ozdil2014

18
1793 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 Ozdil a and Halil Murat Aydin a,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; scaffolds; tissue engineering INTRODUCTION Regenerative medicine has seen great advancements over the past decade with progress in polymer science offering new materi- als and strategies for the fields of tissue engineering, drug delivery, and cosmetic surgery. For tissue engineering research, the primary focus has been on developing substitutes for the natural extra- cellular matrix (ECM) – scaffolds capable of acting as temporary three-dimensional support structures that promote and guide cor- rect 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-specific use and the biodegradable nature of most of them. As poly- meric scaffolds are generally subjected to in vivo conditions they are also generally required to possess good cell adhesive- ness, promote cell–biomaterial interactions, exhibit structural and mechanical integrity, and have a microstructure suitable for tissue integration and the exchange of factors critical to cell survival. Scaffold design parameters and manufacturing methods for multi-faceted polymer systems ultimately rely on the funda- mental chemical nature of the polymer. For example, while collagen is degradable and highly suitable as temporary scaffold- ing 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 biomed- ical applications can be separated into two broad categories: naturally-occurring polymers and synthetic polymers. As biomed- ical 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. NATURALLY-OCCURRING POLYMERS The first polymers to be used in biomedical applications, naturally-occurring polymers, 1 far out-date the use of syn- thetic polymers which first appeared around the 1960s. Frequently-utilized, naturally-occurring polymers can be divided into the following three groups: proteins (e.g. silk, collagen, soy, fibrin 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 scaffold–tissue compatibility due to the positive bio- logical 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. Hyaluronic acid A ubiquitous proteoglycan, and one that is often incorporated into tissue regenerative scaffolds, is the linear, anionic polymer, hyaluronic acid. This co-polymer of D-glucuronic acid and N- acetyl-D-glucosamine can be found in several body tissues includ- ing connective, epithelial and neural tissue and synovial fluid. 3 Correspondence to: Halil Murat Aydin, Environmental Engineering Department and Bioengineering Division, Hacettepe University, 06800, Ankara, Turkey. E-mail: [email protected] 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 J Chem Technol Biotechnol 2014; 89: 1793–1810 www.soci.org © 2014 Society of Chemical Industry

Upload: princesschemist

Post on 12-Jan-2016

6 views

Category:

Documents


2 download

DESCRIPTION

k

TRANSCRIPT

Page 1: ozdil2014

1793

ReviewReceived: 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 engineeringapplicationsDeniz Ozdila and Halil Murat Aydina,b*

Abstract

Recent decades have seen great advancements in medical research into materials, both natural and synthetic, that facilitate therepair and regeneration of compromised tissues through the delivery and support of cells and/or biomolecules. Biocompatiblepolymeric materials have become the most heavily investigated materials used for such purposes. Naturally-occurring andsynthetic 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 biomaterialresearch combined with the developments in manufacturing techniques have expanded capabilities in tissue engineeringand other medical applications of these materials. This review will present an overview of the major classes of polymericbiomaterials, highlight their key properties, advantages, limitations and discuss their applications.© 2014 Society of Chemical Industry

Keywords: polymers; tissues and organs; scaffolds; tissue engineering

INTRODUCTIONRegenerative medicine has seen great advancements over thepast decade with progress in polymer science offering new materi-als and strategies for the fields of tissue engineering, drug delivery,and cosmetic surgery. For tissue engineering research, the primaryfocus has been on developing substitutes for the natural extra-cellular matrix (ECM) – scaffolds capable of acting as temporarythree-dimensional support structures that promote and guide cor-rect tissue pattern formation. Polymers, both naturally-occurringand synthetic, have become the natural choice of biomaterialfor such applications for a number of reasons including theiravailability as a resource and ability to be easily reproduced,their versatility in the range of applications for which they can beadapted, their generally tuneable properties for function-specificuse and the biodegradable nature of most of them. As poly-meric scaffolds are generally subjected to in vivo conditionsthey are also generally required to possess good cell adhesive-ness, promote cell–biomaterial interactions, exhibit structuraland mechanical integrity, and have a microstructure suitablefor tissue integration and the exchange of factors critical to cellsurvival.

Scaffold design parameters and manufacturing methods formulti-faceted polymer systems ultimately rely on the funda-mental chemical nature of the polymer. For example, whilecollagen is degradable and highly suitable as temporary scaffold-ing material for repair at defect sites, the mechanical strengthand non-degradability of polyurethanes are sought for loadbearing applications. Polymers used as biomaterials in biomed-ical applications can be separated into two broad categories:naturally-occurring polymers and synthetic polymers. As biomed-ical polymers are usually combined with other materials formaximized performance and targeted tissue response, thisreview will discuss each type of polymer separately under theaforementioned categories.

NATURALLY-OCCURRING POLYMERSThe first polymers to be used in biomedical applications,naturally-occurring polymers,1 far out-date the use of syn-thetic polymers which first appeared around the 1960s.Frequently-utilized, naturally-occurring polymers can be dividedinto the following three groups: proteins (e.g. silk, collagen, soy,fibrin gels), polysaccharides (e.g. chitin/chitosan, alginate andhyaluronic acid derivatives) and polynucleotides (e.g. DNA andRNA).2 The main advantage of natural polymers is their highdegree of scaffold–tissue compatibility due to the positive bio-logical recognition of their chemical make-up. However, the use ofthese polymers also carries the potential for inciting an immuneresponse due to any impurities in the material gained duringprocessing. Clinical applications require the use of medical gradematerials and, in any instances where immunogenic fragmentsare detected within the material, it is imperative that they areremoved.

Hyaluronic acidA ubiquitous proteoglycan, and one that is often incorporatedinto tissue regenerative scaffolds, is the linear, anionic polymer,hyaluronic acid. This co-polymer of D-glucuronic acid and N-acetyl-D-glucosamine can be found in several body tissues includ-ing connective, epithelial and neural tissue and synovial fluid.3

∗ Correspondence to: Halil Murat Aydin, Environmental Engineering Departmentand Bioengineering Division, Hacettepe University, 06800, Ankara, Turkey.E-mail: [email protected]

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

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

J Chem Technol Biotechnol 2014; 89: 1793–1810 www.soci.org © 2014 Society of Chemical Industry

Page 2: ozdil2014

1794

www.soci.org D Ozdil, HM Aydin

Hyaluronic acid not only has a key role in ensuring joint lubricationbut is also known to promote cell motility and proliferation whichare properties critical for tissue regeneration. These features,alongside the enzymatic degradability, bioresorbability and theability to add further functionality via its side chains has led to thedevelopment of hyaluronic-based scaffolds of all forms includinghydrogels for wound healing applications,4 sponges for cartilagerepair,5 and meshes for bone tissue engineering.6 It is perhaps themore specific features of hyaluronic acid such as its ability to orga-nize and retain aggrecan and sulphated glycosaminoglycans,7

its ability to adhere chondrocytes through cell surface receptorswhich then leads to the production of signals for cell migration,proliferation and differentiation,8,9 and its ability to stimulate pro-teoglycan synthesis10,11 that has made it so popular in cartilagetissue.

Hyaluronic acid is also known to take part in the stimulation ofa local inflammatory response which is particularly useful for theconduction of local tissue repair and remodelling.12 In addition,the angiogenic effects13 of hyaluronic acid have been documentedand add further appeal to the natural biomaterial as local angio-genesis is considered critical for tissue formation and survival. Inrecent years chemical modifications to hyaluronic acid have beenexplored to chemically enhance its physical qualities, rheologicalqualities and tissue–material interactive qualities. Such upgradeswill continue to inspire future research into the use hyaluronic acidin tissue engineering applications.

CollagenCollagens are extra cellular matrix proteins. Type I collagen, afibrillar, rod-shaped molecule,14 is the most abundant of the 27different types of collagen. Type I collagen can be purified froma number of tissues including tendon, ligament, bone, skin andcornea via relatively simple biochemical processing methods.15

The high content of collagen Type I in the extracellular matrix hasbeen a key reason why it has become one of the most investigatedproteins for use in medical applications.16 As it normally providesthe structural framework and tensile strength in body tissues17 itis considered an excellent biological resource for the constructionof tissue engineering scaffolds.

The mechanical performance of collagen relies heavily onthe fundamental crystalline assembly of the protein which hasa helical quaternary structure.18 At the nanoscale, the helicalself-assembly of tropocollagen subunits form the collagen fib-rils that are cross-linked at the microscale to give the ECM andtissues its tensile strength. The interactions of these fibrils withother proteins and extracellular matrix components also play arole in defining the engineered tissue matrix. At this level, fibrildiameter, length, density and orientation can all be modified toenhance the biological and mechanical activity of the scaffold.The covalent bonding between fibrils to form larger fibrils or fibrebundles can be attained through various fabrication methods,including electro-spinning modules and moulding. The appli-cation of mechanical stimulation such as tensile strain19 andcyclic circumferential strain20,21 on collagen Type I-based scaffoldshave proven to assist with the control of collagen deposition bythe recruited cells and therefore serves as a tool for directing thestructural formation of the scaffold.

Collagen carries the advantage of having simulative chemicalcharacteristics, such as integrin receptors, required for adequatecell attachment and proper tissue formation.22,23 Furthermore, thetypes of cells that are attracted to and attach to this biomaterialnot only secrete specific enzymes that degrade the collagen

without creating cytotoxic degradation products, but they arealso the chief synthesizers of new collagen that is deposited in tothe extracellular space. This gives collagen scaffolds behaviouralcharacteristics that are ideal and capable of closely following thebone remodelling process that governs bone tissue engineeringapplications. Collagen can be forged to exhibit the physical andmorphological features required for local connective tissue regen-eration, combined with other materials (for example 𝛽-tricalciumphosphate for bone tissue engineering) to form composites andprepared in many different physical forms (beads, membranes,shaped objects, etc.).24,25 Currently, the main issue with collagenuse from a regulatory perspective is the challenges that are asso-ciated with ensuring the safety with using such animal-derivedmaterials (in particular the risk of transmitting prions and BSE).

Co-polymerization capabilities with other biomaterials raiseeven more options for customizing scaffold properties.

Chitin/ChitosanThe second most abundant polymer in nature is chitosan, a lin-ear cationic polysaccharide that is attained from the deacety-lation of chitin (a linear polysaccharide built from 𝛽-1,4 linkedN-acetylglucosamine units). Owing to the insolubility of chitin incommon solvents and the difficulties this poses for processing,deacetylation is necessitated and chitosan becomes the usableproduct. Chitosan is a promising degradable polymer that is oneof the only two non-human origin natural polymers (along withalginic acid) used in biomedical engineering. Its base unit, chitin,has characteristics, such as wound healing capacity,26 that are par-ticularly favourable in tissue engineering cases. The crystallinityand molecular weight of the chitosan polymer depends on thedegree of deacetylation.27 The degradation of chitosan is alsoprimarily influenced by the degree of acetylation.28 The alterna-tive method for controlling degradation profiles is by altering theside-groups of the polymer such that extensive hydrogen bond-ing is prevented via the attachment of bulky side groups.29 In vitrodegradation of chitosan is performed by several enzymes includ-ing chitosanase, lysozyme and papain30 with lysozyme being thephysiologically relevant enzyme for in vivo degradation.

The poor mechanical strength of chitosan and its high degreeof hydrophilicity has traditionally been resolved by cross-linkingit 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 withother polymers include semi- or full-interpenetrating polymernetworks, chitosan–chitosan networks and hybrid polymernetworks.37,38 The structural stability of chitosan-based scaffoldscan thus be varied through these cross-links.

This cationic polymer is able to form electrostatic interactionswith negatively charged cell surfaces and, depending on its con-figuration, it can also display great hydrophilicity and cell prolifer-ation effects.39,40 The ability of this bioactive polymer to interactwith glycosaminoglycans (GAGs) in particular give scaffolds basedon chitosan a direct influence on the modulation of cytokinesand growth factors41 and thus local tissue regeneration activities.Furthermore, chitosan–DNA complexes have also been exploredas a means of facilitating the regeneration process via scaffoldswith amplified bioactivity at the molecular level.42

The biodegradability, biocompatibility, bio-adhesiveness, andnon-toxicity of chitosan41,43 account for the popularity of thisbiomaterial in medicine. This is topped by the possibility ofprocessing chitosan and chitosan-composites into numerous

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 3: ozdil2014

1795

Polymers for medical and tissue engineering applications www.soci.org

shapes; membranes, sponges, fibres, microspheres and hydro-gels. Some recent research into chitosan-based scaffolds areaimed at further improving surface characteristics of scaffolds forhard tissue regeneration,44 exploring its anti-bacterial qualitieswhen combined with other polymers and anti-bacterial agents,45

increasing stability of chitosan-based cartilage repair scaffoldsby co-polymerization with other natural polymers,46 and intro-ducing chitosan microspheres embedded in calcium sulphate aspoint-of-care drug delivery systems.47 Bovine derived collagen,much like the innovative uptake of fungi-derived chitosan, can beused as an example by which solutions for the abovementionedissues can be found, paving the way for the safe and effectivefuture use of this material.

AlginateThe other non-human derived natural polymer used to fab-ricate tissue engineering scaffolds is the linear copolymer of𝛽-D-manuronic acid and 𝛼-L-glucuronic acid linked by a 1–4 gly-cosidic bond, alginate. Commonly sourced from the cell wall ofbrown algae, extraction of the polysaccharide is through the useof a basic solution followed by acidic precipitation. The molecularweight of the polysaccharide can reach up to 500 kDa.

The simplicity of creating alginate hydrogels with the poly-mer undergoing spontaneous gelation in the presence of diva-lent cations and carboxylate side groups makes it an attractiveand promising biomaterial with tissue engineering applicationpotential. Traditionally, along with drug delivery and cell deliveryscaffolds,48 alginate has also been used in wound dressings.

Most research and development in hydrogel based systemshave been centred on composite constructs that incorporatedifferent materials such as collagen49,50 PLGA,51,52 Poly-L-lysine(PLL),53,54 PCL,55 polyethers,56 and chitosan,57,58 mainly for supportof alginate’s mechanical weakness. Scaffold designs are as variedas the types of body tissues that are aimed to be restored fromgels,59,60,50 porous networks,61,62 and sponges63 to films64 andmicrospheres.65,49,54,53 Along with its design flexibility is the abilityto transform the responsiveness of alginate biomaterials towardpH and cell adhesion through alterations to its carboxylate acidside group.66,67

Among these advantages, however, stand alginate’s natural poorcell adhesion and poor in vivo degradation performance. Thisnecessitates either the gamma irradiation or periodates oxidationof alginate biomaterials. Several types of alginate-based scaffoldshave been used in tissue regeneration attempts of different bodytissues including bone and cartilage,61,59,63,60 nerve,54 and connec-tive tissue.50

FibrinFibrinogen, the building block of fibrin, is a large (340 kD) glycopro-tein that has three pairs of polypeptide chains, namely A𝛼, B𝛽 , and𝛾 which are bound by disulphide bridges. A unique polymerisa-tion process involving thrombin and activated factor XIII convertsfibrinogen into fibrin and forms a fibrin network. This polypeptideis commonly known for its physiological role as a haemostaticplug in tissue injuries. The options for cross-linking are one wayin which diverse microstructural and mechanical features of fibrinnetworks can be achieved. In addition, autologous scaffolds canbe manufactured by using fibrin made from a patient’s own blood.

Injectable fibrin hydrogels are the most frequently utilized formsof the polypeptide as tissue regeneration support biomaterial68

Fibrin can adhere cells both directly via integrin receptors and

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

The polymer also binds growth factors that further promote cellproliferation and differentiation.71 Furthermore, the angiogeniceffects of fibrin-based materials have also been established.72 Thisbioactivity gives fibrin significant appeal and use in medicine assealants or heamostatic agents, which are perhaps the more com-monly known applications of the biopolymer. However, fibrin hasalso shown success as a biomaterial with tissue engineering appli-cability. Orthopaedic, cardiovascular and skin tissue engineeringare key examples of areas with fibrin-based scaffold use.73 – 78

The main impediment to the use of fibrin as scaffolding mate-rial is its morphological deconfiguration (shrinkage) and rapiddegradation in physiological conditions.73,74 Fibrin-only cellcarriers79 are weak mechanically and thus require combinationswith strength enhancers, e.g. hyaluronic acid,80 PLGA81 andPCL/polyurethane.82 Like all successful 3D scaffolds for tissueengineering, fibrin-based scaffolds can exhibit interconnectedpores that allow cell infiltration, the exchange of nutrients andwastes and that support vascularization.83 Fibrinogen concen-tration is the fundamental determining factor of pore structurewhere a low concentration creates appropriately dense matricesthat can mediate the aforementioned points critical for scaffoldperformance.84 – 86

AlbuminAlbumin, a protein that contributes 50% to the total mass ofblood plasma has been investigated for use in drug deliveryapplications. The role of this water-soluble protein in the bloodis to carry hydrophobic fatty acids as well as maintain the pHbalance. Since albumin is ubiquitous in the human body, almostall body tissues can enzymatically degrade the protein. This highdegree of biocompatibility and biodegradability gives albuminhigh potential as a biomaterial. Albumin is particularly successfulas a tissue engineering constructs with drug delivery capabilitiesand is usually preferred in studies determining protein and growthfactor release kinetics.

The weak mechanical properties of albumin has narrowed itsuse to drug delivery,87 coatings,88,89 and suturing90,91 applications.The protein can be processed into several different forms includingnanoparticles,87,92 microparticles,93 and fibres.94,95

Chondrotin sulphateChondrotin sulphate (CS) has shown great promise as a biomate-rial. This sulphated glycosaminoglycan has a structure very similarto HA. Found predominantly in articular cartilage, CS plays a rolein the matrices produced by fibroblasts in wound healing.96 It hasbeen shown to have anti-inflammatory effects and is a bindingfactor for extracellular matrix components.97 Chondrotin sulphatecan be commonly encountered particularly in cartilage tissueengineering combined with other cartilage ECM components toform multi-layered scaffold Its natural role in chondrogenic activ-ities has led to its investigation in cartilage tissue engineering98

both alone99 and featuring in composite materials that alsocontain poly(𝜖-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 branchedinto two groups: natural poly(amino acids) and syntheticpoly(amino acids) (discussed later).

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 4: ozdil2014

1796

www.soci.org D Ozdil, HM Aydin

Natural poly(amino acids) are biodegradable and, unlike pro-teins, are composed of only one type of amino acid bonded byamide linkages. Poly(𝛾-glutamic acid) and poly(L-lysine) are themost commonly utilized polymers within the natural poly(aminoacid) category.

Poly(𝛾-glutamic acid) (𝛾-PGA) is a polyamide that is composed ofD- and L-glutamic acid enantomeric units. It is soluble in water andwith a reactive carboxylate group, there exists a wide variety ofoptions for further development with different functional groupattachments. Previously, 𝛾-PGA has been coupled with chemother-apeutic agents,106 antibiotics,107 DNA,108 and proteins107 to aidtissue healing and recovery. As scaffold material, however, the𝛾-PGA homopolymer is not preferred as it does not have adequatephysical properties. It has seen some application in soft tissue engi-neering within cross-linked hydrogels.109 Attempts to equip thispolyamide with characteristics suited to tissue engineering havebeen 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 𝛾-PGA.

Poly(L-lysine) research has revealed antibacterial,115 and anti-tumor activity116 effects that this polymer carries. This particularionic polymer, however, has also been found to have high toxicityowing to its extremely high positive charge and therefore is lim-ited, with in vivo use such as that with tissue engineering scaffolds.Attempts to appropriate it for use as a biomaterial have beenthrough blending it with 𝛾-PGA117 and other polymers118 – 120

although poly(L-lysine) remains a blending option that stillrequires further research and development before clinical uptake.

SYNTHETIC POLYMERS AS BIOMATERIALSSynthetic polymeric biomaterials may not always come as acheaper alternative to biological polymers due to less availability,however, they are much more easily reproducible and have alonger shelf-life and therefore are often more viable sources ofpolymeric biomaterials. Uniformity of microstructure, degrada-tion rate and mechanical structure with large-scale productionare the key advantages of these polymers. The most exten-sively used synthetic biomedical polymers is the family of linearaliphatic polyesters: polyvinyl alcohol (PVA), (PCL), poly(lacticacid), poly(glycolic acid), poly(hydroxybutyrate) (PHB).121 – 124

Numerous combination options of these polymers, both witheach other and/or various other elements, generate a great assort-ment of composite scaffolds with varying chemical, structural andmechanical properties that can be suited to specific tissues andtasks.

Regulatory bodies have presented affirmative guidance docu-ments for the use of synthetic polymeric biomaterials and as such,in terms of product validity, have supported the more efficient uti-lization of such synthetic materials. The future will thus continueto see synthetic polymers as indispensable tools for medical andtissue engineering applications.

Polyetheretherketone (PEEK)One of biomedical engineering’s most frequently utilized poly-mers is polyetheretherketone (PEEK). This linear, aliphatic,31 semicrystalline polymer is a high performance polymer with a highmelting point of 334 ∘C and a high resistance to wear. PEEK hasa maximum tensile strength of 100 MPa, a maximum elongationof 50–150% and a Young’s Modulus of about 3.7 GPa.125 Withits mechanical properties, chemical resistance and radiolucencyclosely matching those of natural bone,126 the popularity of PEEKin orthopaedic biomedical applications can be understood.

The biocompatibility of PEEK and PEEK composites127 hasbeen confirmed through numerous in vitro cytotoxicity128 – 130

and mutagenecity131 studies as well as in vivo studies that haveshown that the biomaterial triggers a minimal inflammatoryresponse.132 – 134 Adding to the benefits of using PEEK is its MRIcompatibility. These excellent chemical, mechanical and ther-mal properties make PEEK an obvious choice for load-bearingorthopaedic devices including uses in knee joints,135 spinalfusion,136 and craniofacial repair137 which have been the initialand traditional uses for it. Recent research,138,139 however, isseeking to transform PEEK-based biomaterials so that furtherfunctionality such as stimulation and integration of local tissuecan be achieved with it.

Due to the chemical intertness of PEEK, it has a limited abilityto form strong attachments to native tissues.140 Attempts to over-come this in orthopaedic applications introduced the inclusionof osteoinductive or osteoconductive factors, such as hydroxyap-atite. Such additives, however, have not provided a complete solu-tion as the mechanical strength of the polymeric material is quicklycompromised owing to the additional lack of binding betweenPEEK and reinforcement/filler elements.141 – 143 For the purposes oforthopaedic tissue engineering scaffold design, efforts have beenfocused on developing certain manufacturing methods for PEEKscaffolds such as knitting and braiding,144,145 acid-etching, alkalitreatment, anionic oxidation, shot-blasting,146 and sulfonation147

for some modification of its mechanical properties to producemore flexible, deformable, and matrix-like structures suitable fortissue ingrowth and local mechanics. Tissue anchorage with PEEKmaterials have been largely achieved through interconnectedporous scaffold designs146 that allow tissue integration in thematrix. It has been shown that pore sizes of 200–300 μm141 anda porosity value of 75%148 have been considered successful para-metrical values for attaining sufficient bone tissue fixation.

Poly(hydroxyalkanoates)Polyhyrdroxyalkanoates are another large class of naturally-occurring aliphatic polyesters. In terms of both availabil-ity and suitability to tissue engineering research, how-ever, poly 3-hydroxybutyrate (PHB), 3-hydroxybutyrate and3-hydroxyvalerate (PHBV) co-polymers, poly 4-hydroxybutyrate(P4HB), 3-hydroxybutyrate and 3-hydroxyhexanoate (PHBHHx)co-polymers and poly 3-hydroxyoctanoate (PHO) have been themain members of interest.

The simplest and most widely investigated of this family ofthermoplastics is poly(hydroxybutyrate), which has been exploredmainly in the construction of nerve149 and bone150 tissue engi-neering scaffolds. The biocompatibility of this biomaterial foundnaturally in the body151 is quite high, with its completely non-toxicdegradation products150 that are absorbable via natural metabolicpathways.

The main properties of PHB that limit its use are its brittleness,tendency to acquire a high degree of crystallinity,150 poor stiffnessand hydrophobic character and, compared with similar biomedi-cally utilized polymers, its slow degradation rate.152

The difficulties that these properties pose have been shownto be overcome with various scaffold fabrication methods, sur-face property modification procedures and other techniques. Aprimary example of such attempts includes the functionaliza-tion of PHB matrices by blending it with co-polymers such ashydroxyvalerate (PHV).153 The incorporation of hydroxyapatiteparticles in PHB-based scaffolds have also highlighted the contri-bution of such factors in enhancing the success of these scaffolds

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 5: ozdil2014

1797

Polymers for medical and tissue engineering applications www.soci.org

in tissue engineering approaches by the ability of hydroxyap-atite to not only attract osteoblastic activity but also modifythe microarchitecture for a more porous material. Similarly, theporosity achievable with use of aqueous emulsions have alsobeen shown to positively impact PHB scaffold performance.154

Another example is the use of polyethylene glycol, which has beenshown to enhance the hydrophilic properties of PHB nano-fibrillarscaffolds.155 Polyhydroxyalkonoates will continue to maintainsignificance in the field of medical polymers with the contin-uing progress in recombinant, purification and modificationtechnologies.

Poly(𝜶-hydroxyacids) (Poly(glycolic acid), Poly(lactic acid)and their co-polymers)Perhaps the most heavily researched category of syntheticbiopolymers is the linear aliphatic polyester family which includespoly(lactic acid), poly(glycolic acid), their co-polymers poly(lacticacid-co-glycolic acid), poly(𝜖-caprolactone), poly(propylenefumarate) and poly(hydroxy butyrate) (the last three willbe discussed separately later).156 As the most widely used,FDA-approved materials with long-term clinical data from awide range of clinical indications for which they have been uti-lized, this category of synthetic polymers are usually the first tocome to mind as synthetic biomaterials.

The poly(𝛼-hydroxy acids), poly(glycolic acid) (PGA), poly(lacticacid) (PLA) and their copolymer poly(lactic-co-glycolic acid)(PLGA), are broken down to their monomeric units lactic acid andglycolic acid through hydrolysis of the ester bonds in the back-bone of their chains. These breakdown products are then simplycleared by natural metabolic pathways. In order to maintain thehydrolytic stability of the ester bond, the length of the aliphaticchains must be limited. This is also important for keeping thedegradability characteristics of the polymer.

The highly crystalline thermoplastic, PGA, has impressive proper-ties such as a melting temperature >200 ∘C and a tensile strengthof about 12.5 GPa.157 These properties have led to the use ofPGA predominantly as suture material. As scaffold material PGAhas some unique and significant shortcomings. Its high molecu-lar weight form has poor solubility in most organic solvents whileits low molecular weight forms are more soluble. The more solu-ble forms of PGA, however, lack sufficient mechanical integrity invivo with significant decline in its performance observed at 2–4weeks158 – 161 – this may not be sufficient time for adequate regen-eration of certain body tissues such as bone. Although glycolicacid is a natural metabolite, high levels are known to stimulate anexcessive inflammatory response.162,163 Also, because of its highhydrolytic sensitivity, processing of PGA materials must be carriedout in carefully controlled conditions.

Poly(lactic acid) chains are assembled from chiral molecules andmay 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 enantiomersin the PLA chain, rather than the molecular weight, that deter-mine the ultimate properties of PLA-based materials. It is morehydrophobic than PGA which means that degradation rates aremuch slower, making it highly suitable for both in vitro and in vivoapplications164 – 167 where long-term mechanical integrity is criti-cal. The general values for Young’s Modulus, tensile strength andimpact strength for PLA are around 3 GPa, 1.5–2.7 GPa and 50-70MPa,168 respectively.

PLGA, on the other hand, presents a combined, myriad of prop-erty advantages over its polymeric counterparts. The copolymer

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

composition is the main determinant of the chemical, mechanicaland structural variations that can be achieved. Figure 1 depictsthe influence of the co-polymer ratio on degradation rates. Whileincreasing the amount of the slower degrading PLA increasesdegradation time, counteractively increasing the amount of PGAwill slow down this process. Degradation times, for example, canbe from 1–2 months, 4–5 months and 5–6 months, for 50:50,75:25 and 85:15 co-polymer ratios of PLGA, respectively.169 Thebulk erosion of PLGA makes controlled release of therapeuticand other factors difficult. Therefore PLGA microspheres,170 – 173

microcapsules,174,175 nano-spheres,176 and nano-fibres177,178 areoften used.

Poly(𝝐-caprolactone)Another aliphatic polyester used as scaffold material in tissueengineering is the semi-crystalline polymer, poly(𝜖-caprolactone)(PCL).180,181 With great organic solvent solubility, a melting tem-perature of about 55–60 ∘C, and glass transition temperature of−54∘C182 this polymer is easily processed into tissue regenerationsupport structures.

Degradation profiles of the polymer make it suitable for use intissues that have a longer regeneration process. In particular PCLhas been shown to carry a similar rate of degradation to the rate ofnew bone formation.183,184,185 Compared with its family membersPLA and PGA, PCL takes much longer to degrade186 via the samenon-enzymatic cleavage of ester linkages.189 – 191 This is the mainreason why PCL is usually adopted for long-term in vivo systemsrather than short-term systems.192 Enzymatic degradation of PCLis known to occur to some extent as well, although this is not theprimary breakdown mechanism of the polymer.193

The innate biological inactivity of PCL makes it a promisingbiomaterial for safe and controlled in vivo applications. The invivo performance of PCL scaffolds have been commended fortheir particular success in carrying chondrogenic additives thatsuccessfully induce chondrogenesis in mesenchymal stem cells.194

The method by which such an inert polyester is functionalizedand ‘activated’ to produce desired effects is primarily throughsurface modification methods that focus on creating negativelycharged hydrophobic surfaces that promote cell attachment viaproteins such as fibronectin and vitronectin.195

Furthermore, blending and copolymerization196 with otherpolymers are methods by which scaffold characteristics can be

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 6: ozdil2014

1798

www.soci.org D Ozdil, HM Aydin

controlled even further for desired outcomes. Owing to thisflexibility, the pore sizes achievable with PCL scaffolds and therelatively inexpensive manufacturing methods with this material,PCL serves as a viable material solution for tissue engineeringendeavours.

Poly(urethanes)Linear aliphatic polyesters dominate the hard-tissue engineeringsynthetic biomaterials category whereas the main biomate-rial investigated for soft-tissue engineering purposes has beenpolyurethane. A hard segment (diisocyanate), the soft segment(poly(ethers) or poly(esters)) and chain extenders are the fun-damental components of polyurethanes.197 The ratios of thesecomponents involved in the chemical reaction that produces thepolyurethane will define its ultimate properties.

The historical use of polyurethanes had experienced the draw-backs of the polymer’s toxic degradation products. To overcomethis issue, polyurethanes with diisocyanate replacements havebeen developed. One example is using 1,4-diisocyanatobutane(BDI) and a putrescine chain extender where the degradationof BDI leads to the release of putrescine – a polyamine whichencourages cell growth and proliferation.198 Another example ispolymerizing highly pure lysine diisocyanate with glucose so thatbreakdown 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 bemade via electrospinning, water foaming and thermally-inducedphase separation methods. Tensile strength and breaking strainscan reach 13 MPa and 280%, respectively.199 Polyurethanes havealso displayed cell adhesion and proliferation qualities which alsoadd to their suitability for in vivo use.200 However, an importantdrawback for polyurethanes in tissue engineering or drug deliveryuse is that their degradation rates are found to be not suitable forthese applications. For this reason blends and combinations ofpolyurethane with other biomaterials are made to assist degra-dation profiles of polyurethane-based scaffolds. Ongoing studiesare looking into chemically modifying polyurethanes to equipthem with the appropriate mechanical characteristics requiredat the various stages of healing and regeneration, subsequentlyfollowed by its biodegradation. Biodegradable polyurethanes,therefore, continue to hold promise over highly porous scaffoldsthat are disadvantaged by mechanical weakness.

Poly(propylene fumarate)Poly(propylene fumarate) (PPF) is a unique, injectable, linearpolyester that contains multiple unsaturated double bondsthat are also available for covalent cross-linking in the pres-ence of free-radical initiators using benzoyl peroxide and NN dimethyl-p-toluidine. The biocompatibility of PFF has beenestablished by various studies.201,202

The cross-linking capability of the polyester means that degrada-tion 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 scaf-folds to assume scaffold designs that may not be achievable bynon-cross-linkable biodegradable polymers. In fact, this flexibilityhas made PPF a common bone defect filling material.204 – 207

The crosslinking reaction is propagated using free-radicalpolymerization.

PPF-based porous scaffolds.201,208 – 210 and microsphere-embedded scaffolds211 – 214 have been studied in vivo or in vitro

extensively, and have proved to be biocompatible. The traditionalmethod of synthesizing porous PPF scaffolds was by embeddinginto the polymeric material sodium chloride crystals which areleached out upon implantation – a technique that carries the riskof creating a hypertonic local environment and offers minimal con-trol over interconnected porosity. Current methods are based oninitiating foaming reactions during polymerization215,216 such thatthe supercritical CO2 treatment will generate the porosity essentialfor the success of PPF scaffolds in tissue repair and regeneration.The inclusion of ceramics and other enhancement factors in thesescaffolds mechanically strengthens them217 as well as improvingtheir biocompatibility and material–tissue relationships.218 – 220

Synthetic poly(amino acids)Synthetic poly(amino acids) include homo-and co-poly(aminoacids), and display high crystallinity, low degradation rate,poor mechanical properties and immunogenicity221 have beenreported for these polymers.

Poly(L-glutamic acid) (L-PGA) is a flexible synthetic poly(aminoacid) that is relatively easy to fabricate into differentarchitectures.222 It is also non-immunogenic and enzymaticallydegradable.223,224 It has featured as cancer drug delivery225 andMRI contrast agents226 and as a copolymer with other polymers intissue engineering scaffolds.

Poly(aspartic acid) (PAA) is a highly water-soluble polymer thatalso exists as a hydrogel and undergoes enzymatic degradation.227

It can be copolymerized with other polymers (e.g. PLA,228 PCL229

and PEG228). Micellar configurations formed by these co-polymershave seen interest for use in payload delivery.

Poly(ortho esters)The poly(ortho ester) (POE) family of hydrophobic polymers230 arepolymers consisting of three geminal ether bonds. The uniquequalities of POEs are its surface-erosion mechanism of degrada-tion and the sensitivity of the process to pH. Ortho-ester bondsare stable at neutral pH but rapidly hydrolyse at about pH 5.5.Under physiological conditions these highly hydrophobic poly-mers 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 tissueengineering. POE IV, modified from POE II, consists of short seg-ments of lactic acid or glycolic acid incorporated into its backbone.This not only speeds up degradation but makes POE IV more suit-able for tissue regeneration materials than POE I–III, which possessdegradation rates much too slow for these purposes. Varying thebackbone chemistry of POEs will vary the degradation and mate-rial properties.

This new generation of POEs is an evolutionary developmentof the second generation polymers232 and is recognized asself-catalytic poly(ortho esters). In this new variation of POE, shortdimer segments of glycolic acid or lactic acid are incorporatedinto the polymer backbone. Owing to the susceptibility of POEto acid-catalysed hydrolysis, varying the concentration of the𝛼-hydroxy acid dimer segments in the polymer backbone cancontrol the erosion rate of the bulk polymer. These polymersoffer significant advantages over the second-generation POEs,which required the addition of acidic excipients to control erosionrate.

Poly(anhyrides)Polyanhydrides are either aromatic, aliphatic or a mixture of thetwo polymers that are constructed from two carbonyl groups

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 7: ozdil2014

1799

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 unsta-ble polymer is via the dehydration of the diacid or mixtureof diacids by melt polycondensation produces this class ofbiodegradable and biocompatible polymer. Polyanhydrides havepredominantly featured as biomaterials that are practical fordrug delivery systems. Polyanhydride-based microparticles233 – 235

and nanoparticles236 – 238 developed for these purposes can beinjected, administered orally or with aerosol delivery.

These polymers undergo surface erosion that is pH-dependentand degradation is based on the backbone chemistry of the poly-mer, with the specific tuning of degradation rate. Polyanhydrideson their own are usually not suitable for tissue engineering appli-cations that require load bearing scaffolds as their low molecularweights limit their mechanical strength. The Young’s modulus ofpoly(anhydride)s has been determined to be around 1.3 MPa239

which would not, for example, be suitable in bone tissue engi-neering approaches. Dimethacrylated anhydrides, however, isone sub-class that offers a stronger polyanhydride-based mate-rial. This is due to the photo cross-linkability and variability ofmonomers which allow for the attainment of higher structuraland mechanical integrity of these networks.240,241 Cross-linkedpolyanhydrides have shown effective performance in bone tissueengineering approaches as well as drug delivery materials.242

Monomer flexibility also gives promise of applications in othertissue engineering fields.

Poly(anhydrides-co-imides) have been developed as mechan-ically stronger brands of polyanhydride-based materials thatalso undergo surface erosion.243 Compressive strengths up to50–60 MPa have been reported for poly(anhydrides-co-imides)based on succinic acid, trimellitylimidoglycine, andtrimellitylimidoalanine.244

Poly(anhydride-esters), are particularly attractive as they com-bine the properties of polyesters and polyanhydrides. There are, forexample, poly(anhydride esters) that are based on salicyclic acidthat release the non-steroidal anti-inflammatory (NSAID) (i.e. sali-cyclic acid) upon hydrolytic degradation.

The opportunity to tailor polyanhydrides in such ways make ita versatile material that can be modelled into a diverse range oftissue restoration systems.

Poly(glycerol sebacate)Poly(glycerol sebacate) (PGS) is a biodegradable polymer synthe-sized by the polycondensation of glycerol and sebacic acid. PGSwas first used in a soft-tissue engineering applications in 2002.245

PGS is completely bioresorbable with degradation productseliminated through natural metabolic pathways.246

The hydrophilic, partially semi-crystalline PGS polymer has ther-moset elastomeric properties that can be tuned via the alterationof curing temperature, curing time and molar ratio of glycerol tosebacic acid.247 – 251

An additional level of control over these properties inpoly(glycerol sebacate acrylate) (PGSA) is through varying thenumber of acrylate moieties, which translates to varying thenumber 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 amor-phous. Another interesting feature of PGS is its shape–memorybehaviour. With heat, PGS materials can be stimulated tore-conform to ‘memorized’ shapes and this property is attributedto its ability to switch from its fixed three-dimensional networksat cooler temperatures to its amorphous phase with increasedtemperature.253

PGS undergoes surface erosion with the cleavage of ester link-ages. This type of degradation is useful in that unlike bulk ero-sion, mass loss in surface erosion occurs linearly with time, so thatgeometrical features supporting tissue regeneration are main-tained for longer periods.245,254,255 Although PGS has an acceler-ated degradation rate in vivo than in vitro, the biopolymer hasshown complete biocompatibility in several studies.256 – 258 Onelimitation to PGS use in tissue engineering applications, however,is reported to be the acidic local environments that can be gen-erated when the hydrolysis of the ester groups release carboxylicacids.259

Further functionalisation of PGS has been investigated with theuse of cell migration, adhesion, differentiation and proliferationmediators such as laminin, fibronectin, fibrin, collagen types I/III,and elastin.260

To synergize the favourable characteristics of various poly-mers available for combination with PGS several compositePGS-based scaffolds have been explored. Bioglass/PGS mem-branes for cardiac tissue engineering,259 PGS with nano-tubularhalloysite (2SiO2·2Al(OH)2) incorporation,261 micro-structuredfibrous PGS-PCL scaffolds for heart valve regeneration262 are someexamples of where both organic and inorganic biomaterial blendswith PGS have been studied.

It is the modification of textile manufacturing technologiesand their adaptation to production of biodegradable elastomersthat has given rise to some significant biomedical companies ofour day. There are currently intensive research and development

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 8: ozdil2014

1800

www.soci.org D Ozdil, HM Aydin

efforts that are producing, in particular, flexible tubular structuresand woven matrices that are seen in clinical use.

Poly(phosphazenes)Poly(phosphazenes) are a very large group of polymers that havetraditionally been produced with high molecular weights. Theirbackbones are completely inorganic constructed with alternatingphosphorous and nitrogen atoms with two organic side groupsattached to each phosphorous atom. Most phosphazenes aremade by substitution reactions on a poly(dichlorophosphazene)intermediate. The history of poly(phosphazene) use inbiomedicine only extends back to the past two decades.263

In order to functionalize the polymer further, specific side groupscan be introduced onto the primary structure such as amino acidesters, glucosyl, lactate or imidazolyl units.264 – 266 This flexibilityhas led to >500267 types of poly(phosphazenes) produced to dateand as such initiated an exploration of these polymers as biomate-rials for tissue regeneration rather than just their traditional use indrug delivery.

The aforementioned side groups, for example, introducehydrolytic instability to the structure and assist with degradationwhere the non-toxic breakdown products are released. Interest-ingly, when poly(phosphazenes) are combined with polymers, likepolyesters, that have acidic degradation products, the presenceof poly(phosphazene) breakdown products have a pH bufferingeffect.268 Mechanical properties are also varied in this manner too.One study found that such modifications produces significantlylarger value ranges for glass transition temperature (Tg − 0–35 ∘C),contact angle (63∘–107∘), tensile strength (2.4–7.6 MPa), andmodulus of elasticity (31.4–455.9 MPa).269 Poly(phosphazenes)do generally require blends with other polymers in this way toimprove chemical and mechanical characteristics. There has alsobeen some recent concern over the foreign body response270 thatpoly(phosphazene)-based materials have been found to trigger,contrary to previous research which had only found a limitedinflammatory response.271

In various tissue engineering applications such as nerveregeneration272,273 and orthopaedic applications,274,275 poly(phosphazenes) have been used as films,274 fibres,272,275 gels,276,277

and sintered microspheres274 thanks to the ability to increase theirhydrophobicity with side group substitutions.

Poly(dioxanone)Poly(dioxanone) (PDO) is one of the most commonly used mem-bers of the poly(ester ether) family of degradable polymers. Thesepolymers typically have an ether bond integrated into the back-bone of a polyester with the aim of initiating hydrolytic cleavage ofthe ester bond. Ring-opening polymerization of p-dioxanone pro-duces PDO. P-dioxanone has a Tg about −10 to 0 ∘C and a Tm of115 ∘C.278

PDO is considered a slow-degrading polymer that has completemass loss at about 6–12 months.157 PDO does not exhibit themechanical properties that tissue engineering scaffolds wouldgenerally require, with a modulus of (1.5 GPa).157 It does, however,have good flexibility and 1–2 months of considerable strengthmaintenance which is why it has been selected for the productionof monofilament sutures for decades.279

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

are termed poly(ethylene oxide) (PEO). The hydroxyl groups at theend of PEG chains are used for a variety of PEG macromers thathave differing properties and uses.

PEG polymers are biocompatible, biodegradable, non-toxic, andlow-immunogenic synthetic polymers that do not bind proteins orcells.280,281 In fact, the first interest in PEG was to use it as biomate-rial surface coatings owing to its ability to prevent serum proteinadsorption, although the mechanisms by which this occurs is stillnot completely understood and have been attributed to a num-ber of factors.282,283 The mechanical properties of PEG scaffolds intissue engineering depend on the molecular weight, cross-linking,and polymer concentration. Decreasing the molecular weight orincreasing the concentration of the polymer has been shown toimprove the elastic modulus.284

PEG hydrogels have been studied in tissue engineering scaffoldapplications. Of course these scaffolds have been adapted to thesepurposes with the incorporation of cell adhesion RGD peptides toenable cell adhesion and survival.285 For PEG hydrogels that maycarry proteins or cells for implantation and release, photopolymer-ization is preferred to thermal polymerization owing to the sen-sitivity of this method towards such payloads and the better effi-ciency that it offers.286 In fact, one group has been able to demon-strate PEG photopolymerization transdermally.287 PEG is oftenalso preferred for its bio-inactivity toward encapsulated proteinsor cells where such interaction is not desired.285,288 – 291 Functional-isation is also possible where cell adhesive molecules and growthfactors used in PEG hydrogel scaffolds have added to the successof modulated tissue regeneration with these materials.292 – 295

As discussed, there are several synthetic polymer options thatcan be used in various medical applications. The suitability ofeach of these polymers for their intended use is based on theirrespective chemical and physical properties and the capacity towhich they can be processed to assume the required structuralcharacteristics. Table 1 summarizes the key properties of thesynthetic polymers discussed for quick reference.

HydrogelsCross-linked polymer networks that contain 60–90% water, knownas hydrogels, have become attractive materials for regenerativemedicine. Due to their remarkably tuneable properties hydrogelsare emerging as an exciting new material used in a diverse rangeof applications including the development of artificial corneaimplants,306 as fillers for soft tissue engineering307 operations andin cartilage repair materials,308 to name a few.

Degradable three-dimensional hydrogel matrices are made fromeither hydrophilic homopolymers, copolymers, or, they can exist asinsoluble matrices via the introduction of cross-links.

Hydrogels can be fabricated from both natural polymers ofagarose, alginate, chitosan, hyaluronic acid, fibrin, collagen andothers, as well as synthetic polymers such as poly(ethylene gly-col) (PEG), poly(vinyl alcohol) (PVA), and polyacrylates such aspoly(2-hydroxyethyl methacrylate) (PHEMA).309,310 Covalent andnon-covalent bonding in cross-links generate a biocompatible311

matrix with structural similarity to other macromolecules found inthe body.

Among the different methods of hydrogel synthesis, includingMichael (conjugate addition) and click chemistry, free-radicalpolymerisation is the most appealing for tissue engineering dueto the great gelation kinetics and in situ polymerization capabilityassociated with this process.312 Figure 3 is a visual representationof these chemical synthesis processes. Acrylate-based hydrogelsthat have been subjected to this process contain many mechanical

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 9: ozdil2014

1801

Polymers for medical and tissue engineering applications www.soci.org

Table 1. Properties of popular synthetic polymers used in medicine

Polymer Structure

Crystallinity

(%)

Elastic

modulus

(GPa)

Tensile

modulus

(GPa)

Melting

temperature

(∘C)

Glass

transition

temperature

(∘C)

Degradation

products

Time to

degrade

(months) Refs

Polylactic acid 37 2.7 1.5– 2.7 173–178 60–65 L-Lactic acid 12– 18 (296–298)

Polyglycolic acid 45-55 7.0 5–7 225–230 35–40 Glycolic Acid 3– 4 (296,298)

Poly(𝝐-caprolactone)

37 0.4 0.4– 0.6 58–63 −60 Caproic Acid >24 (299–301)

Poly-lactic-co-glycolic acid(50/50)

Amorphous 2.0 1.4– 2.8 Amorphous 50–55 D,L-lactic acidandglycolic acid

3–6 (296–298)

Poly(propylenefumarate)

37 2– 3 2–3 30–50 −60 Fumaric acid,propylene

glycol and

poly(acrylic

acid-cofumaricacid)

Varies (>24) (302,303)

Polyanhydride - - 25–27 50–90 −27 Carboxylic acids 0.14–1.4 (304,305)

Polyhphosphazene 55 - - - - Phosphate,ammonia,correspondingside groups

surface (267,274)

and structural advantages such as increased elastic modulus,313

great cell–biomaterial interactions,314 good tensile modulus,315

prevention of undesired fibrosis,316 enhanced mechanisms ofrelease of payloads embedded in polymeric material,292,294,317 andhigher compressive modulus with GAG incorporation.318 – 322

Mixing or blending hydrogels with micro- and nanoparticles324

which are later removed through various processes or throughnatural dispersion with payload delivery allow porous scaffoldsto form. Successful porous synthetic hydrogels in cornea replace-ments offer an example of this. Such scaffolds are also known topromote cell migration and angiogenesis and with a high watercontent are capable of rapid nutrient diffusion.325

Although porosity is desirable for tissue integration, the biggestchallenge yet for porous hydrogel scaffolds is the poor mechan-ical strength that they display.326 Cross-linking density plays animportant role in appropriating mechanical compliance andmesh size for cell encapsulation and use in tissue augmenta-tion procedures.327 Careful control of chemical and processingparameters can ultimately yield hydrogel scaffolds with numerousbenefits for tissue regeneration efforts.

STIMULI RESPONSIVE POLYMERS‘Smart’ or ‘intelligent’ polymers are those which can alter theirphysio-chemical properties in response to stimuli (pH, tempera-ture, redox, enzymes, light, magnetic, ultrasound) such that a par-ticular functional output of the polymeric system occurs. Amongsome property changes that can occur as a response to stimuli arechanges in surface wettability,328 membrane permeability,329,330

swelling capability (hydrogels),331 aggregation behaviour,332

and sol-gel transition.333 Stimuli-responsive polymers are pro-duced by introducing specific polymeric modalities to traditional

polymeric systems, creating multi-functional systems.334 Assuch, stimuli-responsive polymers have received great interestover the past decade and have been taken up in a number ofmedical applications from drug delivery and tissue engineering toimaging.

Thermo-responsive polymersTo date, temperature- and pH-sensitive polymers have been themost frequently studied type of stimuli-responsive polymer.Thermoresponsive polymers can change their hydrophilic-ity/hydrophobicity, conformation or solubility due to a phasetransition reaction when prompted by elevated temperatures335

to reach a threshold temperature known as critical solution tem-perature (CST). Generally, a lower critical solution temperature(LCST) polymers will reduce the solubility (increase hydrophobic-ity) while a higher critical solution temperature (HCST) will increasesolubility (increase hydrophilicity).336 Poly(N-isopropylacrylamide)(PNIPAAm) in particular has been the most researchedtemperature-responsive polymer where it exhibits a transitionfrom hydrophilicity under 32 ∘C to a hydrophobicity above thistemperature.337 One study demonstrated the utility of the LCSTfeature of PNIPAAm with a thiolated chitosan-PNIPAAm hydro-gel used for wound infections that deliver ciprofloxacin (ananti-bacterial drug) to the wound site that will then swell uponthe addition of cold water for easy removal of the gel. Anotherexample is poly(N-vinylcaprolactam), a lactam-based polymer thatis also thermoresponsive with a LCT of about 35∘C338 and holdspromise in tissue engineering applications as it thermorespon-sivity allows for the formation of a macroporous architecture – acommon limitation for hydrogels intended for use in tissue engi-neering applications. Other temperature-responsive polymersused in biomedical applications include poly(N-vinylcaprolactam)

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 10: ozdil2014

1802

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 acrylategroup. 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 from25–35 ∘C,339 poly(ethylene glycol) (PEG) or poly(ethylene oxide)(PEO) which becomes insoluble above 80 ∘C and poly(propyleneoxide) (PPO) which, when in co-polymer form,340,341 can exhibit awide range of solubility and phase transition temperatures.

pH-responsive polymerspH-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-responsivenessof polymers can be tuned by the incorporation of ionizablemonomer units in to their polymeric backbones. Chitosan-carrageenan-based polyelectrolyte complexes have beenexplored as porous biomaterials that can maintain stabilityat physiological conditions while dissociating upon implan-tation due to chitosan reacting under the lowered pH levelscaused by local inflammation associated with implantation.342

Another study343 was able to show how electrospun poly-electrolyte hydrogel nanofibres were able to display reversibleswelling/contracting with varying pH, highlighting their potentialas artificial muscles, interactive tissue engineering scaffolds ordrug delivery systems. Materials based on poly(acrylic acid) (PAAc)and poly(methacrylic acid) (PMAc) are common polymers usedin pH-responsive systems and can also exhibit reversible swellingwhen pH is varied.344,345 Such acrylic polymers and their unique

properties, for example, increased surface adhesion when inprotonated form, have also inspired interest for their use in drugdelivery systems.

CONCLUDING REMARKSCurrently, the number of donors required for organ and soft tis-sue replacement are well below those numbers that are actually indemand. For this reason, research and development of syntheticmaterials capable of combining cellular components for both thein vitro and in vivo regeneration of tissues are continuing to pro-ceed at a significant pace. The range of different chemical naturesof different polymers, the ability to fabricate them such that theyreplicate tissue or organ morphologies, their ability to biodegradewithout adverse effects, and their biocompatibility has created agreat area of use in the field of tissue engineering. The modifi-cation and enhancement of several processing methods such as3D printing, electrospinning, fibre processing, and foaming for theproduction of tissue scaffolds seen in recent years, the fall of pricesassociated with medical grade products and the improvements inconditions of manufacturing these products in small-scale cleanenvironments have all been factors leading to the uptake of poly-mers in efforts to regenerate bone, cartilage, tendon, skin, cornea,and other tissues that constitute different morphological andmechanical properties. The advancements in growth factor-cell

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 11: ozdil2014

1803

Polymers for medical and tissue engineering applications www.soci.org

studies and stem cell studies that have progressed parallel tothese efforts combined with the widespread use of bioreactors, allwithin the regulative standards of special organizations such as theadvanced technological medicinal products (ATMP), provide fore-sight into the utilization of polymers in multi-component complexsystems that are developed in tissue engineering studies and sub-sequently taken up in clinical use.

REFERENCES1 Lakshmi SN, Laurencin CT, Biodegradable polymers as biomaterials.

Prog Polym Sci 32:762–798 (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 AcademicPress; San Diego, CA, USA (2004).

3 Hahn SK, Jelacic S, Maier RV, Stayton PS and Hoffman AS,Anti-inflammatory drug delivery from hyaluronic acid hydrogels. JBiomater Sci - Polym E 15:1111–1119 (2004).

4 Seidlits SK CTD, Petersen RR, Shear JB, Suggs L J and SchmidtCE, Fibronectin–hyaluronic acid composite hydrogels forthree-dimensional endothelial cell culture. Acta Biomater7:2401–2409 (2011).

5 Kim M, Optimization of macromer density in human MSC-ladenhyaluronic acid (HA) hydrogels, in Bioengineering Conference(NEBEC), 2012 38th Annual Northeast (2012); 16–18 March 2012;Philadelphia, PA: IEEE; 211–212 (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 fabrica-tion of tissue-engineering scaffolds with a poly(lactic-co-glycolicacid) grafted hyaluronic acid conjugate encapsulating an intactbone morphogenetic protein–2/poly(ethylene glycol) complex.Adv Function Mater 21:2906–2912 (2011).

7 Chung C, Erickson IE, Mauck RL and Burdick JA, Differential Behavior ofAuricular and Articular Chondrocytes in Hyaluronic Acid Hydrogels.Tissue Eng Part A 14:1121–1131 (2008).

8 Knudson W, Loeser RF, CD44 and integrin matrix receptors participatein cartilage homeostasis. Cell Mol Life Sci 59:36–44 (2002).

9 Tool BP, Hyaluronan in morphogenesis. Semin Cell Dev Biol 12:79–87(2001).

10 Goodstone NJ, Cartwright A and Ashton B, Effects of high molecularweight hyaluronan on chondrocytes cultured within a resorbablegelatin sponge. Tissue Eng 10:621–631 (2004).

11 Kawasaki K, Ochi M, Uchio Y, Adachi N and Matsusaki M, Hyaluronicacid enhances proliferation and chondroitin sulfate synthesis incultured chondrocytes embedded in collagen gels. J Cell Physiol179:142–148 (1999).

12 Duflo S, Thibeault SL, Li WH, Shu XZ and Prestwich G, Effect of asynthetic extracellular matrix on vocal fold lamina propria geneexpression in early wound healing. Tissue Eng 12:3201–3207(2006).

13 Slevin M, Krupinski J, Gaffney J, Matou S, West D, Delisser H, et al.,Hyaluronan-mediated angiogenesis in vascular disease: uncover-ing RHAMM and CD44 receptor signaling pathways. Matrix Biol26:58–68 (2007).

14 Birk DE and Bruckner P, Collagen suprastructures. Top Curr Chem247:185–205 (2005).

15 Francis G and Thomas J, Isolation and chemical characterization ofcollagen in bovine pulmonary tissues. Biochem J 145:287–297(1975).

16 Lee CH, Singla A and Lee Y, Biomedical applications of collagen. Int JPharm 221:1–22 (2001).

17 Sweeney SM, Orgel JP, Fertala A, McAuliffe JD, Turner KR, Di LulloGA, et al., Candidate cell and matrix interaction domains on thecollagen fibril, the predominant protein of vertebrates. J Biol Chem283:21187–21197 (2008).

18 Rich A, Solving the structure of collagen. Self-assembling peptidesystems in biology. Med Eng 235–241 (2001).

19 Voge CM, Kariolis M, MacDonald RA and Stegemann JP, Direc-tional conductivity in SWNT-collagen-fibrin composite biomateri-als through strain-induced matrix alingment. J Biomed Mater Res A86A:269–277 (2008).

20 Seliktar D, Black RA, Vito RP and Nerem RM, Dynamic mechanical con-ditioning of collagen-gel blood vessel constructs induces remodel-ing in vitro. Ann Biomed Eng 28:351–362 (2000).

21 Isenberg BC and Tranquillo RT, Long-term cyclic distention enhancesthe mechanical properties of collagen-based media-equivalents.Ann Biomed Eng 31:937–949 (2003).

22 Heino J, The collagen receptor integrins have distinct ligand recogni-tion and signaling functions. Matrix Biol 19:319–323 (2000).

23 White DJ, Puranen S, Johnson MS and Heino J, The collagen receptorsubfamily of the integrins. Int J Biochem Cell B 36:1405–1410(2004).

24 Sarikaya B and Aydin HM, Development of collagen/beta-TCP basedsynthetic bone grafts. J Tiss Eng Reg Med 8:410–410 (2014).

25 Aydin HM, Yang Y, Kohler T, El Haj A, Müller R, Piskin E, Interac-tion of osteoblasts with macroporous scaffolds made of PLLA/PCLblends modified with collagen and hydroxyapatite. Adv Eng Mater11:B83–B88 (2009).

26 Kojima K, Okamoto Y, Kojima K, Miyatake K, Fujise H, Shigemasa Y,et al., Effects of chitin and chitosan on collagen synthesis in woundhealing. J Vet Med Sci 66:1595–1598 (2004).

27 Islam A and Yasin T, Controlled delivery of drug from pH sensitivechitosan/poly (vinyl alcohol) blend. Carbohyd Polym 88:1055–1060(2012).

28 Ren DW, Yi HF, Wang W and Ma XJ, The enzymatic degradation andswelling properties of chitosan matrices with different degrees ofN-acetylation. Carbohyd Res 340:2403–2410(2005).

29 Li DH, Liu LM, Tian KL, Liu JC and Fan XQ, Synthesis, biodegradabilityand cytotoxicity of water-soluble isobutylchitosan. Carbohyd Polym67:40–45 (2007).

30 Xia WS, Liu P and Liu J, Advance in chitosan hydrolysis by non-specificcellulases. Bioresource Technol 99:6751–6762 (2008).

31 Chen RN, Wang GM, Chen CH, Ho HO and Sheu MT, Development ofN,O-(carboxymethyl)chitosan/collagen matrixes as a wound dress-ing. Biomacromolecules7:1058–1064 (2006).

32 Liu BS, Yao CH and Fang SS, Evaluation of a non-woven fabric coatedwith a chitosan Bi-layer composite for wound dressing. MacromolBiosci 8:432–440 (2008).

33 Wang JP, Feng SS, Wang S and Chen ZY, Evaluation of cationicnanoparticles of biodegradable copolymers as siRNA deliv-ery system for hepatitis B treatment. Int J Pharm 400:194–200(2010).

34 Yang R, Shim WS, Cui FD, Cheng G, Han X, Jin QR, et al., Enhanced elec-trostatic interaction between chitosan-modified PLGA nanoparti-cle and tumor. Int J Pharm 371:142–147 (2009).

35 Zhang YQ, Chen JJ, Zhang YD, Pan YF, Zhao JF, Ren LF, et al., A novelPEGylation of chitosan nanoparticles for gene delivery. BiotechnolAppl Biochem 46:197–204 (2007).

36 Matsusaki M, Sakaguchi H, Serizawa T and Akashi M, Controlledrelease of vascular endothelial growth factor from alginate hydro-gels nano-coated with polyelectrolyte multilayer films. J Biomat Sci- Polym E18:775–783 (2007).

37 Berger J, Reist M, Mayer JM, Felt O, Peppas NA and Gurny R, Struc-ture and interactions in covalently and ionically crosslinked chi-tosan hydrogels for biomedical applications. Eur J Pharm Biopharm57:19–34 (2004).

38 Rodrigues IR, Forte MMD, Azambuja DS and Castagno KRL, Synthesisand characterization of hybrid polymeric networks (HPN) basedon polyvinyl alcohol/chitosan. React Funct Polym 67:708–715(2007).

39 Ma GP, Yang DZ, Li QZ, Wang KM, Chen BL, Kennedy JF, et al.,Injectable hydrogels based on chitosan derivative/polyethyleneglycol dimethacrylate/N,N-dimethylacrylamide as bone tissueengineering matrix. Carbohyd Polym 79:620–627 (2010).

40 Hsieh CY, Hsieh HJ, Liu HC, Wang DM and Hou LT, Fabrication andrelease behavior of a novel freeze-gelled chitosan/gamma-PGAscaffold as a carrier for rhBMP-2. Dent Mater 22:622–629 (2006).

41 Di Martino A, Sittinger M and Risbud MV, Chitosan: a versatilebiopolymer for orthopaedic tissue-engineering. Biomaterials26:5983–5990 (2005).

42 Peng L, Cheng XR, Zhuo RX, Lan J, Wang YN, Shi B, et al., Novelgene-activated matrix with embedded chitosan/plasmid DNAnanoparticles encoding PDGF for periodontal tissue engineering.J Biomed Mater Res A 90A:564–576 (2009).

43 Martins AM, Alves CM, Kasper FK, Mikos AG and Reis RL, Respon-sive and in situ-forming chitosan scaffolds for bone tissue engi-neering applications: an overview of the last decade. J Mater Chem20:1638–1645 (2010).

44 Azhar FO, A ; Salehi, R. Fabrication and characterization ofchitosan-gelatin/nanohydroxyapatite-polyaniline composite with

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 12: ozdil2014

1804

www.soci.org D Ozdil, HM Aydin

potential application in tissue engineering scaffolds. DesignedMonom Polym 17:654–667 (2014).

45 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 fibersheets containing silver sulfadiazine for use as an antimicrobialwound dressing. J Nanosci Nanotechnol (2014)14:7488–7494.

46 Mekhail M, Jahan K and Tabrizian M, Genipin-crosslinkedchitosan/poly-L-lysine gels promote fibroblast adhesion andproliferation. Carbohyd Polym 108:91–98 (2014).

47 Doty HL, Leedy MR, Courtney HS, Haggard WO and Bumgardner JD,Composite chitosan and calcium sulfate scaffold for dual deliveryof vancomycin and recombinant human bone morphogeneticprotein-2. J Mater Sci - Mater M 25:1449–1459 (2014).

48 Tan CS, Jejurikar A, Rai B, Bostrom T, Lawrie G and Grondahl L,Encapsulation of a glycosaminoglycan in hydroxyapatite/alginatecapsules. J Biomed Mater Res A 91A:866–877 (2009).

49 Jay SM, Shepherd BR, Andrejecsk JW, Kyriakides TR, Pober JS and Saltz-man WM, Dual delivery of VEGF and MCP-1 to support endothe-lial cell transplantation for therapeutic vascularization. Biomaterials31:3054–3062 (2010).

50 Hahn MS, Teply BA, Stevens MM, Zeitels SM and Langer R, Collagencomposite hydrogels for vocal fold lamina propria restoration.Biomaterials 27:1104–1109 (2006).

51 Wang YZ, Kim HJ, Vunjak-Novakovic G and Kaplan DL, Stemcell-based tissue engineering with silk biomaterials. Biomaterials27:6064–6082 (2006).

52 Kim JH, Kim YS, Kim S, Park JH, Kim K, Choi K, et al., Hydrophobicallymodified glycol chitosan nanoparticles as carriers for paclitaxel. JControl Release 111:228–234 (2006).

53 Liu XY, Nothias JM, Scavone A, Garfinkel M and Millis JM,Biocompatibility investigation of polyethylene glycol andalginate-poly-l-lysine for islet encapsulation. ASAIO J 56:241–245(2010).

54 Purcell EK, Singh A and Kipke DR, Alginate composition effects on aneural stem cell-seeded scaffold. Tissue Eng Part C - Me 15:541–550(2009).

55 Gou ML, Wei XW, Men K, Wang BL, Luo F, Zhao X, et al., PCL/PEGcopolymeric nanoparticles: potential nanoplatforms for anticanceragent delivery. Curr Drug Targets 12:1131–1150 (2011).

56 Lim SM, Oh SH, Lee HH, Yuk SH, Im GI and Lee JH, Dual growthfactor-releasing nanoparticle/hydrogel system for cartilage tissueengineering. J Mater Sci - Mater M 21:2593–6000 (2010).

57 Meng X, Li P, Wei Q and Zhang HX, pH sensitive alginate-chitosanhydrogel beads for carvedilol delivery. Pharm Dev Technol16:22–28 (2011).

58 Wang JZ, Huang XB, Xiao J, Yu WT, Wang W, Xie WY, et al.,Hydro-spinning: a novel technology for making alginate/chitosanfibrous scaffold. J Biomed Mater Res A 93A:910–919 (2010).

59 Marsich E, Borgogna M, Donati I, Mozetic P, Strand BL, Salvador SG,et al., Alginate/lactose-modified chitosan hydrogels: a bioactivebiomaterial for chondrocyte encapsulation. J Biomed Mater Res A84A:364–376 (2008).

60 Wang Q, Jamal S, Detamore MS and Berkland C,PLGA-chitosan/PLGA-alginate nanoparticle blends as biodegrad-able colloidal gels for seeding human umbilical cord mesenchymalstem cells. J Biomed Mater Res A 96A:520–527 (2011).

61 Qi XP, Ye JD and Wang YJ, Alginate/poly (lactic-co-glycolicacid)/calcium phosphate cement scaffold with oriented porestructure for bone tissue engineering. J Biomed Mater Res A89A:980–987 (2009).

62 Qi J, Chen AM, You HB, Li KP, Zhang D and Guo FJ, Proliferationand chondrogenic differentiation of CD105-positive enriched ratsynovium-derived mesenchymal stem cells in three-dimensionalporous scaffolds. Biomed Mater 6:015006 (2011).

63 Olmez SS, Korkusuz P, Bilgili H and Senel S, Chitosan and alginate scaf-folds for bone tissue regeneration. Pharmazie 62:423–431 (2007).

64 Wittmer CR, Phelps JA, Lepus CM, Saltzman WM, Harding MJ andVan Tassel PR, Multilayer nanofilms as substrates for hepatocellularapplications. Biomaterials 29:4082–4090 (2008).

65 Jin XB, Sun YS, Zhang K, Wang J, Shi TP, Ju XD, et al., Tissue engineeredcartilage from hTGF beta 2 transduced human adipose derivedstem cells seeded in PLGA/alginate compound in vitro and in vivo.J Biomed Mater Res A 86A:1077–1087 (2008).

66 Chan AW, Whitney RA and Neufeld RJ, Semisynthesis of a con-trolled stimuli-responsive alginate hydrogel. Biomacromolecules10:609–616 (2009).

67 Yamada Y, Hozumi K, Katagiri F, Kikkawa Y and Nomizu M, Biologicalactivity of laminin peptide-conjugated alginate and chitosan matri-ces. Biopolymers 94:711–720 (2010).

68 Ryu JH, Kim IK, Cho SW, Cho MC, Hwang KK, Piao H, et al., Implantationof bone marrow mononuclear cells using injectable fibrin matrixenhances neovascularization in infarcted myocardium. Biomateri-als 26:319–326 (2005).

69 Makogonenko E, Tsurupa G, Ingham K and Medved L, Interactionof fibrin(ogen) with fibronectin: further characterization andlocalization of the fibronectin-binding site. Biochemistry - Us41:7907–7913 (2002).

70 Preissner KT and Jenne D, Vitronectin - a new molecular connectionin hemostasis. Thromb Haemostasis 66:189–194 (1991).

71 Sahni A and Francis CW, Vascular endothelial growth factor binds tofibrinogen and fibrin and stimulates endothelial cell proliferation.Blood 96:3772–3778 (2000).

72 Prasad CK and Krishnan LK, Regulation of endothelial cell phenotypeby biomimetic matrix coated on biomaterials for cardiovasculartissue engineering. Acta Biomater 4:182–191 (2008).

73 Mol A, van Lieshout MI, Veen CGD, Neuenschwander S, Hoerstrup SP,Baaijens FPT, et al., Fibrin as a cell carrier in cardiovascular tissueengineering applications. Biomaterials 26:3113–3121 (2005).

74 Jockenhoevel S, Zund G, Hoerstrup SP, Chalabi K, Sachweh JS, Demir-can L, et al., Fibrin gel-advantages of a new scaffold in cardiovascu-lar tissue engineering. Eur J Cardio -Thorac 19:424–430 (2001).

75 Janmey PA, Winer JP and Weisel JW, Fibrin gels and their clinical andbioengineering applications. J R Soc Interface 6:1–10 (2009).

76 Le Nihouannen D, Le Guehennec L, Rouillon T, Pilet P, Bilban M, Lay-rolle P, et al., Micro-architecture of calcium phosphate granules andfibrin glue composites for bone tissue engineering. Biomaterials27:2716–2722 (2006).

77 Eyrich D, Brandl F, Appel B, Wiese H, Maier G, Wenzel M, et al.,Long-term stable fibrin gels for cartilage engineering. Biomaterials28:55–65 (2007).

78 Bruns H, Kneser U, Holzhuter S, Roth B, Kluth J, Kaufmann PM, et al.,Injectable liver: a novel approach using fibrin gel as a matrix forculture and intrahepatic transplantation of hepatocytes. Tissue Eng11:1718–1726 (2005).

79 Peura M, Siltanen A, Saarinen I, Soots A, Bizik J, Vuola J, et al.,Paracrine factors from fibroblast aggregates in a fibrin-matrix car-rier enhance keratinocyte viability and migration. J Biomed MaterRes A 95A:658–664 (2010).

80 Park SH, Park SR, Chung SI, Pai KS and Min BH, Tissue-engineeredcartilage using fibrin/hyaluronan composite gel and its in vivoimplantation. Artif Organs 29:838–845 (2005).

81 Zhao HG, Ma L, Gao CY and Shen JC, A composite scaffold of PLGAmicrospheres/fibrin gel for cartilage tissue engineering: fabrica-tion, physical properties, and cell responsiveness. J Biomed MaterRes B 88B:240–249 (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 combina-tion of chondrocyte-seeded long-term stable fibrin gels andpolycaprolactone-based polyurethane scaffolds. Tissue Eng13:2207–2218 (2007).

83 Jiang B, Waller TM, Larson JC, Appel AA and Brey EM, Fibrin-loadedporous poly(ethylene glycol) hydrogels as scaffold materials forvascularized tissue formation. Tissue Eng Part A 19:224–234 (2013).

84 Zhao HG, Ma L, Zhou J, Mao ZW, Gao CY and Shen JC, Fabrication andphysical and biological properties of fibrin gel derived from humanplasma. Biomed Mater 3:015001 (2008).

85 Vavken P, Joshi SM and Murray MM, Fibrin concentration affectsACL fibroblast proliferation and collagen synthesis. Knee 18:42–46(2011).

86 Cox S, Cole M and Tawil B, Behavior of human dermal fibroblastsin three-dimensional fibrin clots: Dependence on fibrinogen andthrombin concentration. Tissue Eng 10:942–954 (2004).

87 Li JM, Chen W, Wang H, Jin C, Yu XJ, Lu WY, et al., Preparation ofalbumin nanospheres loaded with gemcitabine and their cytotoxi-city against BXPC-3 cells in vitro. Acta Pharmacol Sin 30:1337–1343(2009).

88 Khan W, Kapoor M and Kumar N, Covalent attachment of proteins tofunctionalized polypyrrole-coated metallic surfaces for improvedbiocompatibility. Acta Biomater 3:541–549 (2007).

89 Wei Q, Li BJ, Yi N, Su BH, Yin ZH, Zhang FL, et al., Improving the bloodcompatibility of material surfaces via biomolecule-immobilizedmussel-inspired coatings. J Biomed Mater Res A 96A:38–45 (2011).

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 13: ozdil2014

1805

Polymers for medical and tissue engineering applications www.soci.org

90 Zehr KJ, Use of bovine albumin-glutaraldehyde glue in cardiovascularsurgery. Ann Thorac Surg 84:1048–1052 (2007).

91 De Somer F, Delanghe J, Somers P, Debrouwere M and Van NootenG, Mechanical and chemical characteristics of an autologous glue.J Biomed Mater Res A 86A:1106–1112 (2008).

92 Shen ZY, Li Y, Kohama K, Oneill B and Bi JX, Improved drug targetingof cancer cells by utilizing actively targetable folic acid-conjugatedalbumin nanospheres. Pharmacol Res 63:51–58 (2011).

93 Gayakwad SG, Bejugam NK, Akhavein N, Uddin NA, Oettinger CEand D’Souza MJ, Formulation and in vitro characterization ofspray-dried antisense oligonucleotide to NF-kappa B encap-sulated albumin microspheres. J Microencapsul 26:692–700(2009).

94 Dror Y, Ziv T, Makarov V, Wolf H, Admon A and Zussman E, Nanofibersmade of globular proteins. Biomacromolecules 9:2749–2754(2008).

95 Regev O, Khalfin R, Zussman E and Cohen Y, About the albuminstructure in solution and related electro-spinnability issues. Int JBiol Macromol 47:261–265 (2010).

96 Kosir MA, Quinn CCV, Wang WL and Tromp G, Matrix glycosaminogly-cans in the growth phase of fibroblasts: more of the story in woundhealing. J Surg Res 92:45–52 (2000).

97 Malavaki C, Mizumoto S, Karamanos N and Sugahara K, Recentadvances in the structural study of functional chondroitin sulfateand dermatan sulfate in health and disease. Connect Tissue Res49:133-139 (2008).

98 van Susante JLC, Pieper J, Buma P, van Kuppevelt TH, van BeuningenH, van der Kraan PM, et al., Linkage of chondroitin-sulfate to typeI collagen scaffolds stimulates the bioactivity of seeded chondro-cytes in vitro. Biomaterials 22:2359–2369 (2001).

99 Park JS, Yang HJ, Woo DG, Yang HN, Na K and Park KH, Chondrogenicdifferentiation of mesenchymal stem cells embedded in a scaffoldby long-term release of TGF-beta 3 complexed with chondroitinsulfate. J Biomed Mater Res A 92A:806–816 (2010).

100 Chang KY, Hung LH, Chu IM, Ko CS and Lee YD, The application oftype II collagen and chondroitin sulfate grafted PCL porous scaffoldin cartilage tissue engineering. J Biomed Mater Res A 92A:712–723(2010).

101 Villanueva I, Gladem SK, Kessler J and Bryant SJ, Dynamic loading stim-ulates chondrocyte biosynthesis when encapsulated in chargedhydrogels prepared from poly(ethylene glycol) and chondroitinsulfate. Matrix Biol 29:51–62 (2010).

102 Nguyen LH, Kudva AK, Guckert NL, Linse KD and Roy K, Unique bio-material compositions direct bone marrow stem cells into specificchondrocytic phenotypes corresponding to the various zones ofarticular cartilage. Biomaterials 32:1327–1338 (2011).

103 Liang WH, Kienitz BL, Penick KJ, Welter JF, Zawodzinski TA andBaskaran H, Concentrated collagen-chondroitin sulfate scaf-folds for tissue engineering applications. J Biomed Mater Res A94A:1050-1060 (2010).

104 Shu XZ, Ahmad S, Liu YC and Prestwich GD, Synthesis and evaluationof injectable, in situ crosslinkable synthetic extracellular matricesfor tissue engineering. J Biomed Mater Res A 79A:902–912 (2006).

105 Chen YL, Lee HP, Chan HY, Sung LY, Chen HC and Hu YC, Compositechondroitin-6-sulfate/dermatan sulfate/chitosan scaffolds for car-tilage tissue engineering. Biomaterials 28:2294–2305 (2007).

106 Van S, Das SK, Wang XH, Feng ZL, Jin Y, Hou Z, et al., Syn-thesis, characterization, and biological evaluation ofpoly(L-gamma-glutamyl-glutamine)-paclitaxel nanoconjugate.Int J Nanomed 5:825–837 (2010).

107 Portilla-Arias JA, Camargo B, Garcia-Alvarez M, de IlarduyaAM and Munoz-Guerra S, Nanoparticles made of microbialpoly(gamma-glutamate)s for encapsulation and delivery of drugsand proteins. J Biomat Sci - Polym E 20:1065–1079 (2009).

108 Sun Y, Tang YZ, Chu MQ, Song SX and Xin YF, A convenient andadjustable surface-modified complex containing poly-L-glutamicacid conjugates as a vector for gene delivery. Int J Nanomed3:249–256 (2008).

109 Matsusaki M, Yoshida H and Akashi M, The construction of3D-engineered tissues composed of cells and extracellular matri-ces by hydrogel template approach. Biomaterials 28:2729–2737(2007).

110 Hsu FY, Cheng YY, Tsai SW and Tsai WB, Fabrication and evalua-tion of a biodegradable cohesive plug based on reconstitutedcollagen/gamma-polyglutamic acid. J Biomed Mater Res B95B:29–35 (2010).

111 Colonna C, Conti B, Perugini P, Pavanetto F, Modena T, Dorati R, et al.,Chitosan glutamate nanoparticles for protein delivery: develop-ment and effect on prolidase stability. J Microencapsul 24:553–564(2007).

112 Deng C, Tian HY, Zhang PB, Sun J, Chen XS and Jing XB, Synthe-sis and characterization of RGD peptide grafted poly(ethyleneglycol)-b-poly(L-lactide)-b-poly(L-glutamic acid) triblock copoly-mer. Biomacromolecules (2006)7:590–596.

113 Gryparis EC, Mattheolabakis G, Bikiaris D and Avgoustakis K, Effect ofconditions of preparation on the size and encapsulation propertiesof PLGA-mPEG nanoparticles of cisplatin. Drug Deliv 14:371–380(2007).

114 Cao N, Cheng D, Zou SY, Ai H, Gao JM and Shuai XT, The syner-gistic effect of hierarchical assemblies of siRNA and chemother-apeutic drugs co-delivered into hepatic cancer cells. Biomaterials32:2222-2232 (2011).

115 Takehara M, Hibino A, Saimura M and Hirohara H, High-yield produc-tion of short chain length poly(epsilon-l-lysine) consisting of 5-20residues by Streptomyces aureofaciens, and its antimicrobial activ-ity. Biotechnol Lett 32:1299–1303 (2010).

116 Tang CK, Sheng KC, Pouniotis D, Esparon S, Son HY, Kim CW,et al., Oxidized and reduced mannan mediated MUC1 DNAimmunization induce effective anti-tumor responses. Vaccine(2008)26:3827–3834.

117 Isaksson K, Akerberg D, Andersson R and Tingstedt B, Toxic-ity and dose response of intra-abdominally administeredpoly-L-alpha-lysine and poly-L-glutamate for postoperativeadhesion protection. Eur Surg Res 44:17–22 (2010).

118 Bertram JP, Jay SM, Hynes SR, Robinson R, Criscione JM and LavikEB, Functionalized poly(lactic-co-glycolic acid) enhances drugdelivery and provides chemical moieties for surface engineeringwhile preserving biocompatibility. Acta Biomater 5:2860–2871(2009).

119 Tahara K, Furukawa S, Yamamoto H and Kawashima Y,Hybrid-modified poly(D,L-lactide-co-glycolide) nanospheresfor 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 amphiphilicpoly(epsilon-caprolactone)-g-poly(L-lysine) degradable copoly-mers. Biomacromolecules 8:2594–2601 (2007).

121 Chu CFL, Lu A, Liszkowski M and Sipehia R, Enhanced growth of animaland human endothelial cells on biodegradable polymers. Bba - GenSubjects 1472:479–485 (1999).

122 Zhang R and Ma PX, Processing of polymer scaffolds: phase separa-tion, in Methods of Tissue Engineering, ed by Atala A LR. AcademicPress: San Diego; 715 (2001).

123 Pachence JM KJ, Biodegradable polymers, in: Principles of Tissue Engi-neering, 2nd edn, ed by Lanza RP LR, Chick WL. Academic Press: SanDiego; 263 (2000).

124 Bolland BJRF, Kanczler JM, Ginty PJ, Howdle SM, Shakesheff KM,Dunlop DG, et al. The application of human bone marrow stromalcells and poly(DL-lactic acid) as a biological bone graft extender inimpaction bone grafting. Biomaterials 29:3221–3227 (2008).

125 Eschbach L, Nonresorbable polymers in bone surgery. Injury31:S22–S7 (2000).

126 Kurtz SM and Devine JN, PEEK biomaterials in trauma, orthopedic, andspinal implants. Biomaterials 28:4845–4869 (2007).

127 von Wilmowsky C, Vairaktaris E, Pohle D, Rechtenwald T, Lutz R, Mun-stedt H, et al., Effects of bioactive glass and beta-TCP containingthree-dimensional laser sintered polyetheretherketone compos-ites on osteoblasts in vitro. J Biomed Mater Res A 87A:896–902(2008).

128 Sagomonyants KB, Jarman-Smith ML, Devine JN, Aronow MS andGronowicz GA, The in vitro response of human osteoblasts topolyetheretherketone (PEEK) substrates compared to commer-cially pure titanium. Biomaterials 29:1563–1572 (2008).

129 Morrison C, Macnair R, Macdonald C, Wykman A, Goldie I and GrantMH, In vitro biocompatibility testing of polymers for orthopedicimplants using cultured fibroblasts and osteoblasts. Biomaterials16:987–992 (1995).

130 Scotchford CA, Garle MJ, Batchelor J, Bradley J and Grant DM, Use ofa novel carbon fibre composite material for the femoral stem com-ponent of a THR system: in vitro biological assessment. Biomateri-als24:4871–4879 (2003).

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 14: ozdil2014

1806

www.soci.org D Ozdil, HM Aydin

131 Katzer A, Marquardt H, Westendorf J, Wening JV and von FoersterG, Polyetheretherketone - cytotoxicity and mutagenicity in vitro.Biomaterials 23:1749–1759 (2002).

132 von Wilmowsky C, Lutz R, Meisel U, Srour S, Rupprecht S, ToyoshimaT, et al., In vivo evaluation of beta-TCP containing 3D laser sinteredpoly(ether ether ketone) composites in pigs. J Bioact Compat Pol24:169–184 (2009).

133 Nieminen T, Kallela I, Wuolijoki E, Kainulainen H, Hiidenheimo Iand Rantala I, Amorphous and crystalline polyetheretherke-tone: mechanical properties and tissue reactions during a 3-yearfollow-up. J Biomed Mater Res A 84A:377–383 (2008).

134 Jockisch KA, Brown SA, Bauer TW and Merritt K, Biological responseto chopped-carbon-fiber-reinforced peek. J Biomed Mater Res26:133-146 (1992).

135 Scholes SC and Unsworth A, Pitch-based carbon-fibre-reinforced poly(ether-ether-ketone) OPTIMA (R) assessed as a bearing material ina mobile bearing unicondylar knee joint. P I Mech Eng H 223:13–25(2009).

136 Toth JM, Wang M, Estes BT, Scifert JL, Seim HB and Turner AS,Polyetheretherketone as a biomaterial for spinal applications. Bio-materials 27:324–334 (2006).

137 Scolozzi P, Martinez A and Jaques B, Complex orbito-fronto-temporalreconstruction using computer-designed PEEK implant. J CraniofacSurg 18:224–228 (2007).

138 Zhao Y, Wong HM, Wang W, Li P, Xu Z, Chong EY, et al., Cytocom-patibility, osseointegration, and bioactivity of three-dimensionalporous and nanostructured network on polyetheretherketone. Bio-materials 34:9264–9277 (2013).

139 Edwards SL and Werkmeister JA, Mechanical evaluation and cellresponse of woven polyetheretherketone scaffolds. J Biomed MaterRes A 100:3326–3331 (2012).

140 Roeder RK and Conrad TL, Bioactive polyaryletherketone composites,in PEEK Biomaterials Handbook, ed by Kurtz SM. William Andrew:Oxford, UK; 163–79 (2012).

141 Converse GL, Conrad TL, Merrill CH and Roeder RK, Hydroxyapatitewhisker-reinforced polyetherketoneketone bone ingrowth scaf-folds. Acta Biomater 6:856–863 (2012).

142 Yu SC, Hariram KP, Kumar R, Cheang P and Aik KK, In vitroapatite formation and its growth kinetics on hydroxya-patite/polyetheretherketone biocomposites. Biomaterials26:2343–2452 (2005).

143 Wong KL, Wong CT, Liu WC, Pan HB, Fong MK, Lam WM, et al., Mechan-ical properties and in vitro response of strontium-containinghydroxyapatite/polyetheretherketone composites. Biomaterials30:3810–3817 (2009).

144 Fujihara K, Huang ZM, Ramakrishna S, Satknanantham K and HamadaH, Performance study of braided carbon/PEEK composite compres-sion bone plates. Biomaterials 24:2661–2667 (2003).

145 Fujihara K, Huang ZM, Ramakrishna S, Satknanantham K and HamadaH, Feasibility of knitted carbon/PEEK composites for orthopedicbone plates. Biomaterials 25:3877–3885 (2004).

146 Karageorgiou V and Kaplan D, Porosity of 3D biomaterial scaffolds andosteogenesis. Biomaterials 26:5474–5491 (2005).

147 Chu PP, Wu CS, Liu PC, Wang TH and Pan JP, Proton exchangemembrane bearing entangled structure: sulfonated poly (etherether ketone)/bismaleimide hyperbranch. Polymer 51:1386–1394(2010).

148 Ryan G, Pandit A and Apatsidis DP, Fabrication methods ofporous metals for use in orthopaedic applications. Biomaterials27:2651–2670 (2006).

149 Mohanna PN, Young RC, Wiberg M and Terenghi G, A compositepoly-hydroxybutyrate-glial growth factor conduit for long nervegap repairs. J Anat 203:553–565 (2003).

150 Misra SK, Valappil SP, Roy I and Boccaccini AR, Polyhydroxyalkanoate(PHA)/inorganic phase composites for tissue engineering applica-tions. Biomacromolecules 7:2249–2258 (2006).

151 Reusch RN, Sparrow AW and Gardiner J, Transport ofpoly-beta-hydroxybutyrate in human plasma. Biochim BiophysActa 1123:3340 (1992).

152 Bergstrand A, Uppstrom S and Larsson A, Permeability of porouspoly(3-hydroxybutyrate) barriers of single and bilayer type forimplant applications. Int J Polym Sci (2014).

153 Masaeli E, Morshed M, Rasekhian P, Karbasi S, Karbalaie K, Kara-mali F, Abedi D, Razavi S, Jafarian-Dehkordi A, Nasr-Esfahani MHand Baharvand H, Does the tissue engineering architecture of

poly(3-hydroxybutyrate) scaffold affects cell-material interactions?J Biomed Mater Res A 100A:1907–1918 (2012).

154 Cai ZJ, Biocompatibility and biodegradation of novel PHB poroussubstrates with controlled multi-pore size by emulsion templatesmethod. J Mater Sci - Mater M 17:1297–1303 (2006).

155 Karahaliloglu Z, Demirbilek M, Sam M, Erol-Demirbilek M, SaglamN and Denkbas EB, Plasma polymerization-modified bacterialpolyhydroxybutyrate nanofibrillar scaffolds. J Appl Polym Sci128:19041912 (2013).

156 Seal BL, Otero TC and Panitch A, Polymeric biomaterials for tissue andorgan regeneration. Mat Sci Eng R 34:147–230 (2001).

157 Maurus PB and Kaeding CC, Bioabsorbable implant material review.Oper Techn Sport Med 12:158–160 (2004).

158 Ma PX and Langer R, Degradation, structure and properties of fibrousnonwoven poly(glycolic acid) scaffolds for tissue engineering.Mater Res Soc Symp P 394:99–104 (1995).

159 Reed AM and Gilding DK, Biodegradable polymers for use in surgery- poly(glycolic)-poly(lactic acid) homo and co-polymers .2. Invitrodegradation. Polymer 22:494–498 (1981).

160 Gilding DK and Reed AM, Biodegradable polymers for use in surgery- polyglycolic-poly(actic acid) homopolymers and copolymers.P olymer 20:1459–1464 (1979).

161 Aydin HM, A three-layered osteochondral plug: structural,mechanical, and in vitro compatibility analysis. Adv Eng Mater13:B511–B517 (2011).

162 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 InvestSurg 23:190–196 (2010).

163 Ceonzo K, Gaynor A, Shaffer L, Kojima K, Vacanti CA and Stahl GL,Polyglycolic acid-induced inflammation: role of hydrolysis andresulting complement activation. Tissue Eng 12:301–308 (2006).

164 Zhang RY and Ma PX, Degradation behavior of porouspoly(alpha-hydroxy acids)/hydroxyapatite composite scaffolds.Abstr Papers Am Chem S 220:U251 (2000).

165 Eling B, Gogolewski S and Pennings AJ, Biodegradable materials ofpoly(L-lactic acid) .1. Melt-spun and solution-spun fibers. Polymer23:1587–1593 (1982).

166 Aydin HM, El Haj AJ, Piskin E, Yang Y, Improving pore interconnectivityin polymeric scaffolds for tissue engineering. J Tiss Eng Reg Med3:470–476 (2009).

167 Aydin HM and Yang Y, Rapid production of highly interconnedtedporous scaffolds by spheroidized sugar particles for tissue engi-neering. Journal of Hacettepe Biology and Chemistry 39:23–28(2011).

168 Anderson KS, Schreck KM and Hillmyer MA, Toughening polylactide.Polym Rev 48:85–108 (2008).

169 Ueda H and Tabata Y, Polyhydroxyalkanonate derivatives in currentclinical applications and trials. Adv Drug Deliver Rev 55:501–518(2003).

170 Thomas C, Gupta V and Ahsan F, Particle Size influences the immuneresponse produced by Hepatitis B vaccine formulated in inhalableparticles. Pharm Res - Dordr 27:905–919 (2010).

171 Zolnik BS and Burgess DJ, Evaluation of in vivo-in vitro releaseof dexamethasone from PLGA microspheres. J Control Release127:137–145 (2008).

172 Gavenis K, Schneider U, Groll J and Schmidt-Rohlfing B, BMP-7-loadedPGLA microspheres as a new delivery system for the cultivation ofhuman chondrocytes in a collagen type I gel: the common nudemouse model. Int J Artif Organs 33:45–53 (2010).

173 Ye ML, Kim S and Park K, Issues in long-term protein deliveryusing biodegradable microparticles. J Control Release 146:241–260(2010).

174 Berkland C, Pollauf E, Varde N, Pack DW and Kim K, Monodis-perse liquid-filled biodegradable microcapsules. Pharm Res - Dordr24:1007–1013 (2007).

175 Haddadi A, Aboofazeli R, Erfan M and Farboud ES, Topical delivery ofurea encapsulated in biodegradable PLGA microparticles: O/W andW/O creams. J Microencapsul 25:379–386 (2008).

176 Vega E, Gamisans F, Garcia ML, Chauvet A, Lacoulonche F and EgeaMA, PLGA nanospheres for the ocular delivery of flurbiprofen: drugrelease and interactions. J Pharm Sci - Us 97:5306–5317 (2008).

177 Xie JW and Wang CH, Electrospun micro- and nano-fibers for sus-tained delivery of paclitaxel to treat C6 glioma in vitro. Pharm Res- Dordr 23:1817–1826 (2006).

178 Ionescu LC, Lee GC, Sennett BJ, Burdick JA and Mauck RL, Ananisotropic nanofiber/microsphere composite with controlled

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 15: ozdil2014

1807

Polymers for medical and tissue engineering applications www.soci.org

release of biomolecules for fibrous tissue engineering. Biomaterials31:4113–4120 (2010).

179 Miller RA, Brady JM and Cutright DE, Degradation rates of oralresorbable implants (polylactates and polyglycolates) - rate mod-ification with changes in Pla-Pga copolymer ratios. J Biomed MaterRes 11:711–719 (1977).

180 Freed LE, Vunjaknovakovic G, Biron RJ, Eagles DB, Lesnoy DC, BarlowSK, et al., Biodegradable polymer scaffolds for tissue engineering.Bio - Technol 12:689–693 (1994).

181 Patrick JC MA and Mclntire VL, Frontiers in Tissue Engineering. Perga-mon: New York (1998).

182 Patlolla A, Collins G and Arinzeh TL, Solvent-dependent properties ofelectrospun fibrous composites for bone tissue regeneration. ActaBiomater 6:90–101 (2010).

183 Lam CXF, Hutmacher DW, Schantz JT, Woodruff MA and Teoh SH,Evaluation of polycaprolactone scaffold degradation for 6 monthsin vitro and in vivo. J Biomed Mater Res A 90A:906–919 (2009).

184 Woodruff MA and Hutmacher DW, The return of a forgotten polymer -Polycaprolactone in the 21st century. Prog Polym Sci 35:1217–1256(2010).

185 Aydin HM, Calimli A and Piskin E, Microporous scaffolds from poly(lactide-co-e-caprolactone) composites with hydroxyapatite andtricalcium phosphate using supercritical CO2 for bone tissue engi-neering. J Bioact Compat Pol 19:383–394 (2004).

186 Dash TK and Konkimalla VB, Poly-epsilon-caprolactone based formu-lations for drug delivery and tissue engineering: a review. J ControlRelease 158:15–33 (2012).

187 Aydin HM, Korkusuz P, Vargel I, Kilic E, Guzel E, Cavusoglu T, Ucgan Dand Piskin E, A 6-month in vivo study of polymer/mesenchymalstem cell constructs for cranial defects. J Bioact Compat Pol26:207–221 (2011).

188 Aydin HM, Turk M, Calimli A and Piskin E, Attachment and growth offibroblasts on poly(L-lactide/epsilon-caprolactone) scaffolds pre-pared in supercritical CO2 and modified by polyethylene iminegrafting with ethylene diamine-plasma in a glow-discharge appa-ratus. Int J Artif Organs 29:873–880 (2006).

189 Li WJ, Danielson KG, Alexander PG and Tuan RS, Biological responseof chondrocytes cultured in three-dimensional nanofibrouspoly(epsilon-caprolactone) scaffolds. J Biomed Mater Res A67A:1105–1114 (2003).

190 Hutmacher DW, Scaffolds in tissue engineering bone and cartilage.Biomaterials 21:2529–2543 (2000).

191 Li WJ, Tuli R, Okafor C, Derfoul A, Danielson KG, Hall DJ, et al., Athree-dimensional nanofibrous scaffold for cartilage tissue engi-neering using human mesenchymal stem cells. Biomaterials26:599–609 (2005).

192 CG. P. Poly-epsilon-caprolactone and its copolymers, in Biodegrad-able Polymers as Drug Delivery Systems, ed by Chasin M LR. MarcelDekker: New York; 71–120 (1990).

193 Gan ZH, Yu DH, Zhong ZY, Liang QZ and Jing XB, Enzymatic degra-dation of poly(epsilon-caprolactone)/poly(DL-lactide) blends inphosphate buffer solution. Polymer 40:2859–2862 (1999).

194 Assoian RK, Komoriya A, Meyers CA, Miller DM and Sporn MB, Trans-forming growth factor-beta in human-platelets - identification ofa major storage site, purification, and characterization. J Biol Chem258:7155–7160 (1983).

195 Joyce ME, Roberts AB, Sporn MB and Bolander ME, Transform-ing growth-factor-beta and the initiation of chondrogenesis andosteogenesis in the rat femur. J Cell Biol 110:2195–2207 (1990).

196 Ye WP, Du FS, Jin JY, Yang JY and Xu Y, In vitro degradation ofpoly(caprolactone), poly(lactide) and their block copolymers: influ-ence of composition, temperature and morphology. React FunctPolym 32:161–168 (1997).

197 Guelcher SA, Biodegradable polyurethanes: synthesis and applica-tions in regenerative medicine. Tissue Eng Pt B - Rev 14:3–17 (2008).

198 Guan JJ, Fujimoto KL, Sacks MS and Wagner WR, Preparation andcharacterization of highly porous, biodegradable polyurethanescaffolds for soft tissue applications. Biomaterials 26:3961–3971(2005).

199 Shi R, Chen DF, Liu QY, Wu Y, Xu XC, Zhang LQ, et al., Recent advancesin synthetic bioelastomers. Int J Mol Sci 10:4223–4256 (2009).

200 Bonzani IC, Adhikari R, Houshyar S, Mayadunne R, GunatillakeP and Stevens MM, Synthesis of two-component injectablepolyurethanes for bone tissue engineering. Biomaterials28:423–433 (2007).

201 Fisher JP, Vehof JWM, Dean D, van der Waerden JPCM, Holland TA,Mikos AG, et al., Soft and hard tissue response to photocrosslinkedpoly(propylene fumarate) scaffolds in a rabbit model. J BiomedMater Res 59:547–556 (2002).

202 Hedberg EL, Kroese-Deutman HC, Shih CK, Crowther RS, Carney DH,Mikos AG, et al., Effect of varied release kinetics of the osteogenicthrombin peptide TP508 from biodegradable, polymeric scaffoldson bone formation in vivo. J Biomed Mater Res A 72A:343–353(2005).

203 He S, Timmer MD, Yaszemski MJ, Yasko AW, Engel PS and Mikos AG,Synthesis of biodegradable poly(propylene fumarate) networkswith poly(propylene fumarate)-diacrylate macromers as crosslink-ing agents and characterization of their degradation products.Polymer 42:1251–1260 (2001).

204 Christenson EM, Soofi W, Holm JL, Cameron NR and Mikos AG,Biodegradable fumarate-based polyHIPEs as tissue engineeringscaffolds. Biomacromolecules 8:3806–3814 (2007).

205 Kim CW, Talac R, Lu LC, Moore MJ, Currier BL and Yaszemski MJ, Char-acterization of porous injectable poly-(propylene fumarate)-basedbone graft substitute. J Biomed Mater Res A 85A:1114–1119(2008).

206 Young S, Patel ZS, Kretlow JD, Murphy MB, Mountziaris PM, BaggettLS, et al., Dose effect of dual delivery of vascular endothelial growthfactor and bone morphogenetic protein-2 on bone regeneration ina rat critical-size defect model. Tissue Eng Pt A (2009)15:2347–2362.

207 Vehof JWM, Fisher JP, Dean D, van der Waerden JPCM, Spauwen PHM,Mikos AG, et al., Bone formation in transforming growth factorbeta-1-coated porous poly(propylene fumarate) scaffolds. J BiomedMater Res 60:241–251 (2002).

208 Lee KW, Wang SF, Fox BC, Ritman EL, Yaszemski MJ and Lu LC,Poly(propylene fumarate) bone tissue engineering scaffold fabrica-tion using stereolithography: effects of resin formulations and laserparameters. Biomacromolecules8:1077–1084 (2007).

209 Lee J, Kim B, Lim G and Cho DW, Scaffold fabrication with biodegrad-able poly(propyllene fumarate) using microstereolithography. KeyEng Mater 342–343:141–144 (2007).

210 Wolfe MS, Dean D, Chen JE, Fisher JP, Han SH, Rimnac CM, et al. Invitro degradation and fracture toughness of multilayered porouspoly(propylene fumarate)/beta-tricalcium phosphate scaffolds.J Biomed Mater Res 61:159–164 (2002).

211 Kharas GB, Kamenetsky M, Simantirakis J, Beinlich KC, Rizzo AMT, Cay-wood GA, et al., Synthesis and characterization of fumarate-basedpolyesters for use in bioresorbable bone cement composites. J ApplPolym Sci 66:1123–1137 (1997).

212 Hedberg EL, Temenoff JS, Tang A, Crowther RS, Carney DH andMikos AG, Controlled release of a tissue inducing peptide fromoligo(poly(ethylene glycol) fumarate) hydrogels for orthopedic tis-sue engineering. 24th Annual Conference and the Annual FallMeeting of the Biomedical Engineering Society EMBS/BMES Con-ference, 2002. IEEE; 1:472–473 (2002).

213 Wang SF, Kempen DH, Simha NX, Lewis JL, Windebank AJ, Yaszem-ski MJ, et al., Photo-cross-linked hybrid polymer networks consist-ing of poly(propylene fumarate) and poly(caprolactone fumarate):controlled physical properties and regulated bone and nerve cellresponses. Biomacromolecules 9:1229–1241 (2008).

214 Payne RG, McGonigle JS, Yaszemski MJ, Yasko AW and Mikos AG,Development of an injectable, in situ crosslinkable, degradablepolymeric carrier for osteogenic cell populations. Part 2. Viability ofencapsulated marrow stromal osteoblasts cultured on crosslinkingpoly(propylene fumarate). Biomaterials 23:4373–4380 (2002).

215 Nam YS and Park TG, Porous biodegradable polymeric scaffoldsprepared by thermally induced phase separation. J Biomed MaterRes 47:8–17 (1999).

216 Lewandrowski KU, Cattaneo MV, Gresser JD, Wise DL, White RL,Bonassar L, et al. Effect of a poly(propylene fumarate) foamingcement on the healing of bone defects. Tissue Eng 5:305–316(1999).

217 Peter SJ, Miller ST, Zhu GM, Yasko AW and Mikos AG, In vivo degra-dation of a poly(propylene fumarate) beta-tricalcium phosphateinjectable composite scaffold. J Biomed Mater Res 41:1–7 (1998).

218 Hedberg EL, Kroese-Deutman HC, Shih CK, Crowther RS, Carney DH,Mikos AG, et al., In vivo degradation of porous poly(propylenefumarate)/poly(DL-lactic-co-glycolic acid) composite scaffolds. Bio-materials 26:4616–4623 (2005).

219 Lukashev M, Zeng W, Goelz S, Lee D, Linker R, Gold R, et al., Activa-tion of Nrf2 and modulation of disease progression in EAE models

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 16: ozdil2014

1808

www.soci.org D Ozdil, HM Aydin

by BG00012 (dimethyl fumarate) suggests a novel mechanism ofaction combining anti-inflammatory and neuroprotective modali-ties. Mult Scler 13:S149 (2007).

220 Lee KW, Wang S, Yaszemski MJ and Lu LC, Physical proper-ties and cellular responses to crosslinkable poly(propylenefumarate)/hydroxyapatite nanocomposites. Biomaterials 29:2839–2848 (2008).

221 Bourke SL and Kohn J, Polymers derived from the amino acidL-tyrosine: polycarbonates, polyarylates and copolymers withpoly(ethylene glycol). Adv Drug Deliver Rev 55:447–466 (2003).

222 Yuan H, Luo K, Lai YS, Pu YJ, He B, Wang G, et al., A novelpoly(L-glutamic acid) dendrimer based drug delivery systemwith both pH-sensitive and targeting functions. Mol Pharmaceut7:953–962 (2010).

223 Wadhwa S and Mumper RJ, Intracellular delivery of the reactive oxy-gen species generating agent D-penicillamine upon conjugationto poly-L-glutamic acid. Mol Pharmaceut 7:854–862 (2010).

224 Chiu HC, Kopeckova P, Deshmane SS and Kopecek J, Lysosomaldegradability of poly(alpha-amino acids). J Biomed Mater Res34:381–392 (1997).

225 Feng XL, Lv FT, Liu LB, Tang HW, Xing CF, Yang QO, et al. Conjugatedpolymer nanoparticles for drug delivery and imaging. Acs ApplMater Inter 2:2429–2435 (2010).

226 Ke TY, Feng Y, Guo JY, Parker DL and Lu ZR, Biodegradable cystaminespacer facilitates the clearance of Gd(III) chelates in poly(glutamicacid) Gd-DO3A conjugates for contrast-enhanced MR imaging.Magn Reson Imaging 24:931–940 (2006).

227 Kakizawa Y, Furukawa S and Kataoka K, Block copolymer-coated cal-cium phosphate nanoparticles sensing intracellular environmentfor oligodeoxynucleotide and siRNA delivery. J Control Release97:345–356 (2004).

228 Karal-Yilmaz O, Kayaman-Apohan N, Misirli Z, Baysal K and Baysal BM,Synthesis and characterization of poly(L-lactic acid-co-ethyleneoxide-co-aspartic acid) and its interaction with cells. J Mater Sci -Mater Med 17:213–227 (2006).

229 Cha EJ, Kim JE and Ahn CH, Stabilized polymeric micelles by elec-trostatic interactions for drug delivery system. Eur J Pharm Sci38:341–346 (2009).

230 Ng SY, Vandamme T, Taylor MS and Heller J, Synthesis and ero-sion studies of self-catalyzed poly(ortho ester)s. Macromolecules30:770–772 (1997).

231 Heller J, Barr J, Ng SY, Abdellauoi KS and Gurny R, Poly(ortho esters):synthesis, characterization, properties and uses. Adv Drug DeliverRev 54:1015–1039 (2002).

232 Heller J and Barr J, Poly(ortho esters) - from concept to reality.Biomacromolecules 5:1625–1632 (2004).

233 Kipper MJ, Wilson JH, Wannemuehler MJ and Narasimhan B, Sin-gle dose vaccine based on biodegradable polyanhydride micro-spheres can modulate immune response mechanism. J BiomedMater Res A 76A:798–810 (2006).

234 Carrillo-Conde B, Schiltz E, Yu J, Minion FC, Phillips GJ, WannemuehlerMJ, et al., Encapsulation into amphiphilic polyanhydridemicroparticles stabilizes Yersinia pestis antigens. Acta Biomater6:3110–3119 (2010).

235 Sun L, Zhou SB, Wang WJ, Su QX, Li XH and Weng J, Preparation andcharacterization of protein-loaded polyanhydride microspheres.J Mater Sci - Mater M 20:2035–2042 (2009).

236 Salman HH, Irache JM and Gamazo C, Immunoadjuvant capacity offlagellin and mannosamine-coated poly(anhydride) nanoparticlesin oral vaccination. Vaccine 27:4784–4790 (2009).

237 Tamayo I, Irache JM, Mansilla C, Ochoa-Reparaz J, Lasarte JJ andGamazo C, Poly(anhydride) nanoparticles act as active Th1 adju-vants through toll-Like receptor exploitation. Clin Vaccine Immunol17:1356–1362 (2010).

238 Petersen LK, Sackett CK and Narasimhan B, High-throughput analysisof protein stability in polyanhydride nanoparticles. Acta Biomater6:3873–3881 (2010).

239 Ibim SEM, Uhrich KE, Attawia M, Shastri VR, El-Amin SF, BronsonR, et al., Preliminary in vivo report on the osteocompatibility ofpoly(anhydride-co-imides) evaluated in a tibial model. J BiomedMater Res 43:374–379 (1998).

240 Weiner AA, Shuck DM, Bush JR and Shastri VP, In vitro degradationcharacteristics of photocrosslinked anhydride systems for boneaugmentation applications. Biomaterials 28:5259–5270 (2007).

241 Determan AS, Trewyn BG, Lin VSY, Nilsen-Hamilton M and NarasimhanB, Encapsulation, stabilization, and release of BSA-FITC frompolyanhydride microspheres. J Control Release 100:97–109 (2004).

242 Weiner AA, Bock EA, Gipson ME and Shastri VP, Photocrosslinkedanhydride systems for long-term protein release. Biomaterials29:2400–2407 (2008).

243 Attawia MA, Uhrich KE, Botchwey E, Fan M, Langer R and Laurencin CT,Cytotoxicity testing of poly(anhydride-co-imides) for orthopedicapplications. J Biomed Mater Res 29:1233–1240 (1995).

244 Uhrich KE, Gupta A, Thomas TT, Laurencin CT and Langer R, Synthe-sis and characterization of degradable poly(anhydride-co-imides).Macromolecules 28:2184–2193 (1995).

245 Wang YD, Ameer GA, Sheppard BJ and Langer R, A tough biodegrad-able elastomer. Nat Biotechnol 20:602–606 (2002).

246 Vert M, Li SM, Spenlehauer G and Guerin P, Bioresorbability andbiocompatibility of aliphatic polyesters. J Mater Sci - Mater M3:432–446 (1992).

247 Aydin HM, Kourush S, Rzayev Z, Piskin E, Microwave-assistedRapid Synthesis of Poly(glycerol-sebacate) Elastomers. BiomatSci 1:503–509 (2013).

248 Aydin HM, Kourush S, Yilmaz M, Turk M, Rzayev Z, Piskin E,The Catalyst Assisted Two Stage Synthesis of Poly(glycerol-co-sebacate-co-e-caprolactone) Elastomers As Potential TissueEngineering Materials. J Tiss Eng Reg DOI: 10.1002/term.1759(2012).

249 Kemppainen JM and Hollister SJ, Tailoring the mechanical proper-ties of 3D-designed poly(glycerol sebacate) scaffolds for cartilageapplications. J Biomed Mater Res A 94A:9–18 (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 29:47–57 (2008).

251 Liu QY, Tian M, Shi R, Zhang LQ, Chen DF and Tian W, Structureand properties of thermoplastic poly(glycerol sebacate) elastomersoriginating from prepolymers with different molecular weights.J Appl Polym Sci 104:1131–1137 (2007).

252 Ifkovits JL, Padera RF and Burdick JA, Biodegradable and radicallypolymerized elastomers with enhanced processing capabilities.Biomed Mater 3:034104 (2008).

253 Cai W and Liu LL, Shape-memory effect of poly (glycerol-sebacate)elastomer. Mater Lett 62:2171–2173 (2008).

254 Jaafar IH, Ammar MM, Jedlicka SS, Pearson RA and Coulter JP,Spectroscopic evaluation, thermal, and thermomechanicalcharacterization of poly(glycerol-sebacate) with variations incuring temperatures and durations. J Mater Sci 45:2525–2529(2010).

255 Komatsu K and Chiba M, The effect of velocity of loading on thebiomechanical responses of the periodontal-ligament in trans-verse sections of the rat molar in vitro. Arch Oral Biol 38:369–375(1993).

256 Sundback CA, Shyu JY, Wang YD, Faquin WC, Langer RS, Vacanti JP,et al., Biocompatibility analysis of poly(glycerol sebacate) as a nerveguide material. Biomaterials 26:5454–5464 (2005).

257 Crapo PM and Wang YD, Physiologic compliance in engineeredsmall-diameter arterial constructs based on an elastomeric sub-strate. Biomaterials 31:1626–1635 (2010).

258 Gao J, Ensley AE, Nerem RM and Wang YD, Poly(glycerol sebacate)supports the proliferation and phenotypic protein expres-sion of primary baboon vascular cells. J Biomed Mater Res A83A:1070–1075 (2007).

259 Chen QZ, Jin LY, Cook WD, Mohn D, Lagerqvist EL, Elliott DA, et al.,Elastomeric nanocomposites as cell delivery vehicles and cardiacsupport devices. Soft Matter. 6:4715–4726 (2010).

260 Sales VL, Engelmayr GC, Johnson JA, Gao J, Wang YD, Sacks MS,et al., Protein precoating of elastomeric tissue-engineering scaf-folds increased cellularity, enhanced extracellular matrix proteinproduction, and differentially regulated the phenotypes of circu-lating endothelial progenitor cells. Circulation 116:I55–I63 (2007).

261 Chen QZ, Liang SL, Wang J and Simon GP, Manipulation of mechan-ical compliance of elastomeric PGS by incorporation of halloysitenanotubes for soft tissue engineering applications. J Mech BehavBiomed 4:1805–1818 (2011).

262 Sant S, Hwang CM, Lee SH and Khademhosseini A, Hybrid PGS-PCLmicrofibrous scaffolds with improved mechanical and biologicalproperties. J Tissue Eng Regen M 5:283–291 (2011).

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810

Page 17: ozdil2014

1809

Polymers for medical and tissue engineering applications www.soci.org

263 Laurencin CT, Koh HJ, Neenan TX, Allcock HR and Langer R, Controlledrelease using a new bioerodible polyphosphazene matrix system.J Biomed Mater Res 21:1231–1246 (1987).

264 Qiu LY and Zhu KJ, Novel biodegradable polyphosphazenes contain-ing glycine ethyl ester and benzyl ester of amino acethydroxamicacid as cosubstituents: syntheses, characterization, and degrada-tion properties. J Appl Polym Sci 77:2987–2995 (2000).

265 Behravesh E, Yasko AW, Engel PS and Mikos AG, Synthetic biodegrad-able polymers for orthopaedic applications. Clin Orthop Relat R367:S118–S129 (1999).

266 Allcock HR, Inorganic-organic polymers as a route to biodegradablematerials. Macromol Symp 144:33–46 (1999).

267 Lakshmi S, Katti DS and Laurencin CT, Biodegradable polyphos-phazenes for drug delivery applications. Adv Drug Deliver Rev55:467–482 (2003).

268 Weikel AL, Owens SG, Morozowich NL, Deng M, Nair LS, LaurencinCT, et al., Miscibility of choline-substituted polyphosphazenes withPLGA and osteoblast activity on resulting blends. Biomaterials31:8507–8515 (2010).

269 Singh A, Krogman NR, Sethuraman S, Nair LS, Sturgeon JL, BrownPW, et al., Effect of side group chemistry on the propertiesof biodegradable L-alanine cosubstituted polyphosphazenes.Biomacromolecules. 7:914–918 (2006).

270 Verret V, Wassef M, Pelage JP, Ghegediban SH, Jouneau L, MoineL, et al., Influence of degradation on inflammatory profile ofpolyphosphazene coated PMMA and trisacryl gelatin micro-spheres in a sheep uterine artery embolization model. Biomaterials32:339–351 (2011).

271 Stampfl U, Sommer CM, Thierjung H, Stampfl S, Lopez-Benitez R,Radeleff B, et al. Reduction of Late In-Stent Stenosis in a PorcineCoronary Artery Model by Cobalt Chromium Stents with aNanocoat of Polyphosphazene (Polyzene-F). Cardiovasc Inter Rad31:1184–1192 (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 poly-meric scaffolds. J Biomed Mater Res A 71A:669–674 (2004).

273 Zhang QS, Yan YH, Li SP and Feng T, The synthesis and characteriza-tion of a novel biodegradable and electroactive polyphosphazenefor nerve regeneration. Mat Sci Eng C - Mater 30:160–166 (2010).

274 Deng M, Nair LS, Nukavarapu SP, Kumbar SG, Jiang T, Krogman NR,et al., Miscibility and in vitro osteocompatibility of biodegradableblends of poly[(ethyl alanato) (p-phenyl phenoxy) phosphazene]and poly(lactic acid-glycolic acid). Biomaterials 29:337–349 (2008).

275 Nair LS, Bhattacharyya S, Bender JD, Greish YE, Brown PW, AllcockHR, et al., Fabrication and optimization of methylphenoxy substi-tuted polyphosphazene nanofibers for biomedical applications.Biomacromolecules 5:2212–2220 (2004).

276 Potta T, Chun C and Song SC, Chemically crosslinkable thermosensi-tive polyphosphazene gels as injectable materials for biomedicalapplications. Biomaterials 30:6178–6192 (2009).

277 Park MR, Chun CJ, Ahn SW, Ki MH, Cho CS and Song SC, Sus-tained delivery of human growth hormone using a polyelec-trolyte complex-loaded thermosensitive polyphosphazenehydrogel. J Control Release 147:359–367 (2010).

278 Qiu ZC, Zhang JJ, Zhou Y, Song BY, Chang JJ, Yang KK, et al.,Biodegradable poly(p-dioxanone) reinforced and toughened byorgano-modified vermiculite. Polym Advan Technol 22:993–1000(2011).

279 Malnati GA and Stone EA, Clinical-experience with polydioxanonesuture material. Vet Surg 12:24–25 (1983).

280 Elisseeff J, McIntosh W, Anseth K, Riley S, Ragan P and Langer R, Pho-toencapsulation of chondrocytes in poly(ethylene oxide)-basedsemi-interpenetrating networks. J Biomed Mater Res 51:164–171(2000).

281 Alcantar NA, Aydil ES and Israelachvili JN, Polyethylene glycol-coatedbiocompatible surfaces. J Biomed Mater Res 51:343–351 (2000).

282 Gombotz WR, Guanghui W, Horbett TA and Hoffman AS, Proteinadsorption to poly(ethylene oxide) surfaces. J Biomed Mater Res25:1547–1562 (1991).

283 Ostuni E, Chapman RG, Holmlin RE, Takayama S and Whitesides GM,A survey of structure-property relationships of surfaces that resistthe adsorption of protein. Langmuir 17:5605–5620 (2001).

284 Zhu JM, Bioactive modification of poly(ethylene glycol) hydrogels fortissue engineering. Biomaterials 31:4639–4656 (2010).

285 Burdick JA and Anseth KS, Photoencapsulation of osteoblasts ininjectable RGD-modified PEG hydrogels for bone tissue engineer-ing. Biomaterials 23:4315–4323 (2002).

286 Papadopoulos A, Bichara DA, Zhao X, Ibusuki S, Randolph MA, AnsethKS, et al., Injectable and photopolymerizable tissue-engineeredauricular cartilage using poly(ethylene glycol) dimethacrylatecopolymer hydrogels. Tissue Eng Pt A 17:161–169 (2011).

287 Elisseeff J, Anseth K, Sims D, McIntosh W, Randolph M, Yarem-chuk M, et al., Transdermal photopolymerization of poly(ethyleneoxide)-based injectable hydrogels for tissue-engineered cartilage.Plast Reconstr Surg 104:1014–1022 (1999).

288 Burdick JA, Mason MN, Hinman AD, Thorne K and Anseth KS, Deliveryof osteoinductive growth factors from degradable PEG hydrogelsinfluences osteoblast differentiation and mineralization. J ControlRelease 83:53–63 (2002).

289 Elisseeff J, McIntosh W, Fu K, Blunk T and Langer R, Controlled-releaseof IGF-I and TGF-beta 1 in a photopolymerizing hydrogel for carti-lage tissue engineering. J Orthopaed Res 19:1098–1104 (2001).

290 Williams CG, Kim TK, Taboas A, Malik A, Manson P and Elisseeff J,In vitro chondrogenesis of bone marrow-derived mesenchymalstem cells in a photopolymerizing hydrogel. Tissue Eng 9:679–688(2003).

291 Cruise GM, Hegre OD, Scharp DS and Hubbell JA, A sensitivity studyof the key parameters in the interfacial photopolymerization ofpoly(ethylene glycol) diacrylate upon porcine islets. BiotechnolBioeng 57:655–665 (1998).

292 Yang F, Williams CG, Wang DA, Lee H, Manson PN and Elisseeff J, Theeffect of incorporating RGD adhesive peptide in polyethylene gly-col diacrylate hydrogel on osteogenesis of bone marrow stromalcells. Biomaterials 26:5991–5998 (2005).

293 Varghese S, Hwang NS, Canver AC, Theprungsirikul P, Lin DW andElisseeff J, Chondroitin sulfate based niches for chondrogenicdifferentiation of mesenchymal stem cells. Matrix Biol 27:12–21(2008).

294 Sharma B, Williams CG, Khan M, Manson P and Elisseeff JH, In vivochondrogenesis of mesenchymal stem cells in a photopolymerizedhydrogel. Plast Reconstr Surg 119:112–120 (2007).

295 Hwang NS, Varghese S, Theprungsirikul P, Canver A and Elisseeff J,Enhanced chondrogenic differentiation of murine embryonic stemcells in hydrogels with glucosamine. Biomaterials 27:6015-6023(2006).

296 Shin H, Jo S and Mikos AG, Biomimetic materials for tissue engineer-ing. Biomaterials 24:4353–4364 (2003).

297 Lepoittevin B, Devalckenaere M, Pantoustier N, Alexandre M, Kubies D,Calberg C, et al., Poly(epsilon-caprolactone)/clay nanocompositesprepared by melt intercalation: mechanical, thermal and rheologi-cal properties. Polymer 43:4017–4023 (2002).

298 Koegler WS and Griffith LG, Osteoblast response to PLGA tissueengineering scaffolds with PEO modified surface chemistriesand demonstration of patterned cell response. Biomaterials 25:2819–2830 (2004).

299 Li G, Gill TJ, DeFrate LE, Zayontz S, Glatt V and Zarins B, Biomechanicalconsequences of PCL deficiency in the knee under simulatedmuscle loads - an in vitro experimental study. J Orthopaed Res20:887–892 (2002).

300 Kang XH, Xie YB, Powell HM, Lee LJ, Belury MA, Lannutti JJ, et al., Adi-pogenesis of murine embryonic stem cells in a three-dimensionalculture system using electrospun polymer scaffolds. Biomaterials28:450–458 (2007).

301 Griffith LG, Polymeric biomaterials. Acta Mater 48:263–277 (2000).302 Shi XF, Hudson JL, Spicer PP, Tour JM, Krishnamoorti R and Mikos

AG, Rheological behaviour and mechanical characterization ofinjectable poly(propylene fumarate)/single-walled carbon nan-otube composites for bone tissue engineering. Nanotechnology16:S531–S538 (2005).

303 Sitharaman B, Shi XF, Tran LA, Spicer PP, Rusakova I, Wilson LJ, et al.,Injectable in situ cross-linkable nanocomposites of biodegradablepolymers and carbon nanostructures for bone tissue engineering.J Biomat Sci-Polym E 18:655–671 (2007).

304 Temenoff JS and Mikos AG, Injectable biodegradable materials fororthopedic tissue engineering. Biomaterials 21:2405–2412 (2000).

305 Gopferich A and Tessmar J, Polyanhydride degradation and erosion.Adv Drug Deliver Rev 54:911–931 (2002).

306 Zhang Q, Su K, Chan-Park MB, Wu H, Wang D and Xu R, Developmentof high refractive ZnS/PVP/PDMAA hydrogel nanocomposites forartificial cornea implants. Acta Biomater 10:1167–1176 (2014).

J Chem Technol Biotechnol 2014; 89: 1793–1810 © 2014 Society of Chemical Industry wileyonlinelibrary.com/jctb

Page 18: ozdil2014

1810

www.soci.org D Ozdil, HM Aydin

307 Tran RT, Choy WM, Cao H, Qattan I, Chiao JC, Ip WY, et al., Fab-rication and characterization of biomimetic multichanneledcrosslinked-urethane-doped polyester tissue engineered nerveguides. J Biomed Mater Res A 102:2793–2804 (2014).

308 Sharma B, Fermanian S, Gibson M, Unterman S, Herzka DA, CascioB, et al., Human cartilage repair with a photoreactive adhesive-hydrogel composite. Sci Translational Med 5:167ra6 (2013).

309 Malafaya PB, Silva GA and Reis RL, Natural-origin polymers as carriersand scaffolds for biomolecules and cell delivery in tissue engineer-ing applications. Adv Drug Deliver Rev 59:207–233 (2007).

310 Lee KY and Mooney DJ, Hydrogels for tissue engineering. Chem Rev101:1869–1879 (2001).

311 Jhon MS and Andrade JD, Water and Hydrogels. J Biomed Mater Res7:509–522 (1973).

312 Ward JH and Peppas NA, Kinetic gelation modeling of controlledradical polymerizations. Macromolecules 33:5137–5142 (2000).

313 Jeon O, Bouhadir KH, Mansour JM and Alsberg E, Photocrosslinkedalginate hydrogels with tunable biodegradation rates and mechan-ical properties. Biomaterials 30:2724–2734 (2009).

314 Chung C, Mesa J, Miller GJ, Randolph MA, Gill TJ and Burdick JA,Effects of auricular chondrocyte expansion on neocartilage for-mation in photocrosslinked hyaluronic acid networks. Tissue Eng12:2665–2673 (2006).

315 Almany L and Seliktar D, Biosynthetic hydrogel scaffolds made fromfibrinogen and polyethylene glycol for 3D cell cultures. Biomaterials26:2467–2477 (2005).

316 Elbert DL and Hubbell JA, Reduction of fibrous adhesion formationby a copolymer possessing an affinity for anionic surfaces. J BiomedMater Res 42:55–65 (1998).

317 Stahl PJ, Romano NH, Wirtz D and Yu SM, PEG-based hydrogelswith collagen mimetic peptide-mediated and tunable physicalcross-links. Biomacromolecules 11:2336–2344 (2010).

318 Melrose J, Chuang C and Whitelock J, Tissue engineering logcartilages using biomatrices. J Chem Technol Biot 83:444–463(2008).

319 Balakrishnan B and Banerjee R, Biopolymer-based hydrogels for carti-lage tissue engineering. Chem Rev 111:4453–4474 (2011).

320 Ng KW, Ateshian GA and Hung CT, Zonal chondrocytes seededin a layered agarose hydrogel create engineered cartilage withdepth-dependent cellular and mechanical inhomogeneity. TissueEng Pt A 15:2315–2324 (2009).

321 Lu HH, Subramony SD, Boushell MK and Zhang XZ, Tissue engineer-ing strategies for the regeneration of orthopedic interfaces. AnnBiomed Eng 38:2142–2154 (2010).

322 Chen JN, Chen HA, Li P, Diao HJ, Zhu SY, Dong L, et al., Simultaneousregeneration of articular cartilage and subchondral bone in vivousing MSCs induced by a spatially controlled gene delivery sys-tem in bilayered integrated scaffolds. Biomaterials 32:4793–4805(2011).

323 Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A and PeppasNA, Hydrogels in regenerative medicine. Adv Mater 21:3307–3329(2009).

324 Park H, Temenoff JS, Holland TA, Tabata Y, Mikos AG. Delivery ofTGF-beta 1 and chondrocytes via injectable, biodegradable hydro-gels for cartilage tissue engineering applications. Biomaterials26:7095–7103 (2005).

325 Bryant SJ and Anseth KS, The effects of scaffold thickness on tis-sue engineered cartilage in photocrosslinked poly(ethylene oxide)hydrogels. Biomaterials 22:619–626 (2001).

326 Omidian H, Rocca JG and Park K, Advances in superporous hydrogels.J Control Release 102:3–12 (2005).

327 Bryant SJ, Anseth KS, Lee DA and Bader DL, Crosslinking densityinfluences the morphology of chondrocytes photoencapsulatedin PEG hydrogels during the application of compressive strain.J Orthopaed Res 22:1143–1149 (2004).

328 Xia F, Ge H, Hou Y, Sun TL, Chen L, Zhang GZ, et al., Multirespon-sive surfaces change between superhydrophilicity and superhy-drophobicity. Adv Mater 19:2520 (2007).

329 Peng T and Cheng YL, Temperature-responsive permeability ofporous PNIPAAm-g-PE membranes. J Appl Polym Sci 70:2133–2142(1998).

330 Lue SJ, Hsu JJ, Chen CH and Chen BC, Thermally on-off switch-ing membranes of poly(N-isopropylacrylamide) immobilized intrack-etched polycarbonate films. J Membrane Sci 301:142–150(2007).

331 Kopecek J and Yang JY, Revie - hydrogels as smart biomaterials. PolymInt 56:1078–1098 (2007).

332 Nath N and Chilkoti A, Creating ’Smart’ surfaces using stimuli respon-sive polymers. Adv Mater 14:1243 (2002).

333 Jeong B, Kim SW and Bae YH, Thermosensitive sol–gel reversiblehydrogels. Adv Drug Deliver Rev 54:37–51 (2002).

334 Joglekar M and Trewyn BG, Polymer-based stimuli-responsivenanosystems for biomedical applications. Biotechnol J 8:931–945(2013).

335 Ju C, Sun J, Zi P, Jin X and Zhang C, Thermosensitive micelles-hydrogelhybrid system based on poloxamer 407 for localized delivery ofpaclitaxel. J Pharm Sci 102:2707–2717 (2013).

336 Fleige E, Quadir MA and Haag R, Stimuli-responsive polymericnanocarriers for the controlled transport of active compounds:concepts and applications. Adv Drug Deliv Rev 64:866–884(2012).

337 Schild HG, Poly (N-Isopropylacrylamide) - experiment, theory andapplication. Prog Polym Sci 17:163–249 (1992).

338 Srivastava A and Kumar A, Thermoresponsive poly(N-vinylcaprolactam) cryogels: synthesis and its biophysical evalua-tion for tissue engineering applications. J Mater Sci Mater Med 21:2937–2945 (2010).

339 Ivanov AE, Galaev IY, Kazakov SV and Mattiasson B, Thermosensi-tive copolymers of N-vinylimidazole as displacers of proteins inimmobilised metal affinity chromatography. J Chromatogr A 907:115–130 (2001).

340 Weber CH, R; Schubert, US. Temperature responsive bio-compatiblepolymers based on poly(ethylene oxide) and poly(2-oxazoline)s.Prog Polym Sci 37:686–714 (2012).

341 Niu G, Djaoui AB and Cohn D, Crosslinkable PEO-PPO-PEO triblocksas building blocks of thermo-responsive nanoshells. Polym Int52:2524–2530 (2011).

342 Araujo JV, Davidenko N, Danner M, Cameron RE and Best SM, Novelporous scaffolds of pH responsive chitosan/carrageenan-basedpolyelectrolyte complexes for tissue engineering. J Biomed MaterRes A (2014).

343 Adrian Gestos PGW, Spinks GM and Wallace GG, Crosslinking neatultrathin films and nanofibres of pH-responsive poly(acrylic acid)by UV radiation. Soft Matter 6:1045–1052 (2010).

344 Kyriakides TR, Cheung CY, Murthy N, Bornstein P, Stayton PS andHoffman AS, pH-sensitive polymers that enhance intracellular drugdelivery in vivo. J Control Release 78:295–303

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 pharmaceuticalapplications. Int J Pharm 245:83–91 (2002).

wileyonlinelibrary.com/jctb © 2014 Society of Chemical Industry J Chem Technol Biotechnol 2014; 89: 1793–1810