You are on page 1of 14

Advanced Drug Delivery Reviews 65 (2013) 457–470

Contents lists available at SciVerse ScienceDirect

Advanced Drug Delivery Reviews


journal homepage: www.elsevier.com/locate/addr

Silk fibroin biomaterials for tissue regenerations☆


Banani Kundu a, 1, Rangam Rajkhowa b, 1, Subhas C. Kundu a,⁎, Xungai Wang b, c,⁎⁎
a
Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
b
Australian Future Fibres Research and Innovation Centre, Deakin University, Geelong, Victoria3217, Australia
c
School of Textile Science and Engineering, Wuhan Textile University, Wuhan, China

a r t i c l e i n f o a b s t r a c t

Article history: Regeneration of tissues using cells, scaffolds and appropriate growth factors is a key approach in the treat-
Accepted 25 September 2012 ments of tissue or organ failure. Silk protein fibroin can be effectively used as a scaffolding material in
Available online 5 November 2012 these treatments. Silk fibers are obtained from diverse sources such as spiders, silkworms, scorpions, mites
and flies. Among them, silk of silkworms is a good source for the development of biomedical device. It pos-
Keywords:
sesses good biocompatibility, suitable mechanical properties and is produced in bulk in the textile sector.
Silk
Fibroin
The unique combination of elasticity and strength along with mammalian cell compatibility makes silk fibro-
Biomaterials in an attractive material for tissue engineering. The present article discusses the processing of silk fibroin into
Scaffolds different forms of biomaterials followed by their uses in regeneration of different tissues. Applications of silk
Tissue regeneration for engineering of bone, vascular, neural, skin, cartilage, ligaments, tendons, cardiac, ocular, and bladder tis-
sues are discussed. The advantages and limitations of silk systems as scaffolding materials in the context of
biocompatibility, biodegradability and tissue specific requirements are also critically reviewed.
© 2012 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458
2. Silks of silkworms: source, chemistry and structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458
3. Characteristics of silk fibroin as biomaterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
3.1. Mechanical properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
3.2. Biocompatibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
3.3. Biodegradation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
3.4. Water based processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
3.5. Manipulation of silk properties through structural re-adjustments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
4. Morphological diversification of silk biomaterials for tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
4.1. Native silk structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
4.2. Regenerated silk morphologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
4.2.1. Films . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
4.2.2. Electro-spun and wet-spun fibers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 462
4.2.3. Hydrogels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
4.2.4. 3-D porous scaffolds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
4.2.5. Particles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
5. Applications of silk fibroin biomaterials for tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
5.1. Vascular tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
5.2. Neural tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464
5.3. Skin tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464
5.4. Bone tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464
5.5. Cartilage tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Bionics — biologically inspired smart materials”.
⁎ Corresponding author. Tel.:+91 3222 283764; fax: +91 3222 278433.
⁎⁎ Correspondence to: Australian Future Fibres Research and Innovation Centre, Deakin University, Geelong, Victoria3217, Australia. Tel.:+61 613 5227 2894; fax: +61 613 5227
2167.
E-mail addresses: kundu@hijli.iitkgp.ernet.in (S.C. Kundu), xwang@deakin.edu.au (X. Wang).
1
Authors contributed equally.

0169-409X/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.addr.2012.09.043
458 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

5.6. Ligament and tendon tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465


5.7. Cardiac tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.8. Ocular tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.9. Hepatic tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.10. Spinal cord tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.11. Inter-vertebral tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.12. Bladder tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.13. Tracheal tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
5.14. Eardrum tissue regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466
6. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466

1. Introduction This review article is focused on recent research based on silk fi-


broin in the field of tissue regeneration and evaluates its prospects
The limited supply of donors and increasing morbidity have put for further development in therapeutic related applications. The re-
new demands on tissue engineering (TE) as a treatment of organ fail- view starts with a brief overview of silk protein. The silk protein fibro-
ures [1]. The TE approach involves regenerating tissue within suitable in structure and morphologies are included as these aspects are
scaffold with the goal of implanting the constructed tissue at the highly relevant to the applications of silk biomaterials in tissue engi-
target site. The regeneration of functional tissue requires a suitable neering. The different silk based platforms are discussed, followed by
microenvironment that closely mimics the host site for desired an account of their applications in tissue regeneration.
cellular responses [1]. Such an environment is typically provided by
3-D tissue engineering scaffold that acts as an architectural template 2. Silks of silkworms: source, chemistry and structure
[2]. Apart from biocompatibility, which is the essential prerequisite
for any biomaterial, matching the degradation time with that of the Silk proteins are present in glands of silk producing arthropods
tissue regeneration is also a critical requirement for a cell scaffolding (such as silkworms, spiders, scorpions, mites and bees) and spun into
material. Such a match can maintain the mechanical properties and fibers during their metamorphosis. Silkworm's silk is an established
structural integrity of the engineered tissue in all stages of its regen- fiber extensively used in the textile industry. On the other hand, the
eration process. In addition, degraded products of the biomaterial cannibalistic nature of spiders restricts the commercial production of
should be safely metabolized and cleared from the host body. spider silk [5]. Additionally, the yield of fiber from a single silk cocoon
Materials like polymers, metals and ceramics are widely used as is 600–1500 m, compared to only ~137 m from the ampullate gland of
cell scaffolds for tissue engineering. Both synthetic and natural poly- a spider and ~12 m from the spider web [11]. Spider silks are also het-
mers have been trialed, though each has its own limitations. While erogeneous in nature. Therefore, silk based biomaterials are commonly
the former allows easy processing and modifications, the later offers prepared from silkworm silk. Of note is the silk produced by Bombyx
better cyto- and bio-compatibility [3]. There is no universal biomate- mori, a member of the Bombycidae family. B. mori silk is also known
rial that meets the scaffolding requirements for all the tissues. Differ- as mulberry silk. Another silk producing family is Saturniidae and the
ent issue constructs require biomaterials with specific physical, silk is known as non-mulberry silk.
mechanical and degradation properties. Hence there is on-going Silk has several major advantages over other protein based bioma-
search for universal biomaterial for regeneration therapy. terials, which are derived from tissues of allogeneic or xenogeneic or-
Protein, being a component of natural tissues, is a rational choice igins. As such, the risk of infection is high for those materials.
for applications in tissue engineering. Structural proteins such as Processing of such materials is also expensive due to the stringent
collagen, elastin, elastin-like-peptides, albumin and fibrin are used protein isolation and purification protocols. In contrast, silk is an
as sutures, tissue scaffolds, haemostatic and drug delivery agents established textile fiber and nearly 1000 metric tons of silk are pro-
[4]. Silk fibroin of silkworms is a commonly available natural biopoly- duced and processed annually. Silk fiber purification is routinely car-
mer with a long history of applications in the human body as sutures. ried out using a simple alkali or enzyme based degumming
Currently silk sutures are used in lips, eyes, oral surgeries and in the procedure, which yields the starting material for sericin free silk
treatment of skin wounds [5]. Increasingly, silk fibroin is exploited based biomaterials. It is also economically advantageous to use silk
in other areas of biomedical science, as a result of new knowledge for biomedical applications, because of available large scale process-
of its processing and properties like mechanical strength, elasticity, ing infrastructure of traditional silk textile industries.
biocompatibility, and controllable biodegradability [5]. These proper- Silk possesses large molecular weight (200–350 kDa or more) with
ties of silk fibroin are particularly useful for tissue engineering. bulky repetitive modular hydrophobic domains, which are interrupted
Furthermore, recent studies evaluates silk as a part of flexible elec- by small hydrophilic groups [12]. The N and C termini of silk fibroin are
tronic devices for real-time physiological and functional recording highly reserved [5]. Silk fibroin of B. mori is composed of a heavy (H),
and optical systems for diagnosis and treatments [6,7]. Silk and a light (L) chain linked together by a disulfide bond [13]. A
possesses excellent (ca. 95%) optical transparency throughout the vis- 25 kDa glycoprotein, named P25, is also non-covalently linked to
ible range with remarkable surface smoothness and aqueous process- these chains [14]. The hydrophobic domains of H chains contain
ing, all of which facilitates its application in optics and photonics Gly-X (X being Ala, Ser, Thr, Val) repeats and can form anti-parallel
biosensor [7,8]. Such silk based systems are implantable and have β-sheets. The L-chain is hydrophilic in nature and relatively elastic.
necessary functionality and sensitivity required for advanced applica- P25 protein is believed to play significant role in maintaining the integ-
tions. Several reviews are published on the fabrication, structure, and rity of the complex [15,16]. H-fibroin, L-fibroin, and P25 are assembled
the application of silk based biomaterials [2,5,9,10]. In view of grow- in the ratio of 6:6:1 in mulberry silk [17]. Non-mulberry silks lack light
ing applications of silk in new areas of tissue engineering and knowl- (L) chain and P25 [2,16]. Instead, they contain heavy (H) chain
edge on characteristics of silk constructs, a further and more detailed homo-dimers with a molecular weight of ~330 kDa formed by individ-
review is now warranted. ual proteins (~160 kDa) [18]. Non-mulberry Saturniidae silks exhibit a
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 459

Fig. 1. Schematic representation of biomaterials fabricated from silk fibroin: (a) hydrogels; (b) lyophilized powder; (c) 3D porous scaffolds; (d) native silk mat; (e) silk microparticles.

higher Ala/Gly ratio and poly-alanine blocks, which form β-sheets [19]. and the presence of easy accessible chemical groups for functional
They also have higher ratios of basic/acid, polar/non-polar, bulky/ modifications. These advantages are elaborated in the following
non-bulky and hydrophilic/hydrophobic ratios of amino acids [20,21]. sections.
As a result of such variations, there are significant differences in the
mechanical properties, bioactivity and the degradation behavior 3.1. Mechanical properties
between mulberry and non-mulberry silks [22].
Apart from the primary organization, secondary structure and the Silk offers an attractive balance of modulus, breaking strength, and
hierarchical organization of silk fibroin determine many of its bioma- elongation, which contributes to its good toughness and ductility
terial properties. The hydrophobic domains of the silk polymeric (Table 1). Silk fibers are tougher than Kevlar, which is used as a
chains consisting of the repeated amino acid sequence which are as- bench mark in high performance fiber technology [25,32]. The
sembled into nano-crystals (β-sheet). The hydrophilic links between strength-to-density ratio of silk is up to ten times higher than that
these hydrophobic domains consist of bulky and polar side chains and of steel [33]. Spider silk fibers in particular have high extensibility
form the amorphous part of the secondary structure [23,24]. The and exhibit marked strain hardening behavior [34]. Strain hardening
chain conformation in amorphous blocks is random coil, which refers to increase in stress as a fiber is extended in the plastic region
gives elasticity to silk [25,26]. The critical factors that determine the beyond the yield point. Such behavior is particularly important for
mechanical properties of any particular silk are the precise control energy absorbing materials. Stress–strain curves of the wild silkworm
of size, number, distribution, orientation and spatial arrangement of silk fibers exhibit shape and strain hardening similar to that of spider
crystalline and non-crystalline domains at the nanometer scale or dragline silks [22,35]. Considering the good strength and tough-
[5,27]. Nano-crystals contribute to the outstanding mechanical prop- ness of silk fibers, it is no surprise that silk has been exploited to
erties of silk, despite of defects in the microstructure in the form of
vacuoles and micro-voids [23,28]. Apart from the secondary struc- Table 1
ture, a hierarchical supra molecular organization is also evident in Tensile properties of silk polymeric fibers.
silk fibers [24]. Spider and silkworm silks are composed of microfila- Source organisms Tensile strength Tensile modulus Breaking References
ment bundles (0.5–2 μm), each of which is made of nano-crystals (g/den) (g/den) strain (%)
and/or semi crystalline domains [29–31]. Despite some variation in
Bombyx mori 4.3–5.2 84–121 10.0–23.4 [22]
the primary organization and structural features at the nanometer Antheraea mylitta 2.5–4.5 66–70 26–39 [22]
scale between silk types, all silkworm silk fibers follow similar hierar- Philosamia cynthia ricini 1.9–3.5 29–31 28.0–24.0 [22]
chical structural arrangements. Coscinocera hercules 5 ± 1.2 87 ± 17 12.1 ± 5.1 [202]
Hyalophora euryalus 2.7 ± 0.9 59 ± 18 11.1 ± 5.8 [202]
Rothschildia hesperis 3.3 ± 0.8 71 ± 16 9.5 ± 4.4 [202]
3. Characteristics of silk fibroin as biomaterial Eupackardia calleta 2.8 ± 0.7 58 ± 18 11.8 ± 5.5 [202]
Rothschildia lebeau 3.1 ± 0.8 54 ± 14 15.5 ± 6.7 [202]
The major advantage of silk compared to other natural biopolymers Antheraea oculea 3.1 ± 0.8 57 ± 15 14.5 ± 6.6 [202]
is its excellent mechanical property. Other important advantages in- Hyalophora gloveri 2.8 ± 0.4 48 ± 13 19.3 ± 6.9 [202]
Copaxa multifenestrata 0.9 ± 0.2 39 ± 6 4.1 ± 2.7 [202]
clude good biocompatibility, water based processing, biodegradability
460 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

develop scaffolds for load bearing tissue engineering. However, in under good laboratory practices (GLP) shows that silk based Seri Fascia
current design of silk based biomaterials, the wide choice of mechan- surgical mesh meets the biocompatibility requirements [46].
ical properties available from using different silk types is not fully ex- Despite the encouraging results, there still remain some questions
plored. A biomaterial implant fails either due to poor mechanical about long term safety of silk biomaterials in the human body. First,
properties needed for the application, or higher than optimum me- silk sutures remain in the body only for a limited time until removed
chanical properties due to inappropriate stress concentration at the depending on the wound healing period. As silk products for tissue
implant tissue interface. Therefore, variations in mechanical proper- engineering are required to be in contact with tissues for a prolonged
ties of different type of silk provide good choice to match material time period, the long term responses of innate and adaptive immune
properties. In general, the wide choice of extensibility and elasticity system based on location of implant site and type of construct using
of silks, good strength and strain hardening from different varieties appropriate in-vivo models warrant further investigation.
of silk provide outstanding advantage to developing a range of silk Second, there may be concerns on the immune reaction in response
based biomaterials. to degraded products of silk biomaterials, depending on their size and
It is important to note that despite excellent mechanical proper- morphology [35]. It is recognized that one of the major causes of failure
ties of native silk fibers, most of the silk materials developed from of any biomaterial implant is the generation of particulate debris,
silk fibroin solution are weak and brittle. For example, dry tensile which may trigger the immune system. Report shows that fractions of
strength of silk film is about 0.02 GPa and elongation at break is less silk fibers are able to induce mild pro-inflammatory cytokine production
than 2% compared to native fibers that have a tensile strength of and increased phagocytosis [37]. Similarly the digestion of C-terminal of
about 0.5–0.6 GPa and elongation at break 10–40% [22,36]. Such A. pernyi silk with α-chimotrypsin results weak cellular attachment and
difference can be attributed to the lack of appropriate secondary low growth, suggesting degradation reduces cyto-compatibility [47].
and hierarchical structure in the regenerated materials as compared The degraded products of silk fibroin may also cause amyloidogenesis
to the native fibers [37,38]. Recent studies show that there are scopes as reported by Lundmark et al. [48]. Their observation suggests the po-
to significantly improve the strength of regenerated silk products to tentiality of B. mori solution to facilitate the accumulation of amyloid,
the level of native fibers or even higher through manipulation of resulting in tissue degeneration. Hence, long term investigations on de-
structure during regeneration [39–41]. Such investigations are still graded products are necessary in order to fully alleviate any concerns
at the proof of concept stage and relevant mostly to regenerated fi- for the use of silk scaffolds in clinical applications.
bers. More attempts are warranted to improve both strength and
elongation of regenerated silk materials. The prospects of application 3.3. Biodegradation
of silk in a range of tissue engineering applications will improve if
mechanical properties can be tailored to the required level based on The silk biodegradation is studied based on mass loss, change in
application needs. morphology and analysis of degraded products in-vitro. Similarly,
degradation is tested in animal models by testing mechanical proper-
ties of silk after implantation for certain time and studying structural
3.2. Biocompatibility integrity by histological examinations, fluorescent staining and
various biochemical assays (Table 2). As implanted construct, regen-
The long history of success of silk sutures has made silk a known erated silk fibroin biomaterial degrades much faster than fibers. The
biocompatible material [31,32]. But, like any other non-autologous bio- degradation rate is dependent upon the secondary structure of silk
materials causing foreign body response, some adverse immunological resulting from preparation of regenerated silk materials [49].
events associated with silk proteins cannot be ruled out, particularly The term biodegradability is often used to discuss the disintegration
due to its non-mammalian origin. Some incidents of delayed hypersen- of silk materials. According to the definition of Vert et al., biodegrad-
sitivity of silk sutures in rare cases are suggested to be due to the pres- ability is the degradability of an implantable polymer by biological ele-
ence of silk gum-like protein sericin [33,34]. Further studies employing ments giving fragments, which can move away from the site through
isolated silk sericin and sericin based biomaterials have provided no fluid transfer but not necessarily from the body [50]. On the other
clear evidence to suggest sericin as the source of adverse effects [35]. hand, bio-sorption is total elimination of the initial foreign material ei-
Detailed investigations are needed to specifically identify the source ther through filtration or metabolization of the degraded bio-products
of any cyto-toxic non fibroin elements in silk and develop appropriate [50]. Evidence of in-vivo degradability of silk is shown by Wang et al.
diagnostic method. [44]. Their study reveals that, upon implantation in Lewis rats, water
The immunogenicity and antigenicity of silk scaffolds are well tested. based 3-D scaffolds were disintegrated in a few weeks and completely
Silk fibroin bio-conjugates during the treatment of musculo-skeletal dis- disappeared after 1 year. Host immune system has significant impact
ease have shown well tolerated response [42]. There are no signs of infec- on the degradation of 3-D silk fibroin porous scaffolds and degradation
tion during the subcutaneous implantation of electro-spun fiber mats in of silk sponge is shown to be mediated by macrophages, suggesting
rats for up to 8 weeks, although some typical accumulation of phagocytes that silk is not only biodegradable but also bio-resorbable [44].
and lymphocytes is observed. Additionally, there is only minimal inflam- In in-vitro models, protease XIV from Streptomyces griseus is the
mation according to microscopy of hematoxylin and eosin stained tissues most widely used model enzyme for silk [51–55], followed by
[43]. Silk 3-D scaffolds when subcutaneously implanted in Lewis rats gen- α-chymotrypsin from bovine pancreas [52,53,56]. The rate of enzy-
erate very mild immune response at the end of one year; overall expres- matic degradation further can be regulated by gamma-radiation
sion levels for all the genes assigned to immune response including [57]. Degradation of the silk systems is shown to be mediated also
TNF-α IFN-δ, IL-4, IL-6 and IL-13, become undetectable for most type of by cells in-vitro. The osteoblasts and osteoclast cells could erode
silk sponges [44]. A pig model used in ligament tissue engineering silk films via expression of metalloproteinases (MMPs) and integrin
shows no evidence of malfunction after 24 weeks of in-vivo culture [45]. [58]. Such results are encouraging because native extracellular ma-
Overall, these studies give a wide spread acceptance that properly trix is continuously remodeled in-vivo by synchronous proteolytic
degummed and sterilized silk products have good biocompatibility degradation via MMPs and matrix regeneration [59]. The in-vitro
and can be compared with other commonly used biomaterials, such studies are useful to compare the biodegradability among different
as poly (lactic acid) and collagen [42]. As a consequence of extensive silk materials and structures. However, the type and the concentra-
studies in recent years, some silk based materials are received regulato- tion of enzymes at the site of the material implant may vary signif-
ry approval for use in expanded biomaterial devices for plastic and re- icantly. This may put limitations on predicting degradation of silk
constructive surgery. For example, ISO 10993 biocompatibility testing compared to synthetic biomaterials that degrade hydrolytically
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 461

Fig. 2. Confocal laser micrographs showing the attachment and morphology of cells on silk fibroin biomaterials. The cells were stained with rhodamine for the actin filaments and
Hoechst for nucleus. (a) Hepato-carcinoma cells on the porous B. mori silk fibroin scaffold; (b) human osteosarcoma cell line (MG-63) on the surface of silk fibroin; (c) feline em-
bryo fibroblasts (AH 927) on the surface of silk fibroin hydrogel. All the cells revealed well stretched healthy morphology on silk biomaterial surfaces.

and have minimum site-to-site or patient-to-patient variations based solution under mild manufacturing conditions such as room
[4]. temperature, neutral pH and without application of high shear force.
The silk has clear advantages over other biomaterials in several Such conditions are favorably exploited for loading sensitive drugs
aspects of biodegradation. For synthetic biomaterials such as into silk implants [62,63]. Mild processing conditions are also helpful
polyglycolides and polylactides, which are approved by regulatory for photonic or electronic devices or biosensors, which may be incorpo-
authorities, degraded products are resorbed through metabolic rated within a silk based system or coated with silk for improved
pathways but release of acidic by-products is an issue of concern. bio-integration in-vivo. Conformational transition of α-helix and ran-
There are no such issues associated with silk. In addition, those dom coil to highly stable β-sheets is required in silk products to
synthetic materials may decrease mechanical properties very provide good resistance to dissolution, thermal and enzymatic degra-
early during degradation [4,60]. On the other hand, retaining dation. This can be achieved through water vapor annealing, mechani-
strength over a long time by many silk systems can be an advan- cal stretching and ultrasonic treatments, hence avoiding the use of
tage for silk, particularly in tissue engineering, where slow degra- harmful chemicals. These processing advantages and good structural
dation and load bearing capacity are required. Despite such stability make silk a promising polymeric system for bio-related
advantages, deep understanding on degradation and clearing applications.
mechanisms of silk requires further investigations, which would
encourage the development of silk as an important biodegradable
3.5. Manipulation of silk properties through structural re-adjustments
and bio-resorbable material.
Silk structure can be adequately tuned during spinning or regener-
3.4. Water based processing ation to obtain different secondary structures to manipulate material
properties. For example, forced extrusion of silk gland protein
The silk fibroin is water soluble in its α-helical and random coil through silkworm spinnerets attain appropriate altered fiber micro-
forms. Solubility can be maintained over days and even weeks structure with significantly high fiber toughness [64]. Such options,
depending on the storage temperature, pH and concentration of silk if optimized for other forms of silk based materials, may offer the ad-
solution [61]. Hence, silk based systems can be prepared using water vantage of matching their load bearing properties with that of the
462 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

Table 2
Investigatios carried out on biodegradation of silk materials.

Material forms In-vitro/In-vivo Type of study Ref.

B. mori/A. pernyi native fiber/surgical mesh • In-vitro using protease XIV/collagenase• Mass loss, microscopy, secondary structure, mechanical [46,52,203]
IA,/α-chymotrypsin properties of substrate
• Implantation in rats • Particle size, amino acids and molecular weight of
degraded products
• Tissue ingrowth and biomechanical analysis tissues
B.mori 3-D sponge • In-vitro using protease XIV • Mass loss [203,204]
• Intramuscular and subcutaneous • Degradation rate vs. cell metabolism and bone
implantation in rats regeneration from MSC
3-D sponge with different pore size and • Intramuscular and subcutaneous • Structural integrity of sponge and invasion by giant [44]
β-sheet contents implantation in Nude and Lewis rats cell and macrophage based on histology and gene
expression of tissue samples by real time RT-PCR
Knitted silk mesh and silk sponge composite • Anterior cruciate ligament of pig • Macroscopic view, tensile strength, μCT scan and [45]
histology
B. mori cast films and thin crystallized films In-vitro using protease XIV, XXI/ trypsin/ • Mass loss, microscopy and secondary structure [43,53–56,205–209]
α-chymotrypsin/ collagenase of substrate
• Mass spectroscopy to determine molecular weight
of degradation products
• Enzyme degradation vs. drug release
B. mori water annealed patterned silk film • Bone cells mediated degradation • Microscopy and essays on metalloproteinases [58]
production by cells
B. mori electro-spun mat and wet spun fibers • In-vitro using protease XIV, K/actinomyces • Mass loss and microscopy [43,210,211]
• Subcutaneous and intra-muscle • Fluorescein assisted microscopy
implantation in rats
B. mori silk solution • Actinase • Molecular weight and amino acid analysis [212]
B. mori silk particles • Proteases • Particle size, morphological imaging, mass loss, [213,214]
secondary structure, amino acid analysis

targeted tissues. Water annealing is also used to induce insolubility in silk filaments directly in tissue engineering is making a knitted silk struc-
silk products [65,66]. Water annealed films are more flexible and de- ture to reinforce 3-D porous tissue engineering scaffolds. Such reinforce-
graded faster than methanol treated films [65]. Excellent opportuni- ment improves the mechanical properties of scaffolds for applications in
ties are available to use the proticionic liquid system to manipulate load bearing tissue engineering, such as ligament [45,72].
the structure and properties of regenerated silk materials [67]. More-
over, process induced variations in structure and surface topography 4.2. Regenerated silk morphologies
may affect properties such as biodegradation [49], cell interaction
[68] and drug release kinetics [69]. For example, a temperature- To prepare silk solution for regenerating different silk formats, con-
controlled water vapor annealing (TCWVA) technique is used to centrated solutions of chaotropic salts such as LiBr, CaCl2/ethanol/
program the crystallinity in order to tune thermal, mechanical and water, LiSCN [73–75] or ionic liquids [67,76] are commonly used. These
biodegradation properties of the silk films [49]. Similarly, slowing solutions usually fail to dissolve non-mulberry silk fibers [77,78]. At-
down the proteolytic degradation by structural adjustments can reduce tempts using Ca(NO3)2 at 105 °C and LiSCN at 40 °C–55 °C to dissolve
the release kinetics of drugs from a silk system [57]. Although windows Chinese oak tasar silk fiber are reported [79,80]. However as the total dis-
of opportunities are demonstrated in all these cases, more studies are solution in such cases still remains a challenge, gland proteins are used
needed to understand the structure-property relationships for ade- mostly for further processing.
quate control of material properties. Such control will be a key to the
success of silk as a natural biopolymer for tissue regeneration.
4.2.1. Films
4. Morphological diversification of silk biomaterials for Silk fibroin films can be produced by casting the aqueous [81],
tissue regeneration acidic [39,81] and ionic [82] silk solution. Fabrication of silk films by
spin coating and Langmuir-Blodgett (LB) process is also reported
Degumming (removal of glue protein sericin) is the first step in silk [82,83]. Additionally, manual or spin assisted layer by layer deposi-
fiber processing. To produce various other material formats, degummed tion techniques have been used to produce very thin films [84]. As
silk fiber is dissolved to obtain silk solution. In some cases, where it is dif- stability of such cast films is low, techniques like controlled drying
ficult to dissolve fibers, fibroin can be extracted directly from glands of [85], water annealing [49], stretching [86], and alcohol immersion
silkworms using an appropriate buffer solution [70]. Different solid are used to improve β-sheet crystallinity. It is often necessary to con-
forms are then prepared from silk solution by liquid solid phase transfer. trol surface properties of silk films for guided and enhanced cell
Schematics of different silk based biomaterials are represented in Fig. 1. growth or to change the optical properties. Lithography and advanced
Both native fibers and materials regenerated from silk solution are printing systems are employed to achieve such features [8,87].
used for tissue engineering.
4.2.2. Electro-spun and wet-spun fibers
4.1. Native silk structures The large surface area and porous structure of electro-spun silk
nano-fiber mats are useful for cell seeding [86]. 3-D constructs of
Degummed silk fibers can be used to form various twisted structures nano-fibers are used as blood vessel grafts and nerve guides [88,89].
including rope, cable, braided and textured yarns for tissue regeneration Wet spinning [39] or micro-fluidic solution spinning [90] are also
[38]. In addition, cocoons are also used to construct non-woven struc- employed in making regenerated silk fibers. Wet-spun fibers are typ-
tures by partially dissolving them to use as a cell supporting template, ically in the micrometer scale in fiber diameter, and can be produced
where the arrangement of filaments in the cocoon is preserved to main- on a much larger scale than nano-fibers. Advantages of such
tain the porous structure of the mat [37,71]. An alternate way of using regenerated fibers over the native silk fibers include the ability to
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 463

tune fiber morphology and properties based on application, and 24 weeks post implantation period, there is uniform distribution of
incorporation of bio-molecules while regenerating from silk solution. cells throughout the construct [72]. The findings suggest the suitabil-
ity of silk composites for applications where mechanical properties
4.2.3. Hydrogels are important.
Silk hydrogels are formed through sol–gel transition of aqueous
silk fibroin solution in the presence of acids, dehydrating agents, 4.2.5. Particles
ions, sonication or lyophilization [44,91–93]. Sol–gel transition can Silk micro and nano-particles are produced from silk solution by
be accelerated by increasing the protein concentration, temperature, freeze drying and grinding [106], spray drying [107], jet breaking
and addition of Ca 2+[94]. Silk hydrogels can be useful for injectable [108], self-assembly [109,110] and freeze-thawing [111]. Milling silk
or non-injectable delivery systems. Mechanical properties of silk fiber is an alternate approach to make silk particles directly from
hydrogels are found to be suitable for preparing scaffolds for load fibers without using any chemicals [104]. While milled particles are
bearing tissue engineering such as cartilage regeneration [95]. used for reinforcing scaffolds to improve mechanical properties and
cellular outcomes, regenerated silk particles are mostly used in drug
4.2.4. 3-D porous scaffolds carrier applications [104,112–115]. Thus it will be of interest to see
Porous 3-D sponges are ideal structures for tissue engineering if the particles can play the dual role of improving mechanical proper-
scaffolds as they closely mimic the in-vivo physiological micro- ties of scaffolds and at the same time act as a carrier of growth factors
environment. Silk scaffolds are prepared by freeze drying, porogen for rapid tissue regeneration. The challenge here is to make very fine
leaching and solid free form fabrication techniques [96–98]. The and uniform silk particles, which is not easy with the milling
freeze dried sponges possess pore sizes below 100 μm and the pore approach.
sizes can be controlled by adjusting the freezing temperature, pH of
the solution and amount of organic solvents [97]. Repeated freezing 5. Applications of silk fibroin biomaterials for tissue regeneration
and thawing processes can increase pore sizes from 60 to 250 μm
[99]. A better control over pore structure can be obtained from Substitution of a human body part with a biomaterial requires good
solvent casting/particle leaching or gas-foaming methods [98]. Due communication between the host and implanted system for a success-
to good control over porosity and pore sizes, porogen leached 3-D ful outcome. The limitations of existing implants are summarized in
silk scaffolds are commonly used in tissue engineering applications, Table 3. To overcome such limitations, silk based tissue engineering
predominantly bone and cartilage [100]. systems are trialed, which are included in the following sections (cellu-
To obtain good mechanical and biological outcomes, composite lar responses on silk biomaterials are represent in Fig. 2.
silk 3-D scaffolds are prepared by incorporating inorganic [101] or or-
ganic [102] fillers. Usually fillers are incorporated during scaffold fab- 5.1. Vascular tissue regeneration
rication process to ensure their homogeneous distribution. However,
particle addition after fabrication is also reported [101]. The challenge Silk based regenerated vascular tissues are clinically used as flow
in composite design is the compatibility between the components. diverting devices and stents [116,117]. In the case of a study related
Poor compatibility between components results in inhomogeneous to flow-devices, two out of the three patients show promising
mixtures, phase separation and adverse tissue reactions [103]. To en- outcome, suggesting silk as an attractive option to treat fragile
sure good compatibility, silk–silk composite scaffolds are fabricated blood-blister like aneurysms. Silk stents are also employed in the re-
by incorporating milled silk particles in porous silk sponge, resulting construction of an intra-cranial aneurysm artery. There has been a
in significant improvement in compressive modulus from less than successful attempt to fabricate a tubular ~ 3 mm blood vessel from
50 kPa to about 2.2 MPa [104]. Further modifications to scaffolds in- silk with a thickness of 0.15 mm having an average tensile strength
clude reinforcement with fine silk fibers to gain further improvement of 2.42 MPa [118]. The burst strength of silk tubular vessels is found
in modulus to about 13 MPa [105]. Such mechanical properties may to be 811 mm Hg compared to 1800 mm Hg of the gold standard sa-
be sufficient for regenerating cancellous bone, but still way short in phenous veins [119,120]. The implantation of vascular graft of silk fi-
meeting the practical requirements of load bearing bone tissue engi- broin composites of B. mori and transgenic silkworm into rat
neering. To obtain implantable tissue construct without using metal abdominal aorta results excellent patency (ca. 85%) after a year
support, further development is still required in design of scaffolds [121]. Composites of silk fibroin and human-like-collagen or
for bone regeneration. Silk composites reinforced with knitted silk double-raschel knitted silk-poly(ethylene glycol diglycoldiglycidyl
mesh are used as ligament scaffolds. It is revealed that after ether) are successfully used to develop vascular constructs [122].

Table 3
The limitations of presently available commercial products or approaches.

Targeted organ or General practice Limitations References


approaches

Autologous grafts Low quality, vasospasam (spasm of blood vessels) , restricted [215–217]
Homograft length
Conventional prostheses of polytetrafluoroethylene (ePTFE) Fast degeneration
and polyethylene terephthalate Grafts of >5 mm in diameter suffer from thrombosis
Peripheral nerve grafts Autologous nerve grafts Limited graft availability [218–221]
Neurolac®, CultiGuide®, SaluBridge®, Neurotube®, Secondary deformities Dissimilarity in tissue organization and size
Surgisis®, restricted to short defects (b3 cm)repair of the small caliber nerves
NeuroMatrix®–Neuroflex® NeuraGen® Revolnerv® Limited short-term clinical studies and patient follow-up available
till date
Cornea Synthetic keratoprosthesis High host rejection rate [222,223]
Skin Allograft Inadequate availability [224]
Xenografts Limited permanent revascularization
Ligament Autograft Compromises normal healthy tissue and prolonged surgical time [225,226]
Allografts and xenografts Risk of disease transmission and immune rejection
Heart Heart transplantation left ventricular assist devices Scarcity of donors [227]
High cost
464 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

While the former composition is able to provide good tensile strength suggesting that selecting appropriate processing route is also impor-
to the construct, the latter is able to avoid early thrombosis. An tant for silk based biomaterial designing [135]. Other silk composites
aqueous gel spinning process is also employed to design a blood successfully used are nano-fibrous silk-chitin [136], silk-collagen
vessel like tubular structure [123]. [137] and intermolecular cross-linked recombinant human-like colla-
Critical requirements for designing blood vessels include survival gen (RHLC) with fibroin [138]. Additionally, silk fibroin-alginate
under the changes in blood pressure, ability to sustain cyclic loading, blended scaffolds sufficiently favor re-epithelization in a full-
compatibility with the adjacent host vessels, and anti-thrombotic lin- thickness rat wound model [139]. These findings suggest that blend-
ing [124]. Silk fibroin possesses an anti-thrombotic surface with good ed silk fibroin may have better prospect than pure silk fibroin for skin
resistance to high shear stress and blood flow pressure [86,125]. tissue regeneration.
Moreover, silk fibers also support the intercellular contact in endo-
thelial cells [126]. The challenge to include a selection of suitable 5.4. Bone tissue regeneration
cell sources still remains. Primary human endothelial cells and endo-
thelial cell lines (HPMEC-ST1.6R and ISO-HAS-1) seeded in silk fiber Bone is a specialized connective tissue composed of calcified ex-
are unable to fill the gaps between two adjacent silk fibers [127]. tracellular matrix, which has collagen type I and hydroxyapatite as
The optimization of co-culture of endothelial cells and smooth muscle the major components [140]. Thus the scaffolding material for bone
cells to get a well-organized endothelium [89], sulfation of silk for tissues must ensure matrix toughness and matrix deposition. In this
better anti-coagulant activity [128] and coating of matrigel to context, silk fibroin is a rational choice for its high toughness and me-
enhance endothelium coverage [89] are suggestive positive develop- chanical strength along with good bio-compatibility. Silk fibroin
ment towards native vascular tissue construction under current based bone tissue engineering is one of the most extensively studied
situation. tissue engineering approaches to date [10]. Porous fibroin scaffold
based bone constructs are able to stimulate advanced development
5.2. Neural tissue regeneration of bone tissues within 5 weeks, when engineered within a bioreactor
[100]. Silk fibroin scaffolds also promote human mesenchymal stem
The human nervous system is broadly classified into (a) the cen- cell based healing of femoral defects in nude mice [141]. Furthermore,
tral nervous system (CNS) and (b) the peripheral nervous system the loading of human mesenchymal stem cells within silk fibroin-
(PNS). The PNS is capable of self-healing minor injuries, while large polyethylene oxide nano-fibrous composite scaffold with bone
damage needs to be treated surgically with nerve grafts harvested morphogenic protein-2 led to regeneration of bone like tissue [142].
elsewhere in the body. Therefore, in such treatments, tissue engineer- Incorporation of hydroxyapatite nano-particles into silk matrix im-
ing is highly relevant and the compatibility of scaffolding material proves bone regeneration in animals [143,144]. The incorporation of
with neuro-progenitor cells assumes significance. Silk fibroin n-Hap within the fibroin sheet and subsequent culture of rat bone
supports the viability of dorsal root ganglia and schwann cells marrow mesenchymal stem cell (BM-MSCs) demonstrates the cell
without affecting their normal phenotype or functionality [129]. supportive nature of embedded Hap crystals and successful osteogen-
Composites of silk fibroin with chitosan or poly(l-lactic acid-co- ic differentiation of BMMSCs [145]. These composite scaffolds
caprolactone) are able to bridge a gap of a 10 mm long sciatic nerve completely heal the segmental bone defect in Sprague–Dawley rats
defects in rats [130–132]. Furthermore, an assembly of B. mori silk after 12 weeks post implantation [143]. Silk fibroin also guides
fibroin and Spider X® fiber (a spider silk like fiber) is effectively full-thickness growth of calvarial defect (8 mm-diameters) in rats
able to bridge up to 13 mm nerve gap within 12 weeks [133]. The within 12 week [100]. Similar results are also obtained when silk hy-
micro-architecture of scaffolds play important role in the regenera- drogel [92,146] or non-woven mats [147] are used. The strategies to
tion of nerves. Improved length and axonal outgrowth of chicken em- improve bone regeneration using silk-silk composites are already
bryo dorsal root ganglion sensory neurons and spiral cord motor discussed in the Section 4.2.4.
neurons are observed on aligned silk nano-fibers, compared to slow The vascularization of in-vitro tissue models is always a problem.
and random orientation of axonal growth on non-aligend fibers The pre-incubation of silk matrices with cells before implantation in
[134]. The orientation of pores and addition of neurotrophic factors animals increases the vascularization in neo-implant [148]. Under
for enhanced neuronal growth are being presently investigated to en- such condition, the rate of vascularization is directly proportional to
hance the outcome of silk based nerve grafts. in-vitro incubation time. Co-culture of osteoblasts with endothelial
cells on silk scaffolds results in the formation of micro-capillary
5.3. Skin tissue regeneration [149] and pre-vascular structures [150]. Furthermore, upon implanta-
tion, these pre-mature micro-capillaries survive the host defense
The skin is the biggest organ in human and acts as barrier to the system and become functional micro-capillaries [151]. Further inves-
infectious organisms. It has limited self-healing capability. Large dam- tigations on scaffold architectures and pore distribution are needed to
age to the skin causes loss of skin integrity, which may lead to death. obtain the completely vascularized 3-D bone tissue.
The adult human skin consists of two major layers: the epidermis (a
keratinized layer) and the dermis (collagen-rich layer). Appendages 5.5. Cartilage tissue regeneration
like hair and hormonal glands are raised from the epidermis but root-
ed deeply within the dermis. This complex structure makes the engi- Cartilage is a non-vascular, non-innervated connective tissue. The
neering of skin a challenging task. Silk fibroin supports well the 3-D porous fibroin scaffolds [152–155], electro-spun silk fibers treat-
human keratinocytes and fibroblasts [86]. However the complex ed with micro-wave induced argon plasma [156], silk fibroin blended
structure of native tissue requires a composite scaffolding material. with chitosan [157] or genipin cross-linked chitosan-fibroin porous
Bio-mimicking approaches with other natural extra-cellular materials sponges [158] provide adequate support to chondrocytes. Non-
are proposed. Layering silk fibroin with collagen-I enhances the at- mulberry gland silk fibroin of A. mylitta [159] also acts as promising
tachment and dispersion of keratinocytes, while fibronectin coating material for cartilage development. Insulin-like growth factor 1
supports both keratinocytes and fibroblasts cells adhesion and disper- (IGF-I) is a regulatory molecule in chondrogenesis [160], thus can
sion within the matrix [135]. Structural integrity of silk matrices in- be incorporated within scaffolds for better chondrogenic outcomes
duced by different treatments also influences greatly the cell [160]. Bioreactors provide mechanical stimulation and maturation of
adherence. Both human keratinocytes and fibroblasts prefer water cartilaginous constructs [161]. Therefore, the hydrodynamic factors
vapor-treated silk nano-fibrous mats over methanol-treated ones, are important in chondrogenic outcome. Other factors to be taken
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 465

into serious consideration for regenerating the cartilaginous tissues sulfate–laminin mixtures enhances the performance of fibroin [175].
are cell sources [162], scaffold architectures, pore sizes and pore Successful pre-clinical reconstruction of human limbal epithelium
distribution. on silk membranes is also reported [176].

5.6. Ligament and tendon tissue regeneration 5.9. Hepatic tissue regeneration

The engineering of ligaments and tendons requires scaffolding Liver plays crucial role in the metabolism of carbohydrates, pro-
material with excellent combination of mechanical strength, elastici- teins, lipids and vitamins. The chief cellular component of hepatic tis-
ty, toughness and structural integrity. The first successful silk based sue is hepatocytes, which is employed in-vitro to reconstruct the 3-D
engineering of anterior cruciate ligament (ACL) employs twisted scaf- tissue or liver patches. The silk composites for hepatic tissue engi-
fold, the mechanical property of which is comparable to human ACL neering includes functionalized silk with lactose and cyanuric acid
[38]. Synergistic incorporation of silk fibers within the collagen [177], silk fibroin blended films with collagen [178], recombinant
matrix [163], coating of silk fibroin on electro-spun poly-lactic- human-like collagen [179], silk fibroin-collagen-heparin scaffolds
co-glycolic acid (PLGA) nano-fibers [164], loading basic fibroblast [180], and silk micro-particle embedded PLA scaffolds [181]. Blending
growth factor (bFGF) and transforming growth factor-β (TGF-β) in of silk fibroin with collagen causes mild inflammation in the omen-
optimal regimen within the scaffold stimulates the biochemical and tum of rats [182]. Furthermore, non-mulberry A. assama fibroin
mechanical pathways for ligament tissue regeneration [165]. based multi-scale scaffolds provide adequate mechanical support
Further approach to design scaffold-free ligament analogs in- and help cellular response [183]. Functionalization of A. assama fibro-
volves incorporation of porcine bone marrow MSCs on laminin coated in with sulfone results enhanced blood-compatibility [184]. However,
silk sutures as anchor points [166]. As a result, the cells within the as hepatocytes carry a degree of structural organization, it forms large
construct are self-organized into cohesive rod-like tissues, which cellular aggregates in long term in-vitro culture. These complex cellu-
mechanically and histologically resemble the native ligament. Silk fi- lar aggregates make it difficult for nutrient diffusion and thus require
broin scaffold of A. pernyi is also able to repair defects in Achilles ten- further investigation in designing silk scaffold for complete tissue
don of an adult New Zealand white rabbit [167]. The recovery of regeneration.
Achilles tendon in the rabbit shows the feasibility of using
non-mulberry silks along with mulberry silk as suitable tendon 5.10. Spinal cord tissue regeneration
scaffolds.
The grafting of olfactory ensheathing cells (OECs) is one of the
5.7. Cardiac tissue regeneration most commonly employed approaches to treat spinal cord injuries.
Silk based spinal cord regeneration is presently at its neo-natal
The loss of cardiomyocytes after injury reduces cardiac function, stage. The culture of OECs on nano-fibrous silk fibroin reveals pros-
which further leads to morbidity and mortality. A possible treatment pects of silk biomaterials in this area [185]. The diameter of the
is engineering an artificial heart or cardiac patch in-vitro followed by nano-fibers possesses regulatory effects on growth of OECs [186];
implantation. Chitosan or hyaluronic acid (HA) loaded silk fibroin smaller diameters result in better cellular responses than the larger
seeded with rat mesenchymal stem cells is able to generate cardiac fibers.
patches [168]. Silk protein fibroin 3-D scaffolds of A. mylitta also
show good outcome without the employment of other extra cellular 5.11. Inter-vertebral tissue regeneration
matrix materials, producing beating patches of rat cardio-myocytes
in-vitro [169]. Critical issues still to be resolved are suitable silk or The treatment of degenerative disk disease involves repair of an-
silk based composite biomaterials supporting the mechanical nulus fibrosus, which is one of the major components of inter-
strength of the heart valves, formation of elastin by employing vertebral disk. Porous scaffolds of silk fibroin allow good growth of
co-culture system with fibroblasts cells, employment of human se- bovine annulus fibrosus cells up to 8 weeks in-vitro [187]. The growth
rums or platelet lysates during in-vitro culture to minimize the tissue of bovine annulus fibrosus cells on silk fibroin is greatly influenced by
engineered graft rejection. culture condition and average pore sizes of the scaffolding material
(≥600 μm) [188]. However, further investigations are required to
5.8. Ocular tissue regeneration completely mimic the high strength, elastic and tough morphology
of naive inter-vertebral tissues.
Allograft approach of corneal tissue regeneration suffers from
bio-burden [170], while the use of silk fibroin results in a much better 5.12. Bladder tissue regeneration
outcome. Optical transparency of silk fibroin films and stability of
aqueous protein solution at neutral pH are the key characteristics In the treatment of stress urinary incontinence, bladder shaped
that favor silk for functional bio-photonic applications over other scaffolds with autologous urothelial and smooth muscle cells are re-
synthetic or inorganic materials [6]. Silk films are stacked into a 3-D quired for successful reconstruction of the bladder [189]. Films of
porous structure mimicking closely the heli-coidal organization of silk fibroin provide good support to transitional epithelial cells of uri-
cornea in-vivo. When these 3-D structures are cultured with human nary bladders of New Zealand rabbits [190]. Silk films in rabbits result
and rabbit corneal fibroblasts, cells reveal natural corneal keratocyte in the successful repair of short-length defects of 1.5 cm [191]. Fur-
morphology [171]. Silk fibroin implants with embedded rabbit cornea ther animal studies include mice by Cannon et al., [192] and murine
epithelial cells become translucent within 4 weeks and results in the by Mauney et al. [193]. The silk based sling engineered with bone
formation of new limbus and blood vessels within 8 weeks marrow mesenchymal stem cells exhibits good control on the
post-implantation. Complete regeneration of rabbit cornea occurs leak-point pressure comparable to negative control [194]. This
within 16 weeks leaving behind a few opaque pieces of degraded suggests a hopeful treatment for stress urinary incontinence.
scaffolds [172]. In addition, the affinity of corneal cells towards a pat-
terned surface [173] and ease in surface patterning in silk films 5.13. Tracheal tissue regeneration
(discussed in detail in another review elsewhere [174]), open up fur-
ther scopes for silk-biomaterials in ocular regenerative medicine. The incidence of pre-mature newborns developing tracheal steno-
Coating of fibroin with collagen IV, fibronectin, and chondroitin sis is on the rise as a result of prolonged incubation. In silk based
466 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

reconstruction of 12 tracheal defects in rabbits, successful generation [10] N. Kasoju, U. Bora, Silk fibroin in tissue engineering, Adv. Healthc. Mater. 1
(2012) 393–412.
of full-thickness fibroblasts layer of 240–302 μm is observed, without [11] R. Lewis, Unraveling the weave of spider silk, Bioscience 46 (1996) 636–638.
foreign-body granuloma and macro-phagocyte infiltration [195]. [12] N.A. Ayoub, J.E. Garb, R.M. Tinghitella, M.A. Collin, C.Y. Hayashi, Blueprint for a
Blending of silk fibroin with chitosan enhances perichondrium high-performance biomaterial: Full-length spider dragline silk genes, PLoS One
2 (2007) e514.
wrapping in nude mice, suggesting the suitability of silk based de- [13] K. Shimura, A. Kikuchi, K. Ohtomo, Y. Katagata, A. Hyodo, Studies on silk fibroin
vices in generation of epithelial lining for tracheal transplants [196]. of bombyx mori. I. Fractionation of fibroin prepared from the posterior silk
gland, J. Biochem. 80 (1976) 693–702.
[14] K. Tanaka, S. Inoue, S. Mizuno, Hydrophobic interaction of P25, containing
5.14. Eardrum tissue regeneration Asn-linked oligosaccharide chains, with the H-L complex of silk fibroin
produced by Bombyx mori, Insect Biochem. Mol. Biol. 29 (1999) 269–276.
[15] C.-Z. Zhou, F. Confalonieri, M. Jacquet, R. Perasso, Z.-G. Li, J. Janin, Silk fibroin:
Eardrum (tympanic membrane) perforation results pain, infection
structural implications of a remarkable amino acid sequence, Proteins Struct.
and even loss of hearing. Surgical treatment to restore chronic Funct. Bioinf. 44 (2001) 119–122.
perforations is myringoplasty, where autologous grafts, allografts, [16] F. Sehnal, M. Žurovec, Construction of silk fiber core in lepidoptera,
and synthetic graft materials are commonly used [197]. Recent at- Biomacromolecules 5 (2004) 666–674.
[17] S. Inoue, K. Tanaka, F. Arisaka, S. Kimura, K. Ohtomo, S. Mizuno, Silk fibroin of
tempts with silk fibroin membrane have resulted good adhesion Bombyx mori is secreted, assembling a high molecular mass elementary unit
and growth kinetics of human tympanic membrane keratinocytes consisting of H-chain, L-chain, and P25, with a 6:6:1 molar ratio, J. Biol. Chem.
[198–200]. Silk membranes provide better healing compared to 275 (2000) 40517–40528.
[18] S.-i. Inoue, H. Tsuda, T. Tanaka, M. Kobayashi, Y. Magoshi, J. Magoshi, Nanostruc-
conventional paper patch [201]. These findings suggest suitability to ture of natural fibrous protein: In vitro nanofabric formation of Samia cynthia
engineer silk based eardrum patches. ricini wild silk fibroin by self-assembling, Nano Lett. 3 (2003) 1329–1332.
[19] F. Lucas, J.T.B. Shaw, S.G. Smith, Comparative studies of fibroins: I. The amino
acid composition of various fibroins and its significance in relation to their
6. Future prospects crystal structure and taxonomy, J. Mol. Biol. 2 (1960) 339–349.
[20] G. Freddi, Y. Gotoh, T. Mori, I. Tsutsui, M. Tsukada, Chemical structure and
physical properties of Antheraea assama silk, J. Appl. Polym. Sci. 52 (1994)
Tissue regeneration for therapeutics is very critical and target
775–781.
specific. To complement the functionality of living systems, the [21] K. Sen, M.K. Babu, Studies on Indian silk. I. Macrocharacterization and analysis of
constructed tissue must successfully interact with the body's immune amino acid composition, J. Appl. Polym. Sci. 92 (2004) 1080–1097.
[22] R. Rajkhowa, V.B. Gupta, V.K. Kothari, Tensile stress–strain and recovery behav-
system. Silk based designs allow easy control on matrix morphology,
ior of Indian silk fibers and their structural dependence, J. Appl. Polym. Sci. 77
degradation rate and conformal adhesion to underlying tissues with (2000) 2418–2429.
low immune-toxicity and good biocompatibility. Recent advance- [23] F. Vollrath, D. Porter, Spider silk as a model biomaterial, Appl. Phys. A: Mater. Sci.
ments in understanding silk structure and processing open up new Process. 82 (2006) 205–212.
[24] T. Lefèvre, M.-E. Rousseau, M. Pézolet, Protein secondary structure and orienta-
opportunities in the use of various forms of silk in tissue regeneration. tion in silk as revealed by raman spectromicroscopy, Biophys. J. 92 (2007)
Silk systems will be particularly useful for applications where slow 2885–2895.
biodegradation and good mechanical properties are critically re- [25] F. Vollrath, D.P. Knight, Liquid crystalline spinning of spider silk, Nature 410
(2001) 541–548.
quired, such as for bone, ligament and muco-skeletal tissues. Success- [26] F. Vollrath, Strength and structure of spiders' silks, J. Biotechnol. 74 (2000)
ful applications of silk based materials in tissue engineering depends 67–83.
on further understanding of the long term biocompatibility, biode- [27] S. Keten, Z. Xu, B. Ihle, M.J. Buehler, Nanoconfinement controls stiffness, strength
and mechanical toughness of [beta]-sheet crystals in silk, Nat. Mater. 9 (2010)
gradability and its degraded products, along with the ability to tune 359–367.
silk morphologies for tissue specific requirements. Hybrid materials, [28] Frische, Maunsbach, Vollrath, Elongate cavities and skin–core structure in
incorporating 100% silk in different matrix morphologies show Nephila spider silk observed by electron microscopy, J. Microsc. 189 (1998)
64–70.
promising results in this regard.
[29] S. Putthanarat, N. Stribeck, S.A. Fossey, R.K. Eby, W.W. Adams, Investigation of
the nanofibrils of silk fibers, Polymer 41 (2000) 7735–7747.
Acknowledgments [30] P. Poza, J. Pérez-Rigueiro, M. Elices, J. Llorca, Fractographic analysis of silkworm
and spider silk, Eng. Fract. Mech. 69 (2002) 1035–1048.
[31] H. Akai, T. Nagashima, S. Aoyagi, Ultrastructure of posterior silk gland cells and
The work is financially supported by the Department of Biotech- liquid silk in Indian tasar silkworm, Antheraea mylitta drury (Lepidoptera:
nology and its Bioinformatics Facility, Department of Science and Saturniidae), Int. J. Insect Morphol. Embryol. 22 (1993) 497–506.
[32] F. Vollrath, Spiders' webs, Curr. Biol. (2005) R364–R365.
Technology (SCK), and Council of Scientific Industrial Research (for [33] T. Giesa, M. Arslan, N.M. Pugno, M.J. Buehler, Nanoconfinement of spider silk
awarding Senior Research Fellowship to B. Kundu), Government of fibrils begets superior strength, extensibility, and toughness, Nano Lett. 11
India, New Delhi. R. Rajkhowa and X. Wang acknowledge the support (2011) 5038–5046.
[34] N. Du, Z. Yang, X.Y. Liu, Y. Li, H.Y. Xu, Structural origin of strain-hardening of
from the Australian Research Council for their silk related work. The spider silk, Adv. Funct. Mater. 21 (2011) 772–778.
authors are thankful to Dr. Tuli Dey, Sunita Nayak and Deboki Naskar [35] Y. Zhang, H. Yang, H. Shao, X. Hu, Antheraea pernyi silk fiber: a potential resource
for providing the photograph of bone cells on silk. for artificially biospinning spider dragline silk, J. Biomed. Biotechnol. (2010)
(2010).
[36] R. Rajkhowa, B. Levin, S.L. Redmond, L. Wang, J.R. kanwar, M.D. Atlas, X. Wang,
References Structure and properties of biomedical films prepared from aqueous and acidic
silk fibroin solutions, J. Biomed. Mater. Res. Part A 97A (2011) 37–45.
[1] R. Langer, J. Vacanti, Tissue engineering, Science 260 (1993) 920–926. [37] K. Gellynck, P. Verdonk, E. Van Nimmen, K. Almqvist, T. Gheysens, G. Schoukens,
[2] B. Kundu, S.C. Kundu, Osteogenesis of human stem cells in silk biomaterial for L. Van Langenhove, P. Kiekens, J. Mertens, G. Verbruggen, Silkworm and spider
regenerative therapy, Prog. Polym. Sci. 35 (2010) 1116–1127. silk scaffolds for chondrocyte support, J. Mater. Sci. Mater. Med. 19 (2008)
[3] B.L. Seal, T.C. Otero, A. Panitch, Polymeric biomaterials for tissue and organ 3399–3409.
regeneration, Mater. Sci. Eng. R 34 (2001) 147–230. [38] G.H. Altman, R.L. Horan, H.H. Lu, J. Moreau, I. Martin, J.C. Richmond, D.L. Kaplan,
[4] L.S. Nair, C.T. Laurencin, Biodegradable polymers as biomaterials, Prog. Polym. Silk matrix for tissue engineered anterior cruciate ligaments, Biomaterials 23
Sci. 32 (2007) 762–798. (2002) 4131–4141.
[5] F.G. Omenetto, D.L. Kaplan, New opportunities for an ancient material, Science [39] S.W. Ha, A.E. Tonelli, S.M. Hudson, Structural studies of Bombyx mori silk fibroin
329 (2010) 528–531. during regeneration from solutions and wet fiber spinning, Biomacromolecules
[6] F.G. Omenetto, D.L. Kaplan, A new route for silk, Nat. Photonics 2 (2008) 6 (2005) 1722–1731.
641–643. [40] B. Zuo, L. Dai, Z. Wu, Analysis of structure and properties of biodegradable
[7] M.K. Hota, M.K. Bera, B. Kundu, S.C. Kundu, C.K. Maiti, A natural silk fibroin regenerated silk fibroin fibers, J. Mater. Sci. 41 (2006) 3357–3361.
protein-based transparent bio-memristor, Adv. Funct. Mater. 22 (2012) 4493–4499. [41] C. Jiang, X. Wang, R. Gunawidjaja, Y.-H. Lin, M.K. Gupta, D.L. Kaplan, R.R. Naik,
[8] H. Tao, D.L. Kaplan, F.G. Omenetto, Silk Materials — a road to sustainable high V.V. Tsukruk, Mechanical properties of robust ultrathin silk fibroin films, Adv.
technology, Adv. Mater. 24 (2012) 2824–2837. Funct. Mater. 17 (2007) 2229–2237.
[9] G.H. Altman, F. Diaz, C. Jakuba, T. Calabro, R.L. Horan, J. Chen, H. Lu, J. Richmond, [42] L. Meinel, D.L. Kaplan, Silk constructs for delivery of musculoskeletal therapeu-
D.L. Kaplan, Silk-based biomaterials, Biomaterials 24 (2003) 401–416. tics, Adv. Drug Deliv. Rev. 64 (2012) 1111–1122.
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 467

[43] J.A. Zhou, C.B. Cao, X.L. Ma, L. Hu, L.A. Chen, C.R. Wang, In vitro and in vivo deg- [74] C. Acharya, B. Hinz, S.C. Kundu, The effect of lactose-conjugated silk biomate-
radation behavior of aqueous-derived electrospun silk fibroin scaffolds, Polym. rials on the development of fibrogenic fibroblast, Biomaterials 29 (2008)
Degrad. Stab. 95 (2010) 1679–1685. 4665–4675.
[44] Y. Wang, D.D. Rudym, A. Walsh, L. Abrahamsen, H.-J. Kim, H.S. Kim, C. [75] N.V. Bhat, S.M. Ahirrao, Investigation of the structure of silk film regenerated
Kirker-Head, D.L. Kaplan, In vivo degradation of three-dimensional silk fibroin with lithium thiocyanate solution, J. Polym. Sci. Part A: Polym. Chem. 21
scaffolds, Biomaterials 29 (2008) 3415–3428. (1983) 1273–1280.
[45] H. Fan, H. Liu, S.L. Toh, J.C.H. Goh, Anterior cruciate ligament regeneration using [76] D.M. Phillips, L.F. Drummy, R.R. Naik, H.C.D. Long, D.M. Fox, P.C. Trulove, R.A.
mesenchymal stem cells and silk scaffold in large animal model, Biomaterials 30 Mantz, Regenerated silk fiber wet spinning from an ionic liquid solution,
(2009) 4967–4977. J. Mater. Chem. 15 (2005) 4206–4208.
[46] R. Horan, D. Bramono, J. Stanley, Q. Simmons, J. Chen, H. Boepple, G. Altman, [77] W. Tao, M. Li, C. Zhao, Structure and properties of regenerated Antheraea pernyi
Biological and biomechanical assessment of a long-term bioresorbable silk fibroin in aqueous solution, Int. J. Biol. Macromol. 40 (2007) 472–478.
silk-derived surgical mesh in an abdominal body wall defect model, Hernia 13 [78] H. Kweon, Y.H. Park, Dissolution and characterization of regenerated Antheraea
(2009) 189–199. pernyi silk fibroin, J. Appl. Polym. Sci. 82 (2001) 750–758.
[47] N. Minoura, S.I. Aiba, M. Higuchi, Y. Gotoh, M. Tsukada, Y. Imai, Attachment and [79] H.Y. Kweon, Y.H. Park, Structural and conformational changes of regenerated
growth of fibroblast cells on silk fibroin, Biochem. Biophys. Res. Commun. 208 Antheraea pernyi silk fibroin films treated with methanol solution, J. Appl.
(1995) 511–516. Polym. Sci. 73 (1999) 2887–2894.
[48] K. Lundmark, G.T. Westermark, A. Olsén, P. Westermark, Protein fibrils in nature [80] M. Li, W. Tao, S. Lu, C. Zhao, Porous 3-D scaffolds from regenerated Antheraea
can enhance amyloid protein A amyloidosis in mice: cross-seeding as a disease pernyi silk fibroin, Polym. Adv. Technol. 19 (2008) 207–212.
mechanism, Proc. Natl. Acad. Sci. 102 (2005) 6098–6102. [81] I.C. Um, H. Kweon, Y.H. Park, S. Hudson, Structural characteristics and properties
[49] Y. Hu, Q. Zhang, R. You, L. Wang, M. Li, The relationship between secondary of the regenerated silk fibroin prepared from formic acid, Int. J. Biol. Macromol.
structure and biodegradation behavior of silk fibroin scaffolds, Adv. Mater. Sci. 29 (2001) 91–97.
Eng. (2012), http://dx.doi.org/10.1155/2012/185905. [82] M.K. Gupta, S.K. Khokhar, D.M. Phillips, L.A. Sowards, L.F. Drummy, M.P. Kadakia,
[50] M. Vert, S.M. Li, G. Spenlehauer, P. Guerin, Bioresorbability and biocompatibility R.R. Naik, Patterned silk films cast from ionic liquid solubilized fibroin as
of aliphatic polyesters, J. Mater. Sci. Mater. Med. 3 (1992) 432–446. scaffolds for cell growth, Langmuir 23 (2006) 1315–1319.
[51] E.M. Pritchard, T. Valentin, D. Boison, D.L. Kaplan, Incorporation of proteinase in- [83] A. Higuchi, M. Yoshida, T. Ohno, T. Asakura, M. Hara, Production of interferon-β
hibitors into silk-based delivery devices for enhanced control of degradation in a culture of fibroblast cells on some polymeric films, Cytotechnology 34
and drug release, Biomaterials 32 (2011) 909–918. (2000) 165–173.
[52] R.L. Horan, K. Antle, A.L. Collette, Y. Wang, J. Huang, J.E. Moreau, V. Volloch, D.L. [84] X. Wang, H.J. Kim, P. Xu, A. Matsumoto, D.L. Kaplan, Biomaterial coatings by
Kaplan, G.H. Altman, In vitro degradation of silk fibroin, Biomaterials 26 (2005) stepwise deposition of silk fibroin, Langmuir 21 (2005) 11335–11341.
3385–3393. [85] Q. Lu, X. Hu, X. Wang, J.A. Kluge, S. Lu, P. Cebe, D.L. Kaplan, Water-insoluble silk
[53] M. Li, M. Ogiso, N. Minoura, Enzymatic degradation behavior of porous silk films with silk I structure, Acta Biomater. 6 (2010) 1380–1387.
fibroin sheets, Biomaterials 24 (2003) 357–365. [86] X. Zhang, M.R. Reagan, D.L. Kaplan, Electrospun silk biomaterial scaffolds for
[54] Q. Lu, B. Zhang, M. Li, B. Zuo, D.L. Kaplan, Y. Huang, H. Zhu, Degradation mechanism regenerative medicine, Adv. Drug Deliv. Rev. 61 (2009) 988–1006.
and control of silk fibroin, Biomacromolecules 12 (2011) 1080–1086. [87] B.D. Lawrence, Z. Pan, M.D. Weber, D.L. Kaplan, M.I. Rosenblatt, Silk film
[55] K. Numata, P. Cebe, D.L. Kaplan, Mechanism of enzymatic degradation of culture system for in vitro analysis and biomaterial Design, J. Vis. Exp. (2012)
beta-sheet crystals, Biomaterials 31 (2010) 2926–2933. 3646.
[56] T. Arai, G. Freddi, R. Innocenti, M. Tsukada, Biodegradation of Bombyx mori silk [88] M.L. Lovett, C.M. Cannizzaro, G. Vunjak-Novakovic, D.L. Kaplan, Gel spinning of
fibroin fibers and films, J. Appl. Polym. Sci. 91 (2004) 2383–2390. silk tubes for tissue engineering, Biomaterials 29 (2008) 4650–4657.
[57] J.N. Wang, Z.-W. Liu, Y.-X. Yang, H.-Y. Huang, Enzymatic degradation behavior of [89] X. Zhang, X. Wang, V. Keshav, X. Wang, J.T. Johanas, G.G. Leisk, D.L. Kaplan,
silk fibroin fiber treated by y-ray irradiation, Textile Res. J. 82 (2012) 1799–1805. Dynamic culture conditions to generate silk-based tissue-engineered vascular
[58] S. Sengupta, S.-H. Park, G.E. Seok, A. Patel, K. Numata, C.-L. Lu, D.L. Kaplan, Quan- grafts, Biomaterials 30 (2009) 3213–3223.
tifying osteogenic cell degradation of silk biomaterials, Biomacromolecules 11 [90] M.E. Kinahan, E. Filippidi, S. Koster, X. Hu, H.M. Evans, T. Pfohl, D.L. Kaplan, J.
(2010) 3592–3599. Wong, Tunable silk: Using microfluidics to fabricate silk fibers with controllable
[59] W.P. Daley, S.B. Peters, M. Larsen, Extracellular matrix dynamics in development properties, Biomacromolecules 12 (2011) 1504–1511.
and regenerative medicine, J. Cell Sci. 121 (2008) 255–264. [91] X. Chen, W. Li, W. Zhong, Y. Lu, T. Yu, pH sensitivity and ion sensitivity of
[60] P.A. Gunatillake, R. Adhikari, Biodegradable synthetic polymer for tissue hydrogels based on complex-forming chitosan/silk fibroin interpenetrating
engineering, Eur. Cells Mater. 5 (2003) 1–16. polymer network, J. Appl. Polym. Sci. 65 (1997) 2257–2262.
[61] T.T. Le Zainuddin, Y. Park, T.V. Chirila, P.J. Halley, A.K. Whittaker, The behavior of [92] A. Motta, C. Migliaresi, F. Faccioni, P. Torricelli, M. Fini, R. Giardino, Fibroin
aged regenerated Bombyx mori silk fibroin solutions studied by 1H NMR and hydrogels for biomedical applications: preparation, characterization and in
rheology, Biomaterials 29 (2008) 4268–4274. vitro cell culture studies, J. Biomater. Sci. Polym. Ed. 15 (2004) 851–864.
[62] V. Karageorgiou, L. Meinel, S. Hofmann, A. Malhotra, V. Volloch, D. Kaplan, Bone [93] N. Guziewicz, A. Best, B. Perez-Ramirez, D.L. Kaplan, Lyophilized silk fibroin
morphogenetic protein-2 decorated silk fibroin films induce osteogenic differ- hydrogels for the sustained local delivery of therapeutic monoclonal antibodies,
entiation of human bone marrow stromal cells, J. Biomed. Mater. Res. Part A Biomaterials 32 (2011) 2642–2650.
71A (2004) 528–537. [94] U.J. Kim, J. Park, C. Li, H.-J. Jin, R. Valluzzi, D.L. Kaplan, Structure and properties of
[63] V. Karageorgiou, M. Tomkins, R. Fajardo, L. Meinel, B. Snyder, K. Wade, J. Chen, G. silk hydrogels, Biomacromolecules 5 (2004) 786–792.
Vunjak-Novakovic, D.L. Kaplan, Porous silk fibroin 3-D scaffolds for delivery of [95] P.H. Chao, S. Yodmuang, X. Wang, L. Sun, D.L. Kaplan, Silk hydrogel for cartilage
bone morphogenetic protein-2 in vitro and in vivo, J. Biomed. Mater. Res. Part tissue engineering, J. Biomed. Mater. Res. B Appl. Biomater. (2010) 84–90.
A 78A (2006) 324–334. [96] M. Li, Z. Wu, C. Zhang, S. Lu, H. Yan, D. Huang, H. Ye, Study on porous silk fibroin
[64] Z. Shao, F. Vollrath, Materials: Surprising strength of silkworm silk, Nature 418 materials. II. Preparation and characteristics of spongy porous silk fibroin
(2002) 741. materials, J. Appl. Polym. Sci. 79 (2001) 2192–2199.
[65] H.J. Jin, J. Park, V. Karageorgiou, U.J. Kim, R. Valluzzi, P. Cebe, D.L. Kaplan, [97] M. Tsukada, G. Freddi, N. Minoura, G. Allara, Preparation and application of
Water-atable silk films with reduced β-sheet content, Adv. Funct. Mater. 15 porous silk fibroin materials, J. Appl. Polym. Sci. 54 (1994) 507–514.
(2005) 1241–1247. [98] L.D. Harris, B.S. Kim, D.J. Mooney, Open pore biodegradable matrices formed
[66] Y. Kawahara, K. Furukawa, T. Yamamoto, Self-expansion behaviour of silk fibroin with gas foaming, J. Biomed. Mater. Res. 42 (1998) 396–402.
film, Macromol. Mater. Eng. 291 (2006) 458–462. [99] M. Li, Z. Wu, C. Zhang, S. Lu, H. Yan, D. Huang, H. Ye, Study on porous silk fibroin
[67] N. Goujon, X. Wang, R. Rajkhowa, N. Byrne, Regenerated silk fibroin using protic materials. II. Preparation and characteristics of spongy porous silk fibroin
ionic liquids solvents : towards an all-ionic-liquid process for producing silk materials, J. Appl. Polym. Sci. 79 (2001) 2129–2199.
with tunable properties, Chem. Commun. 48 (2012) 1278–1280. [100] L. Meinel, R. Fajardo, S. Hofmann, R. Langer, J. Chen, B. Snyder, G.
[68] E. Servoli, D. Maniglio, A. Motta, R. Predazzer, C. Migliaresi, Surface properties of Vunjak-Novakovic, D. Kaplan, Silk implants for the healing of critical size bone
silk fibroin films and their interaction with fibroblasts, Macromol. Biosci. 5 defects, Bone 37 (2005) 688–698.
(2005) 1175–1183. [101] V.V. Seregin, J.L. Coffer, Biomineralization of calcium disilicide in porous
[69] S. Lu, X. Wang, Q. Lu, X. Hu, N. Uppal, F.G. Omenetto, D.L. Kaplan, Stabilization of polycaprolactone scaffolds, Biomaterials 27 (2006) 4745–4754.
enzymes in silk films, Biomacromolecules 10 (2009) 1032–1042. [102] A. Hokugo, T. Takamoto, Y. Tabata, Preparation of hybrid scaffold from fibrin and
[70] B.B. Mandal, S.C. Kundu, A novel method for dissolution and stabilization of biodegradable polymer fiber, Biomaterials 27 (2006) 61–67.
non-mulberry silk gland protein fibroin using anionic surfactant sodium dodecyl [103] M. Wang, Developing bioactive composite materials for tissue replacement,
sulfate, Biotechnol. Bioeng. 99 (2008) 1482–1489. Biomaterials 24 (2003) 2133–2151.
[71] S. Rathbone, P. Furrer, J. Lübben, M. Zinn, S. Cartmell, Biocompatibility of [104] R. Rajkhowa, E.S. Gil, J.A. Kludge, K. Numata, L. Wang, X. Wang, D.L. Kaplan,
polyhydroxyalkanoate as a potential material for ligament and tendon scaffold Reinforcing silk scaffolds with silk particles, Macromol. Biosci. 10 (2010)
material, J. Biomed. Mater. Res. Part A 93A (2010) 1391–1403. 599–611.
[72] H. Fan, H. Liu, E.J.W. Wong, S.L. Toh, J.C.H. Goh, In vivo study of anterior cruciate [105] B.B. Mandal, A. Grinberg, E. Seok Gil, B. Panilaitis, D.L. Kaplan, High-strength silk
ligament regeneration using mesenchymal stem cells and silk scaffold, Biomate- protein scaffolds for bone repair, Proc. Natl. Acad. Sci. 109 (2012) 7699–7704.
rials 29 (2008) 3324–3337. [106] H. Yoshimizu, T. Asakura, Preparation and characterisation of silk fibroin powder
[73] R. Ahmad, A. Kamra, S.E. Hasnain, Fibroin silk proteins from the nonmulberry and its application to enzyme immobilization, J. Appl. Polym. Sci. 40 (1990)
silkworm Philosamia ricini are biochemically and immunologically distinct 127–134.
from those of the mulberry silkworm Bombyx mori, DNA Cell Biol. 23 (2004) [107] J.H. Yeo, K.G. Lee, Y.W. Lee, S.Y. Kim, Simple preparation and characteristics of
149–154. silk fibroin microsphere, Eur. Polym. J. 39 (2003) 1195–1199.
468 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

[108] E. Wenk, A.J. Wandrey, H.P. Merkle, L. Meinel, Silk fibroin spheres as a platform [137] I.S. Yeo, J.E. Oh, L. Jeong, T.S. Lee, S.J. Lee, W.H. Park, B.M. Min, Collagen-based
for controlled drug delivery, J. Control. Release 132 (2008) 26–34. biomimetic nanofibrous scaffolds: preparation and characterization of
[109] A.S. Lammel, X. Hu, S.H. Park, D.L. Kaplan, T.R. Scheibel, Controlling silk fibroin collagen/silk fibroin bicomponent nanofibrous structures, Biomacromolecules
particle features for drug delivery, Biomaterials 31 (2010) 4583–4591. 9 (2008) 1106–1116.
[110] Y.Q. Zhang, S. Wei-De, X. Ru-Li, L.J. Zhuge, W.J. Gao, W.B. Wang, Formation of silk [138] H.L. Khor, K.W. Ng, A.S. Htay, J.T. Schantz, S.H. Teoh, D.W. Hutmacher, Prelimi-
nanoparticles in water-miscible organic solvent and their characterization, nary study of a polycaprolactone membrane utilized as epidermal substrate,
J. Nanopart. Res. 9 (2007) 885–900. J. Mater. Sci. Mater. Med. 14 (2003) 113–120.
[111] Z. Cao, X. Chen, J. Yao, L. Huang, Z. Shao, The preparation of regenerated silk fi- [139] D.H. Roh, S.Y. Kang, J.Y. Kim, Y.B. Kwon, H. Young Kweon, K.G. Lee, Y.H. Park, R.M.
broin microspheres, Soft Matter 3 (2007) 910–915. Baek, C.Y. Heo, J. Choe, J.H. Lee, Wound healing effect of silk fibroin/
[112] D.N. Rockwood, E.S. Gil, S.H. Park, J.A. Kluge, W. Grayson, S. Bhumiratana, R. alginate-blended sponge in full thickness skin defect of rat, J. Mater. Sci.
Rajkhowa, X. Wang, S.J. Kim, G. Vunjak-Novakovic, D.L. Kaplan, Ingrowth of Mater. Med. 17 (2006) 547–552.
human mesenchymal stem cells into porous silk particle reinforced silk compos- [140] L.G. Griffith, G. Naughton, Tissue engineering-current challenges and expanding
ite scaffolds: an in vitro study, Acta Biomater. 7 (2011) 144–151. opportunities, Science 295 (2002) 1009–1014.
[113] A.B. Mathur, V. Gupta, Silk fibroin-derived nanoparticles for biomedical applica- [141] L. Meinel, O. Betz, R. Fajardo, S. Hofmann, A. Nazarian, E. Cory, M. Hilbe, J.
tions, Nanomedicine 5 (2010) 807–820. McCool, R. Langer, G. Vunjak-Novakovic, H.P. Merkle, B. Rechenberg, D.L.
[114] J. Kundu, Y.I. Chung, Y.H. Kim, G. Tae, S.C. Kundu, Silk fibroin nanoparticles for Kaplan, C. Kirker-Head, Silk based biomaterials to heal critical sized femur
cellular uptake and control release, Int. J. Pharm. 388 (2010) 242–250. defects, Bone 39 (2006) 922–931.
[115] X. Wang, T. Yucel, Q. Lu, X. Hu, D.L. Kaplan, Silk nanospheres and microspheres [142] C. Li, C. Vepari, H.J. Jin, H.J. Kim, D.L. Kaplan, Electrospun silk-BMP-2 scaffolds for
from silk/PVA blend films for drug delivery, Biomaterials 31 (2010) 1025–1035. bone tissue engineering, Biomaterials 27 (2006) 3115–3124.
[116] F. Causin, R. Pascarella, G. Pavesi, R. Marasco, G. Zambon, R. Battaglia, M. Munari, [143] G. Wang, H. Yang, M. Li, S. Lu, X. Chen, X. Cai, The use of silk fibroin/
Acute endovascular treatment (b48 hours) of uncoilable ruptured aneurysms at hydroxyapatite composite co-cultured with rabbit bone-marrow stromal cells
non-branching sites using silk flow-diverting devices, Interv. Neuroradiol. 17 in the healing of a segmental bone defect, in: JSBS, British Volume, 92-B, 2010,
(3) (2011) 357–364. pp. 320–325.
[117] M. Leonardi, L. Cirillo, F. Toni, M. Dall'Olio, C. Princiotta, A. Stafa, L. Simonetti, R. Agati, [144] H. Kweon, K.-G. Lee, C.-H. Chae, C. Balázsi, S.-K. Min, J.Y. Kim, J.Y. Choi, S.G. Kim,
Treatment of intracranial aneurysms using flow-diverting silk stents (BALT): a single Development of nano-hydroxyapatite graft with silk fibroin scaffold as a new
centre experience, Interv. Neuroradiol. 17 (3) (2011) 306–315. bone substitute, J. Oral Maxillofac. Surg. 69 (2011) 1578–1586.
[118] L. Soffer, X. Wang, X. Zhang, J. Kluge, L. Dorfmann, D.L. Kaplan, G. Leisk, [145] K. Tanaka, N. Kajiyama, K. Ishikura, S. Waga, A. Kikuchi, K. Ohtomo, T. Takagi, S.
Silk-based electrospun tubular scaffolds for tissue-engineered vascular grafts, Mizuno, Determination of the site of disulfide linkage between heavy and light
J. Biomater. Sci. Polym. Ed. 19 (2008) 653–664. chains of silk fibroin produced by Bombyx mori, Biochim. Biophys. Acta, Protein
[119] J.M. Orban, L.B. Wilson, J.A. Kofroth, M.S. El-Kurdi, T.M. Maul, D.A. Vorp, Struct. Mol. Enzymol. 1432 (1999) 92–103.
Crosslinking of collagen gels by transglutaminase, J. Biomed. Mater. Res. Part A [146] M. Fini, A. Motta, P. Torricelli, G. Giavaresi, N. Nicoli Aldini, M. Tschon, R.
68A (2004) 756–762. Giardino, C. Migliaresi, The healing of confined critical size cancellous de-
[120] T. Nishibe, Y. Kondo, A. Muto, A. Dardik, Optimal prosthetic graft design for small fects in the presence of silk fibroin hydrogel, Biomaterials 26 (2005)
diameter vascular grafts, Vascular 15 (2007) 356–360. 3527–3536.
[121] Y. Nakazawa, M. Sato, R. Takahashi, D. Aytemiz, C. Takabayashi, T. Tamura, S. [147] H.J. Kim, U.J. Kim, H.S. Kim, C. Li, M. Wada, G.G. Leisk, D.L. Kaplan, Bone tissue
Enomoto, M. Sata, T. Asakura, Development of small-diameter vascular grafts engineering with premineralized silk scaffolds, Bone 42 (2008) 1226–1234.
based on silk fibroin fibers from bombyx mori for vascular regeneration, [148] S. Ghanaati, R.E. Unger, M.J. Webber, M. Barbeck, C. Orth, J.A. Kirkpatrick, P.
J. Biomater. Sci. Polym. Ed. 22 (2011) 195–206. Booms, A. Motta, C. Migliaresi, R.A. Sader, C.J. Kirkpatrick, Scaffold vasculariza-
[122] T. Yagi, M. Sato, Y. Nakazawa, K. Tanaka, M. Sata, K. Itoh, Y. Takagi, T. Asakura, tion in vivo driven by primary human osteoblasts in concert with host inflam-
Preparation of double-raschel knitted silk vascular grafts and evaluation matory cells, Biomaterials 32 (2011) 8150–8160.
of short-term function in a rat abdominal aorta, J. Artif. Organs 14 (2011) [149] R.E. Unger, A. Sartoris, K. Peters, A. Motta, C. Migliaresi, M. Kunkel, U. Bulnheim, J.
89–99. Rychly, C. James Kirkpatrick, Tissue-like self-assembly in cocultures of endothelial
[123] M. Lovett, G. Eng, J. Kluge, C. Cannizzaro, G. Vunjak-Novakovic, D.L. Kaplan, cells and osteoblasts and the formation of microcapillary-like structures on
Tubular silk scaffolds for small diameter vascular grafts, Organogenesis 6 three-dimensional porous biomaterials, Biomaterials 28 (2007) 3965–3976.
(2010) 217–224. [150] S. Fuchsa, X. Jianga, H. Schmidtb, E. Dohlea, S. Ghanaatia, C. Ortha, A. Hofmannc,
[124] A. Ratcliffe, Tissue engineering of vascular grafts, Matrix Biol. 19 (2000) A. Mottad, C. Migliaresid, C.J. Kirkpatricka, Dynamic processes involved in the
353–357. pre-vascularization of silk fibroin constructs for bone regeneration using out-
[125] S. Enomoto, M. Sumi, K. Kajimoto, Y. Nakazawa, R. Takahashi, C. Takabayashi, T. growth endothelial cells, Biomaterials 30 (2009) 1329–1338.
Asakura, M. Sata, Long-term patency of small-diameter vascular graft made [151] R.E. Unger, S. Ghanaati, C. Orth, A. Sartoris, M. Barbeck, S. Halstenberg, A. Motta,
from fibroin, a silk-based biodegradable material, J. Vasc. Surg. 51 (2010) C. Migliaresi, C.J. Kirkpatrick, The rapid anastomosis between prevascularized
155–164. networks on silk fibroin scaffolds generated in vitro with cocultures of human
[126] B. Bondar, S. Fuchs, A. Motta, C. Migliaresi, C.J. Kirkpatrick, Functionality of endo- microvascular endothelial and osteoblast cells and the host vasculature, Bioma-
thelial cells on silk fibroin nets: comparative study of micro- and nanometric terials 31 (2010) 6959–6967.
fibre size, Biomaterials 29 (2008) 561–572. [152] Y. Wang, D.J. Blasioli, H.J. Kim, H.S. Kim, D.L. Kaplan, Cartilage tissue engineering
[127] R.E. Unger, K. Peters, M. Wolf, A. Motta, C. Migliaresi, C.J. Kirkpatrick, with silk scaffolds and human articular chondrocytes, Biomaterials 27 (2006)
Endothelialization of a non-woven silk fibroin net for use in tissue engineering: 4434–4442.
growth and gene regulation of human endothelial cells, Biomaterials 25 (2004) [153] Y. Morita, N. Tomita, H. Aoki, M. Sonobe, S. Wakitani, Y. Tamada, T. Suguro, K.
5137–5146. Ikeuchi, Frictional properties of regenerated cartilage in vitro, J. Biomech. 39
[128] H. Liu, X. Li, G. Zhou, H. Fan, Y. Fan, Electrospun sulfated silk fibroin nanofibrous (2006) 103–109.
scaffolds for vascular tissue engineering, Biomaterials 32 (2011) 3784–3793. [154] Y. Wang, U.J. Kim, D.J. Blasioli, H.J. Kim, D.L. Kaplan, In vitro cartilage tissue
[129] Y. Yang, X. Chen, F. Ding, P. Zhang, J. Liu, X. Gu, Biocompatibility evaluation of engineering with 3D porous aqueous-derived silk scaffolds and mesenchymal
silk fibroin with peripheral nerve tissues and cells in vitro, Biomaterials 28 stem cells, Biomaterials 26 (2005) 7082–7094.
(2007) 1643–1652. [155] K. Gellynck, P.C.M. Verdonk, E.V. Nimmen, K.F. Almqvist, T. Gheysens, G.
[130] Y. Yang, F. Ding, J. Wu, W. Hu, W. Liu, J. Liu, X. Gu, Development and evaluation Schokens, L.V. Langenhove, P. Kiekens, J. Mertens, G. Verbruggen, Silkworm
of silk fibroin-based nerve grafts used for peripheral nerve regeneration, and spider silk scaffolds for chondrocyte support, J. Mater. Sci. Mater. Med. 19
Biomaterials 28 (2007) 5526–5535. (2008) 3399–3409.
[131] Y. Wei, K. Gong, Z. Zheng, A. Wang, Q. Ao, Y. Gong, X. Zhang, Chitosan/silk [156] H.S. Baek, Y.H. Park, C.S. Ki, J.C. Park, D.K. Rah, Enhanced chondrogenic responses
fibroin-based tissue-engineered graft seeded with adipose-derived stem cells of articular chondrocytes onto porous silk fibroin scaffolds treated with
enhances nerve regeneration in a rat model, J. Mater. Sci. Mater. Med. 22 microwave-induced argon plasma, Surf. Coat. Technol. 202 (2008) 5794–5797.
(2011) 1947–1964. [157] N. Bhardwaj, Q.T. Nguyen, A.C. Chen, D.L. Kaplan, R.L. Sah, S.C. Kundu, Potential of
[132] C.Y. Wang, K.-H. Zhang, C.Y. Fan, X.M. Mo, H.J. Ruan, F.F. Li, Aligned natural– 3-D tissue constructs engineered from bovine chondrocytes/silk fibroin-chitosan
synthetic polyblend nanofibers for peripheral nerve regeneration, Acta for in vitro cartilage tissue engineering, Biomaterials 32 (2011) 5773–5781.
Biomater. 7 (2011) 634–643. [158] S.S. Silva, A. Motta, M.R.T. Rodrigues, A.F.M. Pinheiro, M.E. Gomes, J.O.F. Mano,
[133] W. Huang, R. Begum, T. Barber, V. Ibba, N.C.H. Tee, M. Hussain, M. Arastoo, Q. R.L. Reis, C. Migliaresi, Novel Genipin-cross-linked chitosan/silk fibroin
Yang, L.G. Robson, S. Lesage, T. Gheysens, N.J.V. Skaer, D.P. Knight, J.V. sponges for cartilage engineering strategies, Biomacromolecules 9 (2008)
Priestley, Regenerative potential of silk conduits in repair of peripheral nerve 2764–2774.
injury in adult rats, Biomaterials 33 (2012) 59–71. [159] S. Talukdar, Q.T. Nguyen, A.C. Chen, R.L. Sah, S.C. Kundu, Effect of initial cell
[134] S. Madduri, M. Papaloïzos, B. Gander, Trophically and topographically function- seeding density on 3D-engineered silk fibroin scaffolds for articular cartilage
alized silk fibroin nerve conduits for guided peripheral nerve regeneration, tissue engineering, Biomaterials 32 (2011) 8927–8937.
Biomaterials 31 (2010) 2323–2334. [160] L. Uebersax, H.P. Merkle, L. Meinel, Insulin-like growth factor I releasing silk fi-
[135] B.M. Min, L. Jeong, K.Y. Lee, W.H. Park, Regenerated silk fibroin nanofibers: broin scaffolds induce chondrogenic differentiation of human mesenchymal
water vapor-induced structural changes and their effects on the behavior of stem cells, J. Control. Release 127 (2008) 12–21.
normal human cells, Macromol. Biosci. 6 (2006) 285–292. [161] C. Shangkai, T. Naohide, Y. Koji, H. Yasuji, N. Masaaki, T. Tomohiro, T. Yasushi,
[136] C.R. Yoo, I.S. Yeo, K.E. Park, J.H. Park, S.J. Lee, W.H. Park, B.M. Min, Effect of Transplantation of allogeneic chondrocytes cultured in fibroin sponge and
chitin/silk fibroin nanofibrous bicomponent structures on interaction with stirring chamber to promote cartilage Regeneration, Tissue Eng. C Methods 13
human epidermal keratinocytes, Int. J. Biol. Macromol. 42 (2008) 324–334. (2007) 483–492.
B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470 469

[162] R. Seda Tigli, S. Ghosh, M.M. Laha, N.K. Shevde, L. Daheron, J. Gimble, M. [192] G.M. Cannon Jr., J.R. Mauney, E.M. Gong, R.N. Yu, R.M. Adam, C.R. Estrada, Silk as
Gümüşderelioglu, D.L. Kaplan, Comparative chondrogenesis of human cell a novel biomaterial in bladder tissue engineering, J. Pediatr. Urol. 6 (Supplement
sources in 3D scaffolds, J. Tissue Eng. Regen. Med. 3 (2009) 348–360. 1) (2010) S82.
[163] X. Chen, Y.Y. Qi, L.L. Wang, Z. Yin, G.L. Yin, X.H. Zou, H.W. Ouyang, Ligament re- [193] J.R. Mauney, G.M. Cannon, M.L. Lovett, E.M. Gong, D. Di Vizio, P. Gomez Iii, D.L.
generation using a knitted silk scaffold combined with collagen matrix, Bioma- Kaplan, R.M. Adam, C.R. Estrada Jr., Evaluation of gel spun silk-based biomate-
terials 29 (2008) 3683–3692. rials in a murine model of bladder augmentation, Biomaterials 32 (2011)
[164] S. Sahoo, S. Lok Toh, J.C. Hong Goh, PLGA nanofiber-coated silk microfibrous 808–818.
scaffold for connective tissue engineering, J. Biomed. Mater. Res. B Appl. [194] X.H. Zou, Y.L. Zhi, X. Chen, H.M. Jin, L.L. Wang, Y.Z. Jiang, Z. Yin, H.W. Ouyang,
Biomater. 95B (2010) 19–28. Mesenchymal stem cell seeded knitted silk sling for the treatment of stress
[165] J.E. Moreau, J. Chen, R.L. Horan, D.L. Kaplan, G.H. Altman, Sequential growth fac- urinary incontinence, Biomaterials 31 (2010) 4872–4879.
tor application in bone marrow stromal cell ligament engineering, Tissue Eng. [195] Y. Ni, X. Zhao, L. Zhou, Z. Shao, W. Yan, X. Chen, Z. Cao, Z. Xue, J.J. Jiang, Radiolog-
11 (2005) 1887–1897. ic and histologic characterization of silk fibroin as scaffold coating for rabbit
[166] M. Hairfield-Stein, C. England, H.J. Paek, K.B. Gilbraith, R. Dennis, E. Boland, P. tracheal defect repair, Otolaryngol. Head Neck Surg. 139 (2008) 256–261.
Kosnik, Development of self-assembled, tissue-engineered ligament from bone [196] M. Zang, Q. Zhang, G. Davis, G. Huang, M. Jaffari, C.N. Ríos, V. Gupta, P. Yu, A.B.
marrow stromal cells, Tissue Eng. 13 (2007) 703–710. Mathur, Perichondrium directed cartilage formation in silk fibroin and
[167] Q. Fang, D. Chen, Z. Yang, M. Li, In vitro and in vivo research on using antheraea chitosan blend scaffolds for tracheal transplantation, Acta Biomater. 7 (2011)
pernyi silk fibroin as tissue engineering tendon scaffolds, Mater. Sci. Eng., C 29 3422–3431.
(2009) 1527–1534. [197] B. Levin, R. Rajkhowa, S.L. Redmond, M.D. Atlas, Grafts in myringplasty :utilizing
[168] M.C. Yang, S.S. Wang, N.K. Chou, N.H. Chi, Y.Y. Huang, Y.L. Chang, M.J. Shieh, T.W. a silk fibroin scaffold as a novel decice, Expert Rev. Med. Devices 6 (2009)
Chung, The cardiomyogenic differentiation of rat mesenchymal stem cells on silk fi- 653–664.
broin–polysaccharide cardiac patches in vitro, Biomaterials 30 (2009) 3757–3765. [198] B. Levin, S.L. Redmond, R. Rajkhowa, R.H. Eikelboom, R.J. Marano, M.D. Atlas,
[169] C. Patra, S. Talukdar, T. Novoyatleva, S.R. Velagala, C. Mühlfeld, B. Kundu, S.C. Preliminary results of the application of a silk fibroin scaffold to otology,
Kundu, F.B. Engel, Silk protein fibroin from Antheraea mylitta for cardiac tissue Otolaryngol. Head Neck Surg. 142 (2010) S33–S35.
engineering, Biomaterials 33 (2012) 2673–2680. [199] R. Ghassemifar, S. Redmond, T.V. Chirila, Zainuddin, Advancing towards a
[170] W. Liu, K. Merrett, M. Griffith, P. Fagerholm, S. Dravida, B. Heyne, J.C. Scaiano, tissue-engineered tympanic membrane: Silk fibroin as a substratum for growing
M.A. Watsky, N. Shinozaki, N. Lagali, R. Munger, F. Li, Recombinant human human eardrum keratinocytes, J. Biomater. Appl. 24 (2010) 591–606.
collagen for tissue engineered corneal substitutes, Biomaterials 29 (2008) [200] B. Levin, S.L. Redmond, R. Rajkhowa, R.H. Eikelboom, M.D. Atlas, Utilising silk
1147–1158. fibroin membranes as scaffolds for the growth of tympanic membrane
[171] B.D. Lawrence, M. Cronin-Golomb, I. Georgakoudi, D.L. Kaplan, F.G. Omenetto, keratinocytes, and application to myringoplasty surgery, J. Laryngol. Otol.
Bioactive silk protein biomaterial systems for optical devices, Biomacromolecules (2012), http://dx.doi.org/10.1017/S0022215112001661.
9 (2008) 1214–1220. [201] J. Kim, C.H. Kim, C.H. Park, J.N. Seo, H. Kweon, S.W. Kang, K.G. Lee, Comparison of
[172] T. Yang, M. Zhang, Biocompatibility of silk fibroin membrane as tissue engineer- methods for the repair of acute tympanic membrane perforations: Silk patch vs.
ing corneal scaffold, Int. J. Ophthalmol. 8 (2008) 1557–1559. paper patch, Wound Repair Regen. 18 (2010) 132–138.
[173] B.D. Lawrence, J.K. Marchant, M.A. Pindrus, F.G. Omenetto, D.L. Kaplan, Silk film [202] N. Reddy, Y. Yang, Morphology and tensile properties of silk fibers produced by
biomaterials for cornea tissue engineering, Biomaterials 30 (2009) 1299–1308. uncommon saturniidae, Int. J. Biol. Macromol. 46 (2010) 419–424.
[174] S.C. Kundu, B. Kundu, S. Talukdar, S. Bano, S. Nayak, J. Kundu, B.B. Mandal, N. [203] C. Zhao, X. Wu, Q. Zhang, S. Yan, M. Li, Enzymatic degradation of Antheraea
Bhardwaj, M. Botlagunta, B.C. Dash, C. Acharya, A.K. Ghosh, Nonmulberry silk pernyi silk fibroin 3-D scaffolds and fibers, Int. J. Biol. Macromol. 48 (2011)
biopolymers, Biopolymers 97 (2012) 455–467. 249–255.
[175] P.W. Madden, J.N.X. Lai, K.A. George, T. Giovenco, D.G. Harkin, T.V. Chirila, [204] S.H. Park, E.S. Gil, H. Shi, H.J. Kim, K. Lee, D.L. Kaplan, Relationships between
Human corneal endothelial cell growth on a silk fibroin membrane, Biomaterials degradability of silk scaffolds and osteogenesis, Biomaterials 31 (2010)
32 (2011) 4076–4084. 6162–6172.
[176] L.J. Bray, K.A. George, S.L. Ainscough, D.W. Hutmacher, T.V. Chirila, D.G. Harkin, [205] X. Hu, Q. Lu, D.L. Kaplan, P. Cebe, Microphase separation controlled beta-sheet
Human corneal epithelial equivalents constructed on Bombyx mori silk fibroin crystallization kinetics in fibrous proteins, Macromolecules 42 (2009) 2079–
membranes, Biomaterials 32 (2011) 5086–5091. 2087.
[177] Y. Gotoh, S. Niimi, T. Hayakawa, T. Miyashita, Preparation of lactose–silk fibroin [206] X. Hu, K. Shmelev, L. Sun, E.S. Gil, S.H. Park, P. Cebe, D.L. Kaplan, Regulation of
conjugates and their application as a scaffold for hepatocyte attachment, Bioma- silk material structure by temperature-controlled water vapor annealing,
terials 25 (2004) 1131–1140. Biomacromolecules 12 (2011) 1686–1696.
[178] B. Cirillo, M. Morra, G. Catapano, Adhesion and function of rat liver cells adher- [207] A. Vasconcelos, G. Freddi, A. Cavaco-Paulo, Biodegradable materials based on silk
ent to silk fibroin/collagen blend films, Int. J. Artif. Organs 27 (2004) 60–68. fibroin and keratin, Biomacromolecules 9 (2008) 1299–1305.
[179] K. Hu, Q. Lv, F.Z. Cui, Q.L. Feng, X.D. Kong, H.L. Wang, L.Y. Huang, T. Li, Biocom- [208] R. Kino, T. Ikoma, S. Yunoki, A. Monkawa, A. Matsuda, G. Kagata, T. Asakura, M.
patible fibroin blended films with recombinant human-like collagen for hepatic Munekata, T. Junzo, Biodegradation of multilayer silk fibroin and hydroxyapatite
tissue engineering, J. Bioact. Compat. Polym. 21 (2006) 23–37. composite material, Key Eng. Mater. 309–311 (2006) 1169–1172.
[180] Q. Lu, S. Zhang, K. Hu, Q. Feng, C. Cao, F. Cui, Cytocompatibility and blood com- [209] E.M. Pritchard, T. Valentin, D. Boison, D.L. Kaplan, Incorporation of proteinase in-
patibility of multifunctional fibroin/collagen/heparin scaffolds, Biomaterials 28 hibitors into silk-based delivery devices for enhanced control of degradation
(2007) 2306–2313. and drug release, Biomaterials 32 (2011) 909–918.
[181] Q. Lv, K. Hu, Q. Feng, F. Cui, C. Cao, Preparation and characterization of [210] B. Zuo, L. Liu, Z. Wu, Effect on properties of regenerated silk fibroin fiber
PLA/fibroin composite and culture of HepG2 (human hepatocellular liver coagulated with aqueous methanol/ethanol, J. Appl. Polym. Sci. 106 (2007)
carcinoma cell line) cells, Compos. Sci. Technol. 67 (2007) 3023–3030. 53–59.
[182] Z. She, W. Liu, Q. Feng, Self-assembly model, hepatocytes attachment and [211] J.H. Kim, C.H. Park, O.J. Lee, J.M. Lee, J.W. Kim, Y.H. Park, C.S. Ki, Preparation and
inflammatory response for silk fibroin/chitosan scaffolds, Biomed. Mater. 4 in vivo degradation of controlled biodegradability of electrospun silk fibroin
(2009) 045014. nanofiber mats, J. Biomed. Mater. Res. A 100 (2012) 3287–3295.
[183] N. Kasoju, R.R. Bhonde, U. Bora, Fabrication of a Biomed. Mater., novel micro– [212] K. Chen, Y. Umeda, K. Hirabayashi, Enzymatic hydrolysis of silk fibroin, J. Seric.
nano fibrous nonwoven scaffold with Antheraea assama silk fibroin for use in tis- Sci. Jpn. 65 (1995) 131–133.
sue engineering, Mater. Lett. 63 (2009) 2466–2469. [213] W. Xu, G. Ke, X. Peng, Studies on the effects of the enzymatic treatment on silk
[184] N. Kasoju, U. Bora, Antheraea assama silk fibroin-based functional scaffold with fine powder, J. Appl. Polym. Sci. 101 (2006) 2967–2971.
enhanced blood compatibility for tissue engineering applications, Adv. Eng. [214] R. Rajkhowa, X. Hu, T. Tsuzuki, D.L. Kaplan, X.G. Wang, Structure and biodegra-
Mater. 12 (2010) B139–B147. dation mechanism of milled B. mori silk particles, Biomacromolecules 13 (2012)
[185] Y. Qian, Y. Shen, Z. Lu, Z. Fan, T. Liu, J. Zhang, F. Zhang, Biocompatibility of silk 2503–2512.
fibroin nanofibers scaffold with olfactory ensheathing cells, Zhongguo Xiu Fu [215] Y. Tomizawa, Vascular prostheses for aortocoronary bypass grafting: a review,
Chong Jian Wai Ke Za Zhi 23 (2009) 1365–1370. Artif. Organs 19 (1995) 39–45.
[186] Y. Shen, Y. Qian, H. Zhang, B. Zuo, Z. Lu, Z. Fan, P. Zhang, F. Zhang, C. Zhou, Guid- [216] A. Callow, Arterial homografts, Eur. J. Vasc. Endovasc. Surg. 12 (1996) 272–281.
ance of olfactory ensheathing cell growth and migration on electrospun silk [217] K.J. Pawlowski, S.E. Rittgers, S.P. Schmidt, G.L. Bowlin, Endothelial cell seeding of
fibroin scaffolds, Cell Transplant. 19 (2010) 147–157. polymeric vascular grafts, Front. Biosci. 9 (2004) 1412–1421.
[187] G. Chang, H.J. Kim, D. Kaplan, G. Vunjak-Novakovic, R. Kandel, Porous silk scaf- [218] G.R.D. Evans, Challenges to nerve regeneration, Semin. Surg. Oncol. 19 (2000)
folds can be used for tissue engineering annulus fibrosus, Eur. Spine J. 16 312–318.
(2007) 1848–1857. [219] A. Beris, M. Lykissas, A. Korompilias, G. Mitsionis, End-to-side nerve repair in
[188] G. Chang, H.J. Kim, G. Vunjak-Novakovic, D.L. Kaplan, R. Kandel, Enhancing an- peripheral nerve injury, J. Neurotrauma 24 (2007) 909–916.
nulus fibrosus tissue formation in porous silk scaffolds, J. Biomed. Mater. Res. [220] M.F. Meek, J.H. Coert, Clinical use of nerve conduits in peripheral-nerve repair:
A 92A (2010) 43–51. review of the literature, J. Reconstr. Microsurg. 18 (2002) 097–110.
[189] A. Atala, Tissue engineering of human bladder, Br. Med. Bull. 97 (2011) 81–104. [221] L. Thomsen, P. Bellemere, T. Loubersac, E. Gaisne, P. Poirier, F. Chaise, Treatment
[190] C.X. Liu, Y.F. Liao, H.L. Li, S.B. Zheng, Cytocompatibility of silk fibroin film with by collagen conduit of painful post-traumatic neuromas of the sensitive digital
rabbit urinary bladder transitional epithelial cells in vitro, Nan Fang Yi Ke Da nerve: a retrospective study of 10 cases, Chir. Main 29 (2010) 255–262.
Xue Xue Bao 28 (2008) 216–218. [222] O. Ilhan-Sarac, E.K. Akpek, Current concepts and techniques in keratoprosthesis,
[191] C.X. Liu, Y.Y. Lin, H.L. Li, S.B. Zheng, Application of silk fibroin film for Curr. Opin. Ophthalmol. 16 (2005) 246–250.
repairing rabbit urethral defect, Nan Fang Yi Ke Da Xue Xue Bao 27 (2007) [223] D. Myung, W. Koh, A. Bakri, F. Zhang, A. Marshall, J. Ko, J. Noolandi, M. Carrasco, J.
184–187. Cochran, C. Frank, C. Ta, Design and fabrication of an artificial cornea based on a
470 B. Kundu et al. / Advanced Drug Delivery Reviews 65 (2013) 457–470

photolithographically patterned hydrogel construct, Biomed. Microdevices 9 [226] S. Calve, R.G. Dennis, P.E. Kosnik II, K. Baar, K. Grosh, E.M. Arruda, Engineering of
(2007) 911–922. functional tendon, Tissue Eng. 10 (2004) 755–761.
[224] S. Geetha Priya, H. Jungvid, A. Kumar, Skin tissue engineering for tissue repair [227] M.P. Prabhakaran, J. Venugopal, D. Kai, S. Ramakrishna, Biomimetic material
and regeneration, Tissue Eng. B Rev. 14 (2008) 105–118. strategies for cardiac tissue engineering, Mater. Sci. Eng., C 31 (2011) 503–513.
[225] G. Vunjak-Novakovic, G. Altman, R. Horan, D.L. Kaplan, Tissue engineering of
ligaments, Annu. Rev. Biomed. Eng. 6 (2004) 131–156.

You might also like