You are on page 1of 18

Review

Received: 20 May 2014

Revised: 24 July 2014

Accepted article published: 4 August 2014

Published online in Wiley Online Library: 3 September 2014

(wileyonlinelibrary.com) DOI 10.1002/jctb.4505

Polymers for medical and tissue engineering


applications
Deniz Ozdila and Halil Murat Aydina,b*
Abstract
Recent decades have seen great advancements in medical research into materials, both natural and synthetic, that facilitate the
repair and regeneration of compromised tissues through the delivery and support of cells and/or biomolecules. Biocompatible
polymeric materials have become the most heavily investigated materials used for such purposes. Naturally-occurring and
synthetic polymers, including their various composites and blends, have been successful in a range of medical applications,
proving to be particularly suitable for tissue engineering (TE) approaches. The increasing advances in polymeric biomaterial
research combined with the developments in manufacturing techniques have expanded capabilities in tissue engineering
and other medical applications of these materials. This review will present an overview of the major classes of polymeric
biomaterials, highlight their key properties, advantages, limitations and discuss their applications.
2014 Society of Chemical Industry
Keywords: polymers; tissues and organs; scaolds; tissue engineering

INTRODUCTION

NATURALLY-OCCURRING POLYMERS

Regenerative medicine has seen great advancements over the


past decade with progress in polymer science oering new materials and strategies for the elds of tissue engineering, drug delivery,
and cosmetic surgery. For tissue engineering research, the primary
focus has been on developing substitutes for the natural extracellular matrix (ECM) scaolds capable of acting as temporary
three-dimensional support structures that promote and guide correct tissue pattern formation. Polymers, both naturally-occurring
and synthetic, have become the natural choice of biomaterial
for such applications for a number of reasons including their
availability as a resource and ability to be easily reproduced,
their versatility in the range of applications for which they can be
adapted, their generally tuneable properties for function-specic
use and the biodegradable nature of most of them. As polymeric scaolds are generally subjected to in vivo conditions
they are also generally required to possess good cell adhesiveness, promote cellbiomaterial interactions, exhibit structural
and mechanical integrity, and have a microstructure suitable
for tissue integration and the exchange of factors critical to cell
survival.
Scaold design parameters and manufacturing methods for
multi-faceted polymer systems ultimately rely on the fundamental chemical nature of the polymer. For example, while
collagen is degradable and highly suitable as temporary scaolding material for repair at defect sites, the mechanical strength
and non-degradability of polyurethanes are sought for load
bearing applications. Polymers used as biomaterials in biomedical applications can be separated into two broad categories:
naturally-occurring polymers and synthetic polymers. As biomedical polymers are usually combined with other materials for
maximized performance and targeted tissue response, this
review will discuss each type of polymer separately under the
aforementioned categories.

The rst polymers to be used in biomedical applications,


naturally-occurring polymers,1 far out-date the use of synthetic polymers which rst appeared around the 1960s.
Frequently-utilized, naturally-occurring polymers can be divided
into the following three groups: proteins (e.g. silk, collagen, soy,
brin gels), polysaccharides (e.g. chitin/chitosan, alginate and
hyaluronic acid derivatives) and polynucleotides (e.g. DNA and
RNA).2 The main advantage of natural polymers is their high
degree of scaoldtissue compatibility due to the positive biological recognition of their chemical make-up. However, the use of
these polymers also carries the potential for inciting an immune
response due to any impurities in the material gained during
processing. Clinical applications require the use of medical grade
materials and, in any instances where immunogenic fragments
are detected within the material, it is imperative that they are
removed.

Correspondence to: Halil Murat Aydin, Environmental Engineering Department


and Bioengineering Division, Hacettepe University, 06800, Ankara, Turkey.
E-mail: hmaydin@hacettepe.edu.tr

a BMT Calsis Co., Hacettepe University Technopolis, 06800, Beytepe, Ankara,


Turkey
b Environmental Engineering Department & Bioengineering Division and Center
for Bioengineering, Hacettepe University, 06800, Ankara, Turkey

www.soci.org

2014 Society of Chemical Industry

1793

J Chem Technol Biotechnol 2014; 89: 17931810

Hyaluronic acid
A ubiquitous proteoglycan, and one that is often incorporated
into tissue regenerative scaolds, is the linear, anionic polymer,
hyaluronic acid. This co-polymer of D-glucuronic acid and Nacetyl-D-glucosamine can be found in several body tissues including connective, epithelial and neural tissue and synovial uid.3

www.soci.org
Hyaluronic acid not only has a key role in ensuring joint lubrication
but is also known to promote cell motility and proliferation which
are properties critical for tissue regeneration. These features,
alongside the enzymatic degradability, bioresorbability and the
ability to add further functionality via its side chains has led to the
development of hyaluronic-based scaolds of all forms including
hydrogels for wound healing applications,4 sponges for cartilage
repair,5 and meshes for bone tissue engineering.6 It is perhaps the
more specic features of hyaluronic acid such as its ability to organize and retain aggrecan and sulphated glycosaminoglycans,7
its ability to adhere chondrocytes through cell surface receptors
which then leads to the production of signals for cell migration,
proliferation and dierentiation,8,9 and its ability to stimulate proteoglycan synthesis10,11 that has made it so popular in cartilage
tissue.
Hyaluronic acid is also known to take part in the stimulation of
a local inammatory response which is particularly useful for the
conduction of local tissue repair and remodelling.12 In addition,
the angiogenic eects13 of hyaluronic acid have been documented
and add further appeal to the natural biomaterial as local angiogenesis is considered critical for tissue formation and survival. In
recent years chemical modications to hyaluronic acid have been
explored to chemically enhance its physical qualities, rheological
qualities and tissuematerial interactive qualities. Such upgrades
will continue to inspire future research into the use hyaluronic acid
in tissue engineering applications.

1794

Collagen
Collagens are extra cellular matrix proteins. Type I collagen, a
brillar, rod-shaped molecule,14 is the most abundant of the 27
dierent types of collagen. Type I collagen can be puried from
a number of tissues including tendon, ligament, bone, skin and
cornea via relatively simple biochemical processing methods.15
The high content of collagen Type I in the extracellular matrix has
been a key reason why it has become one of the most investigated
proteins for use in medical applications.16 As it normally provides
the structural framework and tensile strength in body tissues17 it
is considered an excellent biological resource for the construction
of tissue engineering scaolds.
The mechanical performance of collagen relies heavily on
the fundamental crystalline assembly of the protein which has
a helical quaternary structure.18 At the nanoscale, the helical
self-assembly of tropocollagen subunits form the collagen brils that are cross-linked at the microscale to give the ECM and
tissues its tensile strength. The interactions of these brils with
other proteins and extracellular matrix components also play a
role in dening the engineered tissue matrix. At this level, bril
diameter, length, density and orientation can all be modied to
enhance the biological and mechanical activity of the scaold.
The covalent bonding between brils to form larger brils or bre
bundles can be attained through various fabrication methods,
including electro-spinning modules and moulding. The application of mechanical stimulation such as tensile strain19 and
cyclic circumferential strain20,21 on collagen Type I-based scaolds
have proven to assist with the control of collagen deposition by
the recruited cells and therefore serves as a tool for directing the
structural formation of the scaold.
Collagen carries the advantage of having simulative chemical
characteristics, such as integrin receptors, required for adequate
cell attachment and proper tissue formation.22,23 Furthermore, the
types of cells that are attracted to and attach to this biomaterial
not only secrete specic enzymes that degrade the collagen

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

without creating cytotoxic degradation products, but they are


also the chief synthesizers of new collagen that is deposited in to
the extracellular space. This gives collagen scaolds behavioural
characteristics that are ideal and capable of closely following the
bone remodelling process that governs bone tissue engineering
applications. Collagen can be forged to exhibit the physical and
morphological features required for local connective tissue regeneration, combined with other materials (for example -tricalcium
phosphate for bone tissue engineering) to form composites and
prepared in many dierent physical forms (beads, membranes,
shaped objects, etc.).24,25 Currently, the main issue with collagen
use from a regulatory perspective is the challenges that are associated with ensuring the safety with using such animal-derived
materials (in particular the risk of transmitting prions and BSE).
Co-polymerization capabilities with other biomaterials raise
even more options for customizing scaold properties.
Chitin/Chitosan
The second most abundant polymer in nature is chitosan, a linear cationic polysaccharide that is attained from the deacetylation of chitin (a linear polysaccharide built from -1,4 linked
N-acetylglucosamine units). Owing to the insolubility of chitin in
common solvents and the diculties this poses for processing,
deacetylation is necessitated and chitosan becomes the usable
product. Chitosan is a promising degradable polymer that is one
of the only two non-human origin natural polymers (along with
alginic acid) used in biomedical engineering. Its base unit, chitin,
has characteristics, such as wound healing capacity,26 that are particularly favourable in tissue engineering cases. The crystallinity
and molecular weight of the chitosan polymer depends on the
degree of deacetylation.27 The degradation of chitosan is also
primarily inuenced by the degree of acetylation.28 The alternative method for controlling degradation proles is by altering the
side-groups of the polymer such that extensive hydrogen bonding is prevented via the attachment of bulky side groups.29 In vitro
degradation of chitosan is performed by several enzymes including chitosanase, lysozyme and papain30 with lysozyme being the
physiologically relevant enzyme for in vivo degradation.
The poor mechanical strength of chitosan and its high degree
of hydrophilicity has traditionally been resolved by cross-linking
it to other polymers such as collagen,31,32 poly(lactic acid) (PLA),33
poly(lactic-co-glycolic acid) (PLGA),33,34 polyethylene glycol PEG35
and alginate.36 The various cross-linked forms of chitosan with
other polymers include semi- or full-interpenetrating polymer
networks, chitosanchitosan networks and hybrid polymer
networks.37,38 The structural stability of chitosan-based scaolds
can thus be varied through these cross-links.
This cationic polymer is able to form electrostatic interactions
with negatively charged cell surfaces and, depending on its conguration, it can also display great hydrophilicity and cell proliferation eects.39,40 The ability of this bioactive polymer to interact
with glycosaminoglycans (GAGs) in particular give scaolds based
on chitosan a direct inuence on the modulation of cytokines
and growth factors41 and thus local tissue regeneration activities.
Furthermore, chitosanDNA complexes have also been explored
as a means of facilitating the regeneration process via scaolds
with amplied bioactivity at the molecular level.42
The biodegradability, biocompatibility, bio-adhesiveness, and
non-toxicity of chitosan41,43 account for the popularity of this
biomaterial in medicine. This is topped by the possibility of
processing chitosan and chitosan-composites into numerous

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications


shapes; membranes, sponges, bres, microspheres and hydrogels. Some recent research into chitosan-based scaolds are
aimed at further improving surface characteristics of scaolds for
hard tissue regeneration,44 exploring its anti-bacterial qualities
when combined with other polymers and anti-bacterial agents,45
increasing stability of chitosan-based cartilage repair scaolds
by co-polymerization with other natural polymers,46 and introducing chitosan microspheres embedded in calcium sulphate as
point-of-care drug delivery systems.47 Bovine derived collagen,
much like the innovative uptake of fungi-derived chitosan, can be
used as an example by which solutions for the abovementioned
issues can be found, paving the way for the safe and eective
future use of this material.
Alginate
The other non-human derived natural polymer used to fabricate tissue engineering scaolds is the linear copolymer of
-D-manuronic acid and -L-glucuronic acid linked by a 14 glycosidic bond, alginate. Commonly sourced from the cell wall of
brown algae, extraction of the polysaccharide is through the use
of a basic solution followed by acidic precipitation. The molecular
weight of the polysaccharide can reach up to 500 kDa.
The simplicity of creating alginate hydrogels with the polymer undergoing spontaneous gelation in the presence of divalent cations and carboxylate side groups makes it an attractive
and promising biomaterial with tissue engineering application
potential. Traditionally, along with drug delivery and cell delivery
scaolds,48 alginate has also been used in wound dressings.
Most research and development in hydrogel based systems
have been centred on composite constructs that incorporate
dierent materials such as collagen49,50 PLGA,51,52 Poly-L-lysine
(PLL),53,54 PCL,55 polyethers,56 and chitosan,57,58 mainly for support
of alginates mechanical weakness. Scaold designs are as varied
as the types of body tissues that are aimed to be restored from
gels,59,60,50 porous networks,61,62 and sponges63 to lms64 and
microspheres.65,49,54,53 Along with its design exibility is the ability
to transform the responsiveness of alginate biomaterials toward
pH and cell adhesion through alterations to its carboxylate acid
side group.66,67
Among these advantages, however, stand alginates natural poor
cell adhesion and poor in vivo degradation performance. This
necessitates either the gamma irradiation or periodates oxidation
of alginate biomaterials. Several types of alginate-based scaolds
have been used in tissue regeneration attempts of dierent body
tissues including bone and cartilage,61,59,63,60 nerve,54 and connective tissue.50

J Chem Technol Biotechnol 2014; 89: 17931810

indirectly by binding extracellular matrix proteins in the blood.69,70


The polymer also binds growth factors that further promote cell
proliferation and dierentiation.71 Furthermore, the angiogenic
eects of brin-based materials have also been established.72 This
bioactivity gives brin signicant appeal and use in medicine as
sealants or heamostatic agents, which are perhaps the more commonly known applications of the biopolymer. However, brin has
also shown success as a biomaterial with tissue engineering applicability. Orthopaedic, cardiovascular and skin tissue engineering
are key examples of areas with brin-based scaold use.73 78
The main impediment to the use of brin as scaolding material is its morphological deconguration (shrinkage) and rapid
degradation in physiological conditions.73,74 Fibrin-only cell
carriers79 are weak mechanically and thus require combinations
with strength enhancers, e.g. hyaluronic acid,80 PLGA81 and
PCL/polyurethane.82 Like all successful 3D scaolds for tissue
engineering, brin-based scaolds can exhibit interconnected
pores that allow cell inltration, the exchange of nutrients and
wastes and that support vascularization.83 Fibrinogen concentration is the fundamental determining factor of pore structure
where a low concentration creates appropriately dense matrices
that can mediate the aforementioned points critical for scaold
performance.84 86
Albumin
Albumin, a protein that contributes 50% to the total mass of
blood plasma has been investigated for use in drug delivery
applications. The role of this water-soluble protein in the blood
is to carry hydrophobic fatty acids as well as maintain the pH
balance. Since albumin is ubiquitous in the human body, almost
all body tissues can enzymatically degrade the protein. This high
degree of biocompatibility and biodegradability gives albumin
high potential as a biomaterial. Albumin is particularly successful
as a tissue engineering constructs with drug delivery capabilities
and is usually preferred in studies determining protein and growth
factor release kinetics.
The weak mechanical properties of albumin has narrowed its
use to drug delivery,87 coatings,88,89 and suturing90,91 applications.
The protein can be processed into several dierent forms including
nanoparticles,87,92 microparticles,93 and bres.94,95
Chondrotin sulphate
Chondrotin sulphate (CS) has shown great promise as a biomaterial. This sulphated glycosaminoglycan has a structure very similar
to HA. Found predominantly in articular cartilage, CS plays a role
in the matrices produced by broblasts in wound healing.96 It has
been shown to have anti-inammatory eects and is a binding
factor for extracellular matrix components.97 Chondrotin sulphate
can be commonly encountered particularly in cartilage tissue
engineering combined with other cartilage ECM components to
form multi-layered scaold Its natural role in chondrogenic activities has led to its investigation in cartilage tissue engineering98
both alone99 and featuring in composite materials that also
contain poly(-caprolactone) (PCL),100 PEG,101,102 collagen,100,103
hyaluronic acid,104 and chitosan.105
Naturally-occurring poly(amino acids)
Poly(amino acids) used in biomedical applications are branched
into two groups: natural poly(amino acids) and synthetic
poly(amino acids) (discussed later).

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1795

Fibrin
Fibrinogen, the building block of brin, is a large (340 kD) glycoprotein that has three pairs of polypeptide chains, namely A, B, and
which are bound by disulphide bridges. A unique polymerisation process involving thrombin and activated factor XIII converts
brinogen into brin and forms a brin network. This polypeptide
is commonly known for its physiological role as a haemostatic
plug in tissue injuries. The options for cross-linking are one way
in which diverse microstructural and mechanical features of brin
networks can be achieved. In addition, autologous scaolds can
be manufactured by using brin made from a patients own blood.
Injectable brin hydrogels are the most frequently utilized forms
of the polypeptide as tissue regeneration support biomaterial68
Fibrin can adhere cells both directly via integrin receptors and

www.soci.org

www.soci.org
Natural poly(amino acids) are biodegradable and, unlike proteins, are composed of only one type of amino acid bonded by
amide linkages. Poly(-glutamic acid) and poly(L-lysine) are the
most commonly utilized polymers within the natural poly(amino
acid) category.
Poly(-glutamic acid) (-PGA) is a polyamide that is composed of
D- and L-glutamic acid enantomeric units. It is soluble in water and
with a reactive carboxylate group, there exists a wide variety of
options for further development with dierent functional group
attachments. Previously, -PGA has been coupled with chemotherapeutic agents,106 antibiotics,107 DNA,108 and proteins107 to aid
tissue healing and recovery. As scaold material, however, the
-PGA homopolymer is not preferred as it does not have adequate
physical properties. It has seen some application in soft tissue engineering within cross-linked hydrogels.109 Attempts to equip this
polyamide with characteristics suited to tissue engineering have
been based on blending with natural (collagen110 and chitosan111 )
and synthetic polymers (PLA,112 PLGA,113 and PCL114 ) however,
research has been limited due to the low availability of -PGA.
Poly(L-lysine) research has revealed antibacterial,115 and antitumor activity116 eects that this polymer carries. This particular
ionic polymer, however, has also been found to have high toxicity
owing to its extremely high positive charge and therefore is limited, with in vivo use such as that with tissue engineering scaolds.
Attempts to appropriate it for use as a biomaterial have been
through blending it with -PGA117 and other polymers118 120
although poly(L-lysine) remains a blending option that still
requires further research and development before clinical uptake.

SYNTHETIC POLYMERS AS BIOMATERIALS


Synthetic polymeric biomaterials may not always come as a
cheaper alternative to biological polymers due to less availability,
however, they are much more easily reproducible and have a
longer shelf-life and therefore are often more viable sources of
polymeric biomaterials. Uniformity of microstructure, degradation rate and mechanical structure with large-scale production
are the key advantages of these polymers. The most extensively used synthetic biomedical polymers is the family of linear
aliphatic polyesters: polyvinyl alcohol (PVA), (PCL), poly(lactic
acid), poly(glycolic acid), poly(hydroxybutyrate) (PHB).121 124
Numerous combination options of these polymers, both with
each other and/or various other elements, generate a great assortment of composite scaolds with varying chemical, structural and
mechanical properties that can be suited to specic tissues and
tasks.
Regulatory bodies have presented armative guidance documents for the use of synthetic polymeric biomaterials and as such,
in terms of product validity, have supported the more ecient utilization of such synthetic materials. The future will thus continue
to see synthetic polymers as indispensable tools for medical and
tissue engineering applications.

1796

Polyetheretherketone (PEEK)
One of biomedical engineerings most frequently utilized polymers is polyetheretherketone (PEEK). This linear, aliphatic,31 semi
crystalline polymer is a high performance polymer with a high
melting point of 334 C and a high resistance to wear. PEEK has
a maximum tensile strength of 100 MPa, a maximum elongation
of 50150% and a Youngs Modulus of about 3.7 GPa.125 With
its mechanical properties, chemical resistance and radiolucency
closely matching those of natural bone,126 the popularity of PEEK
in orthopaedic biomedical applications can be understood.

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

The biocompatibility of PEEK and PEEK composites127 has


been conrmed through numerous in vitro cytotoxicity128 130
and mutagenecity131 studies as well as in vivo studies that have
shown that the biomaterial triggers a minimal inammatory
response.132 134 Adding to the benets of using PEEK is its MRI
compatibility. These excellent chemical, mechanical and thermal properties make PEEK an obvious choice for load-bearing
orthopaedic devices including uses in knee joints,135 spinal
fusion,136 and craniofacial repair137 which have been the initial
and traditional uses for it. Recent research,138,139 however, is
seeking to transform PEEK-based biomaterials so that further
functionality such as stimulation and integration of local tissue
can be achieved with it.
Due to the chemical intertness of PEEK, it has a limited ability
to form strong attachments to native tissues.140 Attempts to overcome this in orthopaedic applications introduced the inclusion
of osteoinductive or osteoconductive factors, such as hydroxyapatite. Such additives, however, have not provided a complete solution as the mechanical strength of the polymeric material is quickly
compromised owing to the additional lack of binding between
PEEK and reinforcement/ller elements.141 143 For the purposes of
orthopaedic tissue engineering scaold design, eorts have been
focused on developing certain manufacturing methods for PEEK
scaolds such as knitting and braiding,144,145 acid-etching, alkali
treatment, anionic oxidation, shot-blasting,146 and sulfonation147
for some modication of its mechanical properties to produce
more exible, deformable, and matrix-like structures suitable for
tissue ingrowth and local mechanics. Tissue anchorage with PEEK
materials have been largely achieved through interconnected
porous scaold designs146 that allow tissue integration in the
matrix. It has been shown that pore sizes of 200300 m141 and
a porosity value of 75%148 have been considered successful parametrical values for attaining sucient bone tissue xation.
Poly(hydroxyalkanoates)
Polyhyrdroxyalkanoates are another large class of naturallyoccurring aliphatic polyesters. In terms of both availability and suitability to tissue engineering research, however, poly 3-hydroxybutyrate (PHB), 3-hydroxybutyrate and
3-hydroxyvalerate (PHBV) co-polymers, poly 4-hydroxybutyrate
(P4HB), 3-hydroxybutyrate and 3-hydroxyhexanoate (PHBHHx)
co-polymers and poly 3-hydroxyoctanoate (PHO) have been the
main members of interest.
The simplest and most widely investigated of this family of
thermoplastics is poly(hydroxybutyrate), which has been explored
mainly in the construction of nerve149 and bone150 tissue engineering scaolds. The biocompatibility of this biomaterial found
naturally in the body151 is quite high, with its completely non-toxic
degradation products150 that are absorbable via natural metabolic
pathways.
The main properties of PHB that limit its use are its brittleness,
tendency to acquire a high degree of crystallinity,150 poor stiness
and hydrophobic character and, compared with similar biomedically utilized polymers, its slow degradation rate.152
The diculties that these properties pose have been shown
to be overcome with various scaold fabrication methods, surface property modication procedures and other techniques. A
primary example of such attempts includes the functionalization of PHB matrices by blending it with co-polymers such as
hydroxyvalerate (PHV).153 The incorporation of hydroxyapatite
particles in PHB-based scaolds have also highlighted the contribution of such factors in enhancing the success of these scaolds

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

www.soci.org

in tissue engineering approaches by the ability of hydroxyapatite to not only attract osteoblastic activity but also modify
the microarchitecture for a more porous material. Similarly, the
porosity achievable with use of aqueous emulsions have also
been shown to positively impact PHB scaold performance.154
Another example is the use of polyethylene glycol, which has been
shown to enhance the hydrophilic properties of PHB nano-brillar
scaolds.155 Polyhydroxyalkonoates will continue to maintain
signicance in the eld of medical polymers with the continuing progress in recombinant, purication and modication
technologies.

J Chem Technol Biotechnol 2014; 89: 17931810

Figure 1. Degradation time of PLGA with varying lactic acid and glycolic
acid ratio. Adapted from Ref. (179) By John Wiley & Sons, Inc. Reprinted with
permission.

composition is the main determinant of the chemical, mechanical


and structural variations that can be achieved. Figure 1 depicts
the inuence of the co-polymer ratio on degradation rates. While
increasing the amount of the slower degrading PLA increases
degradation time, counteractively increasing the amount of PGA
will slow down this process. Degradation times, for example, can
be from 12 months, 45 months and 56 months, for 50:50,
75:25 and 85:15 co-polymer ratios of PLGA, respectively.169 The
bulk erosion of PLGA makes controlled release of therapeutic
and other factors dicult. Therefore PLGA microspheres,170 173
microcapsules,174,175 nano-spheres,176 and nano-bres177,178 are
often used.
Poly(-caprolactone)
Another aliphatic polyester used as scaold material in tissue
engineering is the semi-crystalline polymer, poly(-caprolactone)
(PCL).180,181 With great organic solvent solubility, a melting temperature of about 5560 C, and glass transition temperature of
54 C182 this polymer is easily processed into tissue regeneration
support structures.
Degradation proles of the polymer make it suitable for use in
tissues that have a longer regeneration process. In particular PCL
has been shown to carry a similar rate of degradation to the rate of
new bone formation.183,184,185 Compared with its family members
PLA and PGA, PCL takes much longer to degrade186 via the same
non-enzymatic cleavage of ester linkages.189 191 This is the main
reason why PCL is usually adopted for long-term in vivo systems
rather than short-term systems.192 Enzymatic degradation of PCL
is known to occur to some extent as well, although this is not the
primary breakdown mechanism of the polymer.193
The innate biological inactivity of PCL makes it a promising
biomaterial for safe and controlled in vivo applications. The in
vivo performance of PCL scaolds have been commended for
their particular success in carrying chondrogenic additives that
successfully induce chondrogenesis in mesenchymal stem cells.194
The method by which such an inert polyester is functionalized
and activated to produce desired eects is primarily through
surface modication methods that focus on creating negatively
charged hydrophobic surfaces that promote cell attachment via
proteins such as bronectin and vitronectin.195
Furthermore, blending and copolymerization196 with other
polymers are methods by which scaold characteristics can be

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1797

Poly(-hydroxyacids) (Poly(glycolic acid), Poly(lactic acid)


and their co-polymers)
Perhaps the most heavily researched category of synthetic
biopolymers is the linear aliphatic polyester family which includes
poly(lactic acid), poly(glycolic acid), their co-polymers poly(lactic
acid-co-glycolic acid), poly(-caprolactone), poly(propylene
fumarate) and poly(hydroxy butyrate) (the last three will
be discussed separately later).156 As the most widely used,
FDA-approved materials with long-term clinical data from a
wide range of clinical indications for which they have been utilized, this category of synthetic polymers are usually the rst to
come to mind as synthetic biomaterials.
The poly(-hydroxy acids), poly(glycolic acid) (PGA), poly(lactic
acid) (PLA) and their copolymer poly(lactic-co-glycolic acid)
(PLGA), are broken down to their monomeric units lactic acid and
glycolic acid through hydrolysis of the ester bonds in the backbone of their chains. These breakdown products are then simply
cleared by natural metabolic pathways. In order to maintain the
hydrolytic stability of the ester bond, the length of the aliphatic
chains must be limited. This is also important for keeping the
degradability characteristics of the polymer.
The highly crystalline thermoplastic, PGA, has impressive properties such as a melting temperature >200 C and a tensile strength
of about 12.5 GPa.157 These properties have led to the use of
PGA predominantly as suture material. As scaold material PGA
has some unique and signicant shortcomings. Its high molecular weight form has poor solubility in most organic solvents while
its low molecular weight forms are more soluble. The more soluble forms of PGA, however, lack sucient mechanical integrity in
vivo with signicant decline in its performance observed at 24
weeks158 161 this may not be sucient time for adequate regeneration of certain body tissues such as bone. Although glycolic
acid is a natural metabolite, high levels are known to stimulate an
excessive inammatory response.162,163 Also, because of its high
hydrolytic sensitivity, processing of PGA materials must be carried
out in carefully controlled conditions.
Poly(lactic acid) chains are assembled from chiral molecules and
may thus exist as poly(L-lactic acid) (PLLA), poly(D-lactic acid)
(PDLA), poly(D,L-lactic acid) (PDLLA) and meso-poly(lactic acid).
It is the optical impurities that exist between these enantiomers
in the PLA chain, rather than the molecular weight, that determine the ultimate properties of PLA-based materials. It is more
hydrophobic than PGA which means that degradation rates are
much slower, making it highly suitable for both in vitro and in vivo
applications164 167 where long-term mechanical integrity is critical. The general values for Youngs Modulus, tensile strength and
impact strength for PLA are around 3 GPa, 1.52.7 GPa and 50-70
MPa,168 respectively.
PLGA, on the other hand, presents a combined, myriad of property advantages over its polymeric counterparts. The copolymer

www.soci.org
controlled even further for desired outcomes. Owing to this
exibility, the pore sizes achievable with PCL scaolds and the
relatively inexpensive manufacturing methods with this material,
PCL serves as a viable material solution for tissue engineering
endeavours.
Poly(urethanes)
Linear aliphatic polyesters dominate the hard-tissue engineering
synthetic biomaterials category whereas the main biomaterial investigated for soft-tissue engineering purposes has been
polyurethane. A hard segment (diisocyanate), the soft segment
(poly(ethers) or poly(esters)) and chain extenders are the fundamental components of polyurethanes.197 The ratios of these
components involved in the chemical reaction that produces the
polyurethane will dene its ultimate properties.
The historical use of polyurethanes had experienced the drawbacks of the polymers toxic degradation products. To overcome
this issue, polyurethanes with diisocyanate replacements have
been developed. One example is using 1,4-diisocyanatobutane
(BDI) and a putrescine chain extender where the degradation
of BDI leads to the release of putrescine a polyamine which
encourages cell growth and proliferation.198 Another example is
polymerizing highly pure lysine diisocyanate with glucose so that
breakdown products consist of merely glucose and lysine.199
Polyurethane matrices with a wide range of porosities,
surface-to-volume ratios, and three-dimensional structures can be
made via electrospinning, water foaming and thermally-induced
phase separation methods. Tensile strength and breaking strains
can reach 13 MPa and 280%, respectively.199 Polyurethanes have
also displayed cell adhesion and proliferation qualities which also
add to their suitability for in vivo use.200 However, an important
drawback for polyurethanes in tissue engineering or drug delivery
use is that their degradation rates are found to be not suitable for
these applications. For this reason blends and combinations of
polyurethane with other biomaterials are made to assist degradation proles of polyurethane-based scaolds. Ongoing studies
are looking into chemically modifying polyurethanes to equip
them with the appropriate mechanical characteristics required
at the various stages of healing and regeneration, subsequently
followed by its biodegradation. Biodegradable polyurethanes,
therefore, continue to hold promise over highly porous scaolds
that are disadvantaged by mechanical weakness.

1798

Poly(propylene fumarate)
Poly(propylene fumarate) (PPF) is a unique, injectable, linear
polyester that contains multiple unsaturated double bonds
that are also available for covalent cross-linking in the presence of free-radical initiators using benzoyl peroxide and N
N dimethyl-p-toluidine. The biocompatibility of PFF has been
established by various studies.201,202
The cross-linking capability of the polyester means that degradation characteristics will be dependent on the cross-linking density,
the cross-linker and the molecular weight of the polymer.203
This capacity also translates to the ability of PPF-based scaffolds to assume scaold designs that may not be achievable by
non-cross-linkable biodegradable polymers. In fact, this exibility
has made PPF a common bone defect lling material.204 207
The crosslinking reaction is propagated using free-radical
polymerization.
PPF-based porous scaolds.201,208 210 and microsphereembedded scaolds211 214 have been studied in vivo or in vitro

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

extensively, and have proved to be biocompatible. The traditional


method of synthesizing porous PPF scaolds was by embedding
into the polymeric material sodium chloride crystals which are
leached out upon implantation a technique that carries the risk
of creating a hypertonic local environment and oers minimal control over interconnected porosity. Current methods are based on
initiating foaming reactions during polymerization215,216 such that
the supercritical CO2 treatment will generate the porosity essential
for the success of PPF scaolds in tissue repair and regeneration.
The inclusion of ceramics and other enhancement factors in these
scaolds mechanically strengthens them217 as well as improving
their biocompatibility and materialtissue relationships.218 220
Synthetic poly(amino acids)
Synthetic poly(amino acids) include homo-and co-poly(amino
acids), and display high crystallinity, low degradation rate,
poor mechanical properties and immunogenicity221 have been
reported for these polymers.
Poly(L-glutamic acid) (L-PGA) is a exible synthetic poly(amino
acid) that is relatively easy to fabricate into dierent
architectures.222 It is also non-immunogenic and enzymatically
degradable.223,224 It has featured as cancer drug delivery225 and
MRI contrast agents226 and as a copolymer with other polymers in
tissue engineering scaolds.
Poly(aspartic acid) (PAA) is a highly water-soluble polymer that
also exists as a hydrogel and undergoes enzymatic degradation.227
It can be copolymerized with other polymers (e.g. PLA,228 PCL229
and PEG228 ). Micellar congurations formed by these co-polymers
have seen interest for use in payload delivery.
Poly(ortho esters)
The poly(ortho ester) (POE) family of hydrophobic polymers230 are
polymers consisting of three geminal ether bonds. The unique
qualities of POEs are its surface-erosion mechanism of degradation and the sensitivity of the process to pH. Ortho-ester bonds
are stable at neutral pH but rapidly hydrolyse at about pH 5.5.
Under physiological conditions these highly hydrophobic polymers require activation of degradation via an acid.
Of the four classes of POEs, presented diagrammatically in Fig. 2,
it is the fourth group, POE IV, that has particular relevance to tissue
engineering. POE IV, modied from POE II, consists of short segments of lactic acid or glycolic acid incorporated into its backbone.
This not only speeds up degradation but makes POE IV more suitable for tissue regeneration materials than POE IIII, which possess
degradation rates much too slow for these purposes. Varying the
backbone chemistry of POEs will vary the degradation and material properties.
This new generation of POEs is an evolutionary development
of the second generation polymers232 and is recognized as
self-catalytic poly(ortho esters). In this new variation of POE, short
dimer segments of glycolic acid or lactic acid are incorporated
into the polymer backbone. Owing to the susceptibility of POE
to acid-catalysed hydrolysis, varying the concentration of the
-hydroxy acid dimer segments in the polymer backbone can
control the erosion rate of the bulk polymer. These polymers
oer signicant advantages over the second-generation POEs,
which required the addition of acidic excipients to control erosion
rate.
Poly(anhyrides)
Polyanhydrides are either aromatic, aliphatic or a mixture of the
two polymers that are constructed from two carbonyl groups

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

www.soci.org

Figure 2. The four poly(ortho ester) families. Adapted from Ref. (231) By Elsevier Science B.V. Reprinted with permission.

bound by an ether bond. Synthesis of this hydrolytically unstable polymer is via the dehydration of the diacid or mixture
of diacids by melt polycondensation produces this class of
biodegradable and biocompatible polymer. Polyanhydrides have
predominantly featured as biomaterials that are practical for
drug delivery systems. Polyanhydride-based microparticles233 235
and nanoparticles236 238 developed for these purposes can be
injected, administered orally or with aerosol delivery.
These polymers undergo surface erosion that is pH-dependent
and degradation is based on the backbone chemistry of the polymer, with the specic tuning of degradation rate. Polyanhydrides
on their own are usually not suitable for tissue engineering applications that require load bearing scaolds as their low molecular
weights limit their mechanical strength. The Youngs modulus of
poly(anhydride)s has been determined to be around 1.3 MPa239
which would not, for example, be suitable in bone tissue engineering approaches. Dimethacrylated anhydrides, however, is
one sub-class that oers a stronger polyanhydride-based material. This is due to the photo cross-linkability and variability of
monomers which allow for the attainment of higher structural
and mechanical integrity of these networks.240,241 Cross-linked
polyanhydrides have shown eective performance in bone tissue
engineering approaches as well as drug delivery materials.242
Monomer exibility also gives promise of applications in other
tissue engineering elds.
Poly(anhydrides-co-imides) have been developed as mechanically stronger brands of polyanhydride-based materials that
also undergo surface erosion.243 Compressive strengths up to
5060 MPa have been reported for poly(anhydrides-co-imides)
based on succinic acid, trimellitylimidoglycine, and
trimellitylimidoalanine.244
Poly(anhydride-esters), are particularly attractive as they combine the properties of polyesters and polyanhydrides. There are, for
example, poly(anhydride esters) that are based on salicyclic acid
that release the non-steroidal anti-inammatory (NSAID) (i.e. salicyclic acid) upon hydrolytic degradation.
The opportunity to tailor polyanhydrides in such ways make it
a versatile material that can be modelled into a diverse range of
tissue restoration systems.

J Chem Technol Biotechnol 2014; 89: 17931810

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1799

Poly(glycerol sebacate)
Poly(glycerol sebacate) (PGS) is a biodegradable polymer synthesized by the polycondensation of glycerol and sebacic acid. PGS
was rst used in a soft-tissue engineering applications in 2002.245

PGS is completely bioresorbable with degradation products


eliminated through natural metabolic pathways.246
The hydrophilic, partially semi-crystalline PGS polymer has thermoset elastomeric properties that can be tuned via the alteration
of curing temperature, curing time and molar ratio of glycerol to
sebacic acid.247 251
An additional level of control over these properties in
poly(glycerol sebacate acrylate) (PGSA) is through varying the
number of acrylate moieties, which translates to varying the
number of cross-links in the polymer network.252
Typical of elastomeric biomaterials, PGS has a nonlinear stress
strain behaviour and at 37 C the polymer is totally amorphous. Another interesting feature of PGS is its shapememory
behaviour. With heat, PGS materials can be stimulated to
re-conform to memorized shapes and this property is attributed
to its ability to switch from its xed three-dimensional networks
at cooler temperatures to its amorphous phase with increased
temperature.253
PGS undergoes surface erosion with the cleavage of ester linkages. This type of degradation is useful in that unlike bulk erosion, mass loss in surface erosion occurs linearly with time, so that
geometrical features supporting tissue regeneration are maintained for longer periods.245,254,255 Although PGS has an accelerated degradation rate in vivo than in vitro, the biopolymer has
shown complete biocompatibility in several studies.256 258 One
limitation to PGS use in tissue engineering applications, however,
is reported to be the acidic local environments that can be generated when the hydrolysis of the ester groups release carboxylic
acids.259
Further functionalisation of PGS has been investigated with the
use of cell migration, adhesion, dierentiation and proliferation
mediators such as laminin, bronectin, brin, collagen types I/III,
and elastin.260
To synergize the favourable characteristics of various polymers available for combination with PGS several composite
PGS-based scaolds have been explored. Bioglass/PGS membranes for cardiac tissue engineering,259 PGS with nano-tubular
halloysite (2SiO2 2Al(OH)2 ) incorporation,261 micro-structured
brous PGS-PCL scaolds for heart valve regeneration262 are some
examples of where both organic and inorganic biomaterial blends
with PGS have been studied.
It is the modication of textile manufacturing technologies
and their adaptation to production of biodegradable elastomers
that has given rise to some signicant biomedical companies of
our day. There are currently intensive research and development

www.soci.org
eorts that are producing, in particular, exible tubular structures
and woven matrices that are seen in clinical use.
Poly(phosphazenes)
Poly(phosphazenes) are a very large group of polymers that have
traditionally been produced with high molecular weights. Their
backbones are completely inorganic constructed with alternating
phosphorous and nitrogen atoms with two organic side groups
attached to each phosphorous atom. Most phosphazenes are
made by substitution reactions on a poly(dichlorophosphazene)
intermediate. The history of poly(phosphazene) use in
biomedicine only extends back to the past two decades.263
In order to functionalize the polymer further, specic side groups
can be introduced onto the primary structure such as amino acid
esters, glucosyl, lactate or imidazolyl units.264 266 This exibility
has led to >500267 types of poly(phosphazenes) produced to date
and as such initiated an exploration of these polymers as biomaterials for tissue regeneration rather than just their traditional use in
drug delivery.
The aforementioned side groups, for example, introduce
hydrolytic instability to the structure and assist with degradation
where the non-toxic breakdown products are released. Interestingly, when poly(phosphazenes) are combined with polymers, like
polyesters, that have acidic degradation products, the presence
of poly(phosphazene) breakdown products have a pH buering
eect.268 Mechanical properties are also varied in this manner too.
One study found that such modications produces signicantly
larger value ranges for glass transition temperature (Tg 035 C),
contact angle (63 107 ), tensile strength (2.47.6 MPa), and
modulus of elasticity (31.4455.9 MPa).269 Poly(phosphazenes)
do generally require blends with other polymers in this way to
improve chemical and mechanical characteristics. There has also
been some recent concern over the foreign body response270 that
poly(phosphazene)-based materials have been found to trigger,
contrary to previous research which had only found a limited
inammatory response.271
In various tissue engineering applications such as nerve
regeneration272,273 and orthopaedic applications,274,275 poly
(phosphazenes) have been used as lms,274 bres,272,275 gels,276,277
and sintered microspheres274 thanks to the ability to increase their
hydrophobicity with side group substitutions.
Poly(dioxanone)
Poly(dioxanone) (PDO) is one of the most commonly used members of the poly(ester ether) family of degradable polymers. These
polymers typically have an ether bond integrated into the backbone of a polyester with the aim of initiating hydrolytic cleavage of
the ester bond. Ring-opening polymerization of p-dioxanone produces PDO. P-dioxanone has a Tg about 10 to 0 C and a Tm of
115 C.278
PDO is considered a slow-degrading polymer that has complete
mass loss at about 612 months.157 PDO does not exhibit the
mechanical properties that tissue engineering scaolds would
generally require, with a modulus of (1.5 GPa).157 It does, however,
have good exibility and 12 months of considerable strength
maintenance which is why it has been selected for the production
of monolament sutures for decades.279

1800

Poly(ethylene glycol) Poly(ethylene oxide)


Poly(ethylene glycol) (PEG) is polyether that is polymerized from
ethylene oxide condensation. Chains of PEG that are above 10 kDa

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

are termed poly(ethylene oxide) (PEO). The hydroxyl groups at the


end of PEG chains are used for a variety of PEG macromers that
have diering properties and uses.
PEG polymers are biocompatible, biodegradable, non-toxic, and
low-immunogenic synthetic polymers that do not bind proteins or
cells.280,281 In fact, the rst interest in PEG was to use it as biomaterial surface coatings owing to its ability to prevent serum protein
adsorption, although the mechanisms by which this occurs is still
not completely understood and have been attributed to a number of factors.282,283 The mechanical properties of PEG scaolds in
tissue engineering depend on the molecular weight, cross-linking,
and polymer concentration. Decreasing the molecular weight or
increasing the concentration of the polymer has been shown to
improve the elastic modulus.284
PEG hydrogels have been studied in tissue engineering scaold
applications. Of course these scaolds have been adapted to these
purposes with the incorporation of cell adhesion RGD peptides to
enable cell adhesion and survival.285 For PEG hydrogels that may
carry proteins or cells for implantation and release, photopolymerization is preferred to thermal polymerization owing to the sensitivity of this method towards such payloads and the better eciency that it oers.286 In fact, one group has been able to demonstrate PEG photopolymerization transdermally.287 PEG is often
also preferred for its bio-inactivity toward encapsulated proteins
or cells where such interaction is not desired.285,288 291 Functionalisation is also possible where cell adhesive molecules and growth
factors used in PEG hydrogel scaolds have added to the success
of modulated tissue regeneration with these materials.292 295
As discussed, there are several synthetic polymer options that
can be used in various medical applications. The suitability of
each of these polymers for their intended use is based on their
respective chemical and physical properties and the capacity to
which they can be processed to assume the required structural
characteristics. Table 1 summarizes the key properties of the
synthetic polymers discussed for quick reference.
Hydrogels
Cross-linked polymer networks that contain 6090% water, known
as hydrogels, have become attractive materials for regenerative
medicine. Due to their remarkably tuneable properties hydrogels
are emerging as an exciting new material used in a diverse range
of applications including the development of articial cornea
implants,306 as llers for soft tissue engineering307 operations and
in cartilage repair materials,308 to name a few.
Degradable three-dimensional hydrogel matrices are made from
either hydrophilic homopolymers, copolymers, or, they can exist as
insoluble matrices via the introduction of cross-links.
Hydrogels can be fabricated from both natural polymers of
agarose, alginate, chitosan, hyaluronic acid, brin, collagen and
others, as well as synthetic polymers such as poly(ethylene glycol) (PEG), poly(vinyl alcohol) (PVA), and polyacrylates such as
poly(2-hydroxyethyl methacrylate) (PHEMA).309,310 Covalent and
non-covalent bonding in cross-links generate a biocompatible311
matrix with structural similarity to other macromolecules found in
the body.
Among the dierent methods of hydrogel synthesis, including
Michael (conjugate addition) and click chemistry, free-radical
polymerisation is the most appealing for tissue engineering due
to the great gelation kinetics and in situ polymerization capability
associated with this process.312 Figure 3 is a visual representation
of these chemical synthesis processes. Acrylate-based hydrogels
that have been subjected to this process contain many mechanical

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

www.soci.org

Table 1. Properties of popular synthetic polymers used in medicine


Glass
Elastic

Tensile

Melting

transition

Crystallinity

modulus

modulus

(%)

(GPa)

(GPa)

temperature
( C)

temperature
( C)

37

2.7

1.52.7

173178

Polyglycolic acid

45-55

7.0

57

Poly(caprolactone)

37

0.4

Poly-lactic-coglycolic acid
(50/50)

Amorphous

Poly(propylene
fumarate)

Polyanhydride

Polymer

Structure

Polylactic acid

Polyhphosphazene

degrade

products

(months)

6065

L-Lactic acid

1218

( 296 298)

225230

3540

Glycolic Acid

34

( 296,298)

0.40.6

5863

60

Caproic Acid

>24

( 299 301)

2.0

1.42.8

Amorphous

5055

D,L-lactic acid
and
glycolic acid

36

( 296 298)

37

23

23

3050

60

Fumaric acid,
propylene
glycol and
poly(acrylic
acid-cofumaric
acid)

Varies (>24)

( 302,303)

2527

5090

27

Carboxylic acids

0.141.4

( 304,305)

55

Phosphate,
ammonia,
corresponding
side groups

surface

( 267,274)

and structural advantages such as increased elastic modulus,313


great cellbiomaterial interactions,314 good tensile modulus,315
prevention of undesired brosis,316 enhanced mechanisms of
release of payloads embedded in polymeric material,292,294,317 and
higher compressive modulus with GAG incorporation.318 322
Mixing or blending hydrogels with micro- and nanoparticles324
which are later removed through various processes or through
natural dispersion with payload delivery allow porous scaolds
to form. Successful porous synthetic hydrogels in cornea replacements oer an example of this. Such scaolds are also known to
promote cell migration and angiogenesis and with a high water
content are capable of rapid nutrient diusion.325
Although porosity is desirable for tissue integration, the biggest
challenge yet for porous hydrogel scaolds is the poor mechanical strength that they display.326 Cross-linking density plays an
important role in appropriating mechanical compliance and
mesh size for cell encapsulation and use in tissue augmentation procedures.327 Careful control of chemical and processing
parameters can ultimately yield hydrogel scaolds with numerous
benets for tissue regeneration eorts.

STIMULI RESPONSIVE POLYMERS

Refs

polymeric systems, creating multi-functional systems.334 As


such, stimuli-responsive polymers have received great interest
over the past decade and have been taken up in a number of
medical applications from drug delivery and tissue engineering to
imaging.
Thermo-responsive polymers
To date, temperature- and pH-sensitive polymers have been the
most frequently studied type of stimuli-responsive polymer.
Thermoresponsive polymers can change their hydrophilicity/hydrophobicity, conformation or solubility due to a phase
transition reaction when prompted by elevated temperatures335
to reach a threshold temperature known as critical solution temperature (CST). Generally, a lower critical solution temperature
(LCST) polymers will reduce the solubility (increase hydrophobicity) while a higher critical solution temperature (HCST) will increase
solubility (increase hydrophilicity).336 Poly(N-isopropylacrylamide)
(PNIPAAm) in particular has been the most researched
temperature-responsive polymer where it exhibits a transition
from hydrophilicity under 32 C to a hydrophobicity above this
temperature.337 One study demonstrated the utility of the LCST
feature of PNIPAAm with a thiolated chitosan-PNIPAAm hydrogel used for wound infections that deliver ciprooxacin (an
anti-bacterial drug) to the wound site that will then swell upon
the addition of cold water for easy removal of the gel. Another
example is poly(N-vinylcaprolactam), a lactam-based polymer that
is also thermoresponsive with a LCT of about 35 C338 and holds
promise in tissue engineering applications as it thermoresponsivity allows for the formation of a macroporous architecture a
common limitation for hydrogels intended for use in tissue engineering applications. Other temperature-responsive polymers
used in biomedical applications include poly(N-vinylcaprolactam)

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1801

Smart or intelligent polymers are those which can alter their


physio-chemical properties in response to stimuli (pH, temperature, redox, enzymes, light, magnetic, ultrasound) such that a particular functional output of the polymeric system occurs. Among
some property changes that can occur as a response to stimuli are
changes in surface wettability,328 membrane permeability,329,330
swelling capability (hydrogels),331 aggregation behaviour,332
and sol-gel transition.333 Stimuli-responsive polymers are produced by introducing specic polymeric modalities to traditional
J Chem Technol Biotechnol 2014; 89: 17931810

Time to
Degradation

www.soci.org

D Ozdil, HM Aydin

Figure 3. Common methods of hydrogel synthesis A) Free-radical polymerisation of diacrylate macromer. B) Conjugate addition of a thiol and acrylate
group. C) Click bonding of a pendant alkyne and azide via 1,2,3-triazole group. Adapted from Ref. (323) by WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.
Reprinted with permission.

(PVCL) which has increasing hydrophobicity and insolubility from


2535 C,339 poly(ethylene glycol) (PEG) or poly(ethylene oxide)
(PEO) which becomes insoluble above 80 C and poly(propylene
oxide) (PPO) which, when in co-polymer form,340,341 can exhibit a
wide range of solubility and phase transition temperatures.

properties, for example, increased surface adhesion when in


protonated form, have also inspired interest for their use in drug
delivery systems.

CONCLUDING REMARKS

1802

pH-responsive polymers
pH-responsive polymers respond to changes in pH, co-solvent,
and electrolytes such that changes in their solubility, volume,
and chain conformation can be observed. The pH-responsiveness
of polymers can be tuned by the incorporation of ionizable
monomer units in to their polymeric backbones. Chitosancarrageenan-based polyelectrolyte complexes have been
explored as porous biomaterials that can maintain stability
at physiological conditions while dissociating upon implantation due to chitosan reacting under the lowered pH levels
caused by local inammation associated with implantation.342
Another study343 was able to show how electrospun polyelectrolyte hydrogel nanobres were able to display reversible
swelling/contracting with varying pH, highlighting their potential
as articial muscles, interactive tissue engineering scaolds or
drug delivery systems. Materials based on poly(acrylic acid) (PAAc)
and poly(methacrylic acid) (PMAc) are common polymers used
in pH-responsive systems and can also exhibit reversible swelling
when pH is varied.344,345 Such acrylic polymers and their unique

wileyonlinelibrary.com/jctb

Currently, the number of donors required for organ and soft tissue replacement are well below those numbers that are actually in
demand. For this reason, research and development of synthetic
materials capable of combining cellular components for both the
in vitro and in vivo regeneration of tissues are continuing to proceed at a signicant pace. The range of dierent chemical natures
of dierent polymers, the ability to fabricate them such that they
replicate tissue or organ morphologies, their ability to biodegrade
without adverse eects, and their biocompatibility has created a
great area of use in the eld of tissue engineering. The modication and enhancement of several processing methods such as
3D printing, electrospinning, bre processing, and foaming for the
production of tissue scaolds seen in recent years, the fall of prices
associated with medical grade products and the improvements in
conditions of manufacturing these products in small-scale clean
environments have all been factors leading to the uptake of polymers in eorts to regenerate bone, cartilage, tendon, skin, cornea,
and other tissues that constitute dierent morphological and
mechanical properties. The advancements in growth factor-cell

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications


studies and stem cell studies that have progressed parallel to
these eorts combined with the widespread use of bioreactors, all
within the regulative standards of special organizations such as the
advanced technological medicinal products (ATMP), provide foresight into the utilization of polymers in multi-component complex
systems that are developed in tissue engineering studies and subsequently taken up in clinical use.

REFERENCES

J Chem Technol Biotechnol 2014; 89: 17931810

21 Isenberg BC and Tranquillo RT, Long-term cyclic distention enhances


the mechanical properties of collagen-based media-equivalents.
Ann Biomed Eng 31:937949 (2003).
22 Heino J, The collagen receptor integrins have distinct ligand recognition and signaling functions. Matrix Biol 19:319323 (2000).
23 White DJ, Puranen S, Johnson MS and Heino J, The collagen receptor
subfamily of the integrins. Int J Biochem Cell B 36:14051410
(2004).
24 Sarikaya B and Aydin HM, Development of collagen/beta-TCP based
synthetic bone grafts. J Tiss Eng Reg Med 8:410410 (2014).
25 Aydin HM, Yang Y, Kohler T, El Haj A, Mller R, Piskin E, Interaction of osteoblasts with macroporous scaolds made of PLLA/PCL
blends modied with collagen and hydroxyapatite. Adv Eng Mater
11:B83B88 (2009).
26 Kojima K, Okamoto Y, Kojima K, Miyatake K, Fujise H, Shigemasa Y,
et al., Eects of chitin and chitosan on collagen synthesis in wound
healing. J Vet Med Sci 66:15951598 (2004).
27 Islam A and Yasin T, Controlled delivery of drug from pH sensitive
chitosan/poly (vinyl alcohol) blend. Carbohyd Polym 88:10551060
(2012).
28 Ren DW, Yi HF, Wang W and Ma XJ, The enzymatic degradation and
swelling properties of chitosan matrices with dierent degrees of
N-acetylation. Carbohyd Res 340:24032410(2005).
29 Li DH, Liu LM, Tian KL, Liu JC and Fan XQ, Synthesis, biodegradability
and cytotoxicity of water-soluble isobutylchitosan. Carbohyd Polym
67:4045 (2007).
30 Xia WS, Liu P and Liu J, Advance in chitosan hydrolysis by non-specic
cellulases. Bioresource Technol 99:67516762 (2008).
31 Chen RN, Wang GM, Chen CH, Ho HO and Sheu MT, Development of
N,O-(carboxymethyl)chitosan/collagen matrixes as a wound dressing. Biomacromolecules7:10581064 (2006).
32 Liu BS, Yao CH and Fang SS, Evaluation of a non-woven fabric coated
with a chitosan Bi-layer composite for wound dressing. Macromol
Biosci 8:432440 (2008).
33 Wang JP, Feng SS, Wang S and Chen ZY, Evaluation of cationic
nanoparticles of biodegradable copolymers as siRNA delivery system for hepatitis B treatment. Int J Pharm 400:194200
(2010).
34 Yang R, Shim WS, Cui FD, Cheng G, Han X, Jin QR, et al., Enhanced electrostatic interaction between chitosan-modied PLGA nanoparticle and tumor. Int J Pharm 371:142147 (2009).
35 Zhang YQ, Chen JJ, Zhang YD, Pan YF, Zhao JF, Ren LF, et al., A novel
PEGylation of chitosan nanoparticles for gene delivery. Biotechnol
Appl Biochem 46:197204 (2007).
36 Matsusaki M, Sakaguchi H, Serizawa T and Akashi M, Controlled
release of vascular endothelial growth factor from alginate hydrogels nano-coated with polyelectrolyte multilayer lms. J Biomat Sci
- Polym E18:775783 (2007).
37 Berger J, Reist M, Mayer JM, Felt O, Peppas NA and Gurny R, Structure and interactions in covalently and ionically crosslinked chitosan hydrogels for biomedical applications. Eur J Pharm Biopharm
57:1934 (2004).
38 Rodrigues IR, Forte MMD, Azambuja DS and Castagno KRL, Synthesis
and characterization of hybrid polymeric networks (HPN) based
on polyvinyl alcohol/chitosan. React Funct Polym 67:708715
(2007).
39 Ma GP, Yang DZ, Li QZ, Wang KM, Chen BL, Kennedy JF, et al.,
Injectable hydrogels based on chitosan derivative/polyethylene
glycol dimethacrylate/N,N-dimethylacrylamide as bone tissue
engineering matrix. Carbohyd Polym 79:620627 (2010).
40 Hsieh CY, Hsieh HJ, Liu HC, Wang DM and Hou LT, Fabrication and
release behavior of a novel freeze-gelled chitosan/gamma-PGA
scaold as a carrier for rhBMP-2. Dent Mater 22:622629 (2006).
41 Di Martino A, Sittinger M and Risbud MV, Chitosan: a versatile
biopolymer for orthopaedic tissue-engineering. Biomaterials
26:59835990 (2005).
42 Peng L, Cheng XR, Zhuo RX, Lan J, Wang YN, Shi B, et al., Novel
gene-activated matrix with embedded chitosan/plasmid DNA
nanoparticles encoding PDGF for periodontal tissue engineering.
J Biomed Mater Res A 90A:564576 (2009).
43 Martins AM, Alves CM, Kasper FK, Mikos AG and Reis RL, Responsive and in situ-forming chitosan scaolds for bone tissue engineering applications: an overview of the last decade. J Mater Chem
20:16381645 (2010).
44 Azhar FO, A ; Salehi, R. Fabrication and characterization of
chitosan-gelatin/nanohydroxyapatite-polyaniline composite with

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1803

1 Lakshmi SN, Laurencin CT, Biodegradable polymers as biomaterials.


Prog Polym Sci 32:762798 (2007).
2 Yannas IV, Classes of Materials used in Medicine: Natural Materials, ed by
B. D. Ratner ASH, F an. J. Schoen and Lemons J. Elsevier Academic
Press; San Diego, CA, USA (2004).
3 Hahn SK, Jelacic S, Maier RV, Stayton PS and Homan AS,
Anti-inammatory drug delivery from hyaluronic acid hydrogels. J
Biomater Sci - Polym E 15:11111119 (2004).
4 Seidlits SK CTD, Petersen RR, Shear JB, Suggs L J and Schmidt
CE, Fibronectinhyaluronic acid composite hydrogels for
three-dimensional endothelial cell culture. Acta Biomater
7:24012409 (2011).
5 Kim M, Optimization of macromer density in human MSC-laden
hyaluronic acid (HA) hydrogels, in Bioengineering Conference
(NEBEC), 2012 38th Annual Northeast (2012); 1618 March 2012;
Philadelphia, PA: IEEE; 211212 (2012).
6 Park JK, Shim J-H , Kang KS, Yeom J, Jung HS, Kim JY , Lee KH,
Kim T-H, Kim S-Y, Cho D-W and Hahn SK, Solid free-form fabrication of tissue-engineering scaolds with a poly(lactic-co-glycolic
acid) grafted hyaluronic acid conjugate encapsulating an intact
bone morphogenetic protein2/poly(ethylene glycol) complex.
Adv Function Mater 21:29062912 (2011).
7 Chung C, Erickson IE, Mauck RL and Burdick JA, Dierential Behavior of
Auricular and Articular Chondrocytes in Hyaluronic Acid Hydrogels.
Tissue Eng Part A 14:11211131 (2008).
8 Knudson W, Loeser RF, CD44 and integrin matrix receptors participate
in cartilage homeostasis. Cell Mol Life Sci 59:3644 (2002).
9 Tool BP, Hyaluronan in morphogenesis. Semin Cell Dev Biol 12:7987
(2001).
10 Goodstone NJ, Cartwright A and Ashton B, Eects of high molecular
weight hyaluronan on chondrocytes cultured within a resorbable
gelatin sponge. Tissue Eng 10:621631 (2004).
11 Kawasaki K, Ochi M, Uchio Y, Adachi N and Matsusaki M, Hyaluronic
acid enhances proliferation and chondroitin sulfate synthesis in
cultured chondrocytes embedded in collagen gels. J Cell Physiol
179:142148 (1999).
12 Duo S, Thibeault SL, Li WH, Shu XZ and Prestwich G, Eect of a
synthetic extracellular matrix on vocal fold lamina propria gene
expression in early wound healing. Tissue Eng 12:32013207
(2006).
13 Slevin M, Krupinski J, Ganey J, Matou S, West D, Delisser H, et al.,
Hyaluronan-mediated angiogenesis in vascular disease: uncovering RHAMM and CD44 receptor signaling pathways. Matrix Biol
26:5868 (2007).
14 Birk DE and Bruckner P, Collagen suprastructures. Top Curr Chem
247:185205 (2005).
15 Francis G and Thomas J, Isolation and chemical characterization of
collagen in bovine pulmonary tissues. Biochem J 145:287297
(1975).
16 Lee CH, Singla A and Lee Y, Biomedical applications of collagen. Int J
Pharm 221:122 (2001).
17 Sweeney SM, Orgel JP, Fertala A, McAulie JD, Turner KR, Di Lullo
GA, et al., Candidate cell and matrix interaction domains on the
collagen bril, the predominant protein of vertebrates. J Biol Chem
283:2118721197 (2008).
18 Rich A, Solving the structure of collagen. Self-assembling peptide
systems in biology. Med Eng 235241 (2001).
19 Voge CM, Kariolis M, MacDonald RA and Stegemann JP, Directional conductivity in SWNT-collagen-brin composite biomaterials through strain-induced matrix alingment. J Biomed Mater Res A
86A:269277 (2008).
20 Seliktar D, Black RA, Vito RP and Nerem RM, Dynamic mechanical conditioning of collagen-gel blood vessel constructs induces remodeling in vitro. Ann Biomed Eng 28:351362 (2000).

www.soci.org

www.soci.org

45

46
47

48
49

50
51
52
53

54
55
56
57
58
59

60

61

62

63
64
65

66

1804

potential application in tissue engineering scaolds. Designed


Monom Polym 17:654667 (2014).
Lee SH, DN, Moon JH, Park HN, Ko WK, Bae MS, Lee JB, Park SW, Kim EC,
Lee CH, Jung BY and Kwon IK, Chitosan/polyurethane blended ber
sheets containing silver sulfadiazine for use as an antimicrobial
wound dressing. J Nanosci Nanotechnol (2014)14:74887494.
Mekhail M, Jahan K and Tabrizian M, Genipin-crosslinked
chitosan/poly-L-lysine gels promote broblast adhesion and
proliferation. Carbohyd Polym 108:9198 (2014).
Doty HL, Leedy MR, Courtney HS, Haggard WO and Bumgardner JD,
Composite chitosan and calcium sulfate scaold for dual delivery
of vancomycin and recombinant human bone morphogenetic
protein-2. J Mater Sci - Mater M 25:14491459 (2014).
Tan CS, Jejurikar A, Rai B, Bostrom T, Lawrie G and Grondahl L,
Encapsulation of a glycosaminoglycan in hydroxyapatite/alginate
capsules. J Biomed Mater Res A 91A:866877 (2009).
Jay SM, Shepherd BR, Andrejecsk JW, Kyriakides TR, Pober JS and Saltzman WM, Dual delivery of VEGF and MCP-1 to support endothelial cell transplantation for therapeutic vascularization. Biomaterials
31:30543062 (2010).
Hahn MS, Teply BA, Stevens MM, Zeitels SM and Langer R, Collagen
composite hydrogels for vocal fold lamina propria restoration.
Biomaterials 27:11041109 (2006).
Wang YZ, Kim HJ, Vunjak-Novakovic G and Kaplan DL, Stem
cell-based tissue engineering with silk biomaterials. Biomaterials
27:60646082 (2006).
Kim JH, Kim YS, Kim S, Park JH, Kim K, Choi K, et al., Hydrophobically
modied glycol chitosan nanoparticles as carriers for paclitaxel. J
Control Release 111:228234 (2006).
Liu XY, Nothias JM, Scavone A, Garnkel M and Millis JM,
Biocompatibility investigation of polyethylene glycol and
alginate-poly-l-lysine for islet encapsulation. ASAIO J 56:241245
(2010).
Purcell EK, Singh A and Kipke DR, Alginate composition eects on a
neural stem cell-seeded scaold. Tissue Eng Part C - Me 15:541550
(2009).
Gou ML, Wei XW, Men K, Wang BL, Luo F, Zhao X, et al., PCL/PEG
copolymeric nanoparticles: potential nanoplatforms for anticancer
agent delivery. Curr Drug Targets 12:11311150 (2011).
Lim SM, Oh SH, Lee HH, Yuk SH, Im GI and Lee JH, Dual growth
factor-releasing nanoparticle/hydrogel system for cartilage tissue
engineering. J Mater Sci - Mater M 21:25936000 (2010).
Meng X, Li P, Wei Q and Zhang HX, pH sensitive alginate-chitosan
hydrogel beads for carvedilol delivery. Pharm Dev Technol
16:2228 (2011).
Wang JZ, Huang XB, Xiao J, Yu WT, Wang W, Xie WY, et al.,
Hydro-spinning: a novel technology for making alginate/chitosan
brous scaold. J Biomed Mater Res A 93A:910919 (2010).
Marsich E, Borgogna M, Donati I, Mozetic P, Strand BL, Salvador SG,
et al., Alginate/lactose-modied chitosan hydrogels: a bioactive
biomaterial for chondrocyte encapsulation. J Biomed Mater Res A
84A:364376 (2008).
Wang Q, Jamal S, Detamore MS and Berkland C,
PLGA-chitosan/PLGA-alginate nanoparticle blends as biodegradable colloidal gels for seeding human umbilical cord mesenchymal
stem cells. J Biomed Mater Res A 96A:520527 (2011).
Qi XP, Ye JD and Wang YJ, Alginate/poly (lactic-co-glycolic
acid)/calcium phosphate cement scaold with oriented pore
structure for bone tissue engineering. J Biomed Mater Res A
89A:980987 (2009).
Qi J, Chen AM, You HB, Li KP, Zhang D and Guo FJ, Proliferation
and chondrogenic dierentiation of CD105-positive enriched rat
synovium-derived mesenchymal stem cells in three-dimensional
porous scaolds. Biomed Mater 6:015006 (2011).
Olmez SS, Korkusuz P, Bilgili H and Senel S, Chitosan and alginate scaffolds for bone tissue regeneration. Pharmazie 62:423431 (2007).
Wittmer CR, Phelps JA, Lepus CM, Saltzman WM, Harding MJ and
Van Tassel PR, Multilayer nanolms as substrates for hepatocellular
applications. Biomaterials 29:40824090 (2008).
Jin XB, Sun YS, Zhang K, Wang J, Shi TP, Ju XD, et al., Tissue engineered
cartilage from hTGF beta 2 transduced human adipose derived
stem cells seeded in PLGA/alginate compound in vitro and in vivo.
J Biomed Mater Res A 86A:10771087 (2008).
Chan AW, Whitney RA and Neufeld RJ, Semisynthesis of a controlled stimuli-responsive alginate hydrogel. Biomacromolecules
10:609616 (2009).

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

67 Yamada Y, Hozumi K, Katagiri F, Kikkawa Y and Nomizu M, Biological


activity of laminin peptide-conjugated alginate and chitosan matrices. Biopolymers 94:711720 (2010).
68 Ryu JH, Kim IK, Cho SW, Cho MC, Hwang KK, Piao H, et al., Implantation
of bone marrow mononuclear cells using injectable brin matrix
enhances neovascularization in infarcted myocardium. Biomaterials 26:319326 (2005).
69 Makogonenko E, Tsurupa G, Ingham K and Medved L, Interaction
of brin(ogen) with bronectin: further characterization and
localization of the bronectin-binding site. Biochemistry - Us
41:79077913 (2002).
70 Preissner KT and Jenne D, Vitronectin - a new molecular connection
in hemostasis. Thromb Haemostasis 66:189194 (1991).
71 Sahni A and Francis CW, Vascular endothelial growth factor binds to
brinogen and brin and stimulates endothelial cell proliferation.
Blood 96:37723778 (2000).
72 Prasad CK and Krishnan LK, Regulation of endothelial cell phenotype
by biomimetic matrix coated on biomaterials for cardiovascular
tissue engineering. Acta Biomater 4:182191 (2008).
73 Mol A, van Lieshout MI, Veen CGD, Neuenschwander S, Hoerstrup SP,
Baaijens FPT, et al., Fibrin as a cell carrier in cardiovascular tissue
engineering applications. Biomaterials 26:31133121 (2005).
74 Jockenhoevel S, Zund G, Hoerstrup SP, Chalabi K, Sachweh JS, Demircan L, et al., Fibrin gel-advantages of a new scaold in cardiovascular tissue engineering. Eur J Cardio -Thorac 19:424430 (2001).
75 Janmey PA, Winer JP and Weisel JW, Fibrin gels and their clinical and
bioengineering applications. J R Soc Interface 6:110 (2009).
76 Le Nihouannen D, Le Guehennec L, Rouillon T, Pilet P, Bilban M, Layrolle P, et al., Micro-architecture of calcium phosphate granules and
brin glue composites for bone tissue engineering. Biomaterials
27:27162722 (2006).
77 Eyrich D, Brandl F, Appel B, Wiese H, Maier G, Wenzel M, et al.,
Long-term stable brin gels for cartilage engineering. Biomaterials
28:5565 (2007).
78 Bruns H, Kneser U, Holzhuter S, Roth B, Kluth J, Kaufmann PM, et al.,
Injectable liver: a novel approach using brin gel as a matrix for
culture and intrahepatic transplantation of hepatocytes. Tissue Eng
11:17181726 (2005).
79 Peura M, Siltanen A, Saarinen I, Soots A, Bizik J, Vuola J, et al.,
Paracrine factors from broblast aggregates in a brin-matrix carrier enhance keratinocyte viability and migration. J Biomed Mater
Res A 95A:658664 (2010).
80 Park SH, Park SR, Chung SI, Pai KS and Min BH, Tissue-engineered
cartilage using brin/hyaluronan composite gel and its in vivo
implantation. Artif Organs 29:838845 (2005).
81 Zhao HG, Ma L, Gao CY and Shen JC, A composite scaold of PLGA
microspheres/brin gel for cartilage tissue engineering: fabrication, physical properties, and cell responsiveness. J Biomed Mater
Res B 88B:240249 (2009) (2010).
82 Eyrich D, Wiese H, Mailer G, Skodacek D, Appel B, Sarhan H, et al.,
In vitro and in vivo cartilage engineering using a combination of chondrocyte-seeded long-term stable brin gels and
polycaprolactone-based polyurethane scaolds. Tissue Eng
13:22072218 (2007).
83 Jiang B, Waller TM, Larson JC, Appel AA and Brey EM, Fibrin-loaded
porous poly(ethylene glycol) hydrogels as scaold materials for
vascularized tissue formation. Tissue Eng Part A 19:224234 (2013).
84 Zhao HG, Ma L, Zhou J, Mao ZW, Gao CY and Shen JC, Fabrication and
physical and biological properties of brin gel derived from human
plasma. Biomed Mater 3:015001 (2008).
85 Vavken P, Joshi SM and Murray MM, Fibrin concentration aects
ACL broblast proliferation and collagen synthesis. Knee 18:4246
(2011).
86 Cox S, Cole M and Tawil B, Behavior of human dermal broblasts
in three-dimensional brin clots: Dependence on brinogen and
thrombin concentration. Tissue Eng 10:942954 (2004).
87 Li JM, Chen W, Wang H, Jin C, Yu XJ, Lu WY, et al., Preparation of
albumin nanospheres loaded with gemcitabine and their cytotoxicity against BXPC-3 cells in vitro. Acta Pharmacol Sin 30:13371343
(2009).
88 Khan W, Kapoor M and Kumar N, Covalent attachment of proteins to
functionalized polypyrrole-coated metallic surfaces for improved
biocompatibility. Acta Biomater 3:541549 (2007).
89 Wei Q, Li BJ, Yi N, Su BH, Yin ZH, Zhang FL, et al., Improving the blood
compatibility of material surfaces via biomolecule-immobilized
mussel-inspired coatings. J Biomed Mater Res A 96A:3845 (2011).

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

J Chem Technol Biotechnol 2014; 89: 17931810

111 Colonna C, Conti B, Perugini P, Pavanetto F, Modena T, Dorati R, et al.,


Chitosan glutamate nanoparticles for protein delivery: development and eect on prolidase stability. J Microencapsul 24:553564
(2007).
112 Deng C, Tian HY, Zhang PB, Sun J, Chen XS and Jing XB, Synthesis and characterization of RGD peptide grafted poly(ethylene
glycol)-b-poly(L-lactide)-b-poly(L-glutamic acid) triblock copolymer. Biomacromolecules (2006)7:590596.
113 Gryparis EC, Mattheolabakis G, Bikiaris D and Avgoustakis K, Eect of
conditions of preparation on the size and encapsulation properties
of PLGA-mPEG nanoparticles of cisplatin. Drug Deliv 14:371380
(2007).
114 Cao N, Cheng D, Zou SY, Ai H, Gao JM and Shuai XT, The synergistic eect of hierarchical assemblies of siRNA and chemotherapeutic drugs co-delivered into hepatic cancer cells. Biomaterials
32:2222-2232 (2011).
115 Takehara M, Hibino A, Saimura M and Hirohara H, High-yield production of short chain length poly(epsilon-l-lysine) consisting of 5-20
residues by Streptomyces aureofaciens, and its antimicrobial activity. Biotechnol Lett 32:12991303 (2010).
116 Tang CK, Sheng KC, Pouniotis D, Esparon S, Son HY, Kim CW,
et al., Oxidized and reduced mannan mediated MUC1 DNA
immunization induce eective anti-tumor responses. Vaccine
(2008)26:38273834.
117 Isaksson K, Akerberg D, Andersson R and Tingstedt B, Toxicity and dose response of intra-abdominally administered
poly-L-alpha-lysine and poly-L-glutamate for postoperative
adhesion protection. Eur Surg Res 44:1722 (2010).
118 Bertram JP, Jay SM, Hynes SR, Robinson R, Criscione JM and Lavik
EB, Functionalized poly(lactic-co-glycolic acid) enhances drug
delivery and provides chemical moieties for surface engineering
while preserving biocompatibility. Acta Biomater 5:28602871
(2009).
119 Tahara K, Furukawa S, Yamamoto H and Kawashima Y,
Hybrid-modied poly(D,L-lactide-co-glycolide) nanospheres
for a novel cellular drug delivery system. Int J Pharm 392:311-313
(2010).
120 Nottelet B, El Ghzaoui A, Coudane J and Vert M, Novel amphiphilic
poly(epsilon-caprolactone)-g-poly(L-lysine) degradable copolymers. Biomacromolecules 8:25942601 (2007).
121 Chu CFL, Lu A, Liszkowski M and Sipehia R, Enhanced growth of animal
and human endothelial cells on biodegradable polymers. Bba - Gen
Subjects 1472:479485 (1999).
122 Zhang R and Ma PX, Processing of polymer scaolds: phase separation, in Methods of Tissue Engineering, ed by Atala A LR. Academic
Press: San Diego; 715 (2001).
123 Pachence JM KJ, Biodegradable polymers, in: Principles of Tissue Engineering, 2nd edn, ed by Lanza RP LR, Chick WL. Academic Press: San
Diego; 263 (2000).
124 Bolland BJRF, Kanczler JM, Ginty PJ, Howdle SM, Shakeshe KM,
Dunlop DG, et al. The application of human bone marrow stromal
cells and poly(DL-lactic acid) as a biological bone graft extender in
impaction bone grafting. Biomaterials 29:32213227 (2008).
125 Eschbach L, Nonresorbable polymers in bone surgery. Injury
31:S22S7 (2000).
126 Kurtz SM and Devine JN, PEEK biomaterials in trauma, orthopedic, and
spinal implants. Biomaterials 28:48454869 (2007).
127 von Wilmowsky C, Vairaktaris E, Pohle D, Rechtenwald T, Lutz R, Munstedt H, et al., Eects of bioactive glass and beta-TCP containing
three-dimensional laser sintered polyetheretherketone composites on osteoblasts in vitro. J Biomed Mater Res A 87A:896902
(2008).
128 Sagomonyants KB, Jarman-Smith ML, Devine JN, Aronow MS and
Gronowicz GA, The in vitro response of human osteoblasts to
polyetheretherketone (PEEK) substrates compared to commercially pure titanium. Biomaterials 29:15631572 (2008).
129 Morrison C, Macnair R, Macdonald C, Wykman A, Goldie I and Grant
MH, In vitro biocompatibility testing of polymers for orthopedic
implants using cultured broblasts and osteoblasts. Biomaterials
16:987992 (1995).
130 Scotchford CA, Garle MJ, Batchelor J, Bradley J and Grant DM, Use of
a novel carbon bre composite material for the femoral stem component of a THR system: in vitro biological assessment. Biomaterials24:48714879 (2003).

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1805

90 Zehr KJ, Use of bovine albumin-glutaraldehyde glue in cardiovascular


surgery. Ann Thorac Surg 84:10481052 (2007).
91 De Somer F, Delanghe J, Somers P, Debrouwere M and Van Nooten
G, Mechanical and chemical characteristics of an autologous glue.
J Biomed Mater Res A 86A:11061112 (2008).
92 Shen ZY, Li Y, Kohama K, Oneill B and Bi JX, Improved drug targeting
of cancer cells by utilizing actively targetable folic acid-conjugated
albumin nanospheres. Pharmacol Res 63:5158 (2011).
93 Gayakwad SG, Bejugam NK, Akhavein N, Uddin NA, Oettinger CE
and DSouza MJ, Formulation and in vitro characterization of
spray-dried antisense oligonucleotide to NF-kappa B encapsulated albumin microspheres. J Microencapsul 26:692700
(2009).
94 Dror Y, Ziv T, Makarov V, Wolf H, Admon A and Zussman E, Nanobers
made of globular proteins. Biomacromolecules 9:27492754
(2008).
95 Regev O, Khaln R, Zussman E and Cohen Y, About the albumin
structure in solution and related electro-spinnability issues. Int J
Biol Macromol 47:261265 (2010).
96 Kosir MA, Quinn CCV, Wang WL and Tromp G, Matrix glycosaminoglycans in the growth phase of broblasts: more of the story in wound
healing. J Surg Res 92:4552 (2000).
97 Malavaki C, Mizumoto S, Karamanos N and Sugahara K, Recent
advances in the structural study of functional chondroitin sulfate
and dermatan sulfate in health and disease. Connect Tissue Res
49:133-139 (2008).
98 van Susante JLC, Pieper J, Buma P, van Kuppevelt TH, van Beuningen
H, van der Kraan PM, et al., Linkage of chondroitin-sulfate to type
I collagen scaolds stimulates the bioactivity of seeded chondrocytes in vitro. Biomaterials 22:23592369 (2001).
99 Park JS, Yang HJ, Woo DG, Yang HN, Na K and Park KH, Chondrogenic
dierentiation of mesenchymal stem cells embedded in a scaold
by long-term release of TGF-beta 3 complexed with chondroitin
sulfate. J Biomed Mater Res A 92A:806816 (2010).
100 Chang KY, Hung LH, Chu IM, Ko CS and Lee YD, The application of
type II collagen and chondroitin sulfate grafted PCL porous scaold
in cartilage tissue engineering. J Biomed Mater Res A 92A:712723
(2010).
101 Villanueva I, Gladem SK, Kessler J and Bryant SJ, Dynamic loading stimulates chondrocyte biosynthesis when encapsulated in charged
hydrogels prepared from poly(ethylene glycol) and chondroitin
sulfate. Matrix Biol 29:5162 (2010).
102 Nguyen LH, Kudva AK, Guckert NL, Linse KD and Roy K, Unique biomaterial compositions direct bone marrow stem cells into specic
chondrocytic phenotypes corresponding to the various zones of
articular cartilage. Biomaterials 32:13271338 (2011).
103 Liang WH, Kienitz BL, Penick KJ, Welter JF, Zawodzinski TA and
Baskaran H, Concentrated collagen-chondroitin sulfate scaffolds for tissue engineering applications. J Biomed Mater Res A
94A:1050-1060 (2010).
104 Shu XZ, Ahmad S, Liu YC and Prestwich GD, Synthesis and evaluation
of injectable, in situ crosslinkable synthetic extracellular matrices
for tissue engineering. J Biomed Mater Res A 79A:902912 (2006).
105 Chen YL, Lee HP, Chan HY, Sung LY, Chen HC and Hu YC, Composite
chondroitin-6-sulfate/dermatan sulfate/chitosan scaolds for cartilage tissue engineering. Biomaterials 28:22942305 (2007).
106 Van S, Das SK, Wang XH, Feng ZL, Jin Y, Hou Z, et al., Synthesis, characterization, and biological evaluation of
poly(L-gamma-glutamyl-glutamine)-paclitaxel
nanoconjugate.
Int J Nanomed 5:825837 (2010).
107 Portilla-Arias JA, Camargo B, Garcia-Alvarez M, de Ilarduya
AM and Munoz-Guerra S, Nanoparticles made of microbial
poly(gamma-glutamate)s for encapsulation and delivery of drugs
and proteins. J Biomat Sci - Polym E 20:10651079 (2009).
108 Sun Y, Tang YZ, Chu MQ, Song SX and Xin YF, A convenient and
adjustable surface-modied complex containing poly-L-glutamic
acid conjugates as a vector for gene delivery. Int J Nanomed
3:249256 (2008).
109 Matsusaki M, Yoshida H and Akashi M, The construction of
3D-engineered tissues composed of cells and extracellular matrices by hydrogel template approach. Biomaterials 28:27292737
(2007).
110 Hsu FY, Cheng YY, Tsai SW and Tsai WB, Fabrication and evaluation of a biodegradable cohesive plug based on reconstituted
collagen/gamma-polyglutamic acid. J Biomed Mater Res B
95B:2935 (2010).

www.soci.org

www.soci.org

1806

131 Katzer A, Marquardt H, Westendorf J, Wening JV and von Foerster


G, Polyetheretherketone - cytotoxicity and mutagenicity in vitro.
Biomaterials 23:17491759 (2002).
132 von Wilmowsky C, Lutz R, Meisel U, Srour S, Rupprecht S, Toyoshima
T, et al., In vivo evaluation of beta-TCP containing 3D laser sintered
poly(ether ether ketone) composites in pigs. J Bioact Compat Pol
24:169184 (2009).
133 Nieminen T, Kallela I, Wuolijoki E, Kainulainen H, Hiidenheimo I
and Rantala I, Amorphous and crystalline polyetheretherketone: mechanical properties and tissue reactions during a 3-year
follow-up. J Biomed Mater Res A 84A:377383 (2008).
134 Jockisch KA, Brown SA, Bauer TW and Merritt K, Biological response
to chopped-carbon-ber-reinforced peek. J Biomed Mater Res
26:133-146 (1992).
135 Scholes SC and Unsworth A, Pitch-based carbon-bre-reinforced poly
(ether-ether-ketone) OPTIMA (R) assessed as a bearing material in
a mobile bearing unicondylar knee joint. P I Mech Eng H 223:1325
(2009).
136 Toth JM, Wang M, Estes BT, Scifert JL, Seim HB and Turner AS,
Polyetheretherketone as a biomaterial for spinal applications. Biomaterials 27:324334 (2006).
137 Scolozzi P, Martinez A and Jaques B, Complex orbito-fronto-temporal
reconstruction using computer-designed PEEK implant. J Craniofac
Surg 18:224228 (2007).
138 Zhao Y, Wong HM, Wang W, Li P, Xu Z, Chong EY, et al., Cytocompatibility, osseointegration, and bioactivity of three-dimensional
porous and nanostructured network on polyetheretherketone. Biomaterials 34:92649277 (2013).
139 Edwards SL and Werkmeister JA, Mechanical evaluation and cell
response of woven polyetheretherketone scaolds. J Biomed Mater
Res A 100:33263331 (2012).
140 Roeder RK and Conrad TL, Bioactive polyaryletherketone composites,
in PEEK Biomaterials Handbook, ed by Kurtz SM. William Andrew:
Oxford, UK; 16379 (2012).
141 Converse GL, Conrad TL, Merrill CH and Roeder RK, Hydroxyapatite
whisker-reinforced polyetherketoneketone bone ingrowth scaffolds. Acta Biomater 6:856863 (2012).
142 Yu SC, Hariram KP, Kumar R, Cheang P and Aik KK, In vitro
apatite formation and its growth kinetics on hydroxyapatite/polyetheretherketone
biocomposites.
Biomaterials
26:23432452 (2005).
143 Wong KL, Wong CT, Liu WC, Pan HB, Fong MK, Lam WM, et al., Mechanical properties and in vitro response of strontium-containing
hydroxyapatite/polyetheretherketone composites. Biomaterials
30:38103817 (2009).
144 Fujihara K, Huang ZM, Ramakrishna S, Satknanantham K and Hamada
H, Performance study of braided carbon/PEEK composite compression bone plates. Biomaterials 24:26612667 (2003).
145 Fujihara K, Huang ZM, Ramakrishna S, Satknanantham K and Hamada
H, Feasibility of knitted carbon/PEEK composites for orthopedic
bone plates. Biomaterials 25:38773885 (2004).
146 Karageorgiou V and Kaplan D, Porosity of 3D biomaterial scaolds and
osteogenesis. Biomaterials 26:54745491 (2005).
147 Chu PP, Wu CS, Liu PC, Wang TH and Pan JP, Proton exchange
membrane bearing entangled structure: sulfonated poly (ether
ether ketone)/bismaleimide hyperbranch. Polymer 51:13861394
(2010).
148 Ryan G, Pandit A and Apatsidis DP, Fabrication methods of
porous metals for use in orthopaedic applications. Biomaterials
27:26512670 (2006).
149 Mohanna PN, Young RC, Wiberg M and Terenghi G, A composite
poly-hydroxybutyrate-glial growth factor conduit for long nerve
gap repairs. J Anat 203:553565 (2003).
150 Misra SK, Valappil SP, Roy I and Boccaccini AR, Polyhydroxyalkanoate
(PHA)/inorganic phase composites for tissue engineering applications. Biomacromolecules 7:22492258 (2006).
151 Reusch RN, Sparrow AW and Gardiner J, Transport of
poly-beta-hydroxybutyrate in human plasma. Biochim Biophys
Acta 1123:3340 (1992).
152 Bergstrand A, Uppstrom S and Larsson A, Permeability of porous
poly(3-hydroxybutyrate) barriers of single and bilayer type for
implant applications. Int J Polym Sci (2014).
153 Masaeli E, Morshed M, Rasekhian P, Karbasi S, Karbalaie K, Karamali F, Abedi D, Razavi S, Jafarian-Dehkordi A, Nasr-Esfahani MH
and Baharvand H, Does the tissue engineering architecture of

wileyonlinelibrary.com/jctb

154
155

156
157
158
159
160
161
162
163
164
165
166
167

168
169
170
171
172

173
174
175
176
177
178

D Ozdil, HM Aydin
poly(3-hydroxybutyrate) scaold aects cell-material interactions?
J Biomed Mater Res A 100A:19071918 (2012).
Cai ZJ, Biocompatibility and biodegradation of novel PHB porous
substrates with controlled multi-pore size by emulsion templates
method. J Mater Sci - Mater M 17:12971303 (2006).
Karahaliloglu Z, Demirbilek M, Sam M, Erol-Demirbilek M, Saglam
N and Denkbas EB, Plasma polymerization-modied bacterial
polyhydroxybutyrate nanobrillar scaolds. J Appl Polym Sci
128:19041912 (2013).
Seal BL, Otero TC and Panitch A, Polymeric biomaterials for tissue and
organ regeneration. Mat Sci Eng R 34:147230 (2001).
Maurus PB and Kaeding CC, Bioabsorbable implant material review.
Oper Techn Sport Med 12:158160 (2004).
Ma PX and Langer R, Degradation, structure and properties of brous
nonwoven poly(glycolic acid) scaolds for tissue engineering.
Mater Res Soc Symp P 394:99104 (1995).
Reed AM and Gilding DK, Biodegradable polymers for use in surgery
- poly(glycolic)-poly(lactic acid) homo and co-polymers .2. Invitro
degradation. Polymer 22:494498 (1981).
Gilding DK and Reed AM, Biodegradable polymers for use in surgery
- polyglycolic-poly(actic acid) homopolymers and copolymers.
P olymer 20:14591464 (1979).
Aydin HM, A three-layered osteochondral plug: structural,
mechanical, and in vitro compatibility analysis. Adv Eng Mater
13:B511B517 (2011).
Otto J, Binnebosel M, Pietsch S, Anurov M, Titkova S, Ottinger AP, et al.,
Large-pore PDS mesh compared to small-pore PG mesh. J Invest
Surg 23:190196 (2010).
Ceonzo K, Gaynor A, Shaer L, Kojima K, Vacanti CA and Stahl GL,
Polyglycolic acid-induced inammation: role of hydrolysis and
resulting complement activation. Tissue Eng 12:301308 (2006).
Zhang RY and Ma PX, Degradation behavior of porous
poly(alpha-hydroxy acids)/hydroxyapatite composite scaolds.
Abstr Papers Am Chem S 220:U251 (2000).
Eling B, Gogolewski S and Pennings AJ, Biodegradable materials of
poly(L-lactic acid) .1. Melt-spun and solution-spun bers. Polymer
23:15871593 (1982).
Aydin HM, El Haj AJ, Piskin E, Yang Y, Improving pore interconnectivity
in polymeric scaolds for tissue engineering. J Tiss Eng Reg Med
3:470476 (2009).
Aydin HM and Yang Y, Rapid production of highly interconnedted
porous scaolds by spheroidized sugar particles for tissue engineering. Journal of Hacettepe Biology and Chemistry 39:2328
(2011).
Anderson KS, Schreck KM and Hillmyer MA, Toughening polylactide.
Polym Rev 48:85108 (2008).
Ueda H and Tabata Y, Polyhydroxyalkanonate derivatives in current
clinical applications and trials. Adv Drug Deliver Rev 55:501518
(2003).
Thomas C, Gupta V and Ahsan F, Particle Size inuences the immune
response produced by Hepatitis B vaccine formulated in inhalable
particles. Pharm Res - Dordr 27:905919 (2010).
Zolnik BS and Burgess DJ, Evaluation of in vivo-in vitro release
of dexamethasone from PLGA microspheres. J Control Release
127:137145 (2008).
Gavenis K, Schneider U, Groll J and Schmidt-Rohlng B, BMP-7-loaded
PGLA microspheres as a new delivery system for the cultivation of
human chondrocytes in a collagen type I gel: the common nude
mouse model. Int J Artif Organs 33:4553 (2010).
Ye ML, Kim S and Park K, Issues in long-term protein delivery
using biodegradable microparticles. J Control Release 146:241260
(2010).
Berkland C, Pollauf E, Varde N, Pack DW and Kim K, Monodisperse liquid-lled biodegradable microcapsules. Pharm Res - Dordr
24:10071013 (2007).
Haddadi A, Aboofazeli R, Erfan M and Farboud ES, Topical delivery of
urea encapsulated in biodegradable PLGA microparticles: O/W and
W/O creams. J Microencapsul 25:379386 (2008).
Vega E, Gamisans F, Garcia ML, Chauvet A, Lacoulonche F and Egea
MA, PLGA nanospheres for the ocular delivery of urbiprofen: drug
release and interactions. J Pharm Sci - Us 97:53065317 (2008).
Xie JW and Wang CH, Electrospun micro- and nano-bers for sustained delivery of paclitaxel to treat C6 glioma in vitro. Pharm Res
- Dordr 23:18171826 (2006).
Ionescu LC, Lee GC, Sennett BJ, Burdick JA and Mauck RL, An
anisotropic nanober/microsphere composite with controlled

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

179

180
181
182
183
184
185

186
187

188

189

190
191

192
193
194

195
196

197
198

199
200

J Chem Technol Biotechnol 2014; 89: 17931810

201 Fisher JP, Vehof JWM, Dean D, van der Waerden JPCM, Holland TA,
Mikos AG, et al., Soft and hard tissue response to photocrosslinked
poly(propylene fumarate) scaolds in a rabbit model. J Biomed
Mater Res 59:547556 (2002).
202 Hedberg EL, Kroese-Deutman HC, Shih CK, Crowther RS, Carney DH,
Mikos AG, et al., Eect of varied release kinetics of the osteogenic
thrombin peptide TP508 from biodegradable, polymeric scaolds
on bone formation in vivo. J Biomed Mater Res A 72A:343353
(2005).
203 He S, Timmer MD, Yaszemski MJ, Yasko AW, Engel PS and Mikos AG,
Synthesis of biodegradable poly(propylene fumarate) networks
with poly(propylene fumarate)-diacrylate macromers as crosslinking agents and characterization of their degradation products.
Polymer 42:12511260 (2001).
204 Christenson EM, Soo W, Holm JL, Cameron NR and Mikos AG,
Biodegradable fumarate-based polyHIPEs as tissue engineering
scaolds. Biomacromolecules 8:38063814 (2007).
205 Kim CW, Talac R, Lu LC, Moore MJ, Currier BL and Yaszemski MJ, Characterization of porous injectable poly-(propylene fumarate)-based
bone graft substitute. J Biomed Mater Res A 85A:11141119
(2008).
206 Young S, Patel ZS, Kretlow JD, Murphy MB, Mountziaris PM, Baggett
LS, et al., Dose eect of dual delivery of vascular endothelial growth
factor and bone morphogenetic protein-2 on bone regeneration in
a rat critical-size defect model. Tissue Eng Pt A (2009)15:23472362.
207 Vehof JWM, Fisher JP, Dean D, van der Waerden JPCM, Spauwen PHM,
Mikos AG, et al., Bone formation in transforming growth factor
beta-1-coated porous poly(propylene fumarate) scaolds. J Biomed
Mater Res 60:241251 (2002).
208 Lee KW, Wang SF, Fox BC, Ritman EL, Yaszemski MJ and Lu LC,
Poly(propylene fumarate) bone tissue engineering scaold fabrication using stereolithography: eects of resin formulations and laser
parameters. Biomacromolecules8:10771084 (2007).
209 Lee J, Kim B, Lim G and Cho DW, Scaold fabrication with biodegradable poly(propyllene fumarate) using microstereolithography. Key
Eng Mater 342343:141144 (2007).
210 Wolfe MS, Dean D, Chen JE, Fisher JP, Han SH, Rimnac CM, et al. In
vitro degradation and fracture toughness of multilayered porous
poly(propylene fumarate)/beta-tricalcium phosphate scaolds.
J Biomed Mater Res 61:159164 (2002).
211 Kharas GB, Kamenetsky M, Simantirakis J, Beinlich KC, Rizzo AMT, Caywood GA, et al., Synthesis and characterization of fumarate-based
polyesters for use in bioresorbable bone cement composites. J Appl
Polym Sci 66:11231137 (1997).
212 Hedberg EL, Temeno JS, Tang A, Crowther RS, Carney DH and
Mikos AG, Controlled release of a tissue inducing peptide from
oligo(poly(ethylene glycol) fumarate) hydrogels for orthopedic tissue engineering. 24th Annual Conference and the Annual Fall
Meeting of the Biomedical Engineering Society EMBS/BMES Conference, 2002. IEEE; 1:472473 (2002).
213 Wang SF, Kempen DH, Simha NX, Lewis JL, Windebank AJ, Yaszemski MJ, et al., Photo-cross-linked hybrid polymer networks consisting of poly(propylene fumarate) and poly(caprolactone fumarate):
controlled physical properties and regulated bone and nerve cell
responses. Biomacromolecules 9:12291241 (2008).
214 Payne RG, McGonigle JS, Yaszemski MJ, Yasko AW and Mikos AG,
Development of an injectable, in situ crosslinkable, degradable
polymeric carrier for osteogenic cell populations. Part 2. Viability of
encapsulated marrow stromal osteoblasts cultured on crosslinking
poly(propylene fumarate). Biomaterials 23:43734380 (2002).
215 Nam YS and Park TG, Porous biodegradable polymeric scaolds
prepared by thermally induced phase separation. J Biomed Mater
Res 47:817 (1999).
216 Lewandrowski KU, Cattaneo MV, Gresser JD, Wise DL, White RL,
Bonassar L, et al. Eect of a poly(propylene fumarate) foaming
cement on the healing of bone defects. Tissue Eng 5:305316
(1999).
217 Peter SJ, Miller ST, Zhu GM, Yasko AW and Mikos AG, In vivo degradation of a poly(propylene fumarate) beta-tricalcium phosphate
injectable composite scaold. J Biomed Mater Res 41:17 (1998).
218 Hedberg EL, Kroese-Deutman HC, Shih CK, Crowther RS, Carney DH,
Mikos AG, et al., In vivo degradation of porous poly(propylene
fumarate)/poly(DL-lactic-co-glycolic acid) composite scaolds. Biomaterials 26:46164623 (2005).
219 Lukashev M, Zeng W, Goelz S, Lee D, Linker R, Gold R, et al., Activation of Nrf2 and modulation of disease progression in EAE models

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1807

release of biomolecules for brous tissue engineering. Biomaterials


31:41134120 (2010).
Miller RA, Brady JM and Cutright DE, Degradation rates of oral
resorbable implants (polylactates and polyglycolates) - rate modication with changes in Pla-Pga copolymer ratios. J Biomed Mater
Res 11:711719 (1977).
Freed LE, Vunjaknovakovic G, Biron RJ, Eagles DB, Lesnoy DC, Barlow
SK, et al., Biodegradable polymer scaolds for tissue engineering.
Bio - Technol 12:689693 (1994).
Patrick JC MA and Mclntire VL, Frontiers in Tissue Engineering. Pergamon: New York (1998).
Patlolla A, Collins G and Arinzeh TL, Solvent-dependent properties of
electrospun brous composites for bone tissue regeneration. Acta
Biomater 6:90101 (2010).
Lam CXF, Hutmacher DW, Schantz JT, Woodru MA and Teoh SH,
Evaluation of polycaprolactone scaold degradation for 6 months
in vitro and in vivo. J Biomed Mater Res A 90A:906919 (2009).
Woodru MA and Hutmacher DW, The return of a forgotten polymer Polycaprolactone in the 21st century. Prog Polym Sci 35:12171256
(2010).
Aydin HM, Calimli A and Piskin E, Microporous scaolds from poly
(lactide-co-e-caprolactone) composites with hydroxyapatite and
tricalcium phosphate using supercritical CO2 for bone tissue engineering. J Bioact Compat Pol 19:383394 (2004).
Dash TK and Konkimalla VB, Poly-epsilon-caprolactone based formulations for drug delivery and tissue engineering: a review. J Control
Release 158:1533 (2012).
Aydin HM, Korkusuz P, Vargel I, Kilic E, Guzel E, Cavusoglu T, Ucgan D
and Piskin E, A 6-month in vivo study of polymer/mesenchymal
stem cell constructs for cranial defects. J Bioact Compat Pol
26:207221 (2011).
Aydin HM, Turk M, Calimli A and Piskin E, Attachment and growth of
broblasts on poly(L-lactide/epsilon-caprolactone) scaolds prepared in supercritical CO2 and modied by polyethylene imine
grafting with ethylene diamine-plasma in a glow-discharge apparatus. Int J Artif Organs 29:873880 (2006).
Li WJ, Danielson KG, Alexander PG and Tuan RS, Biological response
of chondrocytes cultured in three-dimensional nanobrous
poly(epsilon-caprolactone) scaolds. J Biomed Mater Res A
67A:11051114 (2003).
Hutmacher DW, Scaolds in tissue engineering bone and cartilage.
Biomaterials 21:25292543 (2000).
Li WJ, Tuli R, Okafor C, Derfoul A, Danielson KG, Hall DJ, et al., A
three-dimensional nanobrous scaold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials
26:599609 (2005).
CG. P. Poly-epsilon-caprolactone and its copolymers, in Biodegradable Polymers as Drug Delivery Systems, ed by Chasin M LR. Marcel
Dekker: New York; 71120 (1990).
Gan ZH, Yu DH, Zhong ZY, Liang QZ and Jing XB, Enzymatic degradation of poly(epsilon-caprolactone)/poly(DL-lactide) blends in
phosphate buer solution. Polymer 40:28592862 (1999).
Assoian RK, Komoriya A, Meyers CA, Miller DM and Sporn MB, Transforming growth factor-beta in human-platelets - identication of
a major storage site, purication, and characterization. J Biol Chem
258:71557160 (1983).
Joyce ME, Roberts AB, Sporn MB and Bolander ME, Transforming growth-factor-beta and the initiation of chondrogenesis and
osteogenesis in the rat femur. J Cell Biol 110:21952207 (1990).
Ye WP, Du FS, Jin JY, Yang JY and Xu Y, In vitro degradation of
poly(caprolactone), poly(lactide) and their block copolymers: inuence of composition, temperature and morphology. React Funct
Polym 32:161168 (1997).
Guelcher SA, Biodegradable polyurethanes: synthesis and applications in regenerative medicine. Tissue Eng Pt B - Rev 14:317 (2008).
Guan JJ, Fujimoto KL, Sacks MS and Wagner WR, Preparation and
characterization of highly porous, biodegradable polyurethane
scaolds for soft tissue applications. Biomaterials 26:39613971
(2005).
Shi R, Chen DF, Liu QY, Wu Y, Xu XC, Zhang LQ, et al., Recent advances
in synthetic bioelastomers. Int J Mol Sci 10:42234256 (2009).
Bonzani IC, Adhikari R, Houshyar S, Mayadunne R, Gunatillake
P and Stevens MM, Synthesis of two-component injectable
polyurethanes for bone tissue engineering. Biomaterials
28:423433 (2007).

www.soci.org

www.soci.org

220

221

222

223

224

225

226

227

228

229

230

231

232
233

234

235

236

237

238

239

240

1808

by BG00012 (dimethyl fumarate) suggests a novel mechanism of


action combining anti-inammatory and neuroprotective modalities. Mult Scler 13:S149 (2007).
Lee KW, Wang S, Yaszemski MJ and Lu LC, Physical properties and cellular responses to crosslinkable poly(propylene
fumarate)/hydroxyapatite nanocomposites. Biomaterials 29:
28392848 (2008).
Bourke SL and Kohn J, Polymers derived from the amino acid
L-tyrosine: polycarbonates, polyarylates and copolymers with
poly(ethylene glycol). Adv Drug Deliver Rev 55:447466 (2003).
Yuan H, Luo K, Lai YS, Pu YJ, He B, Wang G, et al., A novel
poly(L-glutamic acid) dendrimer based drug delivery system
with both pH-sensitive and targeting functions. Mol Pharmaceut
7:953962 (2010).
Wadhwa S and Mumper RJ, Intracellular delivery of the reactive oxygen species generating agent D-penicillamine upon conjugation
to poly-L-glutamic acid. Mol Pharmaceut 7:854862 (2010).
Chiu HC, Kopeckova P, Deshmane SS and Kopecek J, Lysosomal
degradability of poly(alpha-amino acids). J Biomed Mater Res
34:381392 (1997).
Feng XL, Lv FT, Liu LB, Tang HW, Xing CF, Yang QO, et al. Conjugated
polymer nanoparticles for drug delivery and imaging. Acs Appl
Mater Inter 2:24292435 (2010).
Ke TY, Feng Y, Guo JY, Parker DL and Lu ZR, Biodegradable cystamine
spacer facilitates the clearance of Gd(III) chelates in poly(glutamic
acid) Gd-DO3A conjugates for contrast-enhanced MR imaging.
Magn Reson Imaging 24:931940 (2006).
Kakizawa Y, Furukawa S and Kataoka K, Block copolymer-coated calcium phosphate nanoparticles sensing intracellular environment
for oligodeoxynucleotide and siRNA delivery. J Control Release
97:345356 (2004).
Karal-Yilmaz O, Kayaman-Apohan N, Misirli Z, Baysal K and Baysal BM,
Synthesis and characterization of poly(L-lactic acid-co-ethylene
oxide-co-aspartic acid) and its interaction with cells. J Mater Sci Mater Med 17:213227 (2006).
Cha EJ, Kim JE and Ahn CH, Stabilized polymeric micelles by electrostatic interactions for drug delivery system. Eur J Pharm Sci
38:341346 (2009).
Ng SY, Vandamme T, Taylor MS and Heller J, Synthesis and erosion studies of self-catalyzed poly(ortho ester)s. Macromolecules
30:770772 (1997).
Heller J, Barr J, Ng SY, Abdellauoi KS and Gurny R, Poly(ortho esters):
synthesis, characterization, properties and uses. Adv Drug Deliver
Rev 54:10151039 (2002).
Heller J and Barr J, Poly(ortho esters) - from concept to reality.
Biomacromolecules 5:16251632 (2004).
Kipper MJ, Wilson JH, Wannemuehler MJ and Narasimhan B, Single dose vaccine based on biodegradable polyanhydride microspheres can modulate immune response mechanism. J Biomed
Mater Res A 76A:798810 (2006).
Carrillo-Conde B, Schiltz E, Yu J, Minion FC, Phillips GJ, Wannemuehler
MJ, et al., Encapsulation into amphiphilic polyanhydride
microparticles stabilizes Yersinia pestis antigens. Acta Biomater
6:31103119 (2010).
Sun L, Zhou SB, Wang WJ, Su QX, Li XH and Weng J, Preparation and
characterization of protein-loaded polyanhydride microspheres.
J Mater Sci - Mater M 20:20352042 (2009).
Salman HH, Irache JM and Gamazo C, Immunoadjuvant capacity of
agellin and mannosamine-coated poly(anhydride) nanoparticles
in oral vaccination. Vaccine 27:47844790 (2009).
Tamayo I, Irache JM, Mansilla C, Ochoa-Reparaz J, Lasarte JJ and
Gamazo C, Poly(anhydride) nanoparticles act as active Th1 adjuvants through toll-Like receptor exploitation. Clin Vaccine Immunol
17:13561362 (2010).
Petersen LK, Sackett CK and Narasimhan B, High-throughput analysis
of protein stability in polyanhydride nanoparticles. Acta Biomater
6:38733881 (2010).
Ibim SEM, Uhrich KE, Attawia M, Shastri VR, El-Amin SF, Bronson
R, et al., Preliminary in vivo report on the osteocompatibility of
poly(anhydride-co-imides) evaluated in a tibial model. J Biomed
Mater Res 43:374379 (1998).
Weiner AA, Shuck DM, Bush JR and Shastri VP, In vitro degradation
characteristics of photocrosslinked anhydride systems for bone
augmentation applications. Biomaterials 28:52595270 (2007).

wileyonlinelibrary.com/jctb

D Ozdil, HM Aydin

241 Determan AS, Trewyn BG, Lin VSY, Nilsen-Hamilton M and Narasimhan
B, Encapsulation, stabilization, and release of BSA-FITC from
polyanhydride microspheres. J Control Release 100:97109 (2004).
242 Weiner AA, Bock EA, Gipson ME and Shastri VP, Photocrosslinked
anhydride systems for long-term protein release. Biomaterials
29:24002407 (2008).
243 Attawia MA, Uhrich KE, Botchwey E, Fan M, Langer R and Laurencin CT,
Cytotoxicity testing of poly(anhydride-co-imides) for orthopedic
applications. J Biomed Mater Res 29:12331240 (1995).
244 Uhrich KE, Gupta A, Thomas TT, Laurencin CT and Langer R, Synthesis and characterization of degradable poly(anhydride-co-imides).
Macromolecules 28:21842193 (1995).
245 Wang YD, Ameer GA, Sheppard BJ and Langer R, A tough biodegradable elastomer. Nat Biotechnol 20:602606 (2002).
246 Vert M, Li SM, Spenlehauer G and Guerin P, Bioresorbability and
biocompatibility of aliphatic polyesters. J Mater Sci - Mater M
3:432446 (1992).
247 Aydin HM, Kourush S, Rzayev Z, Piskin E, Microwave-assisted
Rapid Synthesis of Poly(glycerol-sebacate) Elastomers. Biomat
Sci 1:503509 (2013).
248 Aydin HM, Kourush S, Yilmaz M, Turk M, Rzayev Z, Piskin E,
The Catalyst Assisted Two Stage Synthesis of Poly(glycerolco-sebacate-co-e-caprolactone) Elastomers As Potential Tissue
Engineering Materials. J Tiss Eng Reg DOI: 10.1002/term.1759
(2012).
249 Kemppainen JM and Hollister SJ, Tailoring the mechanical properties of 3D-designed poly(glycerol sebacate) scaolds for cartilage
applications. J Biomed Mater Res A 94A:918 (2010).
250 Chen QZ, Bismarck A, Hansen U, Junaid S, Tran MQ, Harding SE,
et al., Characterisation of a soft elastomer poly(glycerol sebacate)
designed to match the mechanical properties of myocardial tissue.
Biomaterials 29:4757 (2008).
251 Liu QY, Tian M, Shi R, Zhang LQ, Chen DF and Tian W, Structure
and properties of thermoplastic poly(glycerol sebacate) elastomers
originating from prepolymers with dierent molecular weights.
J Appl Polym Sci 104:11311137 (2007).
252 Ifkovits JL, Padera RF and Burdick JA, Biodegradable and radically
polymerized elastomers with enhanced processing capabilities.
Biomed Mater 3:034104 (2008).
253 Cai W and Liu LL, Shape-memory eect of poly (glycerol-sebacate)
elastomer. Mater Lett 62:21712173 (2008).
254 Jaafar IH, Ammar MM, Jedlicka SS, Pearson RA and Coulter JP,
Spectroscopic evaluation, thermal, and thermomechanical
characterization of poly(glycerol-sebacate) with variations in
curing temperatures and durations. J Mater Sci 45:25252529
(2010).
255 Komatsu K and Chiba M, The eect of velocity of loading on the
biomechanical responses of the periodontal-ligament in transverse sections of the rat molar in vitro. Arch Oral Biol 38:369375
(1993).
256 Sundback CA, Shyu JY, Wang YD, Faquin WC, Langer RS, Vacanti JP,
et al., Biocompatibility analysis of poly(glycerol sebacate) as a nerve
guide material. Biomaterials 26:54545464 (2005).
257 Crapo PM and Wang YD, Physiologic compliance in engineered
small-diameter arterial constructs based on an elastomeric substrate. Biomaterials 31:16261635 (2010).
258 Gao J, Ensley AE, Nerem RM and Wang YD, Poly(glycerol sebacate)
supports the proliferation and phenotypic protein expression of primary baboon vascular cells. J Biomed Mater Res A
83A:10701075 (2007).
259 Chen QZ, Jin LY, Cook WD, Mohn D, Lagerqvist EL, Elliott DA, et al.,
Elastomeric nanocomposites as cell delivery vehicles and cardiac
support devices. Soft Matter. 6:47154726 (2010).
260 Sales VL, Engelmayr GC, Johnson JA, Gao J, Wang YD, Sacks MS,
et al., Protein precoating of elastomeric tissue-engineering scaffolds increased cellularity, enhanced extracellular matrix protein
production, and dierentially regulated the phenotypes of circulating endothelial progenitor cells. Circulation 116:I55I63 (2007).
261 Chen QZ, Liang SL, Wang J and Simon GP, Manipulation of mechanical compliance of elastomeric PGS by incorporation of halloysite
nanotubes for soft tissue engineering applications. J Mech Behav
Biomed 4:18051818 (2011).
262 Sant S, Hwang CM, Lee SH and Khademhosseini A, Hybrid PGS-PCL
microbrous scaolds with improved mechanical and biological
properties. J Tissue Eng Regen M 5:283291 (2011).

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

Polymers for medical and tissue engineering applications

J Chem Technol Biotechnol 2014; 89: 17931810

285 Burdick JA and Anseth KS, Photoencapsulation of osteoblasts in


injectable RGD-modied PEG hydrogels for bone tissue engineering. Biomaterials 23:43154323 (2002).
286 Papadopoulos A, Bichara DA, Zhao X, Ibusuki S, Randolph MA, Anseth
KS, et al., Injectable and photopolymerizable tissue-engineered
auricular cartilage using poly(ethylene glycol) dimethacrylate
copolymer hydrogels. Tissue Eng Pt A 17:161169 (2011).
287 Elissee J, Anseth K, Sims D, McIntosh W, Randolph M, Yaremchuk M, et al., Transdermal photopolymerization of poly(ethylene
oxide)-based injectable hydrogels for tissue-engineered cartilage.
Plast Reconstr Surg 104:10141022 (1999).
288 Burdick JA, Mason MN, Hinman AD, Thorne K and Anseth KS, Delivery
of osteoinductive growth factors from degradable PEG hydrogels
inuences osteoblast dierentiation and mineralization. J Control
Release 83:5363 (2002).
289 Elissee J, McIntosh W, Fu K, Blunk T and Langer R, Controlled-release
of IGF-I and TGF-beta 1 in a photopolymerizing hydrogel for cartilage tissue engineering. J Orthopaed Res 19:10981104 (2001).
290 Williams CG, Kim TK, Taboas A, Malik A, Manson P and Elissee J,
In vitro chondrogenesis of bone marrow-derived mesenchymal
stem cells in a photopolymerizing hydrogel. Tissue Eng 9:679688
(2003).
291 Cruise GM, Hegre OD, Scharp DS and Hubbell JA, A sensitivity study
of the key parameters in the interfacial photopolymerization of
poly(ethylene glycol) diacrylate upon porcine islets. Biotechnol
Bioeng 57:655665 (1998).
292 Yang F, Williams CG, Wang DA, Lee H, Manson PN and Elissee J, The
eect of incorporating RGD adhesive peptide in polyethylene glycol diacrylate hydrogel on osteogenesis of bone marrow stromal
cells. Biomaterials 26:59915998 (2005).
293 Varghese S, Hwang NS, Canver AC, Theprungsirikul P, Lin DW and
Elissee J, Chondroitin sulfate based niches for chondrogenic
dierentiation of mesenchymal stem cells. Matrix Biol 27:1221
(2008).
294 Sharma B, Williams CG, Khan M, Manson P and Elissee JH, In vivo
chondrogenesis of mesenchymal stem cells in a photopolymerized
hydrogel. Plast Reconstr Surg 119:112120 (2007).
295 Hwang NS, Varghese S, Theprungsirikul P, Canver A and Elissee J,
Enhanced chondrogenic dierentiation of murine embryonic stem
cells in hydrogels with glucosamine. Biomaterials 27:6015-6023
(2006).
296 Shin H, Jo S and Mikos AG, Biomimetic materials for tissue engineering. Biomaterials 24:43534364 (2003).
297 Lepoittevin B, Devalckenaere M, Pantoustier N, Alexandre M, Kubies D,
Calberg C, et al., Poly(epsilon-caprolactone)/clay nanocomposites
prepared by melt intercalation: mechanical, thermal and rheological properties. Polymer 43:40174023 (2002).
298 Koegler WS and Grith LG, Osteoblast response to PLGA tissue
engineering scaolds with PEO modied surface chemistries
and demonstration of patterned cell response. Biomaterials 25:
28192830 (2004).
299 Li G, Gill TJ, DeFrate LE, Zayontz S, Glatt V and Zarins B, Biomechanical
consequences of PCL deciency in the knee under simulated
muscle loads - an in vitro experimental study. J Orthopaed Res
20:887892 (2002).
300 Kang XH, Xie YB, Powell HM, Lee LJ, Belury MA, Lannutti JJ, et al., Adipogenesis of murine embryonic stem cells in a three-dimensional
culture system using electrospun polymer scaolds. Biomaterials
28:450458 (2007).
301 Grith LG, Polymeric biomaterials. Acta Mater 48:263277 (2000).
302 Shi XF, Hudson JL, Spicer PP, Tour JM, Krishnamoorti R and Mikos
AG, Rheological behaviour and mechanical characterization of
injectable poly(propylene fumarate)/single-walled carbon nanotube composites for bone tissue engineering. Nanotechnology
16:S531S538 (2005).
303 Sitharaman B, Shi XF, Tran LA, Spicer PP, Rusakova I, Wilson LJ, et al.,
Injectable in situ cross-linkable nanocomposites of biodegradable
polymers and carbon nanostructures for bone tissue engineering.
J Biomat Sci-Polym E 18:655671 (2007).
304 Temeno JS and Mikos AG, Injectable biodegradable materials for
orthopedic tissue engineering. Biomaterials 21:24052412 (2000).
305 Gopferich A and Tessmar J, Polyanhydride degradation and erosion.
Adv Drug Deliver Rev 54:911931 (2002).
306 Zhang Q, Su K, Chan-Park MB, Wu H, Wang D and Xu R, Development
of high refractive ZnS/PVP/PDMAA hydrogel nanocomposites for
articial cornea implants. Acta Biomater 10:11671176 (2014).

2014 Society of Chemical Industry

wileyonlinelibrary.com/jctb

1809

263 Laurencin CT, Koh HJ, Neenan TX, Allcock HR and Langer R, Controlled
release using a new bioerodible polyphosphazene matrix system.
J Biomed Mater Res 21:12311246 (1987).
264 Qiu LY and Zhu KJ, Novel biodegradable polyphosphazenes containing glycine ethyl ester and benzyl ester of amino acethydroxamic
acid as cosubstituents: syntheses, characterization, and degradation properties. J Appl Polym Sci 77:29872995 (2000).
265 Behravesh E, Yasko AW, Engel PS and Mikos AG, Synthetic biodegradable polymers for orthopaedic applications. Clin Orthop Relat R
367:S118S129 (1999).
266 Allcock HR, Inorganic-organic polymers as a route to biodegradable
materials. Macromol Symp 144:3346 (1999).
267 Lakshmi S, Katti DS and Laurencin CT, Biodegradable polyphosphazenes for drug delivery applications. Adv Drug Deliver Rev
55:467482 (2003).
268 Weikel AL, Owens SG, Morozowich NL, Deng M, Nair LS, Laurencin
CT, et al., Miscibility of choline-substituted polyphosphazenes with
PLGA and osteoblast activity on resulting blends. Biomaterials
31:85078515 (2010).
269 Singh A, Krogman NR, Sethuraman S, Nair LS, Sturgeon JL, Brown
PW, et al., Eect of side group chemistry on the properties
of biodegradable L-alanine cosubstituted polyphosphazenes.
Biomacromolecules. 7:914918 (2006).
270 Verret V, Wassef M, Pelage JP, Ghegediban SH, Jouneau L, Moine
L, et al., Inuence of degradation on inammatory prole of
polyphosphazene coated PMMA and trisacryl gelatin microspheres in a sheep uterine artery embolization model. Biomaterials
32:339351 (2011).
271 Stamp U, Sommer CM, Thierjung H, Stamp S, Lopez-Benitez R,
Radele B, et al. Reduction of Late In-Stent Stenosis in a Porcine
Coronary Artery Model by Cobalt Chromium Stents with a
Nanocoat of Polyphosphazene (Polyzene-F). Cardiovasc Inter Rad
31:11841192 (2008).
272 Conconi MT, Lora S, Baiguera S, Boscolo E, Folin M, Scienza R, et al.,
In vitro culture of rat neuromicrovascular endothelial cells on polymeric scaolds. J Biomed Mater Res A 71A:669674 (2004).
273 Zhang QS, Yan YH, Li SP and Feng T, The synthesis and characterization of a novel biodegradable and electroactive polyphosphazene
for nerve regeneration. Mat Sci Eng C - Mater 30:160166 (2010).
274 Deng M, Nair LS, Nukavarapu SP, Kumbar SG, Jiang T, Krogman NR,
et al., Miscibility and in vitro osteocompatibility of biodegradable
blends of poly[(ethyl alanato) (p-phenyl phenoxy) phosphazene]
and poly(lactic acid-glycolic acid). Biomaterials 29:337349 (2008).
275 Nair LS, Bhattacharyya S, Bender JD, Greish YE, Brown PW, Allcock
HR, et al., Fabrication and optimization of methylphenoxy substituted polyphosphazene nanobers for biomedical applications.
Biomacromolecules 5:22122220 (2004).
276 Potta T, Chun C and Song SC, Chemically crosslinkable thermosensitive polyphosphazene gels as injectable materials for biomedical
applications. Biomaterials 30:61786192 (2009).
277 Park MR, Chun CJ, Ahn SW, Ki MH, Cho CS and Song SC, Sustained delivery of human growth hormone using a polyelectrolyte complex-loaded thermosensitive polyphosphazene
hydrogel. J Control Release 147:359367 (2010).
278 Qiu ZC, Zhang JJ, Zhou Y, Song BY, Chang JJ, Yang KK, et al.,
Biodegradable poly(p-dioxanone) reinforced and toughened by
organo-modied vermiculite. Polym Advan Technol 22:9931000
(2011).
279 Malnati GA and Stone EA, Clinical-experience with polydioxanone
suture material. Vet Surg 12:2425 (1983).
280 Elissee J, McIntosh W, Anseth K, Riley S, Ragan P and Langer R, Photoencapsulation of chondrocytes in poly(ethylene oxide)-based
semi-interpenetrating networks. J Biomed Mater Res 51:164171
(2000).
281 Alcantar NA, Aydil ES and Israelachvili JN, Polyethylene glycol-coated
biocompatible surfaces. J Biomed Mater Res 51:343351 (2000).
282 Gombotz WR, Guanghui W, Horbett TA and Homan AS, Protein
adsorption to poly(ethylene oxide) surfaces. J Biomed Mater Res
25:15471562 (1991).
283 Ostuni E, Chapman RG, Holmlin RE, Takayama S and Whitesides GM,
A survey of structure-property relationships of surfaces that resist
the adsorption of protein. Langmuir 17:56055620 (2001).
284 Zhu JM, Bioactive modication of poly(ethylene glycol) hydrogels for
tissue engineering. Biomaterials 31:46394656 (2010).

www.soci.org

www.soci.org
307 Tran RT, Choy WM, Cao H, Qattan I, Chiao JC, Ip WY, et al., Fabrication and characterization of biomimetic multichanneled
crosslinked-urethane-doped polyester tissue engineered nerve
guides. J Biomed Mater Res A 102:27932804 (2014).
308 Sharma B, Fermanian S, Gibson M, Unterman S, Herzka DA, Cascio
B, et al., Human cartilage repair with a photoreactive adhesivehydrogel composite. Sci Translational Med 5:167ra6 (2013).
309 Malafaya PB, Silva GA and Reis RL, Natural-origin polymers as carriers
and scaolds for biomolecules and cell delivery in tissue engineering applications. Adv Drug Deliver Rev 59:207233 (2007).
310 Lee KY and Mooney DJ, Hydrogels for tissue engineering. Chem Rev
101:18691879 (2001).
311 Jhon MS and Andrade JD, Water and Hydrogels. J Biomed Mater Res
7:509522 (1973).
312 Ward JH and Peppas NA, Kinetic gelation modeling of controlled
radical polymerizations. Macromolecules 33:51375142 (2000).
313 Jeon O, Bouhadir KH, Mansour JM and Alsberg E, Photocrosslinked
alginate hydrogels with tunable biodegradation rates and mechanical properties. Biomaterials 30:27242734 (2009).
314 Chung C, Mesa J, Miller GJ, Randolph MA, Gill TJ and Burdick JA,
Eects of auricular chondrocyte expansion on neocartilage formation in photocrosslinked hyaluronic acid networks. Tissue Eng
12:26652673 (2006).
315 Almany L and Seliktar D, Biosynthetic hydrogel scaolds made from
brinogen and polyethylene glycol for 3D cell cultures. Biomaterials
26:24672477 (2005).
316 Elbert DL and Hubbell JA, Reduction of brous adhesion formation
by a copolymer possessing an anity for anionic surfaces. J Biomed
Mater Res 42:5565 (1998).
317 Stahl PJ, Romano NH, Wirtz D and Yu SM, PEG-based hydrogels
with collagen mimetic peptide-mediated and tunable physical
cross-links. Biomacromolecules 11:23362344 (2010).
318 Melrose J, Chuang C and Whitelock J, Tissue engineering log
cartilages using biomatrices. J Chem Technol Biot 83:444463
(2008).
319 Balakrishnan B and Banerjee R, Biopolymer-based hydrogels for cartilage tissue engineering. Chem Rev 111:44534474 (2011).
320 Ng KW, Ateshian GA and Hung CT, Zonal chondrocytes seeded
in a layered agarose hydrogel create engineered cartilage with
depth-dependent cellular and mechanical inhomogeneity. Tissue
Eng Pt A 15:23152324 (2009).
321 Lu HH, Subramony SD, Boushell MK and Zhang XZ, Tissue engineering strategies for the regeneration of orthopedic interfaces. Ann
Biomed Eng 38:21422154 (2010).
322 Chen JN, Chen HA, Li P, Diao HJ, Zhu SY, Dong L, et al., Simultaneous
regeneration of articular cartilage and subchondral bone in vivo
using MSCs induced by a spatially controlled gene delivery system in bilayered integrated scaolds. Biomaterials 32:47934805
(2011).
323 Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A and Peppas
NA, Hydrogels in regenerative medicine. Adv Mater 21:33073329
(2009).
324 Park H, Temeno JS, Holland TA, Tabata Y, Mikos AG. Delivery of
TGF-beta 1 and chondrocytes via injectable, biodegradable hydrogels for cartilage tissue engineering applications. Biomaterials
26:70957103 (2005).
325 Bryant SJ and Anseth KS, The eects of scaold thickness on tissue engineered cartilage in photocrosslinked poly(ethylene oxide)
hydrogels. Biomaterials 22:619626 (2001).
326 Omidian H, Rocca JG and Park K, Advances in superporous hydrogels.
J Control Release 102:312 (2005).

D Ozdil, HM Aydin

327 Bryant SJ, Anseth KS, Lee DA and Bader DL, Crosslinking density
inuences the morphology of chondrocytes photoencapsulated
in PEG hydrogels during the application of compressive strain.
J Orthopaed Res 22:11431149 (2004).
328 Xia F, Ge H, Hou Y, Sun TL, Chen L, Zhang GZ, et al., Multiresponsive surfaces change between superhydrophilicity and superhydrophobicity. Adv Mater 19:2520 (2007).
329 Peng T and Cheng YL, Temperature-responsive permeability of
porous PNIPAAm-g-PE membranes. J Appl Polym Sci 70:21332142
(1998).
330 Lue SJ, Hsu JJ, Chen CH and Chen BC, Thermally on-o switching membranes of poly(N-isopropylacrylamide) immobilized in
track-etched polycarbonate lms. J Membrane Sci 301:142150
(2007).
331 Kopecek J and Yang JY, Revie - hydrogels as smart biomaterials. Polym
Int 56:10781098 (2007).
332 Nath N and Chilkoti A, Creating Smart surfaces using stimuli responsive polymers. Adv Mater 14:1243 (2002).
333 Jeong B, Kim SW and Bae YH, Thermosensitive solgel reversible
hydrogels. Adv Drug Deliver Rev 54:3751 (2002).
334 Joglekar M and Trewyn BG, Polymer-based stimuli-responsive
nanosystems for biomedical applications. Biotechnol J 8:931945
(2013).
335 Ju C, Sun J, Zi P, Jin X and Zhang C, Thermosensitive micelles-hydrogel
hybrid system based on poloxamer 407 for localized delivery of
paclitaxel. J Pharm Sci 102:27072717 (2013).
336 Fleige E, Quadir MA and Haag R, Stimuli-responsive polymeric
nanocarriers for the controlled transport of active compounds:
concepts and applications. Adv Drug Deliv Rev 64:866884
(2012).
337 Schild HG, Poly (N-Isopropylacrylamide) - experiment, theory and
application. Prog Polym Sci 17:163249 (1992).
338 Srivastava A and Kumar A, Thermoresponsive poly(Nvinylcaprolactam) cryogels: synthesis and its biophysical evaluation for tissue engineering applications. J Mater Sci Mater Med 21:
29372945 (2010).
339 Ivanov AE, Galaev IY, Kazakov SV and Mattiasson B, Thermosensitive copolymers of N-vinylimidazole as displacers of proteins in
immobilised metal anity chromatography. J Chromatogr A 907:
115130 (2001).
340 Weber CH, R; Schubert, US. Temperature responsive bio-compatible
polymers based on poly(ethylene oxide) and poly(2-oxazoline)s.
Prog Polym Sci 37:686714 (2012).
341 Niu G, Djaoui AB and Cohn D, Crosslinkable PEO-PPO-PEO triblocks
as building blocks of thermo-responsive nanoshells. Polym Int
52:25242530 (2011).
342 Araujo JV, Davidenko N, Danner M, Cameron RE and Best SM, Novel
porous scaolds of pH responsive chitosan/carrageenan-based
polyelectrolyte complexes for tissue engineering. J Biomed Mater
Res A (2014).
343 Adrian Gestos PGW, Spinks GM and Wallace GG, Crosslinking neat
ultrathin lms and nanobres of pH-responsive poly(acrylic acid)
by UV radiation. Soft Matter 6:10451052 (2010).
344 Kyriakides TR, Cheung CY, Murthy N, Bornstein P, Stayton PS and
Homan AS, pH-sensitive polymers that enhance intracellular drug
delivery in vivo. J Control Release 78:295303
345 Donini C, Robinson DN, Colombo P, Giordano F and Peppas NA,
Preparation of poly(methacrylic acid-g-poly(ethylene glycol))
nanospheres from methacrylic monomers for pharmaceutical
applications. Int J Pharm 245:8391 (2002).

1810
wileyonlinelibrary.com/jctb

2014 Society of Chemical Industry

J Chem Technol Biotechnol 2014; 89: 17931810

You might also like