King,
R. Duncan, Int. J. Pharm. 2006, 317, 175.
[95] K. Nakamae, T. Nizuka, T. Miyata, M. Furukawa, T. Nishino,
K. Kato, T. Inoue, A. S. Hoffman, J. Biomater. Sci., Polym. Ed.
1997, 9, 43.
[96] C. Dong, A. S. Hoffman, J. Control. Rel. 1991, 15, 141.
[97] K. Nakamae, T. Nizuka, T. Miyata, T. Uragami, A. S. Hoffman,
Y. Kanzaki, in: Advanced Biomaterials in Biomedical Engin-
eering and Drug Delivery Systems, (Eds. N. Ogata, S. W. Kim,
J. Feijen, T. Okano), Springer-Verlag, Berlin, Germany 1996.
[98] E. S. Lee, K. Na, Y. H. Bae, J. Controlled Release 2003, 91, 103.
[99] S. Davaran, J. Hanaee, A. Khosravi, J Controlled Release 1999,
58, 279.
[100] A. Gallardo, G. Rodriguez, M. R. Aguilar, M. M. Fernandez,
J. San Roman, Macromolecules 2003, 36, 8876.
[101] G. Van den Mooter, B. Maris, C. Samyn, P. Augustijns,
R. Kinget, J. Pharm. Sci. 1997, 86, 1321.
[102] H. Tozaki, J. Nishioka, J. Komoike, N. Okada, T. Fujita, S.
Muranishi, S. I. Kim, H. Terashima, A. Yamamoto, J. Pharm.
Sci. 2001, 90, 89.
[103] M. Sauer, D. Streich, W. Meier, Nat. Mater. 2004, 3, 209.
[104] K. Kataoka, in: Proceedings of the 2004 International Con-
ference on MEMS, NANO and Smart Systems (ICMENS04),
2004, 4.
[105] W. T. Godbey, A. G. Mikos, J. Controlled Release 2001, 72, 115.
[106] P. Erbacher, S. Zou, T. Bettinger, A. M. Steffan, J. S. Remy,
Pharm Res 1998, 15, 1332.
[107] T. Ishii, Y. Okahata, T. Sato, BiochimBiophys Acta 2001, 1514,
51.
[108] Y. B. Lim, Y. H. Choi, J. S. Park, J. Am. Chem. Soc. 1999, 121,
5633.
[109] P. D. Thornton, G. McConnell, R. V. Ulijin, Chem. Commun.
2005, 5913.
[110] R. V. Ulijin, J. Mater. Chem. 2006, 16, 2217.
[111] Z. Yang, H. Gu, D. Fu, P. Gao, J. K. Lam, B. Xu, Adv. Mater. 2004,
16, 1440.
[112] Z. Yang, B. Xu, Chem. Commun. 2004, 2424.
[113] S. Winkler, D. Wilson, D. L. Kaplan, Biochem. 2000, 39, 12739.
[114] S. Toledano, R. J. Williams, V. Jayawarna, R. V. Ulijin, J. Am.
Chem. Soc. 2006, 128, 1070.
[115] G. Grifth, J. J. Sperinde, Macromolecules 1997, 30, 5255.
[116] J. Sperinde, L. G. Grifth, Macromolecules 2000, 33, 5476.
[117] H. G. Schild, Prog. Polym. Sci. 1992, 17, 163.
[118] M. Shibayama, T. Norisuye, S. Nomura, Macromolecules
1996, 29, 8746.
[119] R. Idziak, D. Avoce, D. Lessard, D. Gravel, X. X. Zhu, Macro-
molecules 1999, 32, 1260.
[120] R. Horne, J. P. Almeida, A. F. Day, N. Yu, J. Colloid. Interface Sci.
1971, 35, 77.
[121] M. Mikheeva, N. V. Grinberg, A. Y. Mashkevich, V. Y. Grinberg,
L. T. M. Thanh, E. E. Makhaeva, A. R. Khokhlov, Macromolecules
1997, 30, 2693.
[122] K. Van Durme, S. Verbrugghe, F. E. Du Prez, B. Van Mele,
Macromolecules 2004, 37, 1054.
[123] E. E. Makhaeva, H. Tenhu, A. R. Khokhlov, Macromolecules
1998, 31, 6112.
[124] T. Aoki, M. Kawashima, H. Katono, K. Sanui, N. Ogata,
T. Okano, Y. Sakurai, Macromolecules 1994, 27, 947.
[125] T. Okano, Y. H. Bae, H. Jacobs, S. W. Kim, J Controlled Release
1990, 11, 255.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
309
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de
[126] S. Fujishige, K. Kubota, I. Ando, J. Phys. Chem. 1989, 93, 3311.
[127] L. Klouda, A. G. Mikos, Eur. J. Pharm Biopharm. 2008, 68, 34.
[128] T. Principi, C. C. E. Goh, R. C. W. Liu, F. M. Winnik, Macro-
molecules 2000, 33, 2958.
[129] A. Serres, M. Baudys, S. W. Kim, Pharm. Res. 1996, 13, 196.
[130] C. Ramkissoon-Ganorkar, F. Liu, M. Baudys, S. W. Kim,
J. Biomater. Sci., Polym. Ed. 1999, 10, 1149.
[131] Y. H. Kim, Y. H. Bae, S. W. Kim, J. Controlled Release 1994, 28,
143.
[132] B. Vernon, S. W. Kim, Y. H. Bae, J. Biomed. Mater. Res. 2000,
51, 69.
[133] T. Miyata, T. Uragami, Biological stimulus-responsive hydro-
gels., in: Polymeric biomaterials (Ed. S. Dumitriu) 2nd Ed. CRC
Press, New York, USA 2001, 959.
[134] Z. Lu, P. Kopeckova, J. Kopecek, Macromol. Biosci. 2003, 3,
296.
[135] T. Miyata, N. Asami, T. Uragami, Macromolecules 1999, 32,
2082.
[136] N. Zelikin, J. F. Quinn, F. Caruso, Biomacromolecules 2006,
7, 27.
[137] F. Meng, W. E. Hennink, Z. Zhong, Biomaterials 2009, 30,
2180.
[138] F. Castellani, E. N. Martinez, M. C. Anon, J. Agric. Food Chem.
1999, 47, 3001.
[139] C. Jocelyn, Methods Enzymol. 1987, 143, 246.
[140] K. Oh, D. J. Siegwart, H. Lee, G. Sherwood, L. Peteanu, J. O.
Hollinger, J. Am. Chem. Soc. 2007, 129, 5939.
[141] D. P. Jones, J. L. Carlson, P. S. Samiec, P. Sternberg, Jr, V. C.
Mody, Jr, R. L. Reed, Clin. Chim. Acta. 1998, 275, 175.
[142] N. Koo, H. J. Lee, S. E. Kim, J. H. Chang, C. Park, C. Kim, Chem.
Commun. 2008, 6570.
[143] R. Lin, L. W. Chen, J. Appl. Polym. Sci. 1998, 69, 1563.
[144] J. R. Lin, L. W. Chen, J. Appl. Polym. Sci. 1998, 69, 1575.
[145] Z. G. Wei, R. Sandstrom, S. Miyazaki, J. Mater. Sci. 1998, 33,
3743.
[146] X. Zheng, S. Zhou, X. Li, J. Weng, Biomaterials 2006, 27,
4288.
[147] M. Behl, U. Ridder, Y. Feng, S. Kelch, A. Lendlein, Soft Matter
2009, 5, 676.
[148] S. Zhang, Y. Feng, L. Zhang, J. Sun, X. Xu, Y. Xu, J. Polym. Sci. ;
Part A: Polym. Chem. 2007, 45, 768.
[149] G. Zhu, S. Xu, J. Wang, L. Zhang, Radiat. Phys. Chem. 2006, 75,
443.
[150] G. Filipcsei, J. Feher, M. Zrinyi, J. Mol. Struct. 2000, 554, 109.
[151] T. Shiga, Adv. Polym. Sci. 1997, 134, 131.
[152] S. Y. Kim, H. S. Shin, Y. M. Lee, C. N. Jeong, J. Appl. Polym. Sci.
1999, 73, 1675.
[153] Y. Gao, S. Xu, R. Wu, J. Wang, J. Wei, J. App. Polym. Sci. 2008,
107, 391.
[154] M. Homma, Y. Seida, Y. Nakano, J. Appl. Polym. Sci. 2000, 75,
111.
[155] A. Bajpai, S. Shulka, S. Bhanu, S. Kankane, Prog. Polym. Sci.
2008, 33, 1088.
[156] S. J. Kim, H. I. Kim, S. R. Shin, S. I. Kim, J. Appl. Polym. Sci. 2004,
92, 915.
[157] M. Jensen, P. Birch Hansen, S. Murdan, S. Frokjaer, A. T.
Florence, Eur. J. Pharm. Sci. 2002, 15, 139.
[158] K. Sutani, I. Kaetsu, K. Uchida, Radiat. Phys. Chem. 2001, 61, 49.
[159] S. J. Kim, S. G. Yoon, Y. H. Lee, S. I. Kim, Polym. Int. 2004, 53,
1456.
[160] S. J. Kim, S. J. Park, M. S. Shin, S. I. Kim, J. Appl. Polym. Sci.
2002, 86, 2290.
[161] S. B. Lin, C. H. Yuan, A. R. Ke, Z. L. Quan, Sens. Actuators B
2008, 134, 281.
[162] H. I. Kim, B. K. Gu, M. K. Shin, S. J. Park, S. G. Yoon, I. Y. Kim,
Proc. SPIE Int. Soc. Opt. Eng. 2005, 5759, 447.
[163] J. Pyun, Polym. Rev. 2007, 47, 231.
[164] D. Szabo, G. Szeghy, M. Zrinyi, Macromolecules 1998, 31,
6541.
[165] D. Szabo, I. Czako-Nagy, M. Zrinyi, A. Vertes, J. Colloid Inter-
face Sci. 2000, 221, 166.
[166] M. Zrinyi, L. Barsi, D. Szabo, H. G. Kilian, J. Chem. Phys. 1997,
106, 5685.
[167] G. Wang, W. J. Tian, J. P. Huang, J. Phys. Chem. B 2006, 110,
10738.
[168] M. Babincova, D. Leszczynska, P. Sourivong, P. Cicmanec,
P. Babinec, J. Magn. Mater. 2001, 225, 109.
[169] S. G. Starodoubtsev, E. V. Saenko, A. R. Khokhlov, V. V.
Volkov, K. A. Dembo, V. V. Klechkovskaya, Microelectron.
Eng. 2003, 69, 324.
[170] P. Norris, M. Noble, I. Francolini, A. M. Vinogradov, P. S.
Stewart, B. D. Ratner, Antimicrob. Agents Chemother. 2005,
49, 4272.
[171] I. Lentacker, B. G. De Geest, R. E. Vandenbroucke, L. Peeters,
J. Demeester, S. C. De Smedt, Langmuir 2006, 22, 7273.
[172] P. Marmottant, S. Hilgenfeldt, Nature 2003, 423, 153.
[173] J. Kost, K. Leong, R. Langer, Proc. Natl. Acad. Sci. USA 1989, 86,
7663.
[174] J. Kost, K. Leong, R. Langer, Polym. Sci. Technol. 1986, 34, 387.
[175] G. S. Kumar, D. C. Neckers, Chem. Rev. 1989, 89, 1915.
[176] S. Dai, P. Ravi, K. C. Tam, Soft Matter 2009, 5, 2513.
[177] O. Pieroni, F. Ciardelli, Trends Polym. Sci. 1995, 3, 282.
[178] A. Natansohn, P. Rochon, Chem. Rev. 2002, 102, 4139.
[179] T. Shimoboji, E. Larenas, T. Fowler, S. Kulkarni, A. S. Hoffman,
P. S. Stayton, Proc. Natl. Acad. Sci. USA 2002, 99, 16592.
[180] Y. Deng, N. Li, Y. He, X. Wang, Macromolecules 2007, 40,
6669.
[181] M. Moniruzzaman, C. J. Sabey, G. F. Fernando, Polymer 2007,
48, 255.
[182] C. Konak, R. C. Rathi, P. Kopeckova, J. Kopecek, Macro-
molecules 1997, 30, 5553.
[183] E. Ivanov, N. L. Eremeev, P. O. Wahlund, I. Y. Galaev,
B. Mattiasson, Polymer 2002, 43, 3819.
[184] T. S. Novikova, N. N. Barashkov, A. Yassar, M. Hmyene, J. P.
Ferraris, Synth. Met. 1996, 83, 47.
[185] H. Von Seggern, P. S. Wnkel, C. Zhang, H. W. Schimdt,
Macromol. J. Chem. Phys. 1994, 195, 2023.
[186] N. Barashkov, T. S. Novikova, D. J. Guerrero, J. P. Ferraris,
Synth. Met. 1995, 75, 241.
[187] Y. Kunugi, L. L. Miller, T. Maki, A. Canavesi, Chem. Mater.
1997, 9, 1061.
[188] L. Akcelrud, Prog. Polym. Sci. 2003, 28, 875.
[189] K. J. Lee, W. K. Oh, J. Song, S. Kim, J. Lee, J. Jang, Chem.
Commun. 2010, 46, 5229.
[190] V. Bulmus, Z. Ding, C. J. Long, P. S. Stayton, A. S. Hoffman,
Bioconjug. Chem. 2000, 11, 78.
[191] D. Kuckling, H. J. P. Adler, K. F. Arndt, L. Ling, W. D. Habicher,
Macromol. Chem. Phys. 2000, 201, 273.
[192] L. Verestiuc, C. Ivanov, E. Barbu, J. Tsibouklis, Int J Pharm
2004, 269, 185.
[193] N. Gonzalez, C. Elvira, J. San Roman, Macromolecules 2005,
38, 9298.
[194] M. F. Leung, J. Zhu, P. Li, F. W. Harris, Macromol. Symp. 2005,
226, 177.
[195] K. Kurata, A. Dobashi, J. Macromol. Sci. Part A- Pure Appl.
Chem. 2004, 41, 143.
[196] H. K. Ju, S. Y. Kim, S. J. Kim, Y. M. Lee, J. Appl. Polym. Sci. 2002,
83, 1128.
310
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.MaterialsViews.com
www.mbs-journal.de
R. Ravichandran, S. Sundarrajan, J. R. Venugopal, S. Mukherjee, S. Ramakrishna
[197] L. Ning, Y. Min, Z. Maolin, L. Jiuqiang, H. Hongfei, Radiat.
Phys. Chem. 2001, 61, 69.
[198] R. Lupitskyy, Y. Roiter, S. Minko, C. Tsitsilianis, Langmuir
2005, 21, 8591.
[199] M. Yamato, C. Konno, M. Utsumi, A. Kikuchi, T. Okano,
Biomaterials 2002, 23, 561.
[200] C. Geismann, M. Ulbricht, Macromol. Chem. Phys. 2005, 206,
268.
[201] J. F. Hester, S. C. Olugebefola, A. M. Mayes, J. Membrane Sci.
2002, 208, 375.
[202] J. C. Rodr guez-Cabello, J. Reguera, A. Girotti, M. Alonso, A. M.
Testera, Prog. Polym. Sci. 2005, 30, 1119.
[203] M. Alonso, V. Reboto, L. Guiscardo, A. S. Martin, J. C. Rodr guez-
Cabello, Macromolecules 2000, 33, 9480.
[204] N. Nath, A. Chilkoti, Adv. Mater. 2002, 14, 1243.
[205] D. Webb, Y. H. An, A. Gutowska, V. A. Mironov, R. J. Friedman,
MUSC Orthop. J. 2000, 3, 18.
[206] Y. Qiu, K. Park, Adv. Drug Deliver. Rev. 2001, 53, 321.
[207] I. Schmolka, J. Biomed. Mater. Res. 1972, 6, 571.
[208] Y. L. Cao, C. Ibarra, C. Vacanti, Preparation and use of thermo-
responsive polymers., in: Tissue engineering: methods and
protocols, (Eds. J. R. Morgan, M. L. Yarmush) Humana Press,
Totowa, N.J., USA 2006, 75.
[209] B. Jeong, S. W. Kim, Y. H. Bae, Adv. Drug Deliver. Rev. 2002, 54, 37.
www.MaterialsViews.com
Macromol. Biosci. 2012, 12, 286311
2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
311
Advances in Polymeric Systems for Tissue Engineering. . .
www.mbs-journal.de

Potrebbero piacerti anche