You are on page 1of 20

Hindawi Publishing Corporation

International Journal of Polymer Science


Volume 2011, Article ID 290602, 19 pages
doi:10.1155/2011/290602

Review Article
Polymeric Scaffolds in Tissue Engineering Application: A Review

Brahatheeswaran Dhandayuthapani, Yasuhiko Yoshida,


Toru Maekawa, and D. Sakthi Kumar
Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe,
Saitama 350-8585, Japan

Correspondence should be addressed to D. Sakthi Kumar, sakthi@toyo.jp

Received 16 May 2011; Revised 29 June 2011; Accepted 9 July 2011

Academic Editor: Shanfeng Wang

Copyright 2011 Brahatheeswaran Dhandayuthapani et al. This is an open access article distributed under the Creative
Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the
original work is properly cited.

Current strategies of regenerative medicine are focused on the restoration of pathologically altered tissue architectures by
transplantation of cells in combination with supportive scaolds and biomolecules. In recent years, considerable interest has
been given to biologically active scaolds which are based on similar analogs of the extracellular matrix that have induced
synthesis of tissues and organs. To restore function or regenerate tissue, a scaold is necessary that will act as a temporary
matrix for cell proliferation and extracellular matrix deposition, with subsequent ingrowth until the tissues are totally restored
or regenerated. Scaolds have been used for tissue engineering such as bone, cartilage, ligament, skin, vascular tissues, neural
tissues, and skeletal muscle and as vehicle for the controlled delivery of drugs, proteins, and DNA. Various technologies come
together to construct porous scaolds to regenerate the tissues/organs and also for controlled and targeted release of bioactive
agents in tissue engineering applications. In this paper, an overview of the dierent types of scaolds with their material properties
is discussed. The fabrication technologies for tissue engineering scaolds, including the basic and conventional techniques to the
more recent ones, are tabulated.

1. Introduction strategies is more frequently associated with the concept


of tissue engineering, that is, the use of living cells seeded
The field of tissue engineering has advanced dramatically on a natural or synthetic extracellular substrate to create
in the last 10 years, oering the potential for regenerat- implantable pieces of the organism [2].
ing almost every tissue and organ of the human body. Scaold design and fabrication are major areas of
Tissue engineering and the related discipline of regenera- biomaterial research, and they are also important subjects
tive medicine remain a flourishing area of research with for tissue engineering and regenerative medicine research
potential new treatments for many more disease states. The [1]. Scaold plays a unique role in tissue regeneration and
advances involve researchers in a multitude of disciplines, repair. During the past two decades, many works have been
including cell biology, biomaterials science, imaging, and done to develop potentially applicable scaold materials for
characterization of surfaces and cell material interactions. tissue engineering. Scaolds are defined as three-dimension
Tissue engineering aims to restore, maintain, or improve porous solid biomaterials designed to perform some or
tissue functions that are defective or have been lost by all of the following functions: (i) promote cell-biomaterial
dierent pathological conditions, either by developing bio- interactions, cell adhesion, and ECM deposition, (ii) permit
logical substitutes or by reconstructing tissues. The general sucient transport of gases, nutrients, and regulatory factors
strategies adopted by tissue engineering can be classified to allow cell survival, proliferation, and dierentiation, (iii)
into three groups [1]: (i) Implantation of isolated cells or biodegrade at a controllable rate that approximates the rate
cell substitutes into the organism, (ii) delivering of tissue- of tissue regeneration under the culture conditions of inter-
inducing substances (such as growth factors), and (iii) est, and (iv) provoke a minimal degree of inflammation or
placing cells on or within dierent matrices. The last of these toxicity in vivo [3]. The developing scaolds with the optimal
2 International Journal of Polymer Science

characteristics, such as their strength, rate of degradation, properties which are significant in the application of tis-
porosity, and microstructure, as well as their shapes and sizes, sue engineering and organ substitution. Researchers have
are more readily and reproducibly controlled in polymeric attempted to grow skin and cartilage [17], bone and cartilage
scaolds [4]. The few scaolds that have displayed biological [18], liver [19], heart valves and arteries [20], bladder [21],
activity have induced regeneration of tissues and organs that pancreas [22], nerves [23], corneas [24], and various other
do not regenerate spontaneously and have been referred soft tissues [25].
as regeneration templates. Biological scaolds are derived Scaold materials can be synthetic or biologic, degrad-
from human, animal tissues and synthetic scaolds from able or nondegradable, depending on the intended use [13].
polymers. The first biologically active scaold was synthe- The properties of polymers depend on the composition,
sized in 1974; its degradation behavior and exceptionally structure, and arrangement of their constituent macro-
low antigenicity in vivo, as well as its thromboresistant molecules. It can be categorized into dierent types in terms
behavior in vitro, were described [5]. The initial patent of their structural, chemical, and biological characteristics,
describing these scaolds was granted in 1977 [6]. Principles for example, ceramics, glasses, polymers, and so forth.
for synthesizing a biologically active scaold, including the Naturally occurring polymers, synthetic biodegradable, and
critical importance of the degradation rate, was described in synthetic nonbiodegradable polymers are the main types of
detail in 1980 [7]. The first reports of induced regeneration polymers used as biomaterials.
of tissue in an adult (dermis) by a scaold in animals [8, 9] Natural polymers can be considered as the first biode-
and humans [10], peripheral nerve regeneration across a gradable biomaterials used clinically [26]. Natural materials
gap of unprecedented length [11], and regeneration of the owing to the bioactive properties have better interactions
conjunctiva [12]. with the cells which allow them to enhance the cells per-
Biomaterials play a critical role in this technology by formance in biological system. Natural polymers can be clas-
acting as synthetic frameworks referred as scaolds, matrices, sified as proteins (silk, collagen, gelatin, fibrinogen, elastin,
or constructs. The state of the art in biomaterials design has keratin, actin, and myosin), polysaccharides (cellulose, amy-
continuously evolved over the past few decades. In recent lose, dextran, chitin, and glycosaminoglycans), or polynu-
years, there has been increasing importance on materials that cleotides (DNA, RNA) [27].
could be used in biomedical areas. Biomaterials intended for Synthetic biomaterial guidance provided by biomaterials
biomedical applications target to develop artificial materials may facilitate restoration of structure and function of dam-
that can be used to renovate or restore function of diseased aged or diseased tissues. Synthetic polymers are highly useful
or traumatized tissues in the human body and thus improve in biomedical field since their properties (e.g., porosity,
the quality of life. After an early empirical phase of bioma- degradation time, and mechanical characteristics) can be tai-
terials selection based on availability, design attempts were lored for specific applications. Synthetic polymers are often
primarily focused on either achieving structural/mechanical cheaper than biologic scaolds; it can be produced in large
performance or on rendering biomaterials inert and thus uniform quantities and have a long shelf time. Many com-
unrecognizable as foreign bodies by the immune system. mercially available synthetic polymers show physicochemical
Biomaterials used as implants in the form of sutures, bone and mechanical properties comparable to those of biological
plates, joint replacements, ligaments, vascular grafts, heart tissues. Synthetic polymers represent the largest group of
valves, intraocular lenses, dental implants, and medical biodegradable polymers, and they can be produced under
devices like pacemakers, biosensors, and so forth [13, 14]. controlled conditions. They exhibit, in general, predictable
In the last four decades, significant advances have been and reproducible mechanical and physical properties such
as tensile strength, elastic modulus, and degradation rate
made in the progress of scaolds for biomedical applications.
[28]. PLA, PGA, and PLGA copolymers are among the most
This paper is intended to illustrate the various scaolds in the
commonly used synthetic polymers in tissue engineering
field of tissue engineering. It covers the most commonly used [29]. PHA belongs to a class of microbial polyesters and is
scaolds fabrication technologies. being increasingly considered for applications in tissue engi-
neering [30].
Bioactive ceramics, such as HAP, TCP, and certain
2. Natural Polymers and Synthetic compositions of silicate and phosphate glasses (bioactive
Polymers for Scaffolds glasses) and glass-ceramics (such as apatite-wollastonite) re-
act with physiological fluids and through cellular activity
Polymers have been widely used as biomaterials for the fab- form tenacious bonds to hard and in some cases soft
rication of medical device and tissue-engineering scaolds tissue engineering [31]. However, their biocompatibility
[15, 16]. In biomedical applications, the criteria for selecting and biodegradability are often insucient, limiting their
the materials as biomaterials are based on their material potential use in the clinical side. We can overcome these
chemistry, molecular weight, solubility, shape and struc- issues by blending synthetic and natural polymers or by using
ture, hydrophilicity/hydrophobicity, lubricity, surface ener- composite materials that improve the scaold properties and
gy, water absorption degradation, and erosion mechanism. thereby allowing controlled degradation [32] and improving
Polymeric scaolds are drawing a great attention due to the biocompatibility in tissue engineering applications [33].
their unique properties such as high surface-to-volume ratio, The combination of degradable polymers and inorganic
high porosity with very small pore size, biodegradation, and bioactive particles represents the approach in terms of
mechanical property. They oer distinct advantages of bio- achievable mechanical and biological performance in hard
compatibility, versatility of chemistry, and the biological tissue [34].
International Journal of Polymer Science 3

3. Three-Dimensional Polymeric Scaffold throughout the scaold. A foam polymeric scaold approach
Fabrication and Different Types of Scaffolds has several potential advantages for proliferating or adherent
cell lines such as (a) provide a physical surface onto which
In an era of decreasing availability of organs for transplan- the cells can lay their own ECM, (b) may inhibit cell growth
tation and a growing need for suitable replacements, the of adherent contact-inhibited cells, (c) provides improved
emerging field of tissue engineering gives hope to patients nutrient transport to the center of the device through
who desperately require tissue and organ substitutes. Since the porous interconnecting channel network, and (d) may
1980, researchers have developed many novel techniques to limit cluster size to the pore size of the foam and thereby
shape polymers into complex architectures that exhibit the eliminating very large clusters that can potentially develop
desired properties for specific tissue-engineering applica- a necrotic center. Depending on the choice of solvent and
tions. These fabrication techniques result in reproducible phase separating conditions, the foams can be controlled to
scaolds for the regeneration of specific tissues. Polymer form either random or oriented pore architectures [141].
scaolds can provide mechanical strength, interconnected Improvement in the structure and increased pore inter-
porosity and surface area, varying surface chemistry, and connectivity of the porous scaold is required for the devel-
unique geometries to direct tissue regeneration [138]. Scaf- opment of artificial blood vessels or peripheral nerve growth.
folding is essential in this endeavor to act as a three- Precise three-dimensional shapes are required which lead
dimensional template for tissue ingrowths by mimicking to the development of sophisticated extrusion technologies
ECM [139]. These key scaold characteristics can be tailored [142] and methods of adhering porous membranes to the
to the application by careful selection of the polymers, desirable shapes [143]. Ideal pore sizes vary for dierent
additional scaold components, and the fabrication tech- cells and tissues [144]. Porous scaolds can be manufactured
nique. Typical scaold designs have included meshes, fibers, with specific pore size, porosity, surface-area-to-volume ratio
sponges and foams, and so forth. These designs are chosen and crystallinity. Porous controlled-release systems contain
because they promote uniform cell distribution, diusion pores that are large enough to enable diusion of the drug
of nutrients, and the growth of organized cell communities [145]. Synthetic biodegradable polymers such as PLLA, PGA,
[140]. The fabrication technique for tissue engineering PLGA [50], PCL [146], PDLLA, PEE based on PEO, and
scaolds depends almost entirely on the bulk and surface PBT [147] are used as porous scaolding materials. For
properties of the material and the proposed function of enhanced control over porosity and pore diameter as com-
the scaold. Most techniques involve the application of pared to most fabrication methods, a solvent casting and par-
heat and/or pressure to the polymer or dissolving it in an ticulate leaching technique was developed. A modern meth-
organic solvent to mold the material into its desired shape. od for creating porous scaolds composed of nano- and
While each method presents distinct advantages and disad- microscale biodegradable fibers by electrospinning is a latest
vantages, the appropriate technique must be selected to meet development in this field.
the requirements for the specific type of tissue. Scaolds
structure development is directly related to many methods, 5. Hydrogel Scaffold
which are listed in Table 1.
Large numbers of scaolds from dierent biomaterials In the last decade, hydrogels have played an ever increas-
are available for clinical use which is listed in Table 2. In ing role in the revolutionary field of tissue engineering
order to repair and regenerate lost or damaged tissue and where they are used as scaolds to guide the growth of
organs, 3D scaolds must be designed, fabricated, and util- new tissues. The design and application of biodegrad-
ized to regenerate the tissue similar in both anatomical able hydrogels has dramatically increased the potential
structure and function to the original tissue or organ to be impact of hydrogel materials in the biomedical field and
replaced or repaired. Dierent types of scaolds, including enabled the development of exciting advances in controlled
porous scaold, microsphere scaold, hydrogel scaold, fi- drug delivery and tissue engineering applications [148].
brous scaold, polymer-bioceramic composite scaold, and Hydrogels comprised of naturally derived macromolecules
acellular scaolds are described in this paper. have potential advantages of biocompatibility, cell-controlled
degradability, and intrinsic cellular interaction. They may
exhibit batch variations and generally exhibit a narrow and
4. Porous Scaffold limited range of mechanical properties. In contrast, synthetic
polymers can be prepared with precisely controlled struc-
The three-dimensional polymeric porous scaolds with tures and functions. Hydrogels have structural similarity to
higher porosities having homogeneous interconnected pore the macromolecular-based components in the body and
network are highly useful for tissue engineering. Sponge or are considered biocompatible [149]. Gels are formed when
foam porous scaold have been used in tissue engineering the network is covalently crosslinked [150]. Hydrogels are
applications [50], especially for growth of host tissue, bone made either from synthetic or natural polymers, which are
regrowth, or organ vascularization. Their porous network crosslinked through either covalent or noncovalent bonds.
simulates the ECM architecture allowing cells to interact ef- Hydrogels in tissue engineering must meet a number of
fectively with their environment. Though foams and sponges design criteria to function appropriately and promote new
are more mechanically stable compared to mesh structures, tissue formation. These criteria include both classical physi-
their use is still limited due to the open spaces present cal parameters (e.g., degradation and mechanics) as well as
4 International Journal of Polymer Science

Table 1: Scaolds fabrication techniques in tissue engineering applications.

Method Polymers Unique factors Application


Biodegradable porous scaold fabrication
Solvent casting/salt leaching Absorbable polymer (PLLA, Biodegradable controlled Bone and cartilage tissue
method [3537] PLGA, collagen, etc.) porous scaolds engineering
Ice particle leaching method Control of pore structure and Porous 3D scaolds for bone
PLLA & PLGA
[3840] production of thicker scaolds tissue engineering
Gas foaming/salt leaching Controlled porosity and pore Drug delivery and tissue
PLLA, PLGA & PDLLA
method [4143] structure sponge engineering
Microsphere fabrication
Solvent evaporation technique High-density cell culture, due
PLGA, PLAGA Bone repair
[4446] to the extended surface area
Bone, cartilage, or
Particle aggregated scaold
Chitosan, HAP High mechanical stability osteochondral tissue
[4749]
engineering
PLGA, PLLA, PGA, PLGA/PPF, 3D porous sponge structure,
Freeze drying method [5052] Tissue engineering scaolds
Collagen, and Chitosan durable and flexible
Thermally induced phase Highly porous scaold for Complicated shapes for tissue
PEG, PLLA
separation [53, 54] cellular transplantation engineering applications
Injectable gel scaold fabrication
Ceramic-based injectable CP ceramics, HAp, TCP, BCP,
Porosity and bioresorbability Cartilage tissue engineering
scaolds [5557] and BG
Hydrophilic/hydrophobic
diblock and triblock copolymer Biomimetically, exhibit
combinations of PLA, PGA, biocompatibility and cause
Hydrogel-basedinjectable Cartilage, bone tissue
PLGA, and PEG. Copolymers of minimal inflammatory
scaolds [5860] engineering, and drug delivery
PEO and PPO and polyoxamer. responses, thrombosis, and
alginates, collagen, chitosan, tissue damage
HA, and fibroin
Hydrogel scaold fabrication
Microgels, biologically Insulin delivery, gene therapy,
Micromolding [6163] Alginate, PMMA, HA, PEG degradable, mechanical and bioreactor, and
physical Complexity immunoisolation
Microdevices, biosensors,
Chitosan, fibronectin, HA,
Microwells, microarrays, growth factors, matrix
Photolithography [6466] PEG, PNIAAm, PAA, PMMA,
controlled size and shape components, forces, and
PAam, and PDMAEM
cell-cell interactions
Sensing, cell separation,
PGS, PEG, calcium alginate, Microbeads, microrods, valves,
Microfluidics [6769] cell-based microreactors, and
silicon and PDMS and pumps
controlled microreactors,
Microgels, microsensors, Sustainable and controllable
Emulsification [7072] Gelatin, HA, and collagen
cell-based diagnostics drug delivery therapies
Acellular scaold fabrication
Retain anatomical structure,
Decellularisation process
Biological tissues native ECM, and similar Tissue engineering
[7375]
biomechanical properties
Keratin scaold fabrication
Drug delivery, wound healing,
soft tissue augmentation,
Self-assembled process [7678] Keratin Biocompatibility synthetic skin, coatings for
implants, and scaolds for
tissue engineering
International Journal of Polymer Science 5

Table 1: Continued.
Method Polymers Unique factors Application
Fibrous scaold fabrication
PGA, PLA, PLGA, PCL High surface area, Drug delivery, wound healing,
Nanofiber electrospinning
copolymers, collagen, elastin, biomechanical, and soft tissue synthetic skin, and
process [7981]
and so forth biocompatibility scaolds for tissue engineering
Solar sails, reinforcement,
Microfiber wet-spinning Biocompatible fibres with good vascular grafts, nonwetting
PLGA, PLA, chitosan, and PCL
process [8284] mechanical properties textile surfaces, and scaolds
for tissue
Filtration, membrane
separation, protective military
Nonwoven fibre by melt-blown Submicron fiber size, highly
Polyesters, PGA, and PDO clothing, biosensors, wound
process [8587] porous scaold
dressings, and scaolds for
tissue engineering
Functional scaold fabrication
Collagen, gelatin, alginate, Angiogenesis, bone
Growth factors release process Membranes, hydrogels, foams,
chitosan, fibrin, PLGA, PLA, regeneration, and wound
[8890] microsphere, and particles
and PEG healing
Ceramic scaold fabrication
Sponge replication method PU sponge, PVA, TCP, BCP or Interconnected porous ceramic
Bone tissue engineering
[9193] calcium sulfate scaolds
Coating on: metals, glasses,
inorganic ceramics and organic
Simple calcium phosphate Improve biocompatibility or
polymers (PLGA, PS, PP, Orthopedic application
coating method [9496] enhance the bioreactivity
silicone, and PTFE), collagens,
fibres of silk, and hairs
Automation and direct organ fabrication
Biosensor development,
To build complex tissues microdeposition of active
Inkjet printing process [97100] Sodium alginate composed of multiple cell types proteins on cellulose, biochips
(Hydrogel scaold) and acellular polymeric
scaolds
Melt-based rapid prototyping Biodegradeable polymers or Complex 3D solid object, good Honey comb structure scaold,
process [101, 102] blends mechanical strength hard-tissue scaolds
Design and fabrication of
Computer-aided design (CAD) Develop a program algorithm
patient-specific scaolds and
data manipulation techniques that can be used to design
automated scaold assembly
[103105] scaold internal architectures
algorithm
Layer by layer deposition of To print complex 3D organs
Organ printing [106, 107] Tubular collagen gel
cells or matrix with computer-controlled,
Scaold sterilization
For degradable polymers and
Absolute freedom from
Ethylene oxide gas (EOG) porous scaolds, high
biological contamination in
[108110] penetration ability, and
scaolds
compatibility
Proven process is safe, reliable, Surgical disposables: surgical
Gamma-radiation sterilization
and highly eective at treating sutures, bandages, dressings,
[111113]
single-use medical devices gauge pads, implants
Commercially successful
Compatibility, low penetration, technology for sterilizing a
Electron beam radiation
in line sterilization of thin variety of disposable medical
[114116]
products devices with a wide range of
densities
Ecacy, speed, process Heat is absorbed by the exterior
Dry-heat sterilization [117, 118] simplicity, and lack of toxic surface of scaold and then
residues passed inward to the next layer
Removal of all contamination, Porous scaold for living cell
Steam sterilization [119, 120]
and scaold can be reused immobilization
6 International Journal of Polymer Science

Table 2: List of commercial polymeric scaolds products.

Polymer Property Biomedical application Trade name


First biodegradable synthetic suture
Regenerate biological tissue [121] DEXON
PGA in 1969
Good mechanical properties [122] Bone internal fixation devices Biofix
Bio-Anchor, Meniscal Stinger,
PLLA Good tensile strength Orthopaedic fixation devices
The Clearfix Meniscal Dart
High-strength fibers (FDA approved
Improved suture [123] DEXON
at 1971)
Ligament replacement or
Nondegradable fibers [124] Dacron
augmentation devices
Fiber-based devices [125] Blood vessel conduits
People with human
Injectable form immunodeficiency virus or
correction of facial fat loss
PLDLA Better property modulation [126] Bioresorbable implant material Resomer
PLGA High degradation Multifilament suture Vicryl, Vicryl Rapid & CRYL
Form of meshes Skin graft Vicryl Mesh
PLGA-collagen Matrix Tissue regeneration membrane CYTOPLAST Resorb
PLGA Prostate cancer Drug delivery vehicle LUPRON DEPOT
First commercially developed
PDS
monofilament suture (1980)
Fixation screws for small bone and
PDS Orthopaedic applications Pins
osteochondral fragments
PCL Long-term zero-order release [26] Long-term contraceptive device Capronor
PDLLA-CL Fibers less sti Monofilament suture MONACRYL
Drug delivery vehicle for small, and
PGCL, PLCL, and
Bioresorbable multiblock medium-sized biologically active SynBiosys
PEG
molecules
PCLTMC multiblock Flexible suture materials Maxon
and PGCL Orthopaedic tacks and screws Acufex
Bone pins and plates and drug
PHBHV Piezoelectricity property [127]
delivery
High porous & no adverse eect
PEU Tissue engineering application Degrapol
[128]
Injectable & good mechanical Orthopaedic applications & bone
LDI-based PU Polynova
property [129] cement
Potential bioresorbable suture Site-specific delivery of small
PEAs CAMEO
materials hydrophobic drugs and peptides
Drug delivery applications and
POE Hydrophobic, surface eroding [130] Alzamer
ocular applications
Surface erosion & biocompatibility Chemotherapeutic, brain cancer
Polyanhydrides Gliadel
Evaluations [131] (FDA approved)
First biodegradable polymers used
Absorb or encapsulate a wide range
PCA for developing nanoparticles for drug
of drug or protein molecules [132]
delivery application
Synthetic surgical glue, skin adhesive, Tissue adhesives for topical skin
Dermabond
and an embolic material application (FDA approved)
Major component of skin and other Bilayer skin substitute (FDA Integra Dermal Regeneration
musculoskeletal tissues [133] approved) Template
Wound dressings Biobrane &Alloderm
Scaolds for cardiovascular,
Collagen musculoskeletal & nervous tissue Bioengineered skin equivalents TransCyte
engineering [133]
International Journal of Polymer Science 7

Table 2: Continued.
Polymer Property Biomedical application Trade name
HA Promote angiogenesis [134] Wound dressing application HYAFF
Sponge as a carrier vehicle for
Synthetic bone graft OSSIGEL
osteoinductive protein [135]
Corneal transplantation and
HMW viscous HA Injectable soft tissue fillers [136] AMVISC & AMVISC Plus
glaucoma surgery
To relieve pain and improve join
Viscous HA Synovial fluid substitute [137] SYNVISC,ORTHOVISC
mobility in osteoarthritis patients

biological performance parameters (e.g., cell adhesion). It [169], gelatin [170], chitosan [171], HA [172], silk fibroin
is commonly believed that the degradation rates of tissue [173], PLA [174], PU [175], PCL [176], PLGA [177], PEVA
scaolds must be matched to the rate of various cellular [178], and PLLA-CL [179] are fibrous scaold in biomedical
processes in order to optimize tissue regeneration [151, 152]. application. The blending (or mixing) technique is a com-
Therefore, the degradation behavior of all biodegradable mon choice for the nanofiber functionalization. However,
hydrogels should be well defined, reproducible, and tunable most of the polymer nanofibers do not possess any specific
via hydrogel chemistry or structure. Biocompatible hydro- functional groups, and they must be specifically functional-
gels are currently used in cartilage wound healing, bone ized for successful applications. The most popular and sim-
regeneration, wound dress, and as carriers for drug delivery plest nanofiber modification methods are physical blending
[153]. Hydrogel with growth factor can act directly to and coating. Surface grafting polymerization has also been
support the development and dierentiation of cells in the used for attaching ligand molecules and adhesive proteins
newly formed tissues [154]. Hydrogels are often favorable on nanofiber surface for application of anity membrane
for promoting cell migration, angiogenesis, high water and tissue engineering scaold, respectively. Drugs, growth
content, and rapid nutrient diusion [155]. The hydrogel factors, and genes can be directly mixed into the polymer
scaolds have received intensive study for their use in the solution and electrospun to prepare drug carriers with
engineering of replacement connective tissues, primarily due controlled release properties [180].
to their biochemical similarity with the highly hydrated GAG
components of connective tissues. Examples of hydrogel-
forming polymers of natural origin are collagen [156],
7. Microsphere Scaffold
gelatin [157], fibrin [158], HA [159], alginate [160], and Microsphere-based tissue engineering scaold designs have
chitosan [161]. The synthetic polymers are PLA [162], PPF- attracted significant attention in recent years [181]. Lau-
derived Copolymers [163], PEG-derivatives, and PVA [164]. rencin et al. [44] initially used a microsphere-based approach
for tissue engineering scaold. Microsphere scaolds are
6. Fibrous Scaffold having spatial extension and temporal duration control
which provides the stiness gradients for interfacial tissue
The development of nanofibers has enhanced the scope for engineering [182]. Microsphere scaolds are increasingly
fabricating scaolds that can potentially mimic the archi- used as drug delivery systems and in advanced tissue
tecture of natural human tissue at the nanometer scale. engineering applications such as gene therapy, antibiotic
Currently, there are three techniques available for the synthe- treatment of infected bone, and so forth [183]. The influence
sis of nanofibers: electrospinning, self-assembly, and phase of nanotechnology on scaold design and the possibility of
separation. Of these, electrospinning is the most widely sustained release formulations of growth factors via micro-
studied technique and also seems to exhibit the most promis- spheres are showing promising developments. Microsphere
ing results for tissue engineering applications. Nanofibers scaolds are generally a polymer matrix used for drug
synthesized by self-assembly [165] and phase separation encapsulation for the release of drugs at a relatively slow rate
[50] have had relatively limited studies that explored their over a prolonged period of time [184]. Polymers with low
application as scaolds for tissue engineering. The high molecular weight used in developing porous microspheres
surface-area-to-volume ratio of the nanofibers combined for the rapid release of the drug, while polymers with high
with their microporous structure favors cell adhesion, pro- molecular weight for developing microspheres for a slower
liferation, migration, and dierentiation, all of which are drug release profile which can be achieved due to its dense
highly desired properties for tissue engineering applications nature [185]. Injectable microspheres have also been devel-
[166, 167]. Nanofibers used as scaolds for musculoskeletal oped for the controlled delivery of drugs [186]. Microspheres
tissue engineering including bone, cartilage, ligament, and as building blocks oer several benefits, including ease
skeletal muscle, skin, vascular, neural tissue engineering, of fabrication, control over morphology, physicochemical
and as vehicle for the controlled delivery of drugs, proteins, characteristics, and its versatility of controlling the release
and DNA [168]. Natural polymers and synthetic polymers kinetics of encapsulated factors [187]. The methods used
explored for the fabrication of nanofibers such as collagen to produce microsphere-based scaolds have utilized heat
8 International Journal of Polymer Science

sintering [188, 189], solvent vapor treatment [190, 191], sol- rates to the repair rates of body tissues developed for hard
vent/nonsolvent sintering method [192, 193] or nonsolvent tissue implants and tissue engineering scaolds, due to their
sintering technique [181]. Particle aggregation methodology excellent biocompatibility, bioactivity, and bioresorption in
is proposed to fabricate bilayered scaolds for osteochondral calcified tissue. Highly porous polymer/ceramic composite
tissue engineering in order to achieve an improved integra- scaolding appears to be a promising substrate for bone
tive bone and cartilage interface which has been needed for tissue engineering due to its excellent mechanical prop-
this application. PLAGA microsphere scaolds are in the erties and osteoconductivity [40]. PLGA/HAP composite
range of trabecular bone, demonstrating the potential of the scaold has excellent biocompatibility with hard tissues and
porous microsphere matrix to be used as a scaold for load- high osteoconductivity and bioactivity [50]. The composite
bearing bone tissue engineering [47]. The sintered micro- scaolds supported uniform cell seeding, cell ingrowth,
sphere matrix shows promise as a bone regeneration scaold. and tissue formation. The major inorganic component of
An advantage of the sintered microsphere structure is its pore natural bone; bioceramics, including CP, HAP, and TCP are
composite with PLLA [46], collagen [202], gelatin [203],
interconnectivity and desirable three-dimension pore size.
chitosan [204] are widely used as scaolding materials for
The gel microsphere matrix and the sintered microsphere bone repair.
matrix were designed using the random packing of PLAGA
microspheres to create a three-dimensional porous structure
for bone regeneration [194]. Composite microspheres are 9. Acellular Scaffold
also used for the fabrication of polymer-ceramic matrices
for bone applications [195]. Chitosan microsphere scaolds Acellular tissue matrices can be prepared by manufacturing
have been produced for cartilage and osteochondral tissue artificial scaolds or by removing cellular components from
engineering [48]. tissues by mechanical and chemical manipulation to produce
collagen-rich matrices [205207]. These matrices slowly
degrade on implantation and are generally replaced by the
8. Polymer-Bioceramic Composite Scaffold ECM proteins secreted by the ingrowing cells. The ultimate
goal of any decellularization protocol is to remove all
Development of composite materials for tissue engineering cellular material without adversely aecting the composition,
is attractive since their properties can be engineered to mechanical integrity, and eventual biological activity of
suit the mechanical and physiological demands of the host the remaining ECM. The decellularized biological scaold
tissue by controlling the volume fraction, morphology, and was introduced to obtain a physiological matrix scaold
arrangement of the reinforcing phase [196]. Ceramics used that resembles that of native blood vessels [208]. Acellular
in fabricating implants can be classified as nonabsorbable tissue matrices have proven to support cell ingrowth and
(relatively inert), bioactive or surface reactive (semi-inert) regeneration of genitourinary tissues, including urethra and
[197], and biodegradable or resorbable (noninert) [198]. bladder, with no evidence of immunogenic rejection [207].
Alumina, zirconia, silicone nitrides, and carbons are inert Ureteral acellular matrices were utilized as a scaold for the
bioceramics. Certain glass ceramics and dense HAP are semi- ingrowth of ureteral tissue in rats [209]. Acellular bladder
inert (bioreactive), and examples of resorbable ceramics are matrix has served as a scaold for the ingrowth of host
aluminum calcium phosphate, coralline, plaster of Paris, bladder wall components in rats. Since the structures of
HAP, and TCP [199]. Ceramics are known for their good the proteins (e.g., collagen and elastin) in acellular matrices
compatibility, corrosion resistance, and high compression are well conserved and normally arranged, the mechanical
resistance. Drawbacks of ceramics include brittleness, low properties of the acellular matrices are not significantly
fracture strength, diculty to fabricate, low mechanical dierent from those of native bladder submucosa [209].
reliability, lack of resilience, and high density. In recent years, The matrix was prepared by mechanically and chemically
removing all cellular components from bladder tissue [210].
humans have realized that ceramics and their composites
To engineer tissues successfully, the selection of scaolds is
can also be used to augment or replace various parts of
critical. Although various synthetic biodegradable polymer
body, particularly bone. Thus, the ceramics used for the latter scaolds have been developed and improved by mimicking
purposes are classified as bioceramics. Polymers by them- biological structures, comparing to other scaolds, acellular
selves are generally flexible and exhibit a lack of mechanical scaolds have the following advantages.
strength and stiness, whereas inorganic materials such as
ceramics and glasses are known to be too sti and brittle. (i) Acellular scaolds retain their correct anatomical
The combination of polymers and inorganic phases leads to structure even after the decellularisation process.
composite materials with improved mechanical properties (ii) Acellular scaolds retain native ECM architecture
due to the inherent higher stiness and strength of the and possess the cell adhesion ligands.
inorganic material. Secondly, addition of bioactive phases to
bioresorbable polymers can alter the polymer degradation (iii) The decellularisation process considerably reduces
behavior of the scaolds [200, 201]. Complications in the immunological responses by completely removing
development of polymer bioceramics composite scaold cellular components.
are (i) maintenance of strength and the stability of the (iv) The decellularisation process facilitates similar biom-
interface during the degradation period and replacement echanical properties as those of native tissues that are
by the natural host tissue and (ii) matching resorption critical for the long-term functionality of the grafts.
International Journal of Polymer Science 9

Various extracellular matrices have been utilized success- smart materials that can mimic natural ECM. ECM plays
fully for tissue engineering in animal models and products a key role in tissue architecture by providing structural
incorporating decellularized heart valves, small intestinal support and tensile strength. Attachment sites for cell surface
submucosa (SIS), and urinary bladder have received regula- receptors are related to a wide variety of processes related
tory approval for use in human patients [211]. The obvious to cell dierentiation, tissue formation, homeostasis, and
advantage of this scaold is that it is composed of ECM regeneration [215, 216]. The fabrication and design of
proteins typically found in the body. When derived from macro- to nanoscale structural architectures have received
a vessel, the three-dimension architecture is very similar to much attention in medical applications. Nano- to macroscale
that of the original, thus conferring appropriate mechanical structure geometrically or topologically mimics the native
and physical properties, which is essential in identifying and state of ECM in living tissues. Three-dimensional scaolds
predicting optimal cell environments in order to develop are capable of regenerating tissue and organs in their normal
scaolds for preliminary analysis and implantation. Nat- physiological shape. Mimicking the ECM using biomaterials
urally derived materials and acellular tissue matrices have would be a logical approach for engineering scaold for a
the potential advantage of biological recognition. Polymer variety of tissue types. As polymer materials permit a most
coating of a tissue-derived acellular scaold can improve versatile variety of surface characteristics, ecient control
the mechanical stability and enhance the hemocompatibility over processes of ECM reconstitution can be achieved by
of the protein matrix. Tissue engineering that has been the interaction with polymeric materials. The importance
introduced is the use of biological/polymeric composite of scaold geometry in maintaining highly interconnected
materials as starter matrices. Such hybrids can be complex porous fabrics of high surface density provides an extremely
structures such as heart valves, for example, fabricated from high surface-to-volume ratio, favoring cell attachment and
decellularized porcine aortic valves and dip coated with a proliferation.
biodegradable polymer [212].
12. Surface Properties
10. Physicochemical Characterization of
Scaffolds Surface properties include both chemical and topographical
characteristics, which can control and aect cellular adhesion
Polymeric scaolds have evolved to serve not merely as and proliferation [214]. The scaold surface is the initial and
carriers of cells and inductive factors, but to actively instruct primary site of interaction with surrounding cells and tissue.
cells and provide step by step guidance of tissue formation. As most cells utilized in tissue engineering are anchorage
To accomplish this goal, a thorough understanding of the dependent, it has been reasoned that the scaold should
chemistry and physicochemical properties of the tissue facilitate their attachment. Thus, scaolds with a large and
to be engineered and the materials used in this process accessible surface area are favorable. For example, high
are required. Several characterizations are required for the internal surface-area-to-volume ratios is essential in order
fabrication of successful 3D scaolds. They are to accommodate the number of cells required to replace
or restore tissue or organ functions. The surface properties
(i) external geometry (e.g., macro-, microstructure, can be selectively modified to enhance the performance
interconnectivity), of the biomaterials. For instance, by altering the surface
(ii) surface properties (e.g., surface energy, chemistry, functionality using thin film deposition, the optimal surface,
charge, surface area), chemical, and physical properties can be attained [217, 218].
Hence, surface modification of biomaterials is becoming an
(iii) porosity and pore size,
increasingly popular method to improve device multifunc-
(iv) interface adherence & biocompatibility, tionality, tribological, and mechanical properties. Most of the
(v) degradation characterization (e.g., biodegradation), surface modifications and immobilizations of biomolecules
are performed to improve the biocompatibility of the
(vi) mechanical competence (e.g., compressive and ten- polymeric scaold; thereby, cells can specifically recognize
sile strength). the scaold. These biomolecules include adhesive proteins
Developing scaolds that mimic the architecture of tissue like collagen, fibronectin, RGD peptides, and growth factors
at the nanoscale is one of the most important challenges like bFGF, EGF, insulin, and so forth. The biomolecules can
in the field of tissue engineering [168]. Polymeric scaolds either be covalently attached, electrostatically adsorbed, or
show excellent potential with mechanical properties and with self-assembled on the biomaterial surfaces to develop brand
wide range of degradation, the qualities which are essential new materials [219].
for a range of tissue engineering applications [213].
13. Porosity and Pore Size
11. External Geometry
Scaolds must possess a highly porous structure with an
Physical characteristics are certainly the important factors open fully interconnected geometry for providing a large
to consider when scaolds are applied for tissue recon- surface area that will allow cell ingrowth, uniform cell
struction [214]. Scaold with proper physical characters are distribution, and facilitate the neovascularization of the
10 International Journal of Polymer Science

construct [220]. Average pore size, pore size distribution, 15. Degradation Rates
pore volume, pore interconnectivity, pore shape, pore throat
size, and pore wall roughness are important parameters to Biodegradable polymers have revolutionized the applications
consider while designing a scaold. It provides a porous of biomaterial in the field of drug delivery and implants
biocompatible network into which the surrounding tissue for tissue engineering applications. Scaold degradation can
is induced and acts as a temporary template for the new occur through mechanisms that involve physical or chemical
tissues growth and reorganization [221]. Pore size is also processes and/or biological processes that are mediated by
a very important issue because if the pores employed biological agents, such as enzymes in tissue remodeling. The
are too small, pore occlusion by the cells will happen, biodegradable scaold gradually degrades by predetermined
which will prevent cellular penetration, extracellular matrix period to be replaced by newly grown tissue from the
production, and neovascularization of the inner areas of the adhered cells [1]. Degradation results in scaold dismantling
scaold. The eects of pore size on tissue regeneration has and material dissolution/resorption through the scaolds
been emphasized by experiments demonstrating optimum bulk and/or surface types of degradation [234]. Polymeric
pore size of 5 m for neovascularization [222], 515 m scaolds that undergo bulk degradation tend to break down
for fibroblast ingrowth [223], 20 m for the ingrowth of the internal structure of the scaold thus reducing the
hepatocytes [224], 200350 m for osteoconduction [225], molecular mass [235]. A polymeric scaold that primarily
and 20125 m for regeneration of adult mammalian skin undergoes surface degradation can be described similarly
[226]. Pore interconnectivity is also critical to ensure that all to the dissolution of soap. The rate at which the surface
cells are within 200 m from blood supply in order to provide degrades is usually constant. Therefore, even though the
for mass transfer of oxygen and nutrients [224, 227]. size of the scaold becomes smaller, the bulk structure
is maintained. These types of degrading scaolds provide
14. Interface Adherence and Biocompatibility longer mechanical stability for the tissue to regenerate.
Biodegradation of polymeric biomaterials involves cleavage
The term biocompatibility has been defined in many and of hydrolytically or enzymatically sensitive bonds in the poly-
dierent ways. Historically, materials that caused minimal mer leading to polymer erosion [131]. The biodegradation
biological responses were considered biocompatible. Bio- rate of a polymer depends mainly on the intrinsic properties
compatibility refers to the ability of a biomaterial to perform of the polymer, including the chemical structure, thev
its desired function with respect to a medical therapy, presence of hydrolytically unstable bonds, the level ofv
without eliciting any undesirable local or systemic eects hydrophilicity/hydrophobicity, crystalline/amorphous mor-
in the recipient or beneficiary of that therapy. It should phology, glass transition temperatures (Tg), the copolymer
generate the most appropriate beneficial cellular or tissue ratio, and the molecular weight [236]. The Controllable
response in that specific situation and optimize the clinically degradation and restoration rates should match the rate
relevant performance of that therapy [15]. Biocompatibility of tissue growth in vitro and in vivo for biodegradable or
of a scaold or matrix for a tissue engineering product restorable materials. The nonbiodegradable polymeric scaf-
refers to the ability to perform as a substrate that will folds are biologically stable, and it can provide a permanent
support the appropriate cellular activity, including the support over time and should ideally perform during the
facilitation of molecular and mechanical signalling systems life time of the patient. For example, PMMA is mainly used
[228]. Some important factors that determine scaolds as bone cements in hip and knee replacements, and high-
biocompatibility are their chemistry, structure, and their density PE forms the articulating surfaces of hip and knee
morphology, which in turn are aected by the polymer joints [13].
synthesis, scaold processing, and sterilization conditions.
Recently, several biodegradable polymers such as PLA, PGA,
PLGA, PDO, PTMC, and so on are extensively used or 16. Mechanical Properties
tested on a wide range of medical applications due to
their good biocompatibility [229]. The behavior of the
The proper mechanical properties for a biomaterial to
adsorption and desorption of adhesion and proliferation of
dierent types of mammalian cells on polymeric materials be used in a tissue engineering application are critical
depends on the surface characteristics such as wettability, to the success of the implant. The biostability of many
hydrophilicity/hydrophobicity ratio, bulk chemistry, surface scaolds depends on the factors such as strength, elasticity,
charge and charge distribution, surface roughness, and and absorption at the material interface and its chemical
rigidity. A number of surface treatments are available to degradation. The scaold should have proper mechanical
optimize the biocompatibility of surfaces in contact with properties and degradation rate with the bioactive surface
living tissue, to seal in undesirable residues or additives to encourage the rapid regeneration of the tissue [26]. It
using a coating and to regulate excretion and/or absorption is highly essential to retain the mechanical strength of the
using a selectively permeable surface [230]. Recently, physical scaolds structure after implantation for the reconstruction
and chemical surface modification methods for polymeric of hard, load bearing tissues such as bone and cartilages. To
biomaterials to influence cell adhesion and growth have been be used successfully in tissue engineering, it is critical that
achieved by oxidized polystyrene surface [231], ammonia a biomaterial scaold temporarily withstands and conducts
plasma-treated surface [232], and plasma-deposited acetone the loads and stresses that the new tissue will ultimately bear.
[233]. It is important, therefore, to evaluate one or more of the
International Journal of Polymer Science 11

following rheological parameters: scaold in a dimensional range that may be adequate for
cells and biomolecules. There are clear indications that as the
(i) elastic modulusmeasured strain in response to a goals of biomedical engineering increase in complexity, there
given tensile or compressive stress along the force;
is need to develop novel scaold structures.
(ii) flexural modulusmeasured the relationship
between a bending stress and the resulting strain
in response to a given tensile or compressive stress Future Directions
perpendicular under load;
Medical research continues to explore new scientific frontiers
(iii) tensile strengthmaximum stress that the material for diagnosing, treating, curing, and preventing diseases at
can withstand before it breaks; the molecular/genetic level. Important advances have been
(iv) maximum strainductility of a material or total made in the clinical use of medical implants and other
strain exhibited prior to fracture. devices. Presently, emphasis is placed on the design of poly-
meric scaold, that is, materials that obtain specific, desired,
The low strength and rigidity of the polysaccharides
limit their use to soft tissue applications. Fortunately, the and timely responses from surrounding cells and tissues.
options for tissue engineering are expanded by the use The need for alternative solutions to meet the demand for
of fibrous proteins, whose normal function is to provide replacement organs and tissue parts will continue to drive
mechanical integrity and stability to biological structures. advances in tissue engineering. Polymer scaolds have all the
Fibrous proteins are responsible for the transduction of prospective to provide a new means to control the physical
external mechanical forces to associated cells in a manner and chemical environment of the biological system. There
that influences the outcome of tissue growth [169]. The are several advantages to use biological polymers over widely
mechanical properties of bulk biomaterials are altered by utilized synthetic polymer in tissue engineering scaold.
their processing into scaolds of various pore sizes and pore Despite these recent improvements to the mechanical prop-
orientations and further that these properties will rapidly
erties, porosity, and bioactivity of scaolds, future researches
diminish as a function of implantation time [237]. The
mechanical rigidity of the surrounding matrix, as well as are needed to overcome many remaining limitations in the
material roughness and physical confinement, determined fabricating process. We believe no one material will satisfy
by three-dimensional microstructure on a subcellular and all design parameters in all applications, but a wide range of
supercellular scale, respectively, may significantly modulate materials will find uses in various tissue engineering applica-
the outcome of the balance between cell matrix forces, tions. The overall challenges in scaold design and fabrica-
leading to the remodeling of cytoarchitecture, cell polar- tion gives opportunity for new exciting application oriented
ization, alteration of downstream intracellular signaling research in scaold design which includes polymer assembly,
events as well as modification of the balance of cell-cell surface topography or chemical cues, nano-/macrostructure,
forces [238240]. The major factor aecting the mechanical biocompatibility, biodegradability, mechanical properties,
properties and structural integrity of scaolds, however, directing cell function and induced formation of natural
is their porosity, for example, pore volume, size, shape, tissue.
orientation, and connectivity.

Abbreviations
Conclusions
PU: Polyurethane
In summary, tissue engineering is one of the most exciting PS: Polysulfone
interdisciplinary and multidisciplinary research areas and is CP: Calcium phosphate
growing exponentially over time. Scaold materials and fab- HA: Hyaluronic acid
rication technologies play a crucial role in tissue engineering. PP: Polypropylene
A wide range of polymeric scaold was used to date in the BG: Bioactive glass
tissue engineering area. Scaolds should meet certain design ECM: Extracellular matrix
parameters to be useful in this area, regardless of whether PVA: Polyvinyl alcohol
they originate from natural resources or are synthetically PGA: Polyglycolide
created. All these techniques for scaold fabrication are PLA: Polylactide
sensitive to the various processing parameters. Innovations PPF: Poly(propylene fumarate)
in the material design and fabrication processes are raising PCA: Polycyanoacrylate
the possibility of production of implants with good perfor- PCL: Poly(-caprolactone)
mance. The scaold should be surface compatible as well PDO: Polydioxanone
as architecturally suitable with the host environment. The PHA: Polyhydroxyalkanoates
interest in the principles and theories of the fabrication POE: Poly(ortho ester)
process with polymers would be useful to develop a new PEE: Poly(ether ester)
design for implants and also to understand the behavior of PEO: Poly(ethylene oxide)
the scaold in the biomedical applications. Nanotechnology PBT: Polybutylene terephthalate
can provide strategies that can help to create features on a HAP: Hydroxyapatite
12 International Journal of Polymer Science

TCP: Tricalcium phosphate [8] I. V. Yannas, J. F. Burke, M. Warpehoski et al., Prompt,


PEG: Poly(ethylene glycol) long-term functional replacement of skin, Transactions
PEU: Poly(ester urethane) American Society for Artificial Internal Organs, vol. 27, pp. 19
23, 1981.
PAA: Poly(acrylic acid)
[9] I. V. Yannas, J. F. Burke, D. P. Orgill, and E. M. Skrabut,
LDI: Lysine diisocyanate
Regeneration of skin following closure of deep wounds with
BCP: Biphasic calcium phosphate
a biodegradable template, Transactions of the Society For
HMW: High molecular weight Biomaterials, vol. 5, pp. 2429, 1982.
PAam: Polyacrylamide [10] J. F. Burke, O. V. Yannas, and W. C. Quinby Jr., Successful use
PMMA: Polymethylmethacrylate of a physiologically acceptable artificial skin in the treatment
PLLA: Poly(L-lactic acid) of extensive burn injury, Annals of Surgery, vol. 194, no. 4,
PLGA: Poly(l-lactide-co-glycolide) pp. 413427, 1981.
PTMC: Poly(trimethylene carbonate) [11] I. V. Yannas, D. P. Orgill, J. Silver, T. V. Norregaard,
PDMS: Polydimethylsiloxane N. T. Zervas, and W. C. Schoene, Polymeric template
PTFE: Polytetrafluoroethylene facilitates regeneration of sciatic nerves across 15-mm gap,
PEVA: Poly(ethylene-co-vinylacetate) Transactions of the Society For Biomaterials, vol. 8, p. 146,
PGCL: Poly(glycolide-co--caprolactone) 1985.
PLCL: Poly(l-lactide-co-caprolactone) [12] W. C. Hsu, M. H. Spilker, I. V. Yannas, and P. A. D. Rubin,
Inhibition of conjunctival scarring and contraction by a
PDLLA: Poly(DL-lactide)
porous collagen-glycosaminoglycan implant, Investigative
PLDLA: Poly-L/D-lactide Ophthalmology and Visual Science, vol. 41, no. 9, pp. 2404
PLAGA: Poly(lactic acid-glycolic acid) 2411, 2000.
PHBHV: Poly(3-hydroxybutyrate)3-hydroxyvalerate [13] S. Ramakrishna, J. Mayer, E. Wintermantel, and K. W. Leong,
PCLTMC: Poly(caprolactone-co-trimethylene Biomedical applications of polymer-composite materials: a
carbonate) review, Composites Science and Technology, vol. 61, no. 9, pp.
PNIPAAm: Poly(N-isopropylacrylamide) 11891224, 2001.
PDMAEM: Poly(dimethylaminoethylmethacrylate) [14] M. Vert, Aliphatic polyesters: great degradable polymers that
hydrochloride cannot do everything, Biomacromolecules, vol. 6, no. 2, pp.
PDLLA-CL: Poly(D,L-lactide-co- caprolactone) 538546, 2005.
PLLA-CL: Poly(l-lactide-co--caprolactone) [15] E. Piskin, Biodegradable polymers as biomaterials, Journal
TCP: Tricalcium phosphate. of Biomaterials Science Polymer Edition, vol. 6, pp. 775795,
1994.
[16] Y. Ji, K. Ghosh, X. Z. Shu et al., Electrospun three-
Acknowledgment dimensional hyaluronic acid nanofibrous scaolds, Bioma-
terials, vol. 27, no. 20, pp. 37823792, 2006.
D. Brahatheeswaran, would like to acknowledge the Japanese [17] W. H. Eaglstein and V. Falanga, Tissue engineering and the
Government, Ministry of Education, Culture, Sports, Science development of Apligraf a human skin equivalent, Advances
and Technology (MEXT) for the scholarships provided for in Wound Care, vol. 11, supplement 4, pp. 18, 1998.
the doctoral studies. [18] B. D. Boyan, C. H. Lohmann, J. Romero, and Z. Schwartz,
Bone and cartilage tissue engineering, Clinics in Plastic
Surgery, vol. 26, no. 4, pp. 629645, 1999.
[19] J. Mayer, E. Karamuk, T. Akaike, and E. Wintermantel,
References Matrices for tissue engineering-scaold structure for a
bioartificial liver support system, Journal of Controlled
[1] R. Langer and J. P. Vacanti, Tissue engineering, Science, vol.
Release, vol. 64, no. 13, pp. 8190, 2000.
260, no. 5110, pp. 920926, 1993. [20] J. E. Mayer, T. Shinoka, and D. Shum-Tim, Tissue engi-
[2] R. M. Nerem, Tissue engineering in the USA, Medical and neering of cardiovascular structures, Current Opinion in
Biological Engineering and Computing, vol. 30, no. 4, pp. Cardiology, vol. 12, no. 6, pp. 528532, 1997.
CE8CE12, 1992. [21] F. Oberpenning, J. Meng, J. J. Yoo, and A. Atala, De novo
[3] R. Langer and D. A. Tirrell, Designing materials for biology reconstitution of a functional mammalian urinary bladder by
and medicine, Nature, vol. 428, no. 6982, pp. 487492, 2004. tissue engineering, Nature Biotechnology, vol. 17, no. 2, pp.
[4] J. R. Fuchs, B. A. Nasseri, and J. P. Vacanti, Tissue engi- 149155, 1999.
neering: a 21st century solution to surgical reconstruction, [22] E. Tziampazis and A. Sambanis, Tissue engineering of a
Annals of Thoracic Surgery, vol. 72, no. 2, pp. 577591, 2001. bioartificial pancreas: modeling the cell environment and
[5] I. V. Yannas, J. F. Burke, C. Huang, and P. L. Gordon, device function, Biotechnology Progress, vol. 11, no. 2, pp.
Suppression of in vivo degradability and of immunogenicity 115126, 1995.
of collagen by reaction with glycosaminoglycans, Polymer [23] J. Mohammad, J. Shenaq, E. Rabinovsky, and S. Shenaq,
Preprints, vol. 16, pp. 209214, 1975. Modulation of peripheral nerve regeneration: a tissue-
[6] I. V. Yannas, J. F. Burke, P. L. Gordon, and C. Huang, engineering approach. The role of amnion tube nerve
Multilayer membrane useful as synthetic skin, US patent conduit across a 1-centimeter nerve gap, Plastic and Recon-
4060081, 1977. structive Surgery, vol. 105, no. 2, pp. 660666, 2000.
[7] I. V. Yannas and J. F. Burke, Design of an artificial skin. [24] L. Germain, F. A. Auger, E. Grandbois et al., Reconstructed
I. Basic design principles, Journal of Biomedical Materials human cornea produced in vitro by tissue engineering,
Research, vol. 14, no. 1, pp. 6581, 1980. Pathobiology, vol. 67, no. 3, pp. 140147, 1999.
International Journal of Polymer Science 13

[25] C. A. Diedwardo, P. Petrosko, T. O. Acarturk, P. A. Dimilia, poly(D,L-lactic-co-glycolic acid) without the use of organic
W. A. Laframboise, and P. C. Johnson, Muscle tissue solvents, Biomaterials, vol. 17, no. 14, pp. 14171422, 1996.
engineering, Clinics in Plastic Surgery, vol. 26, no. 4, pp. 647 [42] J. J. Yoon and T. G. Park, Degradation behaviors of
656, 1999. biodegradable macroporous scaolds prepared by gas foam-
[26] L. S. Nair and C. T. Laurencin, Biodegradable polymers as ing of eervescent salts, Journal of Biomedical Materials
biomaterials, Progress in Polymer Science, vol. 32, no. 8-9, pp. Research, vol. 55, no. 3, pp. 401408, 2001.
762798, 2007. [43] W. L. Murphy, R. G. Dennis, J. L. Kileny, and D. J. Mooney,
[27] I. V. Yannas, Classes of materials used in medicine: natural Salt fusion: an approach to improve pore interconnectivity
materials, in Biomaterials ScienceAn Introduction to Mate- within tissue engineering scaolds, Tissue Engineering, vol.
rials in Medicine, B. D. Ratner, A. S. Homan, F. J. Schoen, 8, no. 1, pp. 4352, 2002.
and J. Lemons, Eds., pp. 127136, Elsevier Academic Press, [44] C. T. Laurencin, M. A. Attawia, H. E. Elgendy, and K. M. Her-
San Diego, Calif, USA, 2004. bert, Tissue engineered bone-regeneration using degradable
[28] P. Gunatillake, R. Mayadunne, and R. Adhikari, Recent polymers: the formation of mineralized matrices, Bone, vol.
developments in biodegradable synthetic polymers, Biotech- 19, no. 1, 1996.
nology Annual Review, vol. 12, pp. 301347, 2006. [45] J. E. Devin, M. A. Attawia, and C. T. Laurencin, Three-
[29] P. X. Ma, Scaolds for tissue fabrication, Materials Today, dimensional degradable porous polymer-ceramic matrices
vol. 7, no. 5, pp. 3040, 2004. for use in bone repair, Journal of Biomaterials Science, vol.
[30] L. J. Chen and M. Wang, Production and evaluation of 7, no. 8, pp. 661669, 1996.
biodegradable composites based on PHB-PHV copolymer, [46] B. H. Woo, J. W. Kostanski, S. Gebrekidan, B. A. Dani, B. C.
Biomaterials, vol. 23, no. 13, pp. 26312639, 2002. Thanoo, and P. P. DeLuca, Preparation, characterization and
[31] L. L. Hench, Bioceramics, Journal of the American Ceramic in vivo evaluation of 120-day poly(D,L-lactide) leuprolide
Society, vol. 81, no. 7, pp. 17051727, 1998. microspheres, Journal of Controlled Release, vol. 75, no. 3,
[32] M. G. Cascone, N. Barbani, C. Cristallini, P. Giusti, G. pp. 307315, 2001.
Ciardelli, and L. Lazzeri, Bioartificial polymeric materials [47] M. Borden, S. F. El-Amin, M. Attawia, and C. T. Lau-
based on polysaccharides, Journal of Biomaterials Science, rencin, Structural and human cellular assessment of a
vol. 12, no. 3, pp. 267281, 2001. novel microsphere-based tissue engineered scaold for bone
[33] G. Ciardelli, V. Chiono, G. Vozzi et al., Blends of poly- repair, Biomaterials, vol. 24, no. 4, pp. 597609, 2003.
(-caprolactone) and polysaccharides in tissue engineering [48] P. B. Malafaya, A. J. Pedro, A. Peterbauer, C. Gabriel, H. Redl,
applications, Biomacromolecules, vol. 6, no. 4, pp. 1961 and R. L. Reis, Chitosan particles agglomerated scaolds for
1976, 2005. cartilage and osteochondral tissue engineering approaches
[34] J. A. Roether, A. R. Boccaccini, L. L. Hench, V. Maquet, S. with adipose tissue derived stem cells, Journal of Materials

Gautier, and R. Jerome, Development and in vitro char- Science: Materials in Medicine, vol. 16, no. 12, pp. 10771085,
acterisation of novel bioresorbable and bioactive composite 2005.
materials based on polylactide foams and Bioglass for tissue [49] P. B. Malafaya, T. C. Santos, M. van Griensven, and R. L.
engineering applications, Biomaterials, vol. 23, no. 18, pp. Reis, Morphology, mechanical characterization and in vivo
38713878, 2002. neo-vascularization of chitosan particle aggregated scaolds
[35] A. G. Mikos, G. Sarakinos, S. M. Leite, J. P. Vacanti, and R. architectures, Biomaterials, vol. 29, no. 29, pp. 39143926,
Langer, Laminated three-dimensional biodegradable foams 2008.
for use in tissue engineering, Biomaterials, vol. 14, no. 5, pp. [50] R. Zhang and P. X. Ma, Porous poly(L-lactic acid)/apatite
323330, 1993. composites created by biomimetic process, Journal of
[36] A. G. Mikos, A. J. Thorsen, L. A. Czerwonka et al., Prepa- Biomedical Materials Research, vol. 45, no. 4, pp. 285293,
ration and characterization of poly(l-lactic acid) foams, 1999.
Polymer, vol. 35, no. 5, pp. 10681077, 1994. [51] Y. Ohya, H. Matsunami, E. Yamabe, and T. Ouchi,
[37] K. Ochi, G. Chen, T. Ushida et al., Use of isolated Cell attachment and growth on films prepared from
mature osteoblasts in abundance acts as desired-shaped bone poly(depsipeptide-co-lactide) having various functional
regeneration in combination with a modified poly-DL-lactic- groups, Journal of Biomedical Materials Research A, vol. 65,
co-glycolic acid (PLGA)-collagen sponge, Journal of Cellular no. 1, pp. 7988, 2003.
Physiology, vol. 194, no. 1, pp. 4553, 2003. [52] Y. Ohya, H. Matsunami, and T. Ouchi, Cell growth on the
[38] C. E. Holy, M. S. Shoichet, and J. E. Davies, Engineering porous sponges prepared from poly(depsipeptide-co-lactide)
three-dimensional bone tissue in vitro using biodegradable having various functional groups, Journal of Biomaterials
scaolds: investigating initial cell-seeding density and culture Science, vol. 15, no. 1, pp. 111123, 2004.
period, Journal of Biomedical Materials Research, vol. 51, no. [53] M. Borden, M. Attawia, Y. Khan, and C. T. Laurencin,
3, pp. 376382, 2000. Tissue engineered microsphere-based matrices for bone
[39] J. M. Karp, M. S. Shoichet, and J. E. Davies, Bone formation repair: design and evaluation, Biomaterials, vol. 23, no. 2,
on two-dimensional poly(DL-lactide-co-glycolide) (PLGA) pp. 551559, 2002.
films and three-dimensional PLGA tissue engineering scaf- [54] J. Guan, K. L. Fujimoto, M. S. Sacks, and W. R. Wag-
folds in vitro, Journal of Biomedical Materials Research A, vol. ner, Preparation and characterization of highly porous,
64, no. 2, pp. 388396, 2003. biodegradable polyurethane scaolds for soft tissue applica-
[40] H. G. Kang, S. Y. Kim, and Y. M. Lee, Novel porous gelatin tions, Biomaterials, vol. 26, no. 18, pp. 39613971, 2005.
scaolds by overrun/particle leaching process for tissue [55] T. J. Blokhuis, M. F. Termaat, F. C. Den Boer, P. Patka, F.
engineering applications, Journal of Biomedical Materials C. Bakker, and H. J. T. M. Haarman, Properties of calcium
Research B, vol. 79, no. 2, pp. 388397, 2006. phosphate ceramics in relation to their in vivo behavior,
[41] D. J. Mooney, D. F. Baldwin, N. P. Suh, J. P. Vacanti, and Journal of TraumaInjury, Infection and Critical Care, vol.
R. Langer, Novel approach to fabricate porous sponges of 48, no. 1, pp. 179186, 2000.
14 International Journal of Polymer Science

[56] T. A. Holland, J. K. V. Tessmar, Y. Tabata, and A. Biotechnology and Bioengineering, vol. 96, no. 5, pp. 977989,
G. Mikos, Transforming growth factor-1 release from 2007.
oligo(poly(ethylene glycol) fumarate) hydrogels in condi- [73] G. Steinho, U. Stock, N. Karim et al., Tissue engineering
tions that model the cartilage wound healing environment, of pulmonary heart valves on allogenic acellular matrix
Journal of Controlled Release, vol. 94, no. 1, pp. 101114, 2004. conduits: In vivo restoration of valve tissue, Circulation, vol.
[57] O. Gauthier, R. Muller, D. Von Stechow et al., In vivo bone 102, no. 19, pp. III50III55, 2000.
regeneration with injectable calcium phosphate biomaterial: [74] D. E. Zhao, R. B. Li, W. Y. Liu et al., Tissue-engineered heart
a three-dimensional micro-computed tomographic, biome- valve on acellular aortic valve scaold: in-vivo study, Asian
chanical and SEM study, Biomaterials, vol. 26, no. 27, pp. Cardiovascular and Thoracic Annals, vol. 11, no. 2, pp. 153
54445453, 2005. 156, 2003.
[58] B. Jeong, Y. H. Bae, and S. W. Kim, Thermoreversible [75] H. C. Liang, Y. Chang, C. K. Hsu, M. H. Lee, and H. W. Sung,
gelation of PEG-PLGA-PEG triblock copolymer aqueous Eects of crosslinking degree of an acellular biological tissue
solutions, Macromolecules, vol. 32, no. 21, pp. 70647069, on its tissue regeneration pattern, Biomaterials, vol. 25, no.
1999. 17, pp. 35413552, 2004.
[59] S. Ibusuki, Y. Fujii, Y. Iwamoto, and T. Matsuda, Tissue- [76] A. Tachibana, Y. Furuta, H. Takeshima, T. Tanabe, and K.
engineered cartilage using an injectable and in situ gelable Yamauchi, Fabrication of wool keratin sponge scaolds for
thermoresponsive gelatin: fabrication and in vitro perfor- long-term cell cultivation, Journal of Biotechnology, vol. 93,
mance, Tissue Engineering, vol. 9, no. 2, pp. 371384, 2003. no. 2, pp. 165170, 2002.
[60] J. Y. Seong, Y. J. Jun, B. Jeong, and Y. S. Sohn, New [77] A. Tachibana, S. Kaneko, T. Tanabe, and K. Yamauchi, Rapid
thermogelling poly(organophosphazenes) with methoxy- fabrication of keratin-hydroxyapatite hybrid sponges toward
poly(ethylene glycol) and oligopeptide as side groups, osteoblast cultivation and dierentiation, Biomaterials, vol.
Polymer, vol. 46, no. 14, pp. 50755081, 2005. 26, no. 3, pp. 297302, 2005.
[61] J. Yeh, Y. Ling, J. M. Karp et al., Micromolding of shape- [78] K. Katoh, T. Tanabe, and K. Yamauchi, Novel approach to
controlled, harvestable cell-laden hydrogels, Biomaterials, fabricate keratin sponge scaolds with controlled pore size
vol. 27, no. 31, pp. 53915398, 2006. and porosity, Biomaterials, vol. 25, no. 18, pp. 42554262,
[62] J. Fukuda, A. Khademhosseini, Y. Yeo et al., Micromold- 2004.
ing of photocrosslinkable chitosan hydrogel for spheroid [79] J. Doshi and D. H. Reneker, Electrospinning process and
microarray and co-cultures, Biomaterials, vol. 27, no. 30, pp. applications of electrospun fibers, Journal of Electrostatics,
52595267, 2006. vol. 35, no. 2-3, pp. 151160, 1995.
[63] A. Khademhosseini, G. Eng, J. Yeh et al., Micromolding of
[80] W. J. Li, K. G. Danielson, P. G. Alexander, and R. S.
photocrosslinkable hyaluronic acid for cell encapsulation and
Tuan, Biological response of chondrocytes cultured in three-
entrapment, Journal of Biomedical Materials Research A, vol.
dimensional nanofibrous poly(-caprolactone) scaolds,
79, no. 3, pp. 522532, 2006.
Journal of Biomedical Materials Research A, vol. 67, no. 4, pp.
[64] D. J. Beebe, J. S. Moore, J. M. Bauer et al., Functional
11051114, 2003.
hydrogel structures for autonomous flow control inside
[81] J. Zeng, A. Aigner, F. Czubayko, T. Kissel, J. H. Wendor,
microfluidic channels, Nature, vol. 404, no. 6778, pp. 588
and A. Greiner, Poly(vinyl alcohol) nanofibers by electro-
590, 2000.
spinning as a protein delivery system and the retardation of
[65] V. A. Liu and S. N. Bhatia, Three-dimensional photopat-
enzyme release by additional polymer coatings, Biomacro-
terning of hydrogels containing living cells, Biomedical
molecules, vol. 6, no. 3, pp. 14841488, 2005.
Microdevices, vol. 4, no. 4, pp. 257266, 2002.
[66] D. Dendukuri, D. C. Pregibon, J. Collins, T. A. Hatton, and P. [82] S. Hirano, M. Zhang, M. Nakagawa, and T. Miyata,
S. Doyle, Continuous-flow lithography for high-throughput Wet spun chitosan-collagen fibers, their chemical N-
microparticle synthesis, Nature Materials, vol. 5, no. 5, pp. modifications, and blood compatibility, Biomaterials, vol.
365369, 2006. 21, no. 10, pp. 9971003, 2000.
[67] T. Nisisako, T. Torii, and T. Higuchi, Droplet formation in a [83] S. J. Pomfret, P. N. Adams, N. P. Comfort, and A. P.
microchannel network, Lab on a ChipMiniaturisation for Monkman, Electrical and mechanical properties of polyani-
Chemistry and Biology, vol. 2, no. 1, pp. 2426, 2002. line fibres produced by a one-step wet spinning process,
[68] J. A. Burdick, A. Khademhosseini, and R. Langer, Fabri- Polymer, vol. 41, no. 6, pp. 22652269, 2000.
cation of gradient hydrogels using a microfluidics/photo- [84] H. Okuzaki, Y. Harashina, and H. Yan, Highly conductive
polymerization process, Langmuir, vol. 20, no. 13, pp. 5153 PEDOT/PSS microfibers fabricated by wet-spinning and dip-
5156, 2004. treatment in ethylene glycol, European Polymer Journal, vol.
[69] S. Xu, Z. Nie, M. Seo et al., Generation of monodisperse 45, no. 1, pp. 256261, 2009.
particles by using microfluidics: control over size, shape, and [85] J. Lyons, C. Li, and F. Ko, Melt-electrospinningpart I:
composition, Angewandte Chemie International Edition, vol. processing parameters and geometric properties, Polymer,
44, no. 5, pp. 724728, 2005. vol. 45, no. 22, pp. 75977603, 2004.
[70] N. A. Peppas and A. R. Khare, Preparation, structure [86] C. J. Ellison, A. Phatak, D. W. Giles, C. W. Macosko, and F. S.
and diusional behavior of hydrogels in controlled release, Bates, Melt blown nanofibers: fiber diameter distributions
Advanced Drug Delivery Reviews, vol. 11, no. 1-2, pp. 135, and onset of fiber breakup, Polymer, vol. 48, no. 11, pp.
1993. 33063316, 2007.
[71] T. Alexakis, K. Boadid, D. Guong et al., Microencapsulation [87] K. Kim, C. Lee, I. W. Kim, and J. Kim, Performance
of DNA within alginate microspheres and crosslinked chi- modification of a melt-blown filter medium via an additional
tosan membranes for in vivo application, Applied Biochem- nano-web layer prepared by electrospinning, Fibers and
istry and Biotechnology, vol. 50, no. 1, pp. 93106, 1995. Polymers, vol. 10, no. 1, pp. 6064, 2009.
[72] C. P. Reis, A. J. Ribeiro, R. J. Neufeld, and F. Veiga, Alginate [88] M. J. B. Wissink, R. Beernink, J. S. Pieper et al., Binding
microparticles as novel carrier for oral insulin delivery, and release of basic fibroblast growth factor from heparinized
International Journal of Polymer Science 15

collagen matrices, Biomaterials, vol. 22, no. 16, pp. 2291 scaolds, CAD Computer Aided Design, vol. 37, no. 1, pp.
2299, 2001. 6572, 2005.
[89] F. Causa, P. A. Netti, and L. Ambrosio, A multi-functional [106] S. Lalan, I. Pomerantseva, and J. P. Vacanti, Tissue engi-
scaold for tissue regeneration: the need to engineer a tissue neering and its potential impact on surgery, World Journal
analogue, Biomaterials, vol. 28, no. 34, pp. 50935099, 2007. of Surgery, vol. 25, no. 11, pp. 14581466, 2001.
[90] Y. C. Ho, F. L. Mi, H. W. Sung, and P. L. Kuo, Heparin- [107] T. Boland, V. Mironov, A. Gutowska, E. A. Roth, and R.
functionalized chitosan-alginate scaolds for controlled R. Markwald, Cell and organ printing 2: fusion of cell
release of growth factor, International Journal of Pharmaceu- aggregates in three-dimensional gels, Anatomical Record A,
tics, vol. 376, no. 1-2, pp. 6975, 2009. vol. 272, no. 2, pp. 497502, 2003.
[91] P. Sepulveda and J. G. P. Binner, Processing of cellular [108] N. Hirata, K. I. Matsumoto, T. Inishita, Y. Takenaka, Y. Suma,
ceramics by foaming and in situ polymerisation of organic and H. Shintani, Gamma-ray irradiation, autoclave and
monomers, Journal of the European Ceramic Society, vol. 19, ethylene oxide sterilization to thermosetting polyurethane:
no. 12, pp. 20592066, 1999. sterilization to polyurethane, Radiation Physics and Chem-
[92] Q. Z. Chen, I. D. Thompson, and A. R. Boccaccini, 45S5 istry, vol. 46, no. 3, pp. 377381, 1995.
Bioglass -derived glass-ceramic scaolds for bone tissue [109] K. A. Hooper, J. D. Cox, and J. Kohn, Comparison of
engineering, Biomaterials, vol. 27, no. 11, pp. 24142425, the eect of ethylene oxide and -irradiation on selected
2006. tyrosine-derived polycarbonates and poly(L-lactic acid),
[93] I. H. Jo, K. H. Shin, Y. M. Soon, Y. H. Koh, J. H. Lee, and H. E. Journal of Applied Polymer Science, vol. 63, no. 11, pp. 1499
Kim, Highly porous hydroxyapatite scaolds with elongated 1510, 1997.
pores using stretched polymeric sponges as novel template, [110] C. E. Holy, C. Cheng, J. E. Davies, and M. S. Shoichet,
Materials Letters, vol. 63, no. 20, pp. 17021704, 2009. Optimizing the sterilization of PLGA scaolds for use in
[94] F. Li, Q. L. Feng, F. Z. Cui, H. D. Li, and H. Schubert, A tissue engineering, Biomaterials, vol. 22, no. 1, pp. 2531,
simple biomimetic method for calcium phosphate coating, 2001.
Surface and Coatings Technology, vol. 154, no. 1, pp. 8893, [111] C. Volland, M. Wol, and T. Kissel, The influence of ter-
2002. minal gamma-sterilization on captopril containing poly(D,L-
[95] J. Chen, B. Chu, and B. S. Hsiao, Mineralization of hydrox- lactide-co-glycolide) microspheres, Journal of Controlled
yapatite in electrospun nanofibrous poly(L-lactic acid) scaf- Release, vol. 31, no. 3, pp. 293304, 1994.
folds, Journal of Biomedical Materials Research A, vol. 79, no. [112] M. B. Sintzel, K. S. Abdellaoui, K. Mader et al., Influence
2, pp. 307317, 2006. of irradiation sterilization on a semi-solid poly(ortho ester),
[96] F. Yang, J. G. C. Wolke, and J. A. Jansen, Biomimetic cal- International Journal of Pharmaceutics, vol. 175, pp. 165176,
cium phosphate coating on electrospun poly(-caprolactone) 1998.
scaolds for bone tissue engineering, Chemical Engineering [113] L. Montanari, M. Costantini, E. C. Signoretti et al., Gamma
Journal, vol. 137, no. 1, pp. 154161, 2008. irradiation eects on poly(DL-lactictide-co-glycolide)
[97] A. V. Lemmo, D. J. Rose, and T. C. Tisone, Inkjet dispensing microspheres, Journal of Controlled Release, vol. 56, no. 13,
technology: applications in drug discovery, Current Opinion pp. 219229, 1998.
in Biotechnology, vol. 9, no. 6, pp. 615617, 1998. [114] M. Ohrlander, R. Erickson, R. Palmgren, A. Wirsen, and A. C.
[98] P. Calvert, Inkjet printing for materials and devices, Albertsson, The eect of electron beam irradiation on PCL
Chemistry of Materials, vol. 13, no. 10, pp. 32993305, 2001. and PDXO-X monitored by luminescence and electron spin
[99] L. Pardo, W. Cris Wilson, and T. Boland, Characterization resonance measurements, Polymer, vol. 41, pp. 12771286,
of patterned self-assembled monolayers and protein arrays 1999.
generated by the ink-jet method, Langmuir, vol. 19, no. 5, [115] J. S. C. Loo, C. P. Ooi, and F. Y. C. Boey, Degradation
pp. 14621466, 2003. of poly(lactide-co-glycolide) (PLGA) and poly(L-lactide)
[100] W. Y. Yeong, C. K. Chua, K. F. Leong, M. Chandrasekaran, (PLLA) by electron beam radiation, Biomaterials, vol. 26, no.
and M. W. Lee, Indirect fabrication of collagen scaold 12, pp. 13591367, 2005.
based on inkjet printing technique, Rapid Prototyping [116] K. Odelius, P. Plikk, and A. C. Albertsson, The influence
Journal, vol. 12, no. 4, pp. 229237, 2006. of composition of porous copolyester scaolds on reactions
[101] V. Mironov, T. Boland, T. Trusk, G. Forgacs, and R. R. induced by irradiation sterilization, Biomaterials, vol. 29, no.
Markwald, Organ printing: computer-aided jet-based 3D 2, pp. 129140, 2008.
tissue engineering, Trends in Biotechnology, vol. 21, no. 4, pp. [117] E. M. Darmady, K. E. Hughes, J. D. Jones, D. Prince, and W.
157161, 2003. Tuke, Sterilization by dry heat, Journal of Clinical Pathology,
[102] W. Sun, A. Darling, B. Starly, and J. Nam, Computer- vol. 14, pp. 3844, 1961.
aided tissue engineering: overview, scope and challenges, [118] Q. Fu, M. N. Rahaman, B. S. Bal, and R. F. Brown, In vitro
Biotechnology and Applied Biochemistry, vol. 39, part 1, pp. cellular response to hydroxyapatite scaolds with oriented
2947, 2004. pore architectures, Materials Science and Engineering C, vol.
[103] W. Sun and P. Lal, Recent development on computer 29, no. 7, pp. 21472153, 2009.
aided tissue engineeringa review, Computer Methods and [119] F. A. Muller, L. Muller, I. Hofmann, P. Greil, M. M. Wenzel,
Programs in Biomedicine, vol. 67, no. 2, pp. 85103, 2002. and R. Staudenmaier, Cellulose-based scaold materials for
[104] W. Sun, B. Starly, A. Darling, and C. Gomez, Computer- cartilage tissue engineering, Biomaterials, vol. 27, no. 21, pp.
aided tissue engineering: application to biomimetic mod- 39553963, 2006.
elling and design of tissue scaolds, Biotechnology and [120] K. Gellynck, P. C. M. Verdonk, E. Van Nimmen et al., Silk-
Applied Biochemistry, vol. 39, no. 1, pp. 4958, 2004. worm and spider silk scaolds for chondrocyte support,
[105] Z. Fang, B. Starly, and W. Sun, Computer-aided character- Journal of Materials Science: Materials in Medicine, vol. 19,
ization for eective mechanical properties of porous tissue no. 11, pp. 33993409, 2008.
16 International Journal of Polymer Science

[121] S. Terasaka, Y. Iwasaki, N. Shinya, and T. Uchida, Fibrin glue weight of HA on elasticity of cartilage matrix, Biorheology,
and polyglycolic acid nonwoven fabric as a biocompatible vol. 43, no. 3-4, pp. 347354, 2006.
dural substitute, Neurosurgery, vol. 58, no. 1, pp. S-134S- [138] D. W. Hutmacher, Scaold design and fabrication tech-
138, 2006. nologies for engineering tissuesState of the art and future
[122] P. B. Maurus and C. C. Kaeding, Bioabsorbable implant perspectives, Journal of Biomaterials Science, vol. 12, no. 1,
material review, Operative Techniques in Sports Medicine, pp. 107124, 2001.
vol. 12, no. 3, pp. 158160, 2004. [139] L. E. Freed, G. Vunjak-Novakovic, R. J. Biron et al.,
[123] H. H. Lu, J. A. Cooper, S. Manuel et al., Anterior cruciate Biodegradable polymer scaolds for tissue engineering,
ligament regeneration using braided biodegradable scaolds: Biotechnology, vol. 12, no. 7, pp. 689693, 1994.
in vitro optimization studies, Biomaterials, vol. 26, no. 23, [140] L. E. Freed and G. Vunjak-Novakovic, Culture of organized
pp. 48054816, 2005. cell communities, Advanced Drug Delivery Reviews, vol. 33,
[124] J. A. Cooper, H. H. Lu, F. K. Ko, J. W. Freeman, and no. 1-2, pp. 1530, 1998.
C. T. Laurencin, Fiber-based tissue-engineered scaold for [141] P. X. Ma and R. Zhang, Microtubular architecture of
ligament replacement: design considerations and in vitro biodegradable polymer scaolds, Journal of Biomedical
evaluation, Biomaterials, vol. 26, no. 13, pp. 15231532, Materials Research, vol. 56, no. 4, pp. 469477, 2001.
2005. [142] S. Freiberg and X. X. Zhu, Polymer microspheres for con-
[125] M. Zilberman, K. D. Nelson, and R. C. Eberhart, Mechanical trolled drug release, International Journal of Pharmaceutics,
properties and in vitro degradation of bioresorbable fibers vol. 282, no. 1-2, pp. 118, 2004.
and expandable fiber-based stents, Journal of Biomedical [143] D. J. Mooney, G. Organ, J. P. Vacanti, and R. Langer, Design
Materials Research B, vol. 74, no. 2, pp. 792799, 2005. and fabrication of biodegradable polymer devices to engineer
[126] S. Leinonen, E. Suokas, M. Veiranto, P. Tormala, T. Waris, tubular tissues, Cell Transplantation, vol. 3, no. 2, pp. 203
and N. Ashammakhi, Healing power of bioadsorbable 210, 1994.
ciprofloxacin- containing self reinforced poly(L/DL-lactide [144] G. Wei and P. X. Ma, Structure and properties of nano-
70/30 bioactive glass 13 miniscrews in human cadaver bone, hydroxyapatite/polymer composite scaolds for bone tissue
Journal of Craniofacial Surgery, vol. 13, pp. 212218, 2002. engineering, Biomaterials, vol. 25, no. 19, pp. 47494757,
[127] C. W. Pouton and S. Akhtar, Biosynthetic polyhydroxyalka- 2004.
noates and their potential in drug delivery, Advanced Drug [145] E. M. Ouriemchi and J. M. Vergnaud, Processes of
Delivery Reviews, vol. 18, no. 2, pp. 133162, 1996. drug transfer with three dierent polymeric systems with
[128] B. Saad, T. D. Hirt, M. Welti, G. K. Uhlschmid, P. Neuen- transdermal drug delivery, Computational and Theoretical
schwander, and U. W. Suter, Development of degradable Polymer Science, vol. 10, no. 5, pp. 391401, 2000.
polyesterurethanes for medical applications: in vitro and in [146] Q. Hou, D. W. Grijpma, and J. Feijen, Preparation
vivo evaluations, Journal of Biomedical Materials Research, of porous poly(-caprolactone) structures, Macromolecular
vol. 36, no. 1, pp. 6574, 1997. Rapid Communications, vol. 23, no. 4, pp. 247252, 2002.
[129] I. C. Bonzani, R. Adhikari, S. Houshyar, R. Mayadunne, [147] Q. Hou, D. W. Grijpma, and J. Feijen, Porous polymeric
P. Gunatillake, and M. M. Stevens, Synthesis of two- structures for tissue engineering prepared by a coagulation,
component injectable polyurethanes for bone tissue engi- compression moulding and salt leaching technique, Bioma-
neering, Biomaterials, vol. 28, no. 3, pp. 423433, 2007. terials, vol. 24, no. 11, pp. 19371947, 2003.
[130] J. Heller, Ocular delivery using poly(ortho esters), [148] M. Cabodi, N. W. Choi, J. P. Gleghorn, C. S. D. Lee, L. J.
Advanced Drug Delivery Reviews, vol. 57, no. 14, pp. 2053 Bonassar, and A. D. Stroock, A microfluidic biomaterial,
2062, 2005. Journal of the American Chemical Society, vol. 127, no. 40, pp.
[131] D. S. Katti, S. Lakshmi, R. Langer, and C. T. Laurencin, Tox- 1378813789, 2005.
icity, biodegradation and elimination of polyanhydrides, [149] M. S. Jhon and J. D. Andrade, Water and hydrogels, Journal
Advanced Drug Delivery Reviews, vol. 54, no. 7, pp. 933961, of Biomedical Materials Research, vol. 7, no. 6, pp. 509522,
2002. 1973.
[132] C. Vauthier, C. Dubernet, C. Chauvierre, I. Brigger, and P. [150] A. S. Homan, Hydrogels for biomedical applications,
Couvreur, Drug delivery to resistant tumors: the potential Annals of the New York Academy of Sciences, vol. 944, pp. 62
of poly(alkyl cyanoacrylate) nanoparticles, Journal of Con- 73, 2001.
trolled Release, vol. 93, no. 2, pp. 151160, 2003. [151] J. A. Hubbell, Bioactive biomaterials, Current Opinion in
[133] P. Sai and M. Babu, Collagen based dressingsa review, Biotechnology, vol. 10, no. 2, pp. 123129, 1999.
Burns, vol. 26, no. 1, pp. 5462, 2000. [152] K. Y. Lee and D. J. Mooney, Hydrogels for tissue engineer-
[134] X. Duan, C. McLaughlin, M. Grith, and H. Sheardown, ing, Chemical Reviews, vol. 101, no. 7, pp. 18691879, 2001.
Biofunctionalization of collagen for improved biological [153] N. A. Peppas and A. R. Khare, Preparation, structure
response: scaolds for corneal tissue engineering, Biomate- and diusional behavior of hydrogels in controlled release,
rials, vol. 28, no. 1, pp. 7888, 2007. Advanced Drug Delivery Reviews, vol. 11, no. 1-2, pp. 135,
[135] D. R. Hunt, S. A. Joanovic, U. M. E. Wikesjo, J. M. Wozney, 1993.
and D. W. Bernard, Hyaluronan supports recombinant [154] Y. Tabata, Tissue regeneration based on growth factor
human bone morphogenetic protein-2 induced bone recon- release, Tissue Engineering, vol. 9, no. 4, pp. 515, 2003.
struction of advanced alveolar ridge defects in dogs. A pilot [155] S. J. Bryant and K. S. Anseth, The eects of scaold
study, Journal of Periodontology, vol. 72, pp. 651657, 2001. thickness on tissue engineered cartilage in photocrosslinked
[136] B. L. Eppley and B. Dadvand, Injectable soft-tissue fillers: poly(ethylene oxide) hydrogels, Biomaterials, vol. 22, no. 6,
clinical overview, Plastic and Reconstructive Surgery, vol. 118, pp. 619626, 2001.
no. 4, pp. 98e106e, 2006. [156] D. G. Wallace and J. Rosenblatt, Collagen gel systems for
[137] Y. Kato, S. Nakamura, and M. Nishimura, Beneficial actions sustained delivery and tissue engineering, Advanced Drug
of hyaluronan (HA) on arthritic joints: eects of molecular Delivery Reviews, vol. 55, no. 12, pp. 16311649, 2003.
International Journal of Polymer Science 17

[157] U. J. Kim, J. Park, C. Li, H. J. Jin, R. Valluzzi, and D. [174] F. Yang, R. Murugan, S. Wang, and S. Ramakrishna, Elec-
L. Kaplan, Structure and properties of silk hydrogels, trospinning of nano/micro scale poly(l-lactic acid) aligned
Biomacromolecules, vol. 5, no. 3, pp. 786792, 2004. fibers and their potential in neural tissue engineering,
[158] D. Eyrich, F. Brandl, B. Appel et al., Long-term stable fibrin Biomaterials, vol. 26, no. 15, pp. 26032610, 2005.
gels for cartilage engineering, Biomaterials, vol. 28, no. 1, pp. [175] S. A. Riboldi, M. Sampaolesi, P. Neuenschwander, G. Cossu,
5565, 2007. and S. Mantero, Electrospun degradable polyesterurethane
[159] L. A. Solchaga, J. Gao, J. E. Dennis et al., Treatment of membranes: potential scaolds for skeletal muscle tissue
osteochondral defects with autologous bone marrow in a engineering, Biomaterials, vol. 26, no. 22, pp. 46064615,
hyaluronan-based delivery vehicle, Tissue Engineering, vol. 2005.
8, no. 2, pp. 333347, 2002. [176] W. J. Li, K. G. Danielson, P. G. Alexander, and R. S.
[160] H. J. Kong, M. K. Smith, and D. J. Mooney, Designing Tuan, Biological response of chondrocytes cultured in three-
alginate hydrogels to maintain viability of immobilized cells, dimensional nanofibrous poly(-caprolactone) scaolds,
Biomaterials, vol. 24, no. 22, pp. 40234029, 2003. Journal of Biomedical Materials Research A, vol. 67, no. 4, pp.
[161] J. K. Francis Suh and H. W. T. Matthew, Application of 11051114, 2003.
chitosan-based polysaccharide biomaterials in cartilage tissue [177] K. Uematsu, K. Hattori, Y. Ishimoto et al., Cartilage regener-
engineering: a review, Biomaterials, vol. 21, no. 24, pp. 2589 ation using mesenchymal stem cells and a three-dimensional
2598, 2000. poly-lactic-glycolic acid (PLGA) scaold, Biomaterials, vol.
[162] R. H. Schmedlen, K. S. Masters, and J. L. West, Pho- 26, no. 20, pp. 42734279, 2005.
tocrosslinkable polyvinyl alcohol hydrogels that can be modi- [178] E. R. Kenawy, G. L. Bowlin, K. Mansfield et al., Release of
fied with cell adhesion peptides for use in tissue engineering, tetracycline hydrochloride from electrospun poly(ethylene-
Biomaterials, vol. 23, no. 22, pp. 43254332, 2002. co-vinylacetate), poly(lactic acid), and a blend, Journal of
[163] E. Behravesh and A. G. Mikos, Three-dimensional culture Controlled Release, vol. 81, no. 1-2, pp. 5764, 2002.
of dierentiating marrow stromal osteoblasts in biomimetic [179] X. M. Mo, C. Y. Xu, M. Kotaki, and S. Ramakrishna,
poly(propylene fumarate-co-ethylene glycol)-based macrop- Electrospun P(LLA-CL) nanofiber: a biomimetic extracel-
orous hydrogels, Journal of Biomedical Materials Research A, lular matrix for smooth muscle cell and endothelial cell
vol. 66, no. 3, pp. 698706, 2003. proliferation, Biomaterials, vol. 25, no. 10, pp. 18831890,
[164] S. J. Bryant, K. A. Davis-Arehart, N. Luo, R. K. Shoemaker, 2004.
J. A. Arthur, and K. S. Anseth, Synthesis and charac- [180] G. Verreck, I. Chun, J. Rosenblatt et al., Incorporation of
terization of photopolymerized multifunctional hydrogels: drugs in an amorphous state into electrospun nanofibers
water-soluble poly(vinyl alcohol) and chondroitin sulfate composed of a water-insoluble, nonbiodegradable polymer,
macromers for chondrocyte encapsulation, Macromolecules, Journal of Controlled Release, vol. 92, no. 3, pp. 349360, 2003.
vol. 37, no. 18, pp. 67266733, 2004. [181] M. Singh, C. P. Morris, R. J. Ellis, M. S. Detamore, and C.
[165] P. Berndt, G. B. Fields, and M. Tirrell, Synthetic lipidation Berkland, Microsphere-based seamless scaolds containing
of peptides and amino acids: monolayer structure and macroscopic gradients of encapsulated factors for tissue
properties, Journal of the American Chemical Society, vol. engineering, Tissue Engineering C, vol. 14, no. 4, pp. 299
117, no. 37, pp. 95159522, 1995. 309, 2008.
[166] S. R. Bhattarai, N. Bhattarai, H. K. Yi, P. H. Hwang, D. I. Cha, [182] M. Singh, B. Sandhu, A. Scurto, C. Berkland, and M. S.
and H. Y. Kim, Novel biodegradable electrospun membrane: Detamore, Microsphere-based scaolds for cartilage tissue
scaold for tissue engineering, Biomaterials, vol. 25, no. 13, engineering: using subcritical CO2 as a sintering agent, Acta
pp. 25952602, 2004. Biomaterialia, vol. 6, no. 1, pp. 137143, 2010.
[167] Z. Ma, M. Kotaki, R. Inai, and S. Ramakrishna, Potential [183] D. Stephens, L. Li, D. Robinson et al., Investigation of the
of nanofiber matrix as tissue-engineering scaolds, Tissue in vitro release of gentamicin from a polyanhydride matrix,
Engineering, vol. 11, no. 1-2, pp. 101109, 2005. Journal of Controlled Release, vol. 63, no. 3, pp. 305317, 2000.
[168] R. Vasita and D. S. Katti, Nanofibers and their applications [184] C. Berkland, M. King, A. Cox, K. Kim, and D. W. Pack,
in tissue engineering, International Journal of Nanomedicine, Precise control of PLG microsphere size provides enhanced
vol. 1, no. 1, pp. 1530, 2006. control of drug release rate, Journal of Controlled Release, vol.
[169] J. A. Matthews, G. E. Wnek, D. G. Simpson, and G. L. Bowlin, 82, no. 1, pp. 137147, 2002.
Electrospinning of collagen nanofibers, Biomacromolecules, [185] H. B. Ravivarapu, K. Burton, and P. P. DeLuca, Polymer and
vol. 3, no. 2, pp. 232238, 2002. microsphere blending to alter the release of a peptide from
[170] Y. Zhang, H. Ouyang, T. L. Chwee, S. Ramakrishna, and Z. PLGA microspheres, European Journal of Pharmaceutics and
M. Huang, Electrospinning of gelatin fibers and gelatin/PCL Biopharmaceutics, vol. 50, no. 2, pp. 263270, 2000.
composite fibrous scaolds, Journal of Biomedical Materials [186] R. A. Jain, C. T. Rhodes, A. M. Railkar, A. W. Malick, and
Research B, vol. 72, no. 1, pp. 156165, 2005. N. H. Shah, Controlled delivery of drugs from a novel
[171] X. Geng, O. H. Kwon, and J. Jang, Electrospinning of injectable in situ formed biodegradable PLGA microsphere
chitosan dissolved in concentrated acetic acid solution, system, Journal of Microencapsulation, vol. 17, no. 3, pp.
Biomaterials, vol. 26, no. 27, pp. 54275432, 2005. 343362, 2000.
[172] I. C. Um, D. Fang, B. S. Hsiao, A. Okamoto, and B. Chu, [187] C. Berkland, K. Kim, and D. W. Pack, PLG microsphere size
Electro-spinning and electro-blowing of hyaluronic acid, controls drug release rate through several competing factors,
Biomacromolecules, vol. 5, no. 4, pp. 14281436, 2004. Pharmaceutical Research, vol. 20, no. 7, pp. 10551062, 2003.
[173] H. J. Jin, J. Chen, V. Karageorgiou, G. H. Altman, and D. [188] M. Borden, M. Attawia, Y. Khan, S. F. El-Amin, and C. T.
L. Kaplan, Human bone marrow stromal cell responses on Laurencin, Tissue-engineered bone formation in vivo using
electrospun silk fibroin mats, Biomaterials, vol. 25, no. 6, pp. a novel sintered polymeric microsphere matrix, Journal of
10391047, 2004. Bone and Joint Surgery B, vol. 86, no. 8, pp. 12001208, 2004.
18 International Journal of Polymer Science

[189] J. Yao, S. Radin, P. S. Leboy P., and P. Ducheyne, The [205] S. E. Dahms, H. J. Piechota, R. Dahiya, T. F. Lue, and E. A.
eect of bioactive glass content on synthesis and bioactivity Tanagho, Composition and biomechanical properties of the
of composite poly (lactic-co-glycolic acid)/bioactive glass bladder acellular matrix graft: comparative analysis in rat, pig
substrate for tissue engineering, Biomaterials, vol. 26, no. 14, and human, British Journal of Urology, vol. 82, no. 3, pp.
pp. 19351943, 2005. 411419, 1998.
[190] A. Jaklenec, E. Wan, M. E. Murray, and E. Mathiowitz, Novel [206] J. J. Yoo, J. Meng, F. Oberpenning, and A. Atala, Bladder
scaolds fabricated from protein-loaded microspheres for augmentation using allogenic bladder submucosa seeded
tissue engineering, Biomaterials, vol. 29, no. 2, pp. 185192, with cells, Urology, vol. 51, no. 2, pp. 221225, 1998.
2008. [207] F. Chen, J. J. Yoo, and A. Atala, Acellular collagen matrix
[191] A. Jaklenec, A. Hinckfuss, B. Bilgen, D. M. Ciombor, R. as a possible o the shelf biomaterial for urethral repair,
Aaron, and E. Mathiowitz, Sequential release of bioactive Urology, vol. 54, no. 3, pp. 407410, 1999.
IGF-I and TGF-1 from PLGA microsphere-based scaolds, [208] G. J. Wilson, D. W. Courtman, P. Klement, J. M. Lee, and
Biomaterials, vol. 29, no. 10, pp. 15181525, 2008. H. Yeger, Acellular matrix: a biomaterials approach for
[192] J. L. Brown, L. S. Nair, and C. T. Laurencin, Solvent/non- coronary artery bypass and heart valve replacement, Annals
solvent sintering: a novel route to create porous microsphere of Thoracic Surgery, vol. 60, supplement 2, pp. S353S358,
scaolds for tissue regeneration, Journal of Biomedical 1995.
Materials Research B, vol. 86, no. 2, pp. 396406, 2008. [209] S. E. Dahms, H. J. Piechota, L. Nunes, R. Dahiya, T. F.
[193] S. P. Nukavarapu, S. G. Kumbar, J. L. Brown et al., Lue, and E. A. Tanagho, Free ureteral replacement in rats:
Polyphosphazene/nano-hydroxyapatite composite micro- regeneration of ureteral wall components in the acellular
sphere scaolds for bone tissue engineering, Biomacro- matrix graft, Urology, vol. 50, no. 5, pp. 818825, 1997.
molecules, vol. 9, no. 7, pp. 18181825, 2008. [210] M. Probst, R. Dahiya, S. Carrier, and E. A. Tanagho,
[194] M. Borden, M. Attawia, and C. T. Laurencin, The sintered Reproduction of functional smooth muscle tissue and
microsphere matrix for bone tissue engineering: in vitro partial bladder replacement, British Journal of Urology, vol.
osteoconductivity studies, Journal of Biomedical Materials 79, no. 4, pp. 505515, 1997.
Research, vol. 61, no. 3, pp. 421429, 2002. [211] T. W. Gilbert, T. L. Sellaro, and S. F. Badylak, Decellulariza-
[195] Y. M. Khan, D. S. Katti, and C. T. Laurencin, Novel polymer- tion of tissues and organs, Biomaterials, vol. 27, no. 19, pp.
synthesized ceramic composite-based system for bone repair: 36753683, 2006.
an in vitro evaluation, Journal of Biomedical Materials [212] C. Stamm, A. Khosravi, N. Grabow et al., Bioma-
Research A, vol. 69, no. 4, pp. 728737, 2004. trix/polymer composite material for heart valve tissue engi-
[196] K. Rezwan, Q. Z. Chen, J. J. Blaker, and A. R. Boccaccini, neering, Annals of Thoracic Surgery, vol. 78, no. 6, pp. 2084
Biodegradable and bioactive porous polymer/inorganic 2093, 2004.
composite scaolds for bone tissue engineering, Biomateri- [213] M. Sokolsky-Papkov, K. Agashi, A. Olaye, K. Shakeshe, and
als, vol. 27, no. 18, pp. 34133431, 2006. A. J. Domb, Polymer carriers for drug delivery in tissue
[197] L. L. Hench, Bioceramics: from concept to clinic, American engineering, Advanced Drug Delivery Reviews, vol. 59, no.
Ceramic Society Bulletin, vol. 72, pp. 9398, 1993. 4-5, pp. 187206, 2007.
[198] R. L. Hentrich Jr., G. A. Graves Jr., H. G. Stein, and P. [214] B. D. Boyan, T. W. Hummert, D. D. Dean, and Z. Schwartz,
K. Bajpai, Evaluation of inert and resorbable ceramics for Role of material surfaces in regulating bone and cartilage
future clinical orthopedic applications, Journal of Biomedical cell response, Biomaterials, vol. 17, no. 2, pp. 137146, 1996.
Materials Research, vol. 5, no. 1, pp. 2551, 1971.
[215] K. T. Tran, L. Grith, and A. Wells, Extracellular matrix
[199] J. B. Park and R. S. Lakes, BiomaterialsAn Introduction, signaling through growth factor receptors during wound
Plenum Press, New York, NY, USA, 2nd edition, 1992. healing, Wound Repair and Regeneration, vol. 12, no. 3, pp.
[200] J. J. Blaker, J. E. Gough, V. Maquet, I. Notingher, and 262268, 2004.
A. R. Boccaccini, In vitro evaluation of novel bioactive [216] K. S. Midwood, L. V. Williams, and J. E. Schwarzbauer,
composites based on Bioglass -filled polylactide foams for Tissue repair and the dynamics of the extracellular matrix,
bone tissue engineering scaolds, Journal of Biomedical International Journal of Biochemistry and Cell Biology, vol. 36,
Materials Research A, vol. 67, no. 4, pp. 14011411, 2003. no. 6, pp. 10311037, 2004.
[201] H. W. Kim, E. J. Lee, I. K. Jun, H. E. Kim, and J. [217] R. N. S. Sodhi, Application of surface analytical and
C. Knowles, Degradation and drug release of phosphate modification techniques to biomaterial research, Journal of
glass/polycaprolactone biological composites for hard-tissue Electron Spectroscopy and Related Phenomena, vol. 81, no. 3,
regeneration, Journal of Biomedical Materials Research B, vol. pp. 269284, 1996.
75, no. 1, pp. 3441, 2005.
[218] D. M. Brewis and D. Briggs, Adhesion to polyethylene and
[202] C. Du, F. Z. Cui, X. D. Zhu, and K. De Groot, polypropylene, Polymer, vol. 22, no. 1, pp. 716, 1981.
Three-dimensional nano-HAp/collagen matrix loading
[219] D. L. Elbert and J. A. Hubbell, Surface treatments of
with osteogenic cells in organ culture, Journal of Biomedical
polymers for biocompatibility, Annual Review of Materials
Materials Research, vol. 44, no. 4, pp. 407415, 1999.
Science, vol. 26, no. 1, pp. 365394, 1996.
[203] A. Bigi, E. Boanini, S. Panzavolta, N. Roveri, and K. Rubini,
New perspectives in mercury porosime-
[220] C. A. Leon y Leon,
Bonelike apatite growth on hydroxyapatite-gelatin sponges
try, Advances in Colloid and Interface Science, vol. 76-77, pp.
from simulated body fluid, Journal of Biomedical Materials
341372, 1998.
Research, vol. 59, no. 4, pp. 709715, 2002.
[221] A. G. A. Coombes, S. C. Rizzi, M. Williamson, J. E. Barralet,
[204] Y. Zhang and M. Zhang, Synthesis and characterization of
S. Downes, and W. A. Wallace, Precipitation casting of
macroporous chitosan/calcium phosphate composite scaf-
polycaprolactone for applications in tissue engineering and
folds for tissue engineering, Journal of Biomedical Materials
drug delivery, Biomaterials, vol. 25, no. 2, pp. 315325, 2004.
Research, vol. 55, no. 3, pp. 304312, 2001.
International Journal of Polymer Science 19

[222] J. H. Brauker, V. E. Carr-Brendel, L. A. Martinson, J. Crudele, [238] P. V. Moghe, F. Berthiaume, R. M. Ezzell, M. Toner, R. G.
W. D. Johnston, and R. C. Johnson, Neovascularization of Tompkins, and M. L. Yarmush, Culture matrix configu-
synthetic membranes directed by membrane micro architec- ration and composition in the maintenance of hepatocyte
ture, Journal of Biomedical Materials Research, vol. 29, pp. polarity and function, Biomaterials, vol. 17, no. 3, pp. 373
15171524, 1995. 385, 1996.
[223] J. J. Klawitter and S. F. Hulbert, Application of porous [239] P. L. Ryan, R. A. Foty, J. Kohn, and M. S. Steinberg, Tissue
ceramics for the attachment of load-bearing internal ortho- spreading on implantable substrates is a competitive out-
pedic applications, Journal of Biomedical Materials Research come of cell-cell vs. cell-substratum adhesivity, Proceedings
A Symposium, vol. 2, pp. 161168, 1971. of the National Academy of Sciences of the United States of
[224] S. Yang, K. F. Leong, Z. Du, and C. K. Chua, The design America, vol. 98, no. 8, pp. 43234327, 2001.
of scaolds for use in tissue engineeringpart I: traditional [240] D. E. Ingber, Mechanical signaling and the cellular response
factors, Tissue Engineering, vol. 7, no. 6, pp. 679689, 2001. to extracellular matrix in angiogenesis and cardiovascular
[225] K. Whang, K. E. Healy, D. R. Elenz et al., Engineering physiology, Circulation Research, vol. 91, no. 10, pp. 877
bone regeneration with bioabsorbable scaolds with novel 887, 2002.
microarchitecture, Tissue Engineering, vol. 5, no. 1, pp. 35
51, 1999.
[226] I. V. Yannas, E. Lee, D. P. Orgill, E. M. Skrabut, and
G. F. Murphy, Synthesis and characterization of a model
extracellular matrix that induces partial regeneration of adult
mammalian skin, Proceedings of the National Academy of
Sciences of the United States of America, vol. 86, no. 3, pp. 933
937, 1989.
[227] A. J. Salgado, O. P. Coutinho, and R. L. Reis, Bone tissue
engineering: state of the art and future trends, Macromolec-
ular Bioscience, vol. 4, no. 8, pp. 743765, 2004.
[228] D. F. Williams, On the mechanisms of biocompatibility,
Biomaterials, vol. 29, no. 20, pp. 29412953, 2008.
[229] G. Khang, J. H. Jeon, J. W. Lee, S. C. Cho, and H. B. Lee, Cell
and platelet adhesions on plasma glow discharge-treated
poly(lactide-co-glycolide), Bio-Medical Materials and Engi-
neering, vol. 7, no. 6, pp. 357368, 1997.
[230] K. D. Colter, M. Shen, and A. T. Bell, Reduction of
progesterone release rate through silicone membranes by
plasma polymerization, Biomaterials Medical Devices and
Artificial Organs, vol. 5, no. 1, pp. 1324, 1977.
[231] M. Sato, M. Ishihara, M. Ishihara et al., Eects of growth
factors on heparin-carrying polystyrene-coated atelocollagen
scaold for articular cartilage tissue engineering, Journal of
Biomedical Materials Research B, vol. 83, no. 1, pp. 181188,
2007.
[232] H. Park, K. Y. Lee, S. J. Lee, K. E. Park, and W. H. Park,
Plasma-treated poly(lactic-co-glycolic acid) nanofibers for
tissue engineering, Macromolecular Research, vol. 15, no. 3,
pp. 238243, 2007.
[233] S. A. Mitchell, M. R. Davidson, and R. H. Bradley, Improved
cellular adhesion to acetone plasma modified polystyrene
surfaces, Journal of Colloid and Interface Science, vol. 281, no.
1, pp. 122129, 2005.
[234] J. C. Middleton and A. J. Tipton, Synthetic biodegradable
polymers as orthopedic devices, Biomaterials, vol. 21, no. 23,
pp. 23352346, 2000.
[235] M. A. Woodru and D. W. Hutmacher, The return of a
forgotten polymerpolycaprolactone in the 21st century,
Progress in Polymer Science, vol. 35, no. 10, pp. 12171256,
2010.
[236] W. P. Ye, F. S. Du, W. H. Jin, J. Y. Yang, and Y. Xu, In vitro
degradation of poly(caprolactone), poly(lactide) and their
block copolymers: influence of composition, temperature
and morphology, Reactive and Functional Polymers, vol. 32,
no. 2, pp. 161168, 1997.
[237] K. S. Anseth, C. N. Bowman, and L. Brannon-Peppas,
Mechanical properties of hydrogels and their experimental
determination, Biomaterials, vol. 17, no. 17, pp. 16471657,
1996.
Journal of International Journal of International Journal of Smart Materials Journal of
Nanotechnology
Hindawi Publishing Corporation
Corrosion
Hindawi Publishing Corporation
Polymer Science
Hindawi Publishing Corporation
Research
Hindawi Publishing Corporation
Composites
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of
Metallurgy

BioMed
Research International
Hindawi Publishing Corporation Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Nanomaterials
Submit your manuscripts at
http://www.hindawi.com

Journal of Journal of
Materials
Hindawi Publishing Corporation
Nanoparticles
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Nanomaterials
Journal of

Advances in The Scientific International Journal of


Materials Science and Engineering
Hindawi Publishing Corporation
Scientifica
Hindawi Publishing Corporation Hindawi Publishing Corporation
World Journal
Hindawi Publishing Corporation
Biomaterials
Hindawi Publishing Corporation
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014

Journal of Journal of Journal of Journal of Journal of

Nanoscience
Hindawi Publishing Corporation
Coatings
Hindawi Publishing Corporation
Crystallography
Hindawi Publishing Corporation
Ceramics
Hindawi Publishing Corporation
Textiles
Hindawi Publishing Corporation
http://www.hindawi.com
http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 http://www.hindawi.com Volume 2014 Volume 2014

You might also like