Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Expert Review of Medical Devices

ISSN: 1743-4440 (Print) 1745-2422 (Online) Journal homepage: www.tandfonline.com/journals/ierd20

Design and preparation of polymeric scaffolds for


tissue engineering

Thomas Weigel, Gregor Schinkel & Andreas Lendlein

To cite this article: Thomas Weigel, Gregor Schinkel & Andreas Lendlein (2006) Design and
preparation of polymeric scaffolds for tissue engineering, Expert Review of Medical Devices,
3:6, 835-851, DOI: 10.1586/17434440.3.6.835

To link to this article: https://doi.org/10.1586/17434440.3.6.835

Published online: 09 Jan 2014.

Submit your article to this journal

Article views: 6365

View related articles

Citing articles: 15 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierd20
For reprint orders, please contact: Review
reprints@future-drugs.com

Design and preparation of


polymeric scaffolds for
tissue engineering
Thomas Weigel, Gregor Schinkel and Andreas Lendlein†
Polymeric scaffolds for tissue engineering can be prepared with a multitude of different
techniques. Many diverse approaches have recently been under development. The
CONTENTS adaptation of conventional preparation methods, such as electrospinning, induced phase
Scaffold preparation from separation of polymer solutions or porogen leaching, which were developed originally for
polymer solution by other research areas, are described. In addition, the utilization of novel fabrication
nonsolvent-induced phase techniques, such as rapid prototyping or solid free-form procedures, with their many
separation or thermally
different methods to generate or to embody scaffold structures or the usage of self-
induced phase separation
assembly systems that mimic the properties of the extracellular matrix are also described.
Scaffold preparation from
These methods are reviewed and evaluated with specific regard to their utility in the area
the polymer melt by a
foaming process of tissue engineering.
Scaffolds by Expert Rev. Med. Devices 3(6), 835–851 (2006)
microsphere sintering
Rapid prototyping/
The technology of tissue engineering aims to formation in vitro and in vivo. Up to now
solid free-form fabrication generate new or substitute damaged or malfunc- scaffolds made from biomaterials have temporar-
tioning tissue or organs and could well become ily substituted the extracellular matrix (ECM).
Indirect methods using RP or
SFF fabrication methods an alternative method to whole organ trans- Such biomaterials can be of natural origin, such
plantation [1–6]. In the last decade particularly, as organic collagen [74–76], fibrin glue [77–79],
Self-assembly systems
enormous advances have been made in the area hyaluronic acid [80–82] or the inorganic-like cor-
Electrospinning of tissue regeneration for therapeutical purposes. alline [83,84]. Synthetic polymeric materials have
Scaffold fabrication Tissue engineering is an interdisciplinary also been investigated, including, for example,
supporting processes research area in which principles of material aliphatic, copolyesters [85–87], polyhydroxyal-
Method comparison science/engineering and cell biology/life science kanoates [88–90] and polyethylene glycol [91,92], or
Summary
are combined. The methods of tissue engineer- inorganic materials, such as hydroxy-
ing have been applied to different types of tissue, apatite [48,93,94], tricalciumphosphate [95,96], glass
Expert commentary
including skin [7–11], bone [12–16], liver [17–21], ceramics and glass [97–99].
Five-year view intestine [22–27], esophagus [28–32], valve The intrinsic material properties, such as
Key issues leaflets [33–36], muscle [37–39] and tongue [40–42], the mechanical [100] or thermal [101,102] behav-
References
the vascular system [43–47], for craniofacial ior, of a polymer play a role in the utilization
defects [48–50], tendons and ligaments [51–55], of tissue engineering and also in the embodi-
Affiliations
cartilage [56–60] and nerve tissue [61–65]. Since the ment and the morphology of the shaped scaf-

Author for correspondence early commercialization of tissue-engineered fold body. By ‘embodiment’, we mean the
Institute of Polymer Research, applications, for example, the work of Bell and preparation with, for example, the use of
GKSS Research Center, Kantstr. 55, colleagues [66], research with stem cells [3,67–69] fibers [103], microspheres [104,105] or flat bod-
D-14513 Teltow, Germany
Tel.: +49 332 835 2450
and the use of growth factors [70–73] that support ies, like particular structured films [106], to
andreas.lendlein@gkss.de cell differentiation and proliferation, have pro- become a scaffold body. The definition of
vided new opportunities in the field of tissue embodiment also includes the computer-
KEYWORDS: engineering. At present, tissue engineering aided construction of these bodies [107]. The
electrospinning, phase separation,
preparation methods, rapid methods generally require the use of a porous morphology of a scaffold has to meet the
prototyping, scaffolds, scaffold that serves as a matrix for initial cell demands of the targeted applications [108].
self-assembly systems,
tissue engineering attachment and subsequently for tissue General requirements for the scaffold

www.future-drugs.com 10.1586/17434440.3.6.835 © 2006 Future Drugs Ltd ISSN 1743-4440 835


Weigel, Schinkel & Lendlein

morphology are interconnected pores in order to assure cell require pore sizes in the upper micrometer range, for example,
growth and the transport flow of nutrients and metabolic for skin (20–150 µm) or bone scaffolds (40–400 µm) [124],
waste [109,110]. The scaffold surface should enable a suitable which cannot be realized with this technique unless it is com-
cell attachment and promote the cell differentiation and pro- bined with other processes, such as salt leaching [125]. Special
liferation. Furthermore, the biocompatible degradation of the shaped bodies prepared by ‘pure’ nonsolvent-induced phase sep-
scaffold material has to proceed appropriately and in sync aration (NIPS) can also be used in biomedical applications [126],
with the formation of the tissue [111]. The scaffold morphol- for example, as a carrier for epidermal substitutes (FIGURE 1) [127].
ogy is determined by the fabrication method and/or the As well as the initiation of phase separation by a nonsolvent,
embodiment. By varying the component dimensions it is also possible to initiate the phase separation thermally by a
(e.g., reducing the diameter of filaments from the micrometer thermally induced phase separation (TIPS). A homogeneous
to the nanometer scale), scaffolds can be formed with porosi- solution of a polymer in a solvent is prepared at an elevated
ties and strengths that are significantly different to their origi- temperature [128,129]. The solution is introduced into the
nal morphology [112]. The mechanical properties of the scaf- desired form and cooled afterwards by removal of heat energy
fold and the tissue, which need to be replaced, should in a quenching process [130]. The solvent is typically removed by
match [113] and, eventually, the material processing properties sublimation [131,132] or extraction [133,134] to yield a micro-
need to be suitable for the scaffold formation. porous scaffold. A liquid–liquid phase separation takes place, in
In this review, different methods for scaffold design are which the solution separates into a polymer-rich phase and a
summarized with specific emphasis on fabrication methods. polymer-poor phase (FIGURE 2) [135].
The preparation of porous scaffold bodies can be realized by a Depending on the nature and strength of the polymer sol-
multitude of different techniques. Especially, new develop- vent interactions, further thermodynamic polymer solvent
ments in the area of electronically controlled processes [114,115] systems are conceivable. Such a constellation is a solid–liquid
and the utilization of new research results, which mimic natu- TIPS [136,137], in which the polymer crystallizes from the solu-
ral systems, such as the ECM [116], induce new progress in tion and, at least, in theory, a liquid–solid TIPS exists, in
scaffold formation. The spectrum of achievable scaffold types which the solvent crystallizes first, before the polymer [138].
with very different properties has been expanded by both the The microstructure of the resulting scaffold depends on
process adaptation, which has been established initially in which of these three sequences is followed and on the kinetics
other applications, such as membrane formation [117], and the of structure development. Liquid–liquid TIPS seems to be the
combination of different scaffold preparation methods [118]. preferred method because it has been proven to be a valuable
method of making scaffolds and it has been studied
Scaffold preparation from polymer solution by extensively [130,139–141].
nonsolvent-induced phase separation or thermally induced
phase separation
In order to fabricate scaffolds from polymer solution, poly-
mers are dissolved in a solvent and brought by casting onto a 50 μm
solid surface to immerse this flat thin solution into a nonsol-
vent, to form a flat membrane-like structure for tissue engi-
neering applications [119]. By casting the solution through a
nozzle into the nonsolvent, a tubular structure can be
created [120,121]. A block structure is also possible by casting
the polymer solution into a mold and bringing the solution
surface into contact with the nonsolvent afterwards [122].
When adding the nonsolvent, the solvent diffuses out while
the nonsolvent diffuses into the polymer solution that will
form the scaffold structure. The nonsolvent concentration in
the polymer solution increases. After a short diffusion time,
which means a few fractions of a second, at the specific work-
ing temperature, the ternary system of solvent, nonsolvent
and polymer is no longer stable. The polymer begins to pre-
cipitate from the solution and it finally forms the
scaffold [119]. At the end of this exchanging process, the
porous morphology has been created and the shape is
solidified [122]. Figure 1. Laser-scanning microscopic picture from poly(ether imide)
The average pore size that can be realized by this process can membranes prepared by nonsolvent-induced phase separation and
be varied only from the nanometer [123] to the few micrometer modified with poly(ethylene imine) (keratinocytes cultured for 4 days
stained for cell cytoskeleton). Reproduced with permission from [127].
range [122]. However, applications in tissue engineering often

836 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

example, closed cell foams and microsizes of approximately


10 µm [156] or a homogeneous nucleation with cell sizes in a
broad range of 30–700 µm [157]. It has been published regularly
that the foams possess closed cells [158–160], however, this means,
at the same time, that there is a low level of interconnectivity
and therefore their application in the area of tissue engineering
is limited. Thus, foaming processes are often coupled with
leaching methods in order to increase the porosity of the result-
ing scaffold [161–163]. Scaffold preparation by a foaming process
also allows the incorporation of bioactive [164] or
pharmaceutical [165] substances.

200 μm Scaffolds by microsphere sintering


Microspheres can be formed by different processes, such as
spraying a polymer solution followed by NIPS [166]. The use of
ultrasound [167] to emulsify water in a polymer solution is also
Figure 2. Scanning electron microscope micrograph of scaffold from a described. It aims to finally obtain a water-in-oil-in-water
multiblock copolymer prepared by thermally induced phase separation.
emulsion. The water droplets may even be loaded with, for
Reproduced with permission from [135].
example, DNA [164]. Then, the emulsion of water-in-polymer
solution is brought into an aqueous poly(vinyl alcohol) (PVA)
Scaffold preparation from the polymer melt by a solution to complete the NIPS by stirring for 3 h. By the use of
foaming process a vortexer [168,169], a water-in-oil-in-water is achievable. Hot
The foaming process of thermoplastics represents a fre- stirring of a paraffin/gelatin–water emulsion at 80°C is also
quently used method for the fabrication of porous materials. reported. The stirring is followed by an ice water quenching to
Methods differ mainly in the type of foaming agents, such as obtain paraffin spheres for paraffin leaching [170]. The simplest
water [142] or CO2 and nitrogen [143], as well as way is to pour the organic polymer solution into the aqueous
fluoroform [144], and the processing procedure, for example, PVA solution and to stir for a few hours. It was chosen to iso-
by an injection molding with a foaming agent based on a late microspheres that could be brought into a 3D shape fol-
carboxylic acid to obtain porous cores [145] or by the use of a lowed by a sintering process to an in vivo characterized scaffold
high-pressure chamber in an autoclave [101]. Expanding sub- body [171,172]. Microsphere sintering is, furthermore, a suitable
stances are generated by chemical reaction, for example, dur- method of obtaining composite structures from polymeric and
ing polyurethane synthesis as well as ammonium bicarbonate inorganic substances for bone tissue engineering [173].
that spontaneously produces CO2 and ammonia gas
bubbles [146] or by a thermal decomposition of specially Rapid prototyping/solid free-form fabrication
added substances, such as azodicarbonamide [147,148]. The Conventional fabrication techniques can not build up a complex
foaming process can also be initiated by ‘physically’ acting scaffold with an exactly predefined shape. Rapid prototyping
agents, which change their physical state via variation in tem- (RP) [174,175] or solid free-form (SFF) [115,176,177] fabrication, are
perature and pressure during the process. Foaming processes common terms for a group of techniques that model a scaffold
are realized as continuous [149] or discontinuous [150,151] directly from a computer-aided design (CAD) data set. RP tech-
working processes. A common method of scaffold prepara- niques mostly build up a specific body shape by the selective
tion is the exposure of polymers to high-pressure CO2 [152]. addition of material, layer-by-layer, guided by a computer
This method has the advantages that CO2 is cheap, simple to program [178]. RP systems were not developed for biomaterials
access, gives, for example, a lower dense foam than originally; for example, the 3D-Printing system Model
nitrogen [73,143] and it does not require removal before cell Maker II™ [179] but the manifold of RP designs also enables
seeding like other pore forming substances. The gas is dis- their biomedical application. The step-by-step construction of
solved in the solid polymer or polymer melt at high tempera- scaffolds facilitates improved reproducibility. In contrast to con-
ture and high pressure, for example, for poly(lactide-co-gly- ventional fabrication methods, parameters, such as porosity,
colide)acid (PLGA) 35–40°C, 10–20 MPa [153], for interconnectivity, pore size and geometric stability, can be con-
poly(ε-caprolactone) (PCL) 70–90°C and 7–32 MPa [154] or trolled more precisely [180]. Furthermore, cells can be printed on
even cyclic olefin copolymer (COC) 100–180°C and surfaces [181] and so it seems to be possible that living biological
30 MPa [155]. substances could be incorporated into the prefabricated layer
The thermodynamic instability is then induced by reducing before the final assembly [114,182]. However, the method is
the CO2 gas pressure to an ambient level [150]. During the restricted in the choice of materials and limited by its resolution
expansion process, the morphology of the scaffold may be gen- determined by the use of engineered precision machine tools and
erated by implementing a heterogeneous nucleation with, for tools like nozzles and the frequent small final size of the scaffold

www.future-drugs.com 837
Weigel, Schinkel & Lendlein

(e.g., 0.4–3.5 cm3) [183–185], depending on the fabrication. In Nonfused liquid deposition modeling
most cases, scaffolds with resolutions in a range of 200–500 µm The deposition of extruded strands, filaments or the plotting of
are described [107] – depending on the RP technology used. The dots in 3D is not necessarily connected with high temperatures
usage of a special RP fabrication technique demands specific and polymer melts. A further possibility is the usage of pastes or
material properties. Although many different applications to slurries, solutions and dispersions of polymers or reactive
embody scaffolds under RP processing conditions are described, oligomers [200]. Special experimental equipment, with adapted
the special requisitions of the polymer material limit the processing options, is required for processing of these materials
utilization of RP fabrication techniques. into 3D scaffolds. The described multiphase jet solidification
(MJS) process for biomaterials [201] generally means the use of a
Shape deposition manufacturing dispersion of different phases, for example, solid ceramic particles
Shape deposition manufacturing involves the fabrication of a lay- and a binder solution [202] in RP. The bioplotter™ [203,204] or
ered scaffold in a tailor-made geometry by a computer-controlled reactive plotter [205] dispenses pastes or dispersions into a match-
cutting machine [114]. The scaffolds are built up incrementally ing plotting liquid with the right density. After leaving the nozzle
from prefabricated cross-sectional layers of formed materials. and contacting the previous layer of the body that is being fabri-
Layers are assembled manually and joined to form 3D bodies cated, the plotting medium is becoming solid. No supports are
using biodegradable or biostable fasteners. The concept of needed owing to the fact that the plotting liquid medium
robotic microassembly follows the same principle [114,182]. Differ- compensates the gravity [107].
ently designed block units are first prepared [186], for example, by
lithographic methods or other previously mentioned techniques. 3D printing
The blocks are assembled with a precision robot with microgrip- In 3D printing technology (3D-P), the shaped bodies are cre-
ping capabilities. However, this interesting and innovative ated by a layering printing process with adhesive bonding,
method is still awaiting broad application in tissue engineering. using powder as a base material [301]. In the beginning, a layer
of powder is spread over a building platform. Then a binder
Fused deposition modeling solution is deposited precisely by an ink-jet print head on the
Compressed polymer melts are starting points for the prepara- powder layer to join single powder particles, resulting in a 2D
tion of porous scaffolds using a 3D-fiber deposition layer profile [206]. A fresh layer of powder is laid down and the
technique [187–190]. In this procedure, a polymer melt is formed process is repeated. After drying of the binder, the nonjoined
into a fiber with a temperature–controlled extruder. Its nozzle powder is removed by an air jet flow [184] and the finished scaf-
deposits the fiber or the filaments on a motor–driven x-y-z table. fold retrieved. The resolution of 3D-P is influenced by the par-
The machine first builds a layer of fibers with a well defined dis- ticle size of the powder, the nozzle size and degree of control
tance. On the top, fiber layers are deposited, which form a 3D over the position controller that defines print-head movement.
scaffold with an exact porous morphology and 100% intercon- An achievable channel diameter of 500 µm has been
nectivity. The positional control of the table is often computer reported [206]. The powder of materials that can be used for bio-
aided and allows for the construction of 3D structures (FIGURE 3). medical applications is not always available in the appropriate
Models could also use data from computer tomography, particle size (usually 80–250 µm) and often requires certain
magnetic resonance imaging scans or data created from 3D pretreatments [207]. Powder particles have to flow well with a
object digitizing systems [191]. In a modification of the fused defined size range and have to demonstrate good interaction
deposition modeling (FDM) process, the filament preparation with the liquid binder in an amount of at least 500 ml granu-
to feed the extrusion head as described, for example, with a late [208]. The successful realization of the 3D-P process also
diameter of 1.78 mm [57], is no longer necessary. Instead, the depends on the adaptation of an appropriate binder for differ-
polymer material can be used in its commercial form as pellets ent base materials. A preferred solvent with regard to biocom-
and granules by precision extruding deposition (PED), which patibility is water, suitable for natural biopolymers, such as
forms, for example, PCL scaffolds with a pore size of starch-based [209], gelatin or collagen. Organic solvent as bind-
250 µm [192] or even 200 µm 193]. ers (e.g., chloroform or methylene chloride) might be harmful
A specific demand on the material is that it demonstrates a suit- to the human body and difficult to remove completely [210].
able thermoplastic behavior. Therefore, meltable polymers are
used frequently, such as the slow biodegrading PCL [110,191,194,195] Selective laser sintering
or the nonbiodegradable acrylonitrile butadiene styrene This technique uses deflected laser beams (infrared laser, CO2
(ABS) [188,196] or polypropylene (PP) [197]. The use of the good laser) to sinter thin layers [211] of powdered materials, such as
biodegradable PLGA or poly(lactic acid) (PLA) in FDM is rarely wax, synthetic polymers (polycarbonate [207], polyamides [210],
reported. A disadvantage of the biodegradability may also be the PCL [212]) or mixtures/coatings with polymers, such as ultra
thickness of the pore walls stemming from the fiber diameters, for high molecular weight polyethylene [213], poly(etherether-
example, approximately 200–300 µm [110,191,198] and the dimen- ketone)/hydroxyapatite [214] or polyvinyl alcohol/hydroxy-
sions of the scaffold bodies are often rectangular and with a size of apatite [213], to prepare solid 3D scaffolds. The laser beam selec-
a few hundred mm3 to several cm3 [110,183,199]. tively scans over the powder surface [210]. The interaction of the

838 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

2 mm 1 mm 1 mm

A B C

2 mm D 2 mm E 1 mm F

Figure 3. Scanning electron microscope photos of scaffolds from fibers with different deposition geometries. (A) Homogeneous fiber spacing of 1 mm
showing typical fiber diameters and pore geometries; (B) top view; (C) staggered fiber spacing of 1 mm; (D) inhomogeneous pore-size gradient;
(E) inhomogeneous pore-size gradient deposited on a dense basal layer of fibers; (F) superficial layer of gradient scaffold. Reproduced with permission from [190].

laser beam with the powder elevates the powder temperature Furthermore, different adaptations and variations concern-
just beyond the glass transition temperature but below the ing the improvement of the hydrogel stability (usage of
melting temperature of the used material. The powder particles polymer mixtures) [216] were developed to incorporate living
that are in contact are deformed and fused together. New layers cells during the formation process (adaptation of processing
of powder are deposited by a roller, building a new sintered conditions to prevent toxic free radicals) [218,219] or to
layer on top of the previous one (FIGURE 4). improve the acuteness of prepared structures – more precise
The laser-beam diameter (∼400 µm) or powder–particle laser beam moves connected with the expansion of the
size limits the dimension of generated scaffold structures. material base [114] or using photolithographic techniques
Owing to Gaussian distribution of the laser energy and the with x-rays, electron or ion beams [220]. The specific property
principle of powder bonding (different powder-particle
forms), it is usually difficult to build up special shaped bodies
with sharp corners or clear boundaries. Improvements in
selective laser sintering are expected, especially concerning
laser spot size, powder size and new ways to remove trapped
loose powder from scaffolds [215]. The restrictions to the mate-
rial properties are given by the demand for it to be available as
a powder and that the powder must demonstrate a suitable
melting and welding behavior.

Stereolithographic working principles


The principle of this technique is based on the initiation of a
chemical reaction (photopolymerization [216] or cross-
linking [217]) in special polymer liquids by electromagnetic radi-
ation. Light from a laser beam is directed onto selected regions
of a layer of liquid polymer causing solidification in the
exposed areas. By lowering of the nascent-shaped body, which
is arranged in a polymer liquid bath, a new layer of polymer Figure 4. Scanning electron microscope micrographs of a 3D structure
solution can be exposed to the laser beam for the initiation of obtained by selective laser sintering from a composite containing
the chain reaction. The stereolithographic working principle is 90 wt% polyetheretherketone and 10 wt% hydroxyapatite. Reproduced
repeated, generating 3D scaffolds (FIGURE 5). with permission from [107].

www.future-drugs.com 839
Weigel, Schinkel & Lendlein

Furthermore, the porosity of the scaffold can be increased by


A
combining the solidification process, which is caused either by
Z axis movement
conventional phase separation or by the TIPS technology, with a
X–Y movable Elevator porogen leaching process in order to form sponge-like
UV laser morphologies [223,225]. Scaffolds prepared by indirect RP or SFF
Resin surface fabrication methods sometimes inherit errors and defects caused
by additional processing steps of mold manufacturing and elimi-
Vat nation. Whenever RP techniques for biomaterials require, for the
SL pattern respective application, specific materials with additional and often
specific properties – this method allows the use of a wider range of
biomaterials and combination of materials. For the mold building
step, it is remitted to select the best material for a precise RP proc-
ess that hardly directly depends on the biological behavior of the
scaffold. The scaffold is made with this mold from a biomaterial,
for example, by a molding or casting process.
B

Self-assembly systems
Molecular self-assembly is also an approach for fabrication of scaf-
folds for tissue engineering [226,227] and scaffolds from self-assem-
bling peptide scaffolds may be used, for example, in nerve tissue
engineering [228] or cartilage repair [229]. Molecular self-assembly
requires the molecules to undergo self-association and thus form
hierarchical structures from the bottom-up, molecule by molecule
without any external manipulations. Self-assembly systems are
omnipresent in nature. There are endeavors to mimic such natural
systems, such as the ECM, for scaffold formation [230]. Materials
used for these systems range from bi- or tri-block copolymers [231],
complex DNA structures [232] and lipids to simple or complex
peptides and proteins. Important principles of self-assembly are
Figure 5. Stereolithography. (A) UV light is used to crosslink the material in chemical complementarity and structural compatibility through
specific regions of a layer. The elevator is then lowered to reveal a new layer of weak and noncovalent interactions. Different construction units,
polymer and the process is repeated to create the desired shape. (B) A such as well ordered nanofiber scaffolds [233,234], peptide
prototype scaffold designed using stereolithographic technique. nanotubes [235–237], segmented surface assembling peptides as a
Reproduced with permission from [217].
‘molecular carpet’ [238–240] and dipolar molecules with changeable
conformation as a ‘molecular switch’ [241–243], can be realized by
that a material requires in order to be employed in specially designed peptide units and sequences. However, the real-
stereolithographic techniques is the ability to react to light, ization of peptide scaffolds by molecular self-assembly is extremely
activating a chemical reaction. extensive and it depends on elaborate techniques.

Indirect methods using RP or SFF methods Electrospinning


The manufacturing methods for scaffold preparation Corresponding to the ambition to mimic the supramolecular
resuming under the terms RP or SFF fabrication allow structure and biological functions of the ECM, the 3D-fiber
manifold usage of different materials. Nevertheless, every network of collagen composed of nanoscale multifibrils is often
technique requires certain material properties as prerequisites. the focus of consideration.
The indirect SFF fabrication technique [221] is an emerging Electrospinning (ELSP) is an established process capable of
method designed to partly overcome these limitations fabricating ultrafine fibers, in the micrometer [244] or nanometer
regarding biomaterials. In this procedure, a negative mold is scale [245,246], by electrically charging a suspended droplet of pol-
generated by one of the SFF fabrication techniques ymer melt [244,247] or solution [248,249]. A high-voltage electro-
(e.g., stereolithographic working principles [220], the 3D static field (10–20 kV) operated between a metallic nozzle of a
printing system Model Maker II [222,223]) as a first step. In the syringe and a metallic collector is used to generate a sufficient
second step, the scaffold is formed by adding the casting surface charge to overcome the surface tension in a pendent
solution [224] to the negative mold using the desired polymeric drop of the polymer fluid. Nanofibers are formed by the nar-
and/or ceramic biomaterials. After solidification, the negative rowing of the ejected jet stream as it undergoes increasing sur-
mold is removed by dissolution, melting or other procedures face charge density due to the evaporation of the solvent [250,251].
(FIGURE 6) [225]. Different processing parameters (e.g., kind of polymer/solution,

840 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

A B C D

1 mm

Figure 6. Scaffold fabrication using rapid prototyped negative replica. (A) A computer-generated, 3D negative replica of a scaffold with a cubical pore shape.
(B) The negative replica of the scaffold fabricated using a rapid prototyping machine. (C) A photograph of the polymer scaffold fabricated using the negative
replica. (D) A scanning electron micrograph of the internal pore structure of the scaffold. Reproduced with permission from [225].

viscosity, surface tension, net charge density, polymer mass flux technique in combination with injection [265] or compression
in the syringe or ambient parameters, such as temperature, molding [210,266], in combination with a NIPS [125], in gas
humidity and air velocity) control the ELSP process, especially foaming processes [150], as addendum of indirect SFF fabrica-
the diameter and morphology of the resulting fibers [252]. The tion techniques [267], in combination with soft lithographic
fibers with diameters ranging from several µm down to 100 nm methods [268], photopolymerization [269] or in combination of
and less are deposited in the form of a nonwoven fabric on a coagulation with thermal processing [176], documents the
collector through a random deposition process (FIGURE 7) [253]. comprehensive character of this technique.
Nonwoven sheets with 2D profiles are usually obtained by
ELSP and can be extended to nonwoven 3D fibrous meshes by Computer-aided tissue engineering technique
the prolongation of processing time [252]. Another supporting utility is computer-aided tissue engineering
Various polymer nanofibers can be deposited together into one (CATE) [270,271]. Sun and colleagues see, in this concept, the
multilayered structure by ELSP of different polymer solutions combination of three major categories [272–274]. These are:
layer-by-layer. If different polymer solutions are ejected by several • Computer-aided tissue modeling, including 3D anatomical
orifices simultaneously, a mat consisting of a composition of dif- visualization with imaging-data acquisition, 3D
ferent nanofibers is realizable [254]. The nanofiber-based scaffolds reconstruction and CAD-based tissue modeling [185];
possess a wide range of pore size distribution, high porosity and a
• Computer-aided tissue informatics [275], including computer-
high surface area:volume ratio, which are favorable parameters
aided tissue classification and the application for tissue
for cell attachment, growth and proliferation [255].
identification and characterization at different tissue
hierarchical levels;
Scaffold fabrication supporting processes
Besides the methods for scaffold preparation discussed above, • Computer-aided tissue scaffold design and manufacturing
the following two techniques are presented. They have a com- [113,276], including SFF of tissue scaffolds and bioblueprint

prehensive character and often act as a supporting method to modeling [275], for 3D cell and organ printing [181,277].
realize the targeted parameters in the fabrication process. Under the term CATE, summarized methods are not
restricted to the control and the design [180] of the internal
Leaching technique architecture [185,278] of scaffolds of the RP technologies by CAD
The leaching technique is described mostly in connection methods. They have opened up new possibilities to match the
with the solvent casting/particulate leaching [12,256,257] to different mechanical and geometrical requirements of the origi-
form tissue engineering materials but it has also been applied nal patient computed tomography data to create, for example,
as a supporting process in combination with many other dif- exact bone scaffolds [275,279,280].
ferent scaffold fabrication methods. Different porogens are
used frequently for the leaching techniques. These are, for Method comparison
example, NaCl [258], as perhaps the most common leaching Different authors have given short [204,281,282] or more
substance, as well as paraffin spheres [170,259,260], sugar detailed [57,124,180] tables showing the advantages and dis-
crystals [261–263] or gelatin [264], which serve as place holders advantages of different fabrication methods. However, such
for pores and their interconnections in the actual scaffold for- tables can only partially provide what the reader requires, such
mation process. These spacers are removed by leaching fol- as: a plan to avoid the loss of time and money in attempting to
lowing the main processing step. Thus, the objective of the create a suitable scaffold for its own application. So it is often
leaching procedure is usually the realization of bigger pore reported in overviews in diverse articles [57,180,204] that the
sizes and increased interconnectivity. The use of the leaching heating to a molten state, for example, in a molding or a FDM

www.future-drugs.com 841
Weigel, Schinkel & Lendlein

The requirements of scaffolds for tissue engineering are com-


plex and tissue specific, regarding structure and function. Even
for a specific tissue, it is not quite clear what defines an ideal scaf-
fold/cell or scaffold/neotissue construct. Thus, only the inclusion
of several different scientific disciplines with a high level of multi-
disciplinarity will enable further successful development of scaf-
fold-based tissue engineering. Manifold different techniques of
scaffold fabrication have been introduced. These methods and
the deduced possible combinations describe a pool of procedures
allowing preparation of scaffolds with defined morphologies and
suitable properties regarding application in tissue engineering.
10 μm Expert commentary
In recent decades, success in modern medicine has mainly influ-
Figure 7. Scanning electron microscope micrograph of electrospun enced the therapy of infectious and cardiovascular diseases. A
poly[(dl-lactide)-co-glycolide] nanofibrous nonwoven composed of constant increase of life expectancy and a relevant increase of life
randomly oriented ultrafine fibers. Reproduced with permission from [253]. quality is connected with this development, including the
understandable demand of the population to accept no loss in
process may badly affect the polymer. But it may also advanced age during the whole life. Because of the shifting mor-
accelerate the designated and demanded degradation of the bidity spectrum from now on, the clinic medicine deals with
biomaterial in a special application. Or the use of organic sol- diseases referring to creeping loss of cell or organ functions.
vents during the prepration seems to be harmful [124,282] but if Mechanical overstressing, chronic inflammations cellular
they can be removed completely, for example, by an annealing degeneration, defects in immunological system or organ damage
step after the scaffold fabrication, so the former employment by injuries and by exogenic-toxic influences can be consequences.
of organic solvents is no longer an issue for cell culturing. The In most cases, these diseases cannot be healed by classical
utilization of CO2 foaming can show a higher amount of therapies. They have to be under permanent treatment causing
closed cells [124]. Then the interconnectivity may be increased considerable efforts. Sometimes organ transplantation might be
frequently by combining with salt leaching. But the leached helpful but limited availability of organs and immunological
NaCl crystals leave a partly cubic porous morphology [283] incompatibility narrows this option. An increasing need for
with a difficult interconnectivity control in comparison to suitable organs and a nearly equal number of transplantations
other leaching processes [170,259]. The TIPS is described in in the last 10 years is also evidence of the recent situation.
overviews as a long-time method [282] but the use of extrac- Thus, the desire for more transplantable cells, tissues and
tion methods [133,134] instead of freeze drying for many organs with good tissue compatibility by reduced rejection or
days [86,130] lowers the fabrication time, increases the produc- aggression reaction are an integral part of clinical strategy in the
tion scale and does not require the expensive equipment future. Nonbiological organ replacement through hybridized
either [281]. RP techniques demand specific material proper- systems will bridge the development to fully biological systems
ties and still show a low resolution in many cases. But the and finally to human regenerative systems in vivo. The forma-
exact building of 100% interconnected porous scaffolds tion and design of scaffolds are an essential part of this
from, for example, CT data is impressive. In the end, the suc- progression of tissue engineering.
cessful application of a scaffold in medicine or biology will
only decide that a preparation method is able to fabricate Five-year view
good materials in tissue engineering. In the future, solutions to problems in the fileds of tissue engi-
neering and regenerative medicine will probably be determined
Summary by a causal treatment mode that expects the complete func-
The field of scaffold-based tissue engineering is still in an early tional replacement of damaged or destroyed tissue. New thera-
state of development but has been growing rapidly over the last pies with this background should be superior to conventional
few years. Many different approaches of scaffold fabrication are approaches owing to the duration and extent of recovery or by
under investigation currently. One strategy is the advancement the reduction of potential side effects. Thus, the mechanism of
and adaptation of conventional fabrication technologies, such regeneration will be a standard therapy approach. Tissue engi-
as ELSP, methods of coagulation agent induced-phase separa- neering also provides great promise for posed tasks but there is
tion or leaching methods developed originally for other also a considerable responsibility to quickly identify the best
research areas. But novel fabrication techniques, like RP or SFF treatements with the greatest potential.
procedures with their many different embodiments or usage of For the preparation of scaffolds, the advancement and
self-assembly systems mimicking properties of the ECM, also adaptation of conventional fabrication technologies
have potential for future developments. (e.g., electrospinning, methods of coagulation agent-induced

842 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

phase separation or leaching methods developed originally for aspects, such as usage of materials with self-assembly proper-
other research areas) will be pursued. But novel fabrication tech- ties. The utilization of external stimuli (biological acting by
niques (e.g., rapid prototyping or solid free form procedures with growth factors, chemical acting by adsorption processes or
their many different embodiments or the usage of self-assembly mechanical acting by alternating material stress) with scaf-
systems) will also have the potential for future developments. folds as alloplastic guide track will enable or improve cell
Progress in tissue engineering can be only achieved by inter- settlement behavior.
actions among natural scientists, engineers and physicians. A It will expect that progress in stem cell investigation will gener-
high level of multidisciplinarity is prerequisite of successful ate essential impulses for in vitro breeding of cells and tissue in
work for the development of scaffold design and functional- the next years. Intermediate goals on the way to fully biological
ity. Future scaffold constructs will only be successful if an implants and finally the human regenerative system in vivo
improved compatibility regarding the ECM is realized. The could be biodegradable tissue support systems and smart mate-
variation or selective modification of prestructured scaffolds, rial implants within 5 years and organ-assist systems based on
as well as the reproduction of natural systems, could be pass- human cell-lines within 10 years. Functional autologous tissue
able procedures that account for both the variation of physical systems, such as implantable autologous heart valves, will need
and chemical properties and the special morphological at least another 15–20 years.

Key issues

• In recent decades successes in modern medicine have contributed to increased life expectancy connecting with the understandable
demand of the population to accept no loss of life quality in advanced age.
• Recently employed tissue engineering will bridge the development to fully biological systems and finally to human regenerative
systems in vivo by nonbiological organ replacement through hybridized systems.
• At present, tissue engineering methods generally require the use of porous scaffolds that serve as a matrix for initial cell attachment
and subsequently for tissue formation in vitro and in vivo and in so far temporarily substitutes the extracellular matrix.
• The advancement and adaptation of conventional fabrication technologies (e.g., electrospinning, methods of coagulation agent
induced phase separation or leaching methods) as well as novel fabrication techniques (e.g. rapid prototyping or solid free-form
procedures or the usage of self-assembly systems) have a potential for future scaffold developments.
• Successful development of scaffold-based tissue engineering demands a high level of multidisciplinarity between natural scientists,
engineers and physicians to answer questions regarding efforts to develop scaffolds for specific tissue function and structures and
the ‘ideal’ scaffold/cell or scaffold/neotissue construct.

References 5 Cima LG, Vacanti JP, Vacanti C, 11 Ng KW, Hutmacher DW, Schantz J-T
Papers of special note have been highlighted as: Ingber D, Mooney D, Langer R. Tissue et al. Evaluation of ultra-thin poly(ε-
• of interest engineering by cell transplantation using caprolactone) films for tissue-engineered
•• of considerable interest degradable polymer substrates. J. Biomech. skin. Tissue Eng. 7(4), 441–455 (2001).
Eng. 113(2), 143–151 (1991). 12 Filipczak K, Janik I, Kozicki M et al.
1 Yang S, Leong KF, Du Z, Chua CK. The
design of scaffolds for use in tissue 6 Vacanti JP. Tissue and organ engineering: Porous polymeric scaffolds for bone
engineering. Part I. Traditional factors. can we build intestine and vital organs? regeneration. In: e-Polymers 011, ISSN
Tissue Eng. 7(6), 679–689 (2001). J. Gastrointest. Surg. 7(7), 831–835 (2003). 1618-7229 (2005).
• Scaffold design including several aspects, 7 Bannasch H, Fohn M, Unterberg T, 13 Liu X, Ma Peter X. Polymeric scaffolds
such as fabrication, materials, Bach AD, Weyand B, Stark GB. Skin tissue for bone tissue engineering. Ann. Biomed.
morphology and so on. engineering. Clinics in Plastic Surgery 30(4), Eng. 32(3), 477–486 (2004).
2 Langer R, Vacanti JP. Tissue engineering. 573–579 (2003). 14 Braddock M, Houston P, Campbell C,
Science 260(5110), 920–926 (1993). 8 Sun T, Mai S, Norton D, Haycock JW, Ryan Ashcroft P. Born again bone: Tissue
3 Weinand C, Pomerantseva I, Neville CM AJ, MacNeil S. Self-organization of skin cells engineering for bone repair. News in
et al. Hydrogel-b-TCP scaffolds and stem in three-dimensional electrospun polystyrene Physiological Sciences 16, 208–213
cells for tissue engineering bone. Bone scaffolds. Tissue Eng. 11(7/8), 1023–1033 (2001).
38(4), 555–563 (2006). (2005). 15 Porter BD, Oldham JB, He SL et al.
4 Griffith LG, Naughton G. Bodybuilding: 9 Hutmacher DW, Vanscheidt W. Matrices for Mechanical properties of a biodegradable
bionic human: engineering – current tissue-engineered skin. Drugs of Today 38(2), bone regeneration scaffold. J. Biomech.
challenges and expanding opportunities. 113–133 (2002). Eng.122(3), 286–288 (2000).
Science 295(5557), 1009–1010, 10 Adekogbe I, Ghanem A. Fabrication and 16 Li C, Vepari C, Jin H-J, Kim HJ,
1012–1014 (2002). characterization of DTBP-crosslinked Kaplan DL. Electrospun silk-BMP-2
chitosan scaffolds for skin tissue engineering. scaffolds for bone tissue engineering.
Biomaterials 26(35), 7241–7250 (2005). Biomaterials 27(16), 3115–3124 (2006).

www.future-drugs.com 843
Weigel, Schinkel & Lendlein

17 Dixit V, Elcin YM. Liver tissue engineering: 30 Nagashima A, Ando N, Sato M, Ozawa S, 43 van Lieshout MI, Vaz CM, Rutten MCM,
successes & limitations. Ad. Ex. Med. Biol. Kitajima M. Basic studies on the Peters GWM, Baaijens FPT.
534, 57–67 (2003). application of an artificial esophagus using Electrospinning versus knitting: scaffolds
18 Feng Q. Materials selection and scaffold cultured epidermal cells. Surg. Today for tissue engineering of the aortic valve.
construction for liver tissue engineering. 27(10), 915–923 (1997). J. Biomater. Sci. Polymer Ed. 17(1–2),
Materials Science Forum 475–479 Pt 3, 31 Beckstead BL, Pan S, Bhrany AD, 77–89 (2006).
PRICM 5: The Fifth Pacific Rim Bratt-Leal AM, Ratner BD, Giachelli CM. 44 Xu CY, Inai R, Kotaki M, Ramakrishna S.
International Conference on Advanced Esophageal epithelial cell interaction with Aligned biodegradable nanofibrous
Materials and Processing 2004, 2391–2394 synthetic and natural scaffolds for tissue structure: a potential scaffold for blood
(2005). engineering. Biomaterials 26(31), vessel engineering. Biomaterials 25(5),
19 Bhandari RNB, Riccalton LA, Lewis AL 6217–6228 (2005). 877–886 (2004).
et al. Liver tissue engineering: a role for co- 32 Bhrany AD, Beckstead BL, Lang TC, 45 Zhang L, Ao Q, Wang A et al. A sandwich
culture systems in modifying hepatocyte Farwell DG, Giachelli CM, Ratner BD. tubular scaffold derived from chitosan for
function and viability. Tissue Eng. 7(3), Development of an esophagus acellular blood vessel tissue engineering. J. Biomed.
345–357 (2001). matrix tissue scaffold. Tissue Eng. 12(2), Mater. Res. 77A(2), 277–284 (2006).
20 Kulig KM, Vacanti JP. Hepatic tissue 319–330 (2006). 46 Hoenig MR, Campbell GR, Rolfe BE,
engineering. Transpl. Immunol. 12(3–4), 33 Sodian R, Hoerstrup SP, Sperling JS et al. Campbell JH. Tissue-engineered blood
303–310 (2004). Early in vivo experience with tissue- vessels. Arterioscl. Thrombosis Vasc. Bio.
21 Yang J, Chung TW, Nagaoka M, Goto M, engineered trileaflet heart valves. 25(6), 1128–1134 (2005).
Cho C-S, Akaike T. Hepatocyte-specific Circulation 102(19 Suppl. 3), III22–29 47 Yang J, Motlagh D, Webb Antonio R,
porous polymer-scaffolds of (2000). Ameer Guillermo A. Novel biphasic
alginate/galactosylated chitosan sponge for 34 Sodian R, Sperling JS, Martin DP et al. elastomeric scaffold for small-diameter
liver-tissue engineering. Biotech. Letters Fabrication of a trileaflet heart valve blood vessel tissue engineering. Tissue Eng.
23(17), 1385–1389 (2001). scaffold from a polyhydroxyalkanoate 11(11–12), 1876–1886 (2005).
22 Vacanti JP. Tissue and organ engineering: biopolyester for use in tissue engineering. 48 Chua CK, Leong KF, Tan KH, Wiria FE,
can we build intestine and vital organs? Tissue Eng. 6(2), 183–187 (2000). Cheah CM. Development of tissue
J. Gastrointest. 7(7), 831–835 (2003). 35 Breuer CK, Shin’oka T, Tanel RE et al. scaffolds using selective laser sintering of
23 Lloyd DAJ, Ansari TI, Gundabolu P et al. Tissue engineering lamb heart valve leaflets. polyvinyl alcohol/hydroxyapatite
A pilot study investigating a novel Biotech. Bioeng. 50(5), 562–567 (1996). biocomposite for craniofacial and joint
subcutaneously implanted precellularised 36 Shinoka T, Shum-Tim D, Ma PX et al. defects. J. Mater. Sci. Mater. Med. 15(10),
scaffold for tissue engineering of intestinal Tissue-engineered heart valve leaflets: does 1113–1121 (2004).
mucosa. Eur. Cells Mater. 11, 27–34 cell origin affect outcome? Circulation 49 Cowan CM, Aalami OO, Shi Y-Y et al.
(2006). 96(9 Suppl.), II-102–107 (1997). Bone morphogenetic protein 2 and retinoic
24 Chen MK, Beierle EA. Animal models for 37 Levenberg S, Rouwkema J, Macdonald M acid accelerate in vivo bone formation,
intestinal tissue engineering. Biomaterials et al. Engineering vascularized skeletal osteoclast recruitment, and bone turnover.
25(9), 1675–1681 (2004). muscle tissue. Nat. Biotechnol. 23(7), Tissue Eng. 11(3/4), 645–658 (2005).
25 Nakase Y, Hagiwara A, Nakamura T et al. 879–884 (2005). 50 Hollister SJ, Lin CY, Saito E et al.
Tissue engineering of small intestinal tissue 38 Saxena AK, Willital GH, Vacanti JP. Engineering craniofacial scaffolds. Orthod.
using collagen sponge scaffolds seeded with Vascularized three-dimensional skeletal Craniofac. Res. 8(3), 162–173 (2005).
smooth muscle cells. Tissue Eng. 12(2), muscle tissue-engineering. Bio-Med. Mater. 51 Calve S, Dennis RG, Kosnik PE II, Baar K,
403–412 (2006). Eng. 11(4), 275–281 (2001). Grosh K, Arruda EM. Engineering of
26 Choi RS, Vacanti JP. Preliminary studies of 39 Neumann T, Hauschka SD, Sanders JE. functional tendon. Tissue Eng. 10(5–6),
tissue-engineered intestine using isolated Tissue engineering of skeletal muscle using 755–761 (2004).
epithelial organoid units on tubular polymer fiber arrays. Tissue Eng. 9(5), 52 Bosch U, Krettek C. Tissue engineering of
synthetic biodegradable scaffolds. Transpl. 995–1003 (2003). tendons and ligaments. A new challenge.
Proc. 29(1–2), 848–851 (1997). 40 Lauer G, Schimming R. Tissue-engineered Der Unfallchirurg 105(2), 88–94 (2002).
27 Stelzner M, Chen DC. To make a new mucosa graft for reconstruction of the 53 Ge Z, Yang F, Goh JCH, Ramakrishna S,
intestinal mucosa. Rejuvenation Res. 9(1), intraoral lining after freeing of the tongue: a Lee EH. Biomaterials and scaffolds for
20–25 (2006). clinical and immunohistologic study. ligament tissue engineering. J. Biomed.
28 Marzaro M, Vigolo S, Oselladore B et al. J. Oral Maxillofacial 59(2), 169–175 Mater. Res. Part A 77A(3), 639–652 (2006).
In vitro and in vivo proposal of an artificial (2001). 54 Sahoo S, Ouyang H, Goh JCH, Tay TE,
esophagus. J. Biomed. Mater. Mater. Res. 41 Bunaprasert T, Hadlock T, Marler J et al. Toh SL. Characterization of a novel
77A(4), 795–801 (2006). Tissue engineered muscle implantation for polymeric scaffold for potential application
29 Zhu Y, Chan-Park MB, Chian KS. Surface tongue reconstruction: a preliminary in tendon/ligament tissue engineering.
modification of poly(dl-lactide-co- report. Laryngoscope 113(10), 1792–1797 Tissue Eng. 12(1), 91–99 (2006).
glycolide) copolymer with covalently (2003). 55 Majima T, Funakosi T, Iwasaki N et al.
immobilized collagen for esophageal tissue 42 Kim J, Hadlock T, Cheney M, Varvares M, Alginate and chitosan polyion complex
engineering. Materials Forum 29, 478–483 Marler J. Muscle tissue engineering for hybrid fibers for scaffolds in ligament and
(2005). partial glossectomy defects. Arch. Facial tendon tissue engineering. J. Orthopaedic
Plast. Surg. 5(5), 403–407 (2003). Science 10(3), 302–307 (2005).

844 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

56 Chen G, Sato T, Ushida T, Ochiai N, 70 Richardson TP, Mooney DJ. Multiple photopolymerized hyaluronic acid
Tateishi T. Tissue engineering of cartilage growth factor delivery from polymer networks. Biomacromolecules 6(1), 386–391
using a hybrid scaffold of synthetic polymer systems for regenerative medicine. Idenshi (2005).
and collagen. Tissue Eng. 10(3/4), 323–330 Igaku 6(3), 373–377 (2002). 82 Funakoshi T, Majima T, Iwasaki N et al.
(2004). 71 Hile DD, Amirpour ML, Akgerman A, Novel chitosan-based hyaluronan hybrid
57 Hutmacher DW. Scaffolds in tissue Pishko MV. Active growth factor delivery polymer fibers as a scaffold in ligament
engineering bone and cartilage. Biomaterials from poly(dl-lactide-co-glycolide) foams tissue engineering. J. Biomed. Mater. Res. A
21(24), 2529–2543 (2000). prepared in supercritical CO2. 74(3), 338–346 (2005).
58 Sato T, Chen G, Ushida T et al. Evaluation J. Controlled Rel. 66(2–3), 177–185 83 Baran ET, Tuzlakoglu K, Salgado AJ,
of PLLA-collagen hybrid sponge as a (2000). Reis RL. Multichannel mould processing
scaffold for cartilage tissue engineering. 72 Yasuda A, Kojima K, Tinsley KW, of 3D structures from microporous
Mat. Sci. Eng. C: Biomimetic Supramol. Sys. Yoshioka H, Mori Y, Vacanti CA. In vitro coralline hydroxyapatite granules and
C24(3), 365–372 (2004). culture of chondrocytes in a novel chitosan support materials for guided
59 Kang Y, Yang J, Khan S, Anissian L, thermoreversible gelation polymer tissue regeneration/engineering. J. Mater.
Ameer GA. A new biodegradable polyester scaffold containing growth factors. Tissue Sci. Mater. Med. 15(2), 161–165 (2004).
elastomer for cartilage tissue engineering. Eng. 12(5), 1237–1245 (2006). 84 Knackstedt MA, Arns CH, Senden TJ,
J. Biomed. Mater. Res. 77A(2), 331–339 73 Sheridan MH, Shea LD, Peters MC, Gross K. Structure and properties of
(2006). Mooney DJ. Bioabsorbable polymer clinical coralline implants measured via 3D
60 Sun J, Wu J, Li H, Chang J. Macroporous scaffolds for tissue engineering capable of imaging and analysis. Biomaterials 27(13),
poly(3-hydroxybutyrate-co-3- sustained growth factor delivery. 2776–2786 (2006).
hydroxyvalerate) matrices for cartilage J. Controlled Rel. 64(1–3), 91–102 85 Pattison MA, Wurster S, Webster TJ,
tissue engineering. Eur. Polymer J. 41(10), (2000). Haberstroh KM. Three-dimensional,
2443–2449 (2005). 74 Chan BP, So KF. Photochemical nano-structured PLGA scaffolds for
61 Yang F, Murugan R, Ramakrishna S, crosslinking improves the bladder tissue replacement applications.
Wang X, Ma YX, Wang S. Fabrication of physicochemical properties of collagen Biomaterials 26(15), 2491–2500 (2005).
nano-structured porous PLLA scaffold scaffolds. J. Biomed. Mater. Res. Part A 86 Kim HD, Bae EH, Kwon IC, Pal RR,
intended for nerve tissue engineering. 75A(3), 689–701 (2005). Nam JD, Lee DS. Effect of PEG-PLLA
Biomaterials 25(10), 1891–1900 (2004). 75 Gelinsky M, Koenig U, Sewing A, diblock copolymer on macroporous PLLA
62 Yu X, Bellamkonda RV. Tissue-engineered Pompe W. Porous scaffolds made from scaffolds by thermally induced phase
scaffolds are effective alternatives to mineralized collagen – a biomimetic bone separation. Biomaterials 25(12),
autografts for bridging peripheral nerve graft material. Materialwissenschaft und 2319–2329 (2004).
gaps. Tissue Eng. 9(3), 421–430 (2003). Werkstofftechnik 35(4), 229–233 (2004). 87 Jeong SI, Kim B-S, Lee YM, Ihn KJ,
63 Yoshii S, Oka M. Collagen filaments as a 76 O’Brien FJ, Harley BA, Yannas IV, Kim SH, Kim YH. Morphology of elastic
scaffold for nerve regeneration. J. Biomed. Gibson L. Influence of freezing rate on poly(L-lactide-co-e-caprolactone)
Mater. Res. 56(3), 400–405 (2001). pore structure in freeze-dried collagen- copolymers and in vitro and in vivo
GAG scaffolds. Biomaterials 25(6), degradation behavior of their scaffolds.
64 Maquet V, Martin D, Malgrange B et al.
1077–1086 (2003). Biomacromolecules 5(4), 1303–1309
Peripheral nerve regeneration using
77 Schantz J-T, Brandwood A, (2004).
bioresorbable macroporous polylactide
scaffolds. J. Biomed. Mater. Res. 52(4), Hutmacher DW, Khor HL, Bittner K. 88 Wang Y-W, Wu Q, Chen J, Chen G-Q.
639–651 (2000). Osteogenic differentiation of Evaluation of three-dimensional scaffolds
mesenchymal progenitor cells in made of blends of hydroxyapatite and
65 Dumont CE, Born W. Stimulation of
computer designed fibrin-polymer- poly(3-hydroxybutyrate-co-3-
neurite outgrowth in a human nerve
ceramic scaffolds manufactured by fused hydroxyhexanoate) for bone
scaffold designed for peripheral nerve
deposition modeling. J. Mat.Sci. Mater. reconstruction. Biomaterials 26(8),
reconstruction. J. Biomed. Mater. Res. Part
Med. 16(9), 807–819 (2005). 899–904 (2005).
B: App. BioMater. 73B(1), 194–202 (2005).
78 Hokugo A, Takamoto T, Tabata Y. 89 Chen G-Q, Wu Q. The application of
66 Bell E, Ehrlich HP, Buttle DJ, Nakatsuji T.
Preparation of hybrid scaffold from fibrin polyhydroxyalkanoates as tissue
Living tissue formed in vitro and accepted
and biodegradable polymer fiber. engineering materials. Biomaterials 26(33),
as skin-equivalent tissue of full thickness.
Biomaterials 27(1), 61–67 (2005). 6565–6578 (2005).
Science 211(4486), 1052–1054 (1981).
79 Wnek GE, Carr ME, Simpson DG, 90 Zhao K, Deng Y, Chen G-Q. Effects of
67 Godbey WT. Polymeric scaffolds for stem
Bowlin GL. Electrospinning of nanofiber surface morphology on the
cell growth. Australian J. Chem. 58(10),
fibrinogen structures. Nano Letters 3(2), biocompatibility of
689–690 (2005).
213–216 (2003). polyhydroxyalkanoates. Biochem. Eng. J.
68 Lindblad WJ. Stem cells in dermal wound 16(2), 115–123 (2003).
80 Ji Y, Ghosh K, Shu XZ et al. Electrospun
healing. In: Stem Cells Handbook. Stewart
three-dimensional hyaluronic acid 91 Bryant SJ, Bender RJ, Durand KL,
Sell (Ed.). Humana Press, NJ, USA
nanofibrous scaffolds. Biomaterials Anseth KS. Encapsulating chondrocytes in
101–105 (2004).
27(20), 3782–3792 (2006). degrading PEG hydrogels with high
69 Riha GM, Lin PH, Lumsden AB, Yao Q, modulus: engineering gel structural
81 Burdick JA, Chung C, Jia X,
Chen C. Review: application of stem cells changes to facilitate cartilaginous tissue
Randolph MA, Langer R. Controlled
for vascular tissue engineering. Tissue Eng, production. Biotech. Bioeng. 86(7),
degradation and mechanical behavior of
11(9–10), 1535–1552 (2005). 747–755 (2004).

www.future-drugs.com 845
Weigel, Schinkel & Lendlein

92 Lee WK, Ichi T, Ooya T, Yamamoto T, 103 Bourke SL, Kohn J, Dunn MG. Preliminary 115 Leong KF, Cheah CM, Chua CK. Solid
Katoh M, Yui N. Novel poly(ethylene development of a novel resorbable synthetic freeform fabrication of three-dimensional
glycol) scaffolds crosslinked by hydrolyzable polymer fiber scaffold for anterior cruciate scaffolds for engineering replacement
polyrotaxane for cartilage tissue engineering. ligament reconstruction. Tissue Eng. tissues and organs. Biomaterials 24(13),
J. Biomed. Mater. Res. Part A 67A(4), 10(1/2), 43–52 (2004). 2363–2378 (2003).
1087–1092 (2003). 104 Meese TM, Hu Y, Nowak RW, Marra KG. 116 Shu XZ, Liu Y, Palumbo F, Prestwich GD.
93 Ebaretonbofa E, Evans JRG. High porosity Surface studies of coated polymer Disulfide-crosslinked hyaluronan-gelatin
hydroxyapatite foam scaffolds for bone microspheres and protein release from hydrogel films: a covalent mimic of the
substitute. J. Porous Mat. 9(4), 257–263 tissue-engineered scaffolds. J. Biomater. Sci. extracellular matrix for in vitro cell growth.
(2003). Polym. Ed. 13(2), 141–151 (2002). Biomaterials 24(21), 3825–3834 (2003).
94 Damien E, Hing K, Saeed S, Revell PA. 105 Mercier NR, Costantino HR, Tracy MA, 117 Barbanti SH, Santos AR Jr, Zavaglia CAC,
A preliminary study on the enhancement of Bonassar LJ. Poly(lactide-co-glycolide) Duek EAR. Porous and dense poly(L-lactic
the osteointegration of a novel synthetic microspheres as a moldable scaffold for acid) and poly(D,L-lactic acid-co-glycolic
hydroxyapatite scaffold in vivo. J. Biomed. cartilage tissue engineering. Biomaterials acid) scaffolds: in vitro degradation in
Mater. Res. Part A 66A(2), 241–246 (2003). 26(14), 1945–1952 (2005). culture medium and osteoblasts culture.
95 Saito A, Suzuki Y, Kitamura M et al. Repair 106 King KR, Wang C, Vacanti JP, J. Mater. Sci. Mater. Med. 15(12),
of 20-mm long rabbit radial bone defects Borenstein JT. Biodegradable polymer 1315–1321 (2004).
using BMP-derived peptide combined with microfluidics for tissue engineering 118 Cai Q, Yang J, Bei J, Wang S. A novel
an α-tricalcium phosphate scaffold. microvasculature. Materials Research Society porous cells scaffold made of polylactide-
J. Biomed. Mater. Res. Part A 77A(4), Symposium Proceedings 729(BioMEMS and dextran blend by combining phase-
700–706 (2006). Bionanotechnology), 3–8 (2002). separation and particle-leaching techniques.
96 Aydin HM, Piskin E, Calimli A. 107 Yeong WY, Chua CK, Leong KF, Biomaterials 23(23), 4483–4492 (2002).
Microporous scaffolds from poly(lactide-co- Chandrasekaran M. Rapid prototyping in 119 Young TH, Lin CW, Cheng LP, Hsieh CC.
e-caprolactone) composites with tissue engineering: challenges and potential. Preparation of EVAL membranes with
hydroxyapatite and tricalcium phosphates Trends Biotechnol. 22(12), 643–652 (2004). smooth and particulate morphologies for
using supercritical CO2 for bone tissue 108 Xu C, Yang F, Wang S, Ramakrishna S. neuronal culture. Biomaterials 22(13),
engineering. J. Bioactive Compatible Polymers In vitro study of human vascular endothelial 1771–1777 (2001).
19(5), 383–394 (2004). cell function on materials with various 120 Albrecht W, Weigel T, Groth T, Hilke R,
97 Damien CJ, Parsons JR. Bone graft and surface roughness. J. Biomed. Mater. Res. Paul D. Formation of porous bilayer hollow
bone graft substitutes: a review of current Part A 71A(1), 154–161 (2004). fiber membranes. Macromol. Symposia
technology and applications. J. App. Biomat. 109 O’Brien FJ, Harley BA, Yannas IV, 188(Polymer Membranes), 131–141
2(3), 187–208 (1991). Gibson LJ. The effect of pore size on cell (2002).
98 Pereira MM, Jones JR, Orefice RL, adhesion in collagen-GAG scaffolds. 121 Albrecht W, Kneifel K, Weigel T et al.
Hench LL. Preparation of bioactive glass- Biomaterials 26(4), 433–441 (2005). Preparation of highly asymmetric hollow
polyvinyl alcohol hybrid foams by the sol-gel 110 Hutmacher DW, Schantz T, Zein I, fiber membranes from poly(ether imide) by
method. J. Mater. Sci. Mater. Med. 16(11), Ng KW, Teoh SH, Tan KC. Mechanical a modified dry-wet phase inversion
1045–1050 (2005). properties and cell cultural response of technique using a triple spinneret. J. Memb.
99 Lu HH, El-Amin SF, Scott KD, polycaprolactone scaffolds designed and Sci. 262(1–2), 69–80 (2005).
Laurencin CT. Three-dimensional, fabricated via fused deposition modeling. 122 Coombes AGA, Rizzi SC, Williamson M,
bioactive, biodegradable, polymer-bioactive J. Biomed. Mater. Res. 55(2), 203–216 Barralet JE, Downes S, Wallace WA.
glass composite scaffolds with improved (2001). Precipitation casting of polycaprolactone
mechanical properties support collagen 111 Ma PX. Scaffolds for tissue fabrication. for applications in tissue engineering and
synthesis and mineralization of human Materials Today 7(5), 30–40 (2004). drug delivery. Biomaterials 25(2), 315–325
osteoblast-like cells in vitro. J. Biomed. (2003).
112 Jayaraman K, Kotaki M, Zhang Y, Mo X,
Mater. Res. Part A 64A(3), 465–474 (2003). 123 Krasteva N, Seifert B, Albrecht W et al.
Ramakrishna S. Recent advances in polymer
100 Gong S, Wang H, Sun Q, Xue S-T, nanofibers. J. Nanosci. Nanotech. 4 (1/2), Influence of polymer membrane porosity
Wang J-Y. Mechanical properties and in vitro 52–65 (2004). on C3A hepatoblastoma cell adhesive
biocompatibility of porous zein scaffolds. interaction and function. Biomaterials
113 Hollister SJ, Maddox RD, Taboas JM.
Biomaterials 27(20), 3793–3799 (2006). 25(13), 2467–2476 (2004).
Optimal design and fabrication of scaffolds
101 Mathieu LM, Mueller TL, Bourban P-E, to mimic tissue properties and satisfy 124 Sarazin P, Roy X, Favis BD. Controlled
Pioletti DP, Mueller R, Manson J-AE. biological constraints. Biomaterials 23(20), preparation and properties of porous
Architecture and properties of anisotropic 4095–4103 (2002). poly(L-lactide) obtained from a co-
polymer composite scaffolds for bone tissue continuous blend of two biodegradable
114 Hutmacher DW, Sittinger M, Risbud MV.
engineering. Biomaterials 27(6), 905–916 polymers. Biomaterials 25(28), 5965–5978
Scaffold-based tissue engineering: rationale
(2006). (2004).
for computer-aided design and solid free-
102 Washburn NR, Simon CG Jr, Tona A, form fabrication systems. Trends Biotechnol. 125 Chowdhury PR, Kumar V. Fabrication and
Elgendy HM, Karim A, Amis EJ. 22(7), 354–362 (2004). evaluation of porous 2,3-
Co-extrusion of biocompatible polymers • Different fabrication techniques of dialdehydecellulose membrane as a
for scaffolds with co-continuous category rapid prototyping or solid free- potential biodegradable tissue-engineering
morphology. J. Biomed. Mater. Res. 60(1), form fabrications. scaffold. J. Biomed. Mater. Res. Part A
20–29 (2002). 76A(2), 300–309 (2006).

846 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

126 Seifert B, Mihanetzis G, Groth T et al. 138 Chiang CY, Starov VM, Hall MS, 149 Gendron R, Daigneault LE. Continuous
Polyetherimide: a new membrane-forming Lloyd DR. Crystallization kinetics of extrusion of microcellular polycarbonate.
polymer for biomedical applications. Artif. polymer-solvent systems: 2. Experimental Polym. Eng. Sci. 43(7), 1361–1377 (2003).
Organs 26(2), 189–199 (2002). verification of the model. Colloid J. 150 Riddle KW, Mooney DJ. Role of
127 Trimpert C, Boese G, Albrecht W et al. (Translation of Kolloidnyi Zhurnal) 59(2), poly(lactide-co-glycolide) particle size on
Poly(ether imide) membranes modified 236–247 (1997). gas-foamed scaffolds. J. BioMater. Sci.,
with poly(ethylene imine) as potential 139 Shang M, Matsuyama H, Maki T, Polym. Ed. 15(12), 1561–1570 (2004).
carriers for epidermal substitutes. Teramoto M, Lloyd DR. Effect of 151 Unger RE, Huang Q, Peters K, Protzer D,
Macromol. Biosci. 6(4), 274–284 (2006). crystallization and liquid-liquid phase Paul D, Kirkpatrick CJ. Growth of human
128 Lloyd DR, Kim SS, Kinzer KE. separation on phase-separation kinetics in cells on polyethersulfone (PES) hollow fiber
Microporous membrane formation via poly(ethylene-co-vinyl alcohol)/glycerol membranes. Biomaterials 26(14),
thermally induced phase separation. II. solution. J. Polym. Sci. Part B: Polym. 1877–1884 (2005).
Liquid-liquid phase separation. J. Memb. Physics 41(2), 194–201 (2002).
152 Barry JJA, Silva MMCG, Popov VK,
Science 64(1–2), 1–11 (1991). 140 Wei G, Ma PX. Structure and properties of Shakesheff KM, Howdle SM. Supercritical
129 Matsuyama H, Berghmans S, Lloyd DR. nano-hydroxyapatite/polymer composite carbon dioxide: putting the fizz into
Formation of anisotropic membranes via scaffolds for bone tissue engineering. biomaterials. Philosophical Transactions of the
thermally induced phase separation. Biomaterials 25(19), 4749–4757 (2004). Royal Society of London, Series A:
Polymer 40(9), 2289–2301 (1999). 141 Hua FJ, Kim GE, Lee JD, Son YK, Lee DS. Mathematical, Physical and Eng. Sciences
130 Shin KC, Kim BS, Kim JH, Park TG, Macroporous poly(L-lactide) scaffold 1. 364(1838), 249–261 (2006).
Nam JD, Lee DS. A facile preparation of Preparation of a macroporous scaffold by 153 Singh L, Kumar V, Ratner BD. Generation
highly interconnected macroporous PLGA liquid-liquid phase separation of a PLLA- of porous microcellular 85/15 poly ((DL)-
scaffolds by liquid-liquid phase dioxane-water system. J. Biomed. Mater. lactide-co-glycolide) foams for biomedical
separation II. Polymer 46(11), 3801–3808 Res. 63(2), 161–167 (2002). applications. Biomaterials 25(13),
(2005). 142 Haugen H, Ried V, Brunner M, Will J, 2611–2617 (2004).
131 Li Z, Zhang M. Chitosan-alginate as Wintermantel E. Water as foaming agent 154 Cotugno S, Di Maio E, Mensitieri G et al.
scaffolding material for cartilage tissue for open cell polyurethane structures. Characterization of microcellular
engineering. J. Biomed. Mater. Res. A J. Mater. Sci Mater. Med 15(4), 343–346 biodegradable polymeric foams produced
75(2), 485–493 (2005). (2004). from supercritical carbon dioxide solutions.
132 Gong Y, Ma Z, Gao C, Wang W, Shen J. 143 Di Maio E, Mensitieri G, Iannace S, Indust. Eng. Chem. Res. 44(6), 1795–1803
Specially elaborated thermally induced Nicolais L, Li W, Flumerfelt RW. Structure (2005).
phase separation to fabricate poly(L-lactic optimization of polycaprolactone foams by 155 Sun XH, Liu HJ, Li G, Liao X, He JS.
acid) scaffolds with ultra large pores and using mixtures of CO2 and N2 as blowing Investigation on the cell nucleation and cell
good interconnectivity. J. App. Polym. Sci. agents. Polym. Eng. Sci. 45(3), 432–441 growth in microcellular foaming by means of
101(5), 3336–3342 (2006). (2005). temperature quenching. J. App. Polym. Sci.
133 Ho M-H, Kuo P-Y, Hsieh H-J et al. 144 Parks KL, Beckman EJ. Generation of 93(1), 163–171 (2004).
Preparation of porous scaffolds by using microcellular polyurethane foams via • Computer-aided tissue engineering (CATE)
freeze-extraction and freeze-gelation polymerization in carbon dioxide. II: foam from the point of view of supporting utility
methods. Biomaterials 25(1), 129–138 formation and characterization. Polym. Eng. with different application areas.
(2003). Sci. 36(19), 2417–2431 (1996). 156 Fu J, Naguib HE. Effect of nanoclay on the
134 Gao CY, Wang DY, Shen JC. Fabrication 145 Gomes ME, Ribeiro AS, Malafaya PB, mechanical properties of PMMA/clay
of porous collagen/chitosan scaffolds with Reis RL, Cunha AM. A new approach nanocomposite foams. J. Cellular Plast.
controlling microstructure for dermal based on injection moulding to produce 42(4), 325–342 (2006).
equivalent. Polym. Adv. Tech. 14(6), biodegradable starch-based polymeric 157 Xu Q, Ren XW, Chang YN, Wang JW, Yu L,
373–379 (2003). scaffolds: morphology, mechanical and Dean K. Generation of microcellular
degradation behavior. Biomaterials 22(9), biodegradable polycaprolactone foams in
135 Lendlein A, Ridder U. Poröse strukturen
883–889 (2001). supercritical carbon dioxide. J. App. Polym.
aus abbaubaren, elastischen polymeren für
das tissue engineering. Deutsche 146 Kim TK, Yoon JJ, Lee DS, Park TG. Gas Sci. 94(2), 593–597 (2004).
Gesellschaft für Chirugie (Kongressband foamed open porous biodegradable 158 Jacobs MA, Kemmere MF, Keurentjes JTF.
2000) 761–763 (2000). polymeric microspheres. Biomaterials 27(2), Foam processing of poly (ethylene-co-vinyl
152–159 (2006). acetate) rubber using supercritical carbon
136 Lloyd DR, Kinzer KE, Tseng HS.
Microporous membrane formation via 147 Cartmell S, Huynh K, Lin A, Nagaraja S, dioxide. Polymer 45(22), 7539–7547 (2004).
thermally induced phase separation. I. Guldberg R. Quantitative microcomputed 159 Krause B, van der Vegt NFA, Wessling M.
Solid-liquid phase separation. J. Memb. tomography analysis of mineralization New ways to produce porous polymeric
Sci. 52(3), 239–261 (1990). within three-dimensional scaffolds in vitro. membranes by carbon dioxide foaming.
J. Biomed. Mater. Res. Part A 69A(1), Desalination 144(1–3), 5–7 (2002).
137 Tu C, Cai Q, Yang J, Wan Y, Bei J, W
97–104 (2004).
ang S. The fabrication and 160 Barry JJA, Gidda HS, Scotchford CA,
characterization of poly(lactic acid) 148 Lin ASP, Barrows TH, Cartmell SH, Howdle SM. Porous methacrylate scaffolds:
scaffolds for tissue engineering by Guldberg RE. Microarchitectural and supercritical fluid fabrication and in vitro
improved solid-liquid phase separation. mechanical characterization of oriented chondrocyte responses. Biomaterials 25(17),
Polym. Adv. Tech. 14(8), 565–573 (2003). porous polymer scaffolds. Biomaterials 3559–3568 (2004).
24(3), 481–489 (2003).

www.future-drugs.com 847
Weigel, Schinkel & Lendlein

161 Yoon JJ, Park TG. Degradation behaviors 173 Khan YM, Katti DS, Laurencin CT. Novel tissue-engineering applications: 3D
of biodegradable macroporous scaffolds polymer-synthesized ceramic composite- bioplotting versus 3D printing. J. Polym. Sci.
prepared by gas foaming of effervescent based system for bone repair: an in vitro Part A: Polym. Chem. 42(3), 624–638
salts. J. Biomed. Mater. Res. 55(3), 401–408 evaluation. J. Biomed. Mater. Res. Part A (2004).
(2001). 69A(4), 728–737 (2004). 185 Cheah CM, Chua CK, Leong KF,
162 Nam YS, Yoon JJ, Park TG. A novel 174 Kochan D, Chua CK, Du ZH. Rapid Cheong CH, Naing MW. Automatic
fabrication method of macroporous prototyping issues in the 21st century. algorithm for generating complex
biodegradable polymer scaffolds using gas Computers in Industry 39(1), 3–10 (1999). polyhedral scaffold structures for tissue
foaming salt as a porogen additive. 175 Chua CK. Three-dimensional rapid engineering. Tissue Eng. 10(3/4), 595–610
J. Biomed. Mater. Res. 53(1), 1–7 (2000). prototyping technologies and (2004).
163 Harris LD, Kim BS, Mooney DJ. Open keydevelopment areas. Comp. Control Eng. J. 186 Dollar AM, Howe RD. A robust compliant
pore biodegradable matrices formed with 5(4), 200–206 (1994). grasper via shape deposition
gas foaming. J. Biomed. Mater. Res. 42(3), 176 Taboas JM, Maddox RD, Krebsbach PH, manufacturing. Ieee-Asme Transactions on
396–402 (1998). Hollister SJ. Indirect solid free form Mechatronics 11(2), 154–161 (2006).
164 Jang J-H, Shea LD. Controllable delivery fabrication of local and global porous, 187 Ang KC, Leong KF, Chua CK,
of non-viral DNA from porous scaffolds. biomimetic and composite 3D polymer- Chandrasekaran M. Compressive
J. Controlled Rel. 86(1), 157–168 (2003). ceramic scaffolds. Biomaterials 24(1), properties and degradability of poly(ε-
165 Yoon JJ, Kim JH, Park TG. 181–194 (2003). caprolatone)/ hydroxyapatite composites
Dexamethasone-releasing biodegradable 177 Das S, Hollister SJ, Flanagan C et al. under accelerated hydrolytic degradation.
polymer scaffolds fabricated by a gas- Computational design, freeform fabrication J. Biomed. Mater. Res.: Part A (2006).
foaming/salt-leaching method. Biomaterials and testing of nylon-6 tissue engineering 188 Ang KC, Leong KF, Chua CK,
24(13), 2323–2329 (2003). scaffolds. Materials Research Society Chandrasekaran M. Investigation of the
166 Albrecht W, Luetzow K, Weigel T, Symposium Proceedings 758(Rapid mechanical properties and porosity
Groth T, Schossig M, Lendlein A. Prototyping Technologies), 205–210 (2003). relationships in fused deposition
Development of highly porous 178 Chua CK, Chou SM, Wong TS. A study of modelling-fabricated porous structures.
microparticles from poly(ether imide) the state-of-the-art rapid prototyping Rapid Prototyping J. 12(2), 100–105
prepared by a spraying/coagulation process. technologies. Intl. J. Adv. Manufac. Tech. (2006).
J. Memb. Sci. 273(1–2), 106–115 (2006). 14(2), 146–152 (1998). 189 Malda J, Woodfield TBF, van der Vloodt F
167 Newman KD, McBurney MW. 179 Chua CK, Feng C, Lee CW, Ang GQ. et al. The effect of PEGT/PBT scaffold
Poly(D,L lactic-co-glycolic acid) Rapid investment casting: direct and architecture on the composition of tissue
microspheres as biodegradable indirect approaches via model maker II. engineered cartilage. Biomaterials 26(1),
microcarriers for pluripotent stem cells. Intl. J. Adv. Manufac. Tech. 25(1–2), 26–32 63–72 (2005).
Biomaterials 25(26), 5763–5771 (2004). (2005). 190 Woodfield TBF, Malda J, de Wijn J,
168 Ruhe PQ, Hedberg EL, Padron NT, 180 Chua CK, Leong KF, Cheah CM, Peters F, Riesle J, van Blitterswijk CA.
Spauwen PHM, Jansen JA, Mikos AG. Chua SW. Development of a tissue Design of porous scaffolds for cartilage
Biocompatibility and degradation of engineering scaffold structure library for tissue engineering using a three-
poly(DL-lactic-co-glycolic acid)/calcium rapid prototyping. Part 1: Investigation and dimensional fiber-deposition technique.
phosphate cement composites. J. Biomed. Classification. Intl. J. Adv. Manufac. Tech. Biomaterials 25(18), 4149–4161 (2004).
Mater. Res. Part A 74A(4), 533–544 21(4), 291–301 (2003). 191 Zein I, Hutmacher DW, Tan KC, Teoh SH.
(2005). 181 Mironov V, Boland T, Trusk T, Forgacs G, Fused deposition modeling of novel
169 Hedberg EL, Tang A, Crowther RS, Markwald RR. Organ printing: computer- scaffold architectures for tissue engineering
Carney DH, Mikos AG. Controlled release aided jet-based 3D tissue engineering. applications. Biomaterials 23(4),
of an osteogenic peptide from injectable Trends Biotech. 21(4), 157–161 (2003). 1169–1185 (2001).
biodegradable polymeric composites. 182 Zhang H, Hutmacher DW, Chollet F, 192 Wang F et al. Precision extruding
J. Controlled Rel. 84(3), 137–150 (2002). Poo AN, Burdet E. Microrobotics and deposition and characterizing of cellular
170 Zhang J, Zhang H, Wu L, Ding J. MEMS-based fabrication techniques for polycarpolactone tissue scaffolds. Rapid
Fabrication of three dimensional polymeric scaffold-based tissue engineering. Prototyping J. 10, 42–49 (2004).
scaffolds with spherical pores. J. Mater. Sci. Macromol. Biosci. 5(6), 477–489 (2005). 193 Darling AL, Sun W. 3D microtomographic
41(6), 1725–1731 (2006). • Different fabrication techniques of characterization of precision extruded poly-
171 Borden M, Attawia M, Khan Y, category rapid prototyping or solid free- ε-caprolactone scaffolds. J. Biomed. Mater.
El-Amin SF, Laurencin CT. Tissue- form fabrications. Res. Part B: App. BioMater. 70B(2),
engineered bone formation in vivo using a 183 Schantz J-T, Ng MM-L, Netto P et al. 311–317 (2004).
novel sintered polymeric microsphere Application of an X-ray microscopy 194 Rohner D, Hutmacher DW, Cheng TK,
matrix. J. Bone Joint Surg. Br. Vol. 86(8), technique to evaluate tissue-engineered Oberholzer M, Hammer B. In vivo efficacy
1200–1208 (2004). bone-scaffold constructs. Mater. Sci. Eng. of bone-marrow-coated polycaprolactone
172 Borden M, Attawia M, Laurencin CT. The C: Biomimetic Supramol. Syst. C20(1–2), scaffolds for the reconstruction of orbital
sintered microsphere matrix for bone tissue 9–17 (2002). defects in the pig. J. Biomed. Mater. Res.
engineering: in vitro osteoconductivity 184 Pfister A, Landers R, Laib A, Huebner U, Part B: App. BioMater. 66B(2), 574–580
studies. J. Biomed. Mater. Res. 61(3), Schmelzeisen R, Muelhaupt R. (2003).
421–429 (2002). Biofunctional rapid prototyping for

848 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

195 Byers BA, Guldberg RE, Hutmacher DW, 206 Leukers B, Guelkan H, Irsen SH et al. 217 Cooke MN, Fisher JP, Dean D, Rimnac C,
Garcia AJ. Effects of Runx2 genetic Hydroxyapatite scaffolds for bone tissue Mikos AG. Use of stereolithography to
engineering and in vitro maturation of engineering made by 3D printing. J. Mater. manufacture critical-sized 3D
tissue-engineered constructs on the repair Sci. Mater. Med. 16(12), 1121–1124 biodegradable scaffolds for bone ingrowth.
of critical size bone defects. J. Biomed. (2005). J. Biomed. Mater. Res. Part B: App. Biomat.
Mater. Res. Part A 76A(3), 646–655 (2006). 207 Curodeau A, Sachs E, Caldarise S. Design 64B(2), 65–69 (2003).
196 Cai H, Azangwe G, Shepherd DET. Skin and fabrication of cast orthopedic implants 218 Matsuyama H, Yuasa M, Kitamura Y,
cell culture on an ear-shaped scaffold with freeform surface textures from 3-D Teramoto M, Lloyd DR. Structure control
created by fused deposition modelling. Bio- printed ceramic shell. J. Biomed. Mater. Res. of anisotropic and asymmetric
Med. Mater. Eng. 15(5), 375–380 (2005). 53(5), 525–535 (2000). polypropylene membrane prepared by
197 Kalita SJ, Bose S, Hosick HL, 208 Irsen SH, Leukers B, Hoeckling C, Tille C, thermally induced phase separation.
Bandyopadhyay A. Development of Seitz H. Bioceramic granulates for use in J. Memb. Sci. 179(1–2), 91–100 (2000).
controlled porosity polymer-ceramic 3D printing: process engineering aspects. 219 Chen J, Cui L, Liu W, Cao Y. Development
composite scaffolds via fused deposition Materialwissenschaft und Werkstofftechnik on solvent casting/particulate leaching.
modeling. Mater. Sci. Eng. C: Biomimetic 37(6), 533–537 (2006). Zhongguo Shengwu Gongcheng Zazhi 23(4),
Supramol. Syst. C23(5), 611–620 (2003). 209 Lam CXF, Mo XM, Teoh SH, 32–35, 42 (2003).
198 Chen Z, Li D, Lu B, Tang Y, Sun M, Xu S. Hutmacher DW. Scaffold development 220 Lawes RA. Future trends in high-resolution
Fabrication of osteo-structure analogous using 3D printing with a starch-based lithography. App. Surface Sci. 154–155,
scaffolds via fused deposition modeling. polymer. Mater. Sci. Eng. C: Biomimetic and 519–526 (2000).
Scripta Materialia 52(2), 157–161 (2004). Supramolecular Systems C20(1–2), 49–56 221 Lin CY, Kikuchi N, Hollister SJ. A novel
199 Huang M-H, Li S, Hutmacher DW et al. (2002). method for biomaterial scaffold internal
Degradation and cell culture studies on 210 Giordano RA, Wu BM, Borland SW, architecture design to match bone elastic
block copolymers prepared by ring opening Cima LG, Sachs EM, Cima MJ. properties with desired porosity. J. Biomech.
polymerization of e-caprolactone in the Mechanical properties of dense polylactic 37(5), 623–636 (2004).
presence of poly(ethylene glycol). acid structures fabricated by three 222 Taylor PM, Sachlos E, Dreger SA,
J. Biomed. Mater. Res. Part A 69A(3), dimensional printing. J. Biomater. Sci. Chester AH, Czernuszka JT, Yacoub MH.
417–427 (2004). Polym. Ed. 8(1), 63–75 (1996). Interaction of human valve interstitial cells
200 Landers R, Mulhaupt R. Desktop 211 Tan KH, Chua CK, Leong KF et al. with collagen matrices manufactured using
manufacturing of complex objects, Selective laser sintering of biocompatible rapid prototyping. Biomaterials 27(13),
prototypes and biomedical scaffolds by polymers for applications in tissue 2733–2737 (2006).
means of computer-assisted design engineering. Bio-Med. Mater. Eng. 15(1–2), 223 Sachlos E, Reis N, Ainsley C, Derby B,
combined with computer-guided 3D 113–124 (2005). Czernuszka JT. Novel collagen scaffolds
plotting of polymers and reactive 212 Williams JM, Adewunmi A, Schek RM with predefined internal morphology made
oligomers. Macromol. Mater. Eng. 282, et al. Bone tissue engineering using by solid free form fabrication. Biomaterials
17–21 (2000). polycaprolactone scaffolds fabricated via 24(8), 1487–1497 (2003).
201 Koch KU, Biesinger B, Arnholz C, selective laser sintering. Biomaterials 224 Yeong W-Y, Chua C-K, Leong K-F. Indirect
Jansson V. Creating of biocompatible, high 26(23), 4817–4827 (2005). fabrication of collagen scaffold based on
stress resistent and resorbable implants 213 Rimell JT, Marquis PM. Selective laser inkjet printing technique. Rapid
using multiphase jet solidification sintering of ultra high molecular weight Prototyping J. 12(4) 229–237 (2006).
technology. Time compression polyethylene for clinical applications. 225 Ma PX. Tissue Engineering. Kroschwitz JI
Technologies ’98 Conference London, GB. J. Biomed. Mater. Res. 53(4), 414–420 (Ed.). John Wiley & Sons, NY, USA
Rapid News Publication 209–214 (1998). (2000). (2004).
202 Lenk R. Rapid prototyping of ceramic 214 Tan KH, Chua CK, Leong KF, Naing MW, 226 Zhang S. Fabrication of novel biomaterials
components. Adv. Eng. Mater. 2(1–2), Cheah CM. Fabrication and through molecular self-assembly. Nat.
40–41, 44–47 (2000). characterization of three-dimensional Biotechnol. 21(10), 1171–1178 (2003).
203 Ruecker M, Laschke MW, Junker D et al. poly(ether-ether-ketone)/-hydroxyapatite • Self-assembly systems.
Angiogenic and inflammatory response to biocomposite scaffolds using laser sintering.
227 Zhang S. Emerging biological materials
biodegradable scaffolds in dorsal skinfold Proceedings of: the Institution of Mechanical
through molecular self-assembly. Biotech.
chambers of mice. Biomaterials 27(29), Engineers Part H-J. Eng. in Medicine
Adv. 20(5–6), 321–339 (2002).
5027–5038 (2006). 219(H3), 183–194 (2005).
228 Holmes TC, De Lacalle S, Su X, Liu G,
204 Sachlos E, Czernuszka JT. Making tissue 215 Tan KH, Chua CK, Leong KF et al.
Rich A, Zhang S. Extensive neurite
engineering scaffolds work. Review on the Scaffold development using selective laser
outgrowth and active synapse formation on
application of solid freeform fabrication sintering of polyetheretherketone-
self-assembling peptide scaffolds. Proc. Natl
technology to the production of tissue hydroxyapatite biocomposite blends.
Acad. Sci. USA 97(12), 6728–6733 (2000).
engineering scaffolds. Euro. Cells Mater. 5, Biomaterials 24(18), 3115–3123 (2003).
29–40 (2003). 229 Kisiday J, Jin M, Kurz B et al.
216 Dhariwala B, Hunt E, Boland T. Rapid
Self-assembling peptide hydrogel fosters
205 Landers R, Pfister A, Huebner U, John H, prototyping of tissue-engineering
chondrocyte extracellular matrix production
Schmelzeisen R, Muelhaupt R. Fabrication constructs, using photopolymerizable
and cell division: implications for cartilage
of soft tissue engineering scaffolds by means hydrogels and stereolithography. Tissue Eng.
tissue repair. Proc. Natl Acad. Scie. USA
of rapid prototyping techniques. J. Mater. 10(9/10), 1316–1322 (2004).
99(15), 9996–10001 (2002).
Sci. 37(15), 3107–3116 (2002).

www.future-drugs.com 849
Weigel, Schinkel & Lendlein

230 Zhu H, Ji J, Shen J. Biomacromolecules 243 Zhang S, Rich A. Direct conversion of an 255 Ma Z, Kotaki M, Inai R, Ramakrishna S.
electrostatic self-assembly on 3-dimensional oligopeptide from a β-sheet to an α-helix: a Potential of nanofiber matrix as tissue-
tissue engineering scaffold. Biomacromolecules model for amyloid formation. Proc. Natl engineering scaffolds. Tissue Eng. 11(1/2),
5(5), 1933–1939 (2004). Acad. Sci. USA 94(1), 23–28 (1997). 101–109 (2005).
231 Lin H-H, Cheng Y-L. In situ 244 Mitchell SB, Sanders JE. A unique device • Tissue engineering under special aspects
thermoreversible gelation of block and star for controlled electrospinning. J. Biomed. of nanofiber matrices.
copolymers of poly(ethylene glycol) and Mater. Res. Part A 78A(1), 110–120 (2006). 256 Sherwood JK, Riley SL, Palazzolo R et al.
poly(N-isopropylacrylamide) of varying 245 Kim K, Luu YK, Chang C et al. A three-dimensional osteochondral
architectures. Macromolecules 34(11), Incorporation and controlled release of a composite scaffold for articular cartilage
3710–3715 (2001). hydrophilic antibiotic using poly(lactide- repair. Biomaterials 23(24), 4739–4751
232 Ren K, Wang Y, Ji J, Lin Q, Shen J. co-glycolide)-based electrospun (2002).
Construction and deconstruction of nanofibrous scaffolds. J. Controlled Rel. 257 Ishaug-Riley SL, Crane-Kruger GM,
PLL/DNA multilayered films for DNA 98(1), 47–56 (2004). Yaszemski MJ, Mikos AG. Three-
delivery: effect of ionic strength. Colloids and 246 Shin M, Yoshimoto H, Vacanti JP. In vivo dimensional culture of rat calvarial
Surfaces, B: Biointerfaces 46(2), 63–69 (2005). bone tissue engineering using mesenchymal osteoblasts in porous biodegradable
233 Klok H-A, Hwang JJ, Hartgerink JD, Stupp stem cells on a novel electrospun polymers. Biomaterials 19(15),
SI. Self-assembling biomaterials: L-lysine- nanofibrous scaffold. Tissue Eng. 10(1/2), 1405–1412 (1998).
dendron-substituted cholesterylg(L-lactic 33–41 (2004). 258 Liao C-J, Chen C-F, Chen J-H,
acid).hivin.n. Macromolecules 35(16), 247 Dalton PD, Klinkhammer K, Salber J, Chiang S-F, Lin Y-J, Chang K-Y.
6101–6111 (2002). Klee D, Moeller M. Direct in vitro Fabrication of porous biodegradable
234 Kisiday JD, Jin M, DiMicco MA, Kurz B, electrospinning with polymer melts. polymer scaffolds using a solvent
Grodzinsky AJ. Effects of dynamic Biomacromolecules 7(3), 686–690 (2006). merging/particulate leaching method.
compressive loading on chondrocyte J. Biomed. Mater. Res. 59(4), 676–681
248 Yoshimoto H, Shin YM, Terai H,
biosynthesis in self-assembling peptide (2002).
Vacanti JP. A biodegradable nanofiber
scaffolds. J. Biomechanics 37(5), 595–604 scaffold by electrospinning and its potential 259 Ma PX, Choi J-W. Biodegradable polymer
(2004). for bone tissue engineering. Biomaterials scaffolds with well-defined interconnected
235 Couet J, Jeyaprakash JD, Samuel S, Kopyshev 24(12), 2077–2082 (2003). spherical pore network. Tissue Eng. 7(1),
A, Santer S, Biesalski M. Peptide-polymer 23–33 (2001).
249 Luu YK, Kim K, Hsiao BS, Chu B,
hybrid nanotubes. Angewandte Chemie, Hadjiargyrou M. Development of a 260 Shum AWT, Li J, Mak AFT. Fabrication
International Edition 44(21), 3297–3301 nanostructured DNA delivery scaffold via and structural characterization of porous
(2005). electrospinning of PLGA and PLA-PEG biodegradable poly(DL-lactic-co-glycolic
236 Gao X, Matsui H. Peptide-based nanotubes block copolymers. J. Controlled Rel. 89(2), acid) scaffolds with controlled range of
and their applications in bionanotechnology. 341–353 (2003). pore sizes. Polym. Degradation and Stability
Adv. Mat. 17(17), 2037–2050 (2005). 87(3), 487–493 (2005).
250 Li W-J, Mauck RL, Tuan RS. Electrospun
237 Hartgerink JD, Clark TD, Ghadiri MR. nanofibrous scaffolds: Production, 261 Capes JS, Ando HY, Cameron RE.
Peptide nanotubes and beyond. Chemistry – A characterization, and applications for tissue Fabrication of polymeric scaffolds with a
European Journal 4(8), 1367–1372 (1998). engineering and drug delivery. J. Biomed. controlled distribution of pores. J. Mater.
Nanotechnol. 1(3), 259–275 (2005). Sci. Mater. Med. 16(12), 1069–1075
238 Harrington DA, Cheng EY, Guler MO et al.
(2005).
Branched peptide-amphiphiles as self- 251 Pham QP, Sharma U, Mikos AG.
assembling coatings for tissue engineering Electrospinning of polymeric nanofibers for 262 Hou Q, Grijpma DW, Feijen J.
scaffolds. J. Biomed. Mater. Res. Part A, tissue engineering applications: a review. Preparation of interconnected highly
78A(1), 157–167 (2006). Tissue Eng. 12(5), 1197–1211 (2006). porous polymeric structures by a
replication and freeze-drying process.
239 Zhang S, Yan L, Altman M et al. Biological 252 Nair LS, Bhattacharyya S, Laurencin CT.
J. Biomed. Mater. Res. Part B: App. Biomat.
surface engineering: a simple system for cell Development of novel tissue engineering
67B(2), 732–740 (2003).
pattern formation. Biomaterials 20(13), scaffolds via electrospinning. Expert Opin.
1213–1220 (1999). Biol.Therapy 4(5), 659–668 (2004). 263 Wei G, Ma PX. Macroporous and
• Developments in electrospinning (ELSP). nanofibrous polymer scaffolds and
240 Chen CS, Mrksich M, Huang S, Whitesides
polymer/bone-like apatite composite
GM, Ingber DE. Geometric control of cell 253 Li W-J, Laurencin CT, Caterson EJ,
scaffolds generated by sugar spheres.
life and death. Science 276(5317), Tuan RS, Ko FK. Electrospun nanofibrous
J. Biomed. Mater. Res. Part A 78A(2),
1425–1428 (1997). structure: a novel scaffold for tissue
306–315 (2006).
241 Altman M, Lee P, Rich A, Zhang S. engineering. J. Biomed. Mater. Res. 60(4),
613–621 (2002). 264 Draghi L, Resta S, Pirozzolo MG,
Conformational behavior of ionic self-
Tanzi MC. Microspheres leaching for
complementary peptides. Protein Sci. 9(6), 254 Kidoaki S, Kwon Il K, Matsuda T.
scaffold porosity control. J. Mater. Sci.
1095–1105 (2000). Mesoscopic spatial designs of nano- and
Mater. Med. 16(12), 1093–1097 (2005).
242 Hamad-Schifferli K, Schwartz JJ, microfiber meshes for tissue-engineering
matrix and scaffold based on newly devised 265 Wu L, Jing D, Ding J. A room-
Santos AT, Zhang S, Jacobson JM. Remote
multilayering and mixing electrospinning temperature injection molding/particulate
electronic control of DNA hybridization
techniques. Biomaterials 26(1), 37–46 leaching approach for fabrication of
through inductive coupling to an attached
(2005). biodegradable three-dimensional porous
metal nanocrystal antenna. Nature
• Developments in ELSP. scaffolds. Biomaterials 27(2), 185–191
415(6868), 152–155 (2002).
(2005).

850 Expert Rev. Med. Devices 3(6), (2006)


Design and preparation of polymeric scaffolds for tissue engineering

266 Wang H, Pieper J, Peters F, 274 Sun W, Yan Y, Lin F, Spector M. 281 Quirk RA, France RM, Shakesheff KM,
van Blitterswijk CA, Lamme EN. Biomanufacturing: a US-China national Howdle SM. Supercritical fluid technologies
Synthetic scaffold morphology controls science foundation-sponsored workshop. and tissue engineering scaffolds. Curr. Opin.
human dermal connective tissue Tissue Eng. 12(5), 1169–1181 (2006). Solid State Mat. Sci. 8(3–4), 313–321 (2005).
formation. J. Biomed. Mater. Res. Part A 275 Sun W, Starly B, Nam J, Darling A. 282 Rezwan K, Chen QZ, Blaker JJ,
74A(4), 523–532 (2005). Bio-CAD modeling and its applications in Boccaccini AR. Biodegradable and bioactive
267 Lee M, Dunn JCY, Wu BM. Scaffold computer-aided tissue engineering. porous polymer/inorganic composite
fabrication by indirect three-dimensional Computer-Aided Design. 37(11), scaffolds for bone tissue engineering.
printing. Biomaterials 26(20), 4281–4289 1097–1114 (2005). Biomaterials 27(18), 3413–3431 (2006).
(2005). 276 Naing MW, Chua CK, Leong KF, Wang Y. 283 Gross KA, Rodriguez-Lorenzo LM.
268 Vozzi G, Flaim C, Ahluwalia A, Bhatia S. Fabrication of customised scaffolds using Biodegradable composite scaffolds with an
Fabrication of PLGA scaffolds using soft computer-aided design and rapid interconnected spherical network for bone
lithography and microsyringe deposition. prototyping techniques. Rapid Prototyping tissue engineering. Biomaterials 25(20),
Biomaterials 24(14), 2533–2540 (2003). J. 11(4), 249–259 (2005). 4955–4962 (2004).
269 Landis FA, Stephens JS, Cooper JA, 277 Wilson WC Jr, Boland T. Cell and organ
Cicerone MT, Lin-Gibson S. Tissue printing 1: protein and cell printers. In: The Patents
engineering scaffolds based on photocured anatomical record. Part A, Discoveries in 301 Sachs EM, Haggerty JS, Cima MJ,
dimethacrylate polymers for in vitro Molecular, Cellular, And Evolutionary Williams PA. Three-dimensional printing
optical imaging. Biomacromolecules 7(6), Biology. Wiley-Liss, Inc., DE, USA 272(2), techniques. US patent 5204055 (1989).
1751–1757 (2006). 491–496 (2003).
270 Semple JL, Woolridge N, Lumsden CJ. 278 Chua CK, Leong KF, Cheah CM, Chua Affiliations
Review: In vitro, in vivo, in silico: SW. Development of a tissue engineering
• Thomas Weigel
computational systems in tissue scaffold structure library for rapid Head of Department of Polymer Technology,
engineering and regenerative medicine. prototyping. Institute of Polymer Research, GKSS Research
Tissue Eng. 11(3/4), 341–356 (2005). Part 2: Parametric library and assembly Center Geesthacht, Kantstr. 55,
271 Sun W, Lal P. Recent development on program. Int. J. Ad. Manufacturing Tech. D-14513 Teltow, Germany
computer aided tissue engineering – a 21(4), 302–312 (2003). Tel.: +49 332 835 2492
review. Comp. Meth. Prog. Biomed. 67(2), 279 Peters F, Groisman D, Davids R, Haenel T, thomas.weigel@gkss.de
85–103 (2002). Duerr H, Klein M. Comparative study of • Gregor Schinkel
272 Sun W, Starly B, Darling A, Gomez C. patient individual implants from b- Research Associate, Institute of Polymer Research,
Computer-aided tissue engineering: tricalcium phosphate made by different GKSS Research Center Geesthacht,
techniques based on CT data. Department of Polymer Technology, Kantstr. 55,
application to biomimetic modelling and
Materialwissenschaft und Werkstofftechnik D-14513 Teltow, Germany
design of tissue scaffolds. Biotechnol Appl
37(6), 457–461 (2006). Tel.: +49 332 835 2321
Biochem 39(Pt 1), 49–58 (2004).
gregor.schinkel@gkss.de
273 Sun W, Darling A, Starly B, Nam J. 280 Warnke PH, Springer IN, Acil Y et al. The
• Andreas Lendlein
Computer-aided tissue engineering: mechanical integrity of in vivo engineered
Director of Institute, Institute of Polymer
overview, scope and challenges. Biotechnol. heterotopic bone. Biomaterials 27(7),
Research, GKSS Research Center Geesthacht,
Appl. Biochem. 39(Pt 1), 29–47 (2004). 1081–1087 (2006).
Kantstr. 55, D-14513 Teltow, Germany
Tel.: +49 332 835 2450
andreas.lendlein@gkss.de

www.future-drugs.com 851

You might also like