silk-based biomaterials - biomedical...

16
Biomaterials 24 (2003) 401–416 Silk-based biomaterials Gregory H. Altman, Frank Diaz, Caroline Jakuba, Tara Calabro, Rebecca L. Horan, Jingsong Chen, Helen Lu, John Richmond, David L. Kaplan* Department of Chemical and Biological Engineering, Bioengineering Center, Tufts University, 4 Colby Street, Medford, MA 02155, USA Received 17 April 2002; accepted 19 June 2002 Abstract Silk from the silkworm, Bombyx mori, has been used as biomedical suture material for centuries. The unique mechanical properties of these fibers provided important clinical repair options for many applications. During the past 20 years, some biocompatibility problems have been reported for silkworm silk; however, contamination from residual sericin (glue-like proteins) was the likely cause. More recent studies with well-defined silkworm silk fibers and films suggest that the core silk fibroin fibers exhibit comparable biocompatibility in vitro and in vivo with other commonly used biomaterials such as polylactic acid and collagen. Furthermore, the unique mechanical properties of the silk fibers, the diversity of side chain chemistries for ‘decoration’ with growth and adhesion factors, and the ability to genetically tailor the protein provide additional rationale for the exploration of this family of fibrous proteins for biomaterial applications. For example, in designing scaffolds for tissue engineering these properties are particularly relevant and recent results with bone and ligament formation in vitro support the potential role for this biomaterial in future applications. To date, studies with silks to address biomaterial and matrix scaffold needs have focused on silkworm silk. With the diversity of silk-like fibrous proteins from spiders and insects, a range of native or bioengineered variants can be expected for application to a diverse set of clinical needs. r 2002 Elsevier Science Ltd. All rights reserved. Keywords: Silk; Sericin; Fibroin; Foreign body response; Suture; Tissue engineering; Biomaterial; Scaffold 1. Background Silks are generally defined as protein polymers that are spun into fibers by some lepidoptera larvae such as silkworms, spiders, scorpions, mites and flies [1–3]. Silk proteins are usually produced within specialized glands after biosynthesis in epithelial cells, followed by secre- tion into the lumen of these glands where the proteins are stored prior to spinning into fibers. Silks differ widely in composition, structure and properties depend- ing on the specific source. The most extensively characterized silks are from the domesticated silkworm, Bombyx mori, and from spiders (Nephila clavipes and Araneus diadematus). Many of the more evolutionarily advanced spiders synthesize different types of silks. Each of these different silks has a different amino acid composition and exhibits mechanical properties tailored to their specific functions: reproduction as cocoon capsular structures, lines for prey capture, lifeline support (dragline), web construction and adhesion. Fibrous proteins, such as silks and collagens, are characterized by a highly repetitive primary sequence that leads to significant homogeneity in secondary structure, i.e., triple helices in the case of collagens and b-sheets in the case of many of the silks. These types of proteins usually exhibit important mechanical proper- ties, in contrast to the catalytic and molecular recogni- tion functions of globular proteins. Because of these impressive mechanical properties, this family of proteins provides an important set of material options in the fields of controlled release, biomaterials and scaffolds for tissue engineering. The relative environmental stability of these families of proteins, in comparison to globular proteins, in combination with their biocompat- ibility, unique mechanical properties, and options for genetic control to tailor sequence provides an important basis to exploit these natural proteins for biomedical applications. *Corresponding author. Tel.: +1-617-627-3251; fax: +1-617-627- 3231. E-mail address: [email protected] (D.L. Kaplan). 0142-9612/02/$ - see front matter r 2002 Elsevier Science Ltd. All rights reserved. PII:S0142-9612(02)00353-8

Upload: others

Post on 26-Jun-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

Biomaterials 24 (2003) 401–416

Silk-based biomaterials

Gregory H. Altman, Frank Diaz, Caroline Jakuba, Tara Calabro, Rebecca L. Horan,Jingsong Chen, Helen Lu, John Richmond, David L. Kaplan*

Department of Chemical and Biological Engineering, Bioengineering Center, Tufts University, 4 Colby Street, Medford, MA 02155, USA

Received 17 April 2002; accepted 19 June 2002

Abstract

Silk from the silkworm, Bombyx mori, has been used as biomedical suture material for centuries. The unique mechanical

properties of these fibers provided important clinical repair options for many applications. During the past 20 years, some

biocompatibility problems have been reported for silkworm silk; however, contamination from residual sericin (glue-like proteins)

was the likely cause. More recent studies with well-defined silkworm silk fibers and films suggest that the core silk fibroin fibers

exhibit comparable biocompatibility in vitro and in vivo with other commonly used biomaterials such as polylactic acid and

collagen. Furthermore, the unique mechanical properties of the silk fibers, the diversity of side chain chemistries for ‘decoration’

with growth and adhesion factors, and the ability to genetically tailor the protein provide additional rationale for the exploration of

this family of fibrous proteins for biomaterial applications. For example, in designing scaffolds for tissue engineering these

properties are particularly relevant and recent results with bone and ligament formation in vitro support the potential role for this

biomaterial in future applications. To date, studies with silks to address biomaterial and matrix scaffold needs have focused on

silkworm silk. With the diversity of silk-like fibrous proteins from spiders and insects, a range of native or bioengineered variants

can be expected for application to a diverse set of clinical needs.

r 2002 Elsevier Science Ltd. All rights reserved.

Keywords: Silk; Sericin; Fibroin; Foreign body response; Suture; Tissue engineering; Biomaterial; Scaffold

1. Background

Silks are generally defined as protein polymers thatare spun into fibers by some lepidoptera larvae such assilkworms, spiders, scorpions, mites and flies [1–3]. Silkproteins are usually produced within specialized glandsafter biosynthesis in epithelial cells, followed by secre-tion into the lumen of these glands where the proteinsare stored prior to spinning into fibers. Silks differwidely in composition, structure and properties depend-ing on the specific source. The most extensivelycharacterized silks are from the domesticated silkworm,Bombyx mori, and from spiders (Nephila clavipes andAraneus diadematus). Many of the more evolutionarilyadvanced spiders synthesize different types of silks. Eachof these different silks has a different amino acidcomposition and exhibits mechanical properties tailored

to their specific functions: reproduction as cocooncapsular structures, lines for prey capture, lifelinesupport (dragline), web construction and adhesion.

Fibrous proteins, such as silks and collagens, arecharacterized by a highly repetitive primary sequencethat leads to significant homogeneity in secondarystructure, i.e., triple helices in the case of collagens andb-sheets in the case of many of the silks. These types ofproteins usually exhibit important mechanical proper-ties, in contrast to the catalytic and molecular recogni-tion functions of globular proteins. Because of theseimpressive mechanical properties, this family of proteinsprovides an important set of material options in thefields of controlled release, biomaterials and scaffoldsfor tissue engineering. The relative environmentalstability of these families of proteins, in comparison toglobular proteins, in combination with their biocompat-ibility, unique mechanical properties, and options forgenetic control to tailor sequence provides an importantbasis to exploit these natural proteins for biomedicalapplications.

*Corresponding author. Tel.: +1-617-627-3251; fax: +1-617-627-

3231.

E-mail address: [email protected] (D.L. Kaplan).

0142-9612/02/$ - see front matter r 2002 Elsevier Science Ltd. All rights reserved.

PII: S 0 1 4 2 - 9 6 1 2 ( 0 2 ) 0 0 3 5 3 - 8

Page 2: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

1.1. Silkworm silk

Silkworm silk has been used commercially as biome-dical sutures for decades, and in textile production forcenturies. The silk from the cocoon of B. mori containsat least two major fibroin proteins, light and heavychains, 25 and 325 kDa, respectively. These core fibersare encased in a sericin coat, a family of glue-likeproteins that holds two fibroin fibers together to formthe composite fibers of the cocoon case to protect thegrowing worm. This structural arrangement contrastswith spider silks where these glue-like proteins aregenerally absent. Silkworm cocoon silk production,known as sericulture, produces high yields since thelarvae can be maintained in high densities. The coresequence repeats in the fibroin heavy chain from B. mori

include alanine–glycine repeats with serine or tyrosine.

1.2. Spider silks

Spider silks have not been commercialized forbiomedical applications primarily due to the predatorynature of spiders and the relatively low levels ofproduction of these silks when compared to silkwormcocoon silk. The molecular weights of spider silkproteins vary depending on source but can range from70 to 700 kDa depending on the method of analysis. Thedragline silk from N. clavipes is characterized bypolyalanine and glycine–glycine-X regions, where X isoften tyrosine, glutamine or leucine. Genetic engineeringis being actively explored to construct, clone and expressnative and synthetic genes encoding recombinant spidersilk proteins to overcome limits to use of the nativeorganisms. This strategy has provided new opportunitiesin fundamental studies of spider silk genetics, silkprotein structure and function, and materials processing[2,4–6]. In general, interest in spider silk has increased inrecent years due to the differences in mechanicalproperties when compared to silkworm silk and thepresence of the multi-gene family encoding this group ofsilks as a basis for the study of protein structure-function relationships [4,6–11].

During the past 10 years a great deal of progress hasbeen made in understanding silk genetic and proteinstructures. Cloning and expression of native andsynthetic silks has been achieved in a variety of hostsystems. The sequences of cDNAs and genomic clonesencoding spider silks illustrate the highly repetitivestructures [4,6,8–11], which can be readily exploited toconstruct genetically engineered spider silk-like proteinsusing synthetic oligonucleotide versions of the consensusrepeats or variants of these repeats. This highlyrepetitive sequence was also recently fully described forthe fibroin heavy chain from the silkworm, B. mori [12].The unique organization of the family of silk genes inmore advanced spiders provides a fertile area for the

exploration of structure-function relationships in pro-tein design. For example, the dragline spider silk fromthe golden orb weaver N. clavipes displays impressivetoughness, and a balance of stiffness, strength andextensibility reflecting the native function of the silk orbweb construction [7,13,14]. Transgenic expression ofspider silks in plants (tobacco and potato) andmammalian epithelial cells has been reported [15,16]and may point the way toward more substantiveproduction of these proteins in the future.

Furthermore, since native and genetically engineeredversions of the silks tend to self-assemble into micro-fibrils, causing precipitation and leading to loss ofprotein during purification [3,17], alternative methods tomaintain solubility of the recombinant versions of theseproteins have been explored. For example, tweakingthe native or consensus sequences of spider silks toinclude encoded ‘triggers’ to regulate molecular-levelassembly of the proteins has been demonstrated [17–21].Chemical redox triggers, or a kinase recognition site forbiochemical phosphorylation, flanking the b-sheet form-ing regions of the proteins have been demonstrated.These modifications encoded in the synthetic genedesigns enabled improved control over solubility of thegenetically engineered silk protein.

1.3. Properties of silk

The enhanced environmental stability of silk fibers incomparison to globular proteins is due to the extensivehydrogen bonding, the hydrophobic nature of much ofthe protein, and the significant crystallinity. Silks areinsoluble in most solvents, including water, dilute acidand alkali. Detailed structural analysis of spider draglinesilk proteins has yielded information on the organiza-tion and orientation of the numerous but very smallb-sheet crystals in the fibers, and a high level oforganization of the protein even in the less crystallinedomains [2]. Liquid crystalline phases and conforma-tional polymorphism have been implicated in thebiological processing of these proteins to contribute tothe architectural features within the fibers [22–24]. Thesenanoscale features, factoring in the small, orientated andnumerous b-sheet crystals, a fuzzy interphase betweenthese crystals and the less crystalline domains, and theshear alignment of the chains, provides a basis for theorigin of the novel mechanical properties exhibited bysilk fibers. A comparison of mechanical properties(Table 1) suggests that they provide a remarkablecombination of strength and toughness. The distinguish-ing features of the spider silks are the very high strengthin combination with excellent elasticity in comparisonwith these other biomaterials. In addition, these fibersdisplay resistance to failure in compression thatdistinguishes them from other high performance fibers,such as Kevlar [13].

G.H. Altman et al. / Biomaterials 24 (2003) 401–416402

Page 3: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

2. Utility in biomedical applications

B. mori silkworm silk fibers have been the primarysilk-like material used in biomedical applications parti-cularly as sutures. During decades of use, silk fibers haveproven to be effective in many clinical applications. Atthe same time, some biological responses to the proteinhave raised questions about biocompatibility. One of themajor difficulties in assessing the biological responsesreported to these silk fibers is the absence of detailedcharacterization of the fibers used including, extent ofextraction of the sericin, the chemical nature of wax-likecoatings sometimes used, and many related processingfactors. This variability in source material has resultedin confusion in the literature and in clinical settingsconcerning the benefits or potential concerns with thisclass of fibrous protein. For example, of greatestimportance is that based on many studies it is clearthat the sericin glue-like proteins are the major cause ofadverse problems with biocompatibility and hypersensi-tivity to silk (Table 2). If sericin is removed, the biolo-gical responses to the core fibroin fibers appear to becomparable to most other commonly used biomaterials(Table 3). An additional major misconception revolvesaround silk’s classification as non-degradable. Thereis clear evidence in the literature that silk, as a protein, issusceptible to proteolytic degradation in vivo and overlonger time periods in vivo will slowly be absorbed.

3. In vivo biocompatibility of virgin silk

Despite centuries of use as sutures [25], silk matricesare now being rediscovered and reconsidered as

potentially useful biomaterials for a range of applica-tions in clinical repairs and in vitro as scaffolds fortissue engineering [26]; some benefits and concerns ofsilk are detailed in Table 4. Silk has been usedmost extensively as sutures for wound ligation andbecame the most common natural suture surpassingcollagen (CatgutTM or Chromic CatgutTM, cross-linkedcollagen) used in the biomedical industry over the past100 years [25]. During the past 20 years, a variety ofdegradable synthetic materials including polyglycolicacid (PGA)(DexonTM), co-polymers of PGA (polygly-conate)(MaxonTM or gylcolide and trimethylene carbo-nate), and co-polymers of PGA and polylactic acid(VicrylTM or polyglactin 910, a co-polymer of 90%glycolide and 10% l-lactide), polydioxanone (PDS),and non-degradable synthetics including braided polye-ster (EthibondTM or MersileneTM), nylon (EthilonTM),Teflon coated polyester (TevdekTM) and polypropylene(ProleneTM or SurgileneTM) have dominated the suturemarket. In general, sutures should be strong, handleeasily, and form secure knots [25]. Sutures require thefollowing characteristics for general surgical applica-tions [27]:

1. Tensile strength—to match the clinical repair.2. Knot strength—the amount of force required to

cause a knot to slip.3. Elasticity—the ability to conform to the current stage

of wound repair.4. Memory—change in stiffness over time; the better the

suture, the less memory.5. Degradability—ability to be metabolized by the host

once its repair function has been completed.6. Tissue Reactivity—non-irritant.

Table 1

Comparison of mechanical properties of common silks (silkworm and spider dragline) to several types of biomaterial fibers and tissues commonly

used today

Material UTS (MPa) Modulus (GPa) % Strain at break Authors

B. mori silk (w/ sericin)a 500 5–12 19 Perez-Rigueiro et al. [68]

B. mori silk (w/o sericin)b 610–690 15–17 4–16 Perez-Rigueiro et al. [68]

B. mori silkc 740 10 20 Cunniff et al. [13]

Spider silkd 875–972 11–13 17–18 Cunniff et al. [13]

Collagene 0.9–7.4 0.0018–0.046 24–68 Pins et al. [69]

Collagen X-linkedf 47–72 0.4–0.8 12–16 Pins et al. [69]

PLAg 28–50 1.2–3.0 2–6 Engelberg and Kohn [70]

Tendon (comprised of mainly collagen) 150 1.5 12 Gosline et al. [71]

Bone 160 20 3 Gosline et al. [71]

Kevlar (49 fiber) 3600 130 2.7 Gosline et al. [71]

Synthetic Rubber 50 0.001 850 Gosline et al. [71]

a Bombyx mori silkworm silk—determined from bave (multithread fibers naturally produced from the silk worm coated in sericin).b Bombyx mori silkworm silk—determined from single brins (individual fibroin filaments following extraction of sericin).c Bombyx mori silkworm silk—average calculated from data in Ref. [13].d Nephila clavipes silk produced naturally and through controlled silking.e Rat-tail collagen Type I extruded fibers tested after stretching from 0% to 50%.f Rat-tail collagen dehydrothermally cross-linked and tested after stretching from 0% to 50%.g Polylactic acid with molecular weights ranging from 50,000 to 300,000.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 403

Page 4: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

7. Free from infection—related to the material’s geo-metry, e.g., multifilament vs. monofilament.

Processing methods for virgin silk (fibroin containingsericin gum) were developed in industry to extractsericin from the inner silk fibroin fibers. As sutures, thesilk fibroin fibers are usually coated with waxes orsilicone to enhance material properties and reducefraying; these sutures are commonly referred to as blackbraided silk (e.g., Perma-HandTM). The transition fromvirgin silk to black braided sutures occurred in the late1970s and early 1980s; however, virgin silk is stillcommercially available today.

Virgin silk, like most proteins, is a potential allergen[28,29] causing a Type I allergic response in some cases[30]. Delayed allergic responses to silk (an average of 10months after initial exposure) induced some patientcomplications including asthma and specific upregula-tion in IgE levels [29,31]. Wen et al. [29] showed theextracted sericin was responsible for sensitization (thedevelopment of a T-cell mediated allergic response) by

skin testing of 64 children with asthma. Furtherbiochemical analysis by Dewair et al. [32] and Zaominget al. [33] concluded that upregulated IgEs wereproduced in response to the sericin, clarifying the roleof these fibroin contaminants as the main allergenicagent in silk and not the core fibroin fibers. Fibroindegradation products could also potentially be involvedin adverse biological responses. For example, one casewas reported where fibroin (black braided silk) mayhave induced a Type I allergic reaction [30]. However,the authors clarified later that there were no cases in theliterature that implicated black braided silk as inducer ofIgE hypersensitivity reactions. In addition, in thepreparation of this review no references were found inthe literature that implicated black braided silk ininducing hypersensitivity and allergic reactions. Kuro-saki et al. [30] concluded that the hypersensitivity intheir single case may have been the result of patientsensitization to silk from a prior surgical procedure 7years earlier. The type of silk used in this earlierexposure was unknown (e.g., virgin vs. black braided).

Table 2

Biological responses to virgin silk fibers

Type of silka Response In vivo or in vitro model/tissue site Authors

? Mucosal edema was greatest around silk and chromic

collagen compared to PGA

Dog/genitourinary tract Morrow et al. [38]

? Chronic inflammation including granulosous and fibrosis Rabbit/median or sciatic nerve

ligation

Nebel et al. [39]

Virgin (a) Acute and chronic inflammation initially including

conjunctival and episcleral hyperemia progressing to

chemosis and nodular episcleritis, peripheral corneal

ulceration and wound necrosis

Human/ocular cataract surgery Soong and Kenyon

[34]

(b) Sensitization to virgin silk during bilateral cataract

surgery following the use of virgin silk on the first eye

Black braided No comparable suture reactions Human/ocular cataract surgery Soong and Kenyon

[34]

Virgin IgE obtain from sera isolated from 8 of 9 patients with

allergy to silk bound specifically to sericin and was

negative towards fibroin

In vitro immunoblot of IgE and IgG

isolated from sera of 9 persons

allergic to silk

Dewair et al. [32]

? (a) Acute and chronic inflammation Rabbit/tracheal anastomosis Peleg et al. [40]

(b) However, the response was not significantly different

from VicrylTM, Chromic Catgut, TevdekTM and

polypropylene

? Severe delayed chronic inflammatory reaction to suture Three human pediatric patients/

neurosurgery

Rossitch et al. [31]

Virgin Delayed hypersensitivity to virgin silk resulting in asthma Children o15 yr old/skin tests Wen et al. [29]

Virgin (a) 90% of patient population developed an allergic

reaction to silk extract (sericin); none of the control

group was allergic

41 humans with asthma and a clinical

history of silk allergy; 4 control

patients with no allergy to silk/sera

and skin tests were used.

Zaoming et al. [33]

(b) IgE from 41% of the patient population sera bound

specifically to sericin in in vitro testing

? (a) Type I allergy response to black braided silk Single case in a human/trachea and

throat region

Kurosaki et al. [30]

(b) Patient sensitized to silk 7 years prior in an

independent surgical procedure

a ?=Type of silk used was not specified, e.g., virgin silk or black braided; Virgin=virgin silk containing sericin; Black braided=black braided

extracted silk fibroin (e.g., no sericin) coated in waxes or silicone.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416404

Page 5: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

Tab

le3

Co

mm

on

sutu

res

that

elic

ita

fore

ign

bo

dy

resp

on

sefo

llo

win

gim

pla

nta

tio

nin

viv

o

Su

ture

trad

en

am

e/m

ate

rial

Mo

del

/im

pla

nta

tio

nsi

teA

bso

rbab

lea

Fo

reig

nb

od

yre

spo

nse

Filam

ent

typ

ebA

uth

ors

Su

rgic

al

con

tro

lR

at/

—8.2

%(7

.3–10.6

)—

cF

osc

hi

etal.

[43]

Vic

rylT

M(p

oly

gla

ctin

910)

Ab

do

min

al

cavit

yY

es;o

60

days

8.7

%(7

.1–9.9

)M

ult

i

Catg

utT

M(C

ollagen

)Y

es;o

60

days

9.4

8%

(8.3

–15)

Mu

lti

Po

lygly

colic

aci

dsa

lt(P

GA

)Y

es;o

60

days

11.2

5%

(9.5

–12.7

)M

ult

i

Sil

k(a

ssu

med

bla

ckb

raid

ed)

Yes

;>

60

da

ys

14

.2%

(10

.9–2

2.3

)M

ult

i

Tit

an

ium

(met

al

clip

)N

o15.8

%(1

2.1

–19.1

)M

on

o

Extr

ud

edT

eflo

nT

MR

ab

bit

/N

oM

ild

(1)

Mo

no

dS

etze

net

al.

[44]

Pro

len

eTM

(po

lyp

rop

yle

ne)

Su

bcu

tan

eou

sN

oM

ild

(1)

Mo

no

Su

rgilen

eTM

(po

lyp

rop

yle

ne)

No

Mild

(1)

Mo

no

PD

ST

M(p

oly

dio

xan

on

e)Y

es;o

60

days

Mo

der

ate

(2)

Mu

lti

Vic

rylT

MY

es;o

60

days

Mo

der

ate

(2)

Mu

lti

Sil

k(b

lack

bra

ided

)Y

es;

>6

0d

ay

sM

od

era

te(2

)M

ult

i

Nylo

nN

oM

od

erate

(2)

Mo

no

Eth

ibo

nd

TM

(bra

ided

po

lyes

ter)

No

Mo

der

ate

(2)

Mu

lti

Mer

silin

eTM

(bra

ided

po

lyes

ter)

No

Mo

der

ate

(2)

Mu

lti

Tev

dek

TM

(Tefl

on

coate

d

po

lyes

ter)

No

Mo

der

ate

(2)

Mu

lti

Ch

rom

icC

atg

utT

M(c

ross

lin

ked

collagen

)

Yes

;o

60

days

Exte

nsi

ve

(3)

Mu

lti

Nylo

nR

at/

No

Min

imal

Mo

no

eB

uck

nall

etal.

[45]

Nylo

nA

bd

om

inal

inci

sio

nN

oM

inim

al

Mu

lti

PG

AY

es;o

60

days

Min

imal

Mu

lti

Sil

k(b

lack

bra

ided

)Y

es;

>6

0d

ay

sM

od

era

teM

ult

i

aA

bso

rbab

leis

defi

ned

as

the

loss

of

ten

sile

stre

ngth

wit

hin

60

days

inviv

o,

bu

td

oes

no

tre

flec

tth

ep

ersi

sten

ceo

fa

fore

ign

bo

dy

resp

on

se(F

BR

)fo

rgre

ate

rth

an

60

days.

bM

on

ofi

lam

ent

sutu

res

con

tain

asi

ngle

fib

eran

dm

ult

ifila

men

tsu

ture

sco

nta

inm

ult

iple

fib

ers

incr

easi

ng

mate

rial

surf

ace

are

ain

viv

oan

dth

ep

ote

nti

al

risk

of

infe

ctio

n(i

.e.,

mo

rep

lace

sfo

r

con

tam

inati

on

toh

ide

inm

ult

ifila

men

tsu

ture

s).

cF

RB

was

chara

cter

ized

by

the

per

cen

tgro

wth

of

neo

ves

sels

(an

gio

gen

esis

)aro

un

dth

esu

ture

7d

ays

po

st-i

mp

lan

tati

on

.dF

BR

was

chara

cter

ized

on

a3

level

scale

30

days

po

st-i

mp

lan

tati

on

:M

ild

(1)

incl

ud

edth

ep

rese

nce

of

his

tio

cyte

san

dfo

reig

n-b

od

ygia

nt

cells,

Mo

der

ate

(2)

ind

icate

sh

isti

ocy

tes

an

dfo

reig

n-b

od

y

gia

nt

cells,

an

dcl

ust

ero

fly

mp

ho

cyte

san

da

few

pla

smacy

tes,

an

dE

xte

nsi

ve

(3)

incl

ud

esh

isti

ocy

tes,

fore

ign

-bo

dy

gia

nt

cells,

an

dd

iffu

se,

scatt

ered

lym

ph

ocy

tes

an

dp

lasm

acy

tes.

eF

BR

was

chara

cter

ized

his

tolo

gic

ally

10

days

inviv

o;

ho

wev

er,

the

au

tho

rsd

on

ot

pro

vid

ea

defi

nit

ion

of

‘min

imal’

or

‘mo

der

ate

’in

flam

mati

on

an

dp

rovid

en

om

ean

sb

yw

hic

hth

ey

syst

emati

cally

chara

cter

ize

the

resp

on

seh

isto

logic

ally.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 405

Page 6: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

In other cases where the type of silk suture wasunknown [31], silk induced a delayed allergic reactionthat necessitated the removal of the suture. The authorsdid not distinguish between the use of virgin silkcontaining sericin versus black braided silk. However,based on the time frame of the surgical procedures(1970s) and the severity of the reaction, sericin can beconsidered as the most likely cause of the response.Soong and Kenyon [34] detailed 12 patients from 1980to 1983 that reported severe reactions to virgin silksutures following cataract surgery. No comparablereactions were noted when black-braided silk sutureswere used. Further, they provided evidence of patientsensitization to virgin silk in bilateral surgical proce-dures; an allergic reaction developed against virgin silkfollowing its use in the first eye procedure.

The use of virgin silk during the 1960s to the early1980s negatively impacted the general acceptance of thisbiomaterial from the surgical practitioner perspective[35–37]. For example, Morrow et al. [38] observed silkand collagen (catgut) to be moderately more reactivethan polyglycolic acid (PGA) in the genitourinary tractfollowing 3 weeks of implantation in vivo, althoughagain the type of silk used in the study was notidentified. Nebel et al. [39] observed cellular infiltration,granulosous and fibrosis for silk sutures that persistedfor more than 4 weeks compared to autologous collagensutures used for peripheral nerve ligation in the rabbit.Again the specific type of silk was not described. Of notein the study was that inflammation was comparable tochromic collagen sutures. Peleg et al. [40] drew a similarconclusion regarding the inflammatory potential ofsilk used in tracheal anastomosis in the rabbit with-out identifying the type of silk used. Of interest was asimilar acute and chronic inflammatory response toVicrylTM and TeflonTM coated polyester sutures in thestudy.

4. Foreign body response to silk

All biomaterials derived from a non-autologoussource will elicit some level of foreign body response(FBR) following implantation in vivo. Absorbable andnon-degradable biomaterials generally do not induce aT-cell mediated hypersensitivity immune response (aswas observed with sericin) [30]. However, while rare,sensitization to fibroin due to pre-exposure or a failedphagocytic response can result in the formation of agranuloma years after the implantation of black braidedsilk [30]. Some common materials that have been shownto induce a FBR are detailed in Table 3.

Biomaterial characteristics, including implantationsite, size, geometry [41], and surface topography caninfluence the level of the foreign body response [42]. Thisresponse can be predicted in part based on the surface tovolume ratio of the biomaterial [42]. This relationship isfrequently not addressed in the relative comparisonbetween different suture types in vivo. Several studieshave shown black braided silk to elicit a greaterFBR when compared to other commonly used sutures[43–45]. However, a single fiber of native silk iscomposed of several individual fibroin filaments and asingle black braided suture comprises several extractedsilk fibers. No studies have attempted to normalize theFBR to the suture surface area, rather equating suturesbased on their final diameter. Thus studies that reportsilk as inducing a heightened FBR may not be reflectiveof the inflammatory potential of the silk but rather amanifestation of the overall suture structure andgeometry.

Silk sutures induced angiogenesis, a limiting step inmounting a foreign body response, in the rat’smesenteric window 7 days post-implantation to anlesser extent as titanium and to a greater extent thancollagen, polyglactin, and PGA; however the differences

Table 4

Benefits and concerns with the use of silks for biomedical applications

Benefits

Novel mechanical properties of some silks that are superior to any other natural fiber and rival many high performance fibers

Natural fiber with a long standing history of use in clinical applications

The ability to process silks in aqueous solutions for subsequent formation of films and other material formats, with relatively simple

insolubilization via exposure to alcohols and other environmental factors

Easily chemically modified with surface decorations, such as adhesion sites or cytokines, due to the availability of amine and acid side chains on

some of the amino acids

Genetically tailorable composition and sequence to moderate specific features, such as molecular weight, crystallinity, solubility

Slow rates of degradation in vitro and in vivo, this is particularly useful in biodegradable scaffolds in which slow tissue ingrowth is desirable

No known risk of bioburden

Concerns

Adequate removal of contaminating sericin from silkworm silk to avoid biocompatibility problems

Slow degradation of crystalline (b-sheet) regions

Aborted proteolytic attack by macrophages and giant cells leading to encapsulation and the formation of a granuloma

Potential sensitization to silk fibroin resulting in an allergic response upon exposure to the biomaterial

G.H. Altman et al. / Biomaterials 24 (2003) 401–416406

Page 7: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

were not statistically significant ðp > 0:05Þ (Table 3) [43].The type of silk suture (virgin versus black braided) wasnot described but it is assumed to be black braidedbased on the year of study. A curiosity about the studywas the choice to close all the wounds of all studygroups with silk suture, potentially sensitizing theanimals to the silk suture under investigation. Bucknallet al. [45] examined four sutures in abdominal woundclosure in a rat model 10, 30, and 70 days post-implantation. Histological examination revealed silkelicited a ‘moderate’ zone of inflammation comparedto a ‘minimal response’ induced by multi- and mono-filament nylon and multifilament PGA (Table 3). Nohistological criteria or ranking system was provided.However, the authors noted that even monofilamentnylon, thought to be the most biocompatible of thematerials evaluated, induced fibrous encapsulation 10days post-implantation. Of interest was the finding thatPGA, despite a 92% decrease in tensile strength 30 dayspost-implantation continued to elicit a FBR over 70days in vivo, the final time point of the study.

Setzen et al. [44] in a comprehensive blind study,statistically analyzed tissue responses to 11 types ofsutures subcutaneously implanted in the rabbit (Table 3).Responses were based on the number of giant cellspresent in the fibrous capsule and within the individualfilaments of the sutures, and the thickness of the fibrouscapsule surrounding the suture. Evaluations wereperformed 30, 60 and 120 days post-implantation. Theinflammatory response surrounding the encapsulationwas graded on a scale of 1–3 based on the type of cellsand the number of vessels found. Silk elicited anequivalent response to absorbable sutures and multi-filament non-absorbable sutures when evaluating thenumber of foreign body cells surrounding each suture 30days post-implantation. Non-degradable EthibondTM

(braided polyester) and TevdekTM(Teflon coated polye-ster) induced a significantly greater accumulation of cellsat the wound site compared to the group of suturesunder investigation. However, when assessing thethickness of the fibrous capsule, black braided silk aswell as EthibondTM induced the formation of signifi-cantly thicker capsules compared to the mean responseof the group. Histological analysis 30 days afterimplantation showed the tissue response to the silkwas equivalent to VicrylTM and PDSTM (two commonsynthetic absorbables), nylon (monofilament), andEthibondTM, MersileneTM and TevdekTM (three com-mon multifilament non-degradables), and less thancross-linked collagen (Chromic CatgutTM). The foreignbody response to VicrylTM lasted in excess of 120 days invivo, long after the loss of tensile integrity. Inconclusion, Setzen et al. [44] confirmed that theinflammatory response induced by silk was no greaterthan that in response to common absorbable sutures 30days post-implantation in a subcutaneous rabbit in vivo

model (Table 3). Furthermore, they found a significantlyhigher response to multifilament sutures than monofila-ment, confirming the influence of higher surface area tovolume ratio as indicative of induction of a greaterinflammatory response.

As a suture, silk is still popular in ocular, neural andcardiovascular surgery, but has also been used in avariety of other tissues in the body. Silk’s knot strength,handling characteristics and ability to lay low to thetissue surface make it a popular suture in cardiovascularapplications where bland tissue reactions are desirablefor the coherence of the sutured structures [36].However, black braided silk is thrombotic followingimplantation within ateriovessels [46,47]. Using a ratmodel, Dahike et al. [47] showed black braided silkbinds fibrin and platelets within 3 days in vivo followingimplantation into arteriovessels. Within 7 days, silk wasencapsulated by a dense thrombus consisting of plateletsand lymphocytes, erythrocytes, and granulocytes. From14 to 28 days, the gradual decline of the thrombus andthe development of a new endothelium layer wasobserved. By 28 days in vivo, silk was completelyendothelialized, indicating the presence of regeneratedendothelial tissue. While silk was initially thrombotic(e.g, 3 and 7 days post-implantation) when compared toProleneTM, EthilonTM, VicrylTM and MersileneTM, by28 days in vivo, silk was less so when compared to bothEthilonTM and VicrylTM. At 56 days post-implantationonly traces of thrombogenicity were detected with blackbraided silk by the presence of loosely bound platelets tothe neoendothelium.

The ability of black braided silk to induce thrombosisimmediately following implantation in blood flow maybe due to the surface properties and ability to bindproteins in the clotting cascade such as fibrin. Alteringthe surface properties of extracted fibroin silk (i.e.,eliminating the wax coating) significantly diminished theinitially high thrombotic response to silk [48]. Sakabeet al. [49] coated polyester suture with solubilized fibroinand demonstrated that the reconstituted fibroin proteindid not induce significant thrombogenicity in vivo. Invitro studies performed by Santin et al. [50] confirmedthe ability of silk fibroin films (extracted sericin-freefibroin dissolved in LiBr salt, dialyzed in dIH2O, castand made insoluble with methanol) to bind fibrinogen asa mechanism by which the alternative complementpathway is activated in vivo. However, compared totwo reference materials, polystyrene and poly(2-hydro-xyethyl methacrylate), fibroin films bound fibrinogen toa lesser extent [50]. C3 and IgG, components of thehuman plasma complement system bound similarlywhen the silk was compared to the reference materials.The authors concluded that C3 binding may be theresult of specific binding to hydrophobic patches (e.g., b-sheets) of fibroin. When compared to the referencematerials after 24 h in vitro, diminished interleukin-1b

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 407

Page 8: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

production from mononuclear cells isolated from hu-man plasma incubated with fibroin was observed [50].

Uff et al. [51] examined the effects of soluble factorsfrom sutures on macrophage activation (e.g., attach-ment, phagacytosis and the production of lysozyme andproinflammatory tumor necrosis factor (TNF)) in vitro.Soluble factors were acquired by subjecting sterilesutures to ultrasound for 1 h in detergent. Solublefactors from silk were found to elicit the highestphagocytic response from macrophages when comparedto catgut, PDSTM, steel, nylon and VicrylTM. However,VicrylTM induced the highest response when character-izing macrophage adherence and TNF production.PDSTM induced the highest production of lysozyme ofthe group of sutures extracted. The authors failed tocharacterize the soluble factors produced by sonificationof the suture materials. In the case of silk, the resultingresponse from macrophages could be due to a variety offactors including residual sericin, waxes or silicones usedin manufacture of the sutures, or the specific size offibroin particles or crystals potentially generated. Thus,the response may be completely independent of actualsilk fibroin material properties and rather a function ofthe coating and/or geometry and size of particlesproduced.

5. Silk degradation

According to the US Pharmacopeia an absorbable(suture) biomaterial is defined as one that ‘loses most ofits tensile strength within 60 days’ post-implantation invivo. Within this definition, silk is correctly classified asnon-degradable. However, according to the literature,silk is degradable but over longer time periods due toproteolytic degradation usually mediated by a foreignbody response [31,34,41,52]. Several studies detailvariable rates of silk absorption in vivo dependent onthe animal model and tissue implantation site (Table 5).In general, silk fibers lose the majority of their tensilestrength within 1 year in vivo, and fail to be recognizedat the site within 2 years [53].

Silk studies in vitro have demonstrated that proteasessuch as chymotrypsin will cleave the less-crystallineregions of the protein to peptides which are then capableof being phagocytosed for further metabolism by thecell. Furthermore, protease cocktails [54] and chymo-trypsin (known to be produced by macrophages) arecapable of enzymatically degrading silk [55]. Of interest,the silkworm B. mori produces a protease inhibitorin the silk gland embedding it within the silk cocoonfor protection against premature proteolytic degrada-tion [56].

In a comparative study of six absorbable and fournon-degradable sutures implanted circumferentiallyunder the skin of rats, silk lost 55% of its tensile T

ab

le5

Evid

ence

of

silk

deg

rad

ati

on

invit

roan

din

viv

o

Typ

eo

fsi

lka

Inviv

o/i

nvit

roM

ech

an

ism

Deg

ree

an

dm

easu

reo

fd

egra

dati

on

Au

tho

r

Extr

act

edfi

bro

infi

lmIn

vit

roP

rote

oly

tic

deg

rad

ati

on

B10%

wei

gh

tlo

ss5

days

follo

win

g

enzy

mati

cd

iges

tio

n

Min

ou

raet

al.

[54]

Un

kn

ow

n/a

ssu

med

bla

ckb

raid

edR

at/

sub

cuta

neo

us

Un

kn

ow

n/a

ssu

med

fore

ign

bo

dy

resp

on

se

55%

loss

inte

nsi

lest

ren

gth

6w

eek

s

inviv

o

Gre

enw

ald

etal.

[57]

Bla

ckb

raid

edR

at/

sub

cuta

neo

us

Un

kn

ow

n/a

ssu

med

fore

ign

bo

dy

resp

on

se

83%

loss

inte

nsi

lest

ren

gth

10

wee

ks

inviv

o

Bu

ckn

all

etal.

[45]

Un

kn

ow

n/a

ssu

med

bla

ckb

raid

edR

at/

ab

do

min

al

wall

mu

scle

Fo

reig

nb

od

yre

spo

nse

(pro

teo

lyti

c

deg

rad

ati

on

)

Fra

gm

enta

tio

nat

6w

eek

s;n

ot

det

ecte

d

at

24

wee

ks

Lam

etal.

[41]

Bla

ckb

raid

edR

ab

bit

/co

rnea

,sc

lera

an

do

cula

rm

usc

leF

ore

ign

bo

dy

resp

on

se(p

rote

oly

tic

deg

rad

ati

on

)

Red

uce

dn

um

ber

of

fila

men

tsan

d

dia

met

erat

42

days;

ab

sorp

tio

nat

90

days

inviv

o

Salt

ho

use

etal.

[52]

Un

kn

ow

n/a

ssu

med

vir

gin

silk

Rab

bit

/ab

do

min

al

wall

mu

scle

Fo

reig

nb

od

yre

spo

nse

(pro

teo

lyti

c

deg

rad

ati

on

)

80%

dec

rease

inte

nsi

lest

ren

gth

at

12

wee

ks;

0%

stre

ngth

at

2yea

rs;

dec

rease

inth

en

um

ber

of

fib

ers

ob

serv

ed

his

tolo

gic

ally;

fragm

enta

tio

nfo

llo

win

g

4w

eek

sin

viv

o

Pro

stle

thw

ait

[36]

aT

yp

eo

fsi

lk:

vir

gin

=ra

wsi

lkco

nta

inin

gse

rici

n;

bla

ckb

raid

ed=

extr

act

edsi

lkfi

bro

infi

ber

s(e

.g.,

no

seri

cin

)co

ate

din

waxes

or

silico

ne;

fib

roin

film

=ex

tract

edan

dso

lub

iliz

edfi

bro

inca

stan

d

inso

lub

iliz

edw

ith

met

han

ol.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416408

Page 9: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

strength and 16% of its elastic modulus 6 weeks post-implantation [57] (Table 6). In a rat model withsubcutaneous implantation, silk fibers lost 29% of theirtensile strength at 10 days, 73% at 30 days and 83% 70days post-implantation (Table 7) [45]. In abdominalwound closures in the rat, silk promoted a moderateforeign body response compared to mono- and multi-filament nylon and polyglycolic acid (PGA) sutures [45].However, common to all sutures, inflammation wasrequired as the main mechanism for degradation.

Lam et al. [41] described silk as biodegradable due toits susceptibility to proteolytic enzymes. The studycompared dry-spun hot-drawn poly(l-lactic acid)(PLLA) fibers to several absorbable and non-degradablesutures in the muscle layer surrounding the abdomen ofrats. PLLA, PDSTM, VicrylTM, and black braided silkshowed signs of degradation after 2 weeks in vivo asdetermined by scanning electron microscopy. Allsutures, in addition to monofilament nylon, provokeda chronic inflammatory response at 2 weeks. At 6 and 12weeks in vivo, the authors were unable to retrieve silkdue to fragmentation. At 24 weeks, neither silk norinflammation was observed in vivo most likely due therapid degradation of silk by proteolytic enzymaticdigestion. Inflammation subsided by 6 weeks in vivofor Vicryl; a chronic inflammatory response was presentsurrounding PLLA at 80 weeks in vivo, the final timepoint of the study.

In general, silk is slowly absorbed in vivo. The rate ofabsorption is dependent upon the implantation site,mechanical environment, and variables related to thehealth and physiological status of the patient, the type(virgin silk versus extracted black braided fibroin), andthe diameter of the silk fiber [31,34,52]. Furthermore,alterations in silk processing may cause conformationalchanges in the protein structure potentially increasing ordecreasing susceptibility to degradation. None ofthese variables has been studied in detail; therefore,it is difficult to gain a clear understanding of therelationships between structure, processing and degrad-ability. Regardless, silk protein fibers will degrade invivo; rates are variable depending on the factors listedabove.T

ab

le6

Mate

rial

pro

per

ties

of

10

com

mo

nsu

ture

sfo

llo

win

g6

wee

ks

of

sub

cuta

neo

us

imp

lan

tati

on

ina

rat

mo

del

.V

alu

esat

tim

e0–6

wee

ks

tak

enfr

om

Tab

le1

of

Ref

.[5

7]

Su

ture

nam

eS

tren

gth

(N/m

2)a

Tan

gen

tel

ast

icm

od

ulu

s(N

/m2)b

0W

eek

s6

Wee

ks

%D

ecre

ase

0W

eek

s6

Wee

ks

%D

ecre

ase

Vic

rylT

M(p

oly

gla

ctin

910)

1.4

2E

14

Fra

gm

ente

dB

100

3.0

0E

8—

100

Dex

on

TM

(po

lygly

colic

aci

d)

3.4

6E

14

Fra

gm

ente

dB

100

2.6

2E

8—

100

Catg

utT

M(c

ollagen

)2.3

0E

14

Fra

gm

ente

dB

100

1.4

1E

8—

100

Ch

rom

icC

atg

utT

M(c

ross

-lin

ked

collagen

)2.2

8E

14

Fra

gm

ente

dB

100

1.5

6E

8—

100

PD

ST

M(p

oly

dio

xan

on

e)7.2

0E

14

9.7

2E

13

86.5

9.8

7E

77.1

3E

785.8

Maxo

nT

M(p

oly

gly

con

ate

)5.9

6E

14

8.2

9E

13

86.1

1.0

0E

87.4

E7

26

TM

Sil

kc

8.4

8E

13

3.8

0E

13

55

.22

.42

E8

2.0

3E

81

6.1

Eth

ilo

nT

M(n

ylo

n)

4.5

7E

14

2.5

4E

14

44.4

1.0

4E

81.0

7E

80

Pro

len

eTM

(po

lyp

rop

yle

ne)

4.0

2E

14

2.9

9E

14

25.6

1.0

0E

81.0

1E

80

Eth

ibo

nd

TM

(po

lyes

ter)

2.4

9E

14

2.3

2E

14

6.8

2.4

7E

82.3

0E

86.9

aS

tren

gth

of

init

ial

sutu

rep

rio

rto

imp

lan

tati

on

(0w

eek

s)an

dat

6w

eek

sfo

llo

win

gin

viv

oci

rcu

mfe

ren

tial

sub

cuta

neo

us

imp

lan

tati

on

ina

rat

mo

del

.F

ragm

ente

dsu

ture

sth

at

wer

eu

nab

leto

be

harv

este

dfo

llo

win

gim

pla

nta

tio

nat

6w

eek

sw

ere

ass

um

edto

have

lost

100%

of

thei

rte

nsi

lein

tegri

ty.

bE

last

icm

od

ulu

so

fsu

ture

s(N

/m2)

pri

or

toim

pla

nta

tio

nan

dfo

llo

win

g6

wee

ks

inviv

o.

cT

yp

eo

fsi

lk(e

.g.,

vir

gin

or

bla

ckb

raid

ed)

was

no

td

escr

ibed

.It

isass

um

edto

be

bla

ck-b

raid

edsi

lk.

Table 7

Percent loss in suture mechanical tensile strength over time sub-

cutaneously implanted in a rat model. Durability data was taken from

Table 3 of Ref. [45]

Suture Time post-implantation

10 days 30 days 70 days

Polyglycolic acid 65% 92% 99%

Silk, black braided 29% 73% 83%

Multifilament nylon 16% 19% 22%

Monofilament nylon 3% 7% 16%

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 409

Page 10: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

It should be recognized that amyloid-like deposits inAlzheimer’s and related diseases are often associatedwith b-sheet structures, particularly cross-beta crystals[58]. Similar structures are also implicated in membrane-associated cytotoxicity [58]. It is unknown if silks, suchas those from silkworm, spiders or genetic variantsthereof, will negatively influence biological function in asimilar mode as these amyloid systems. This issue shouldbe addressed over time as tissue-engineering studiesmove forward. The outcomes of these studies wouldinfluence the types of tissue applications that should beaddressed with silk. It is encouraging that no amyloid-related problems have been reported throughout theliterature to date associated with silk fibers, despite theirextensive use in the clinical setting for many centuries.As the use of silk-based biomaterials broadens, pre-sumably the ASTM would be involved to establishstandard guidelines for this type of assessment.

6. Scaffolds for tissue engineering

The emergence of tissue engineering has increased thedemand for a diverse portfolio of biomaterials tosupport the development of tissues in vitro prior toimplantation in vivo. The biomaterial or matrix plays akey role in communicating or transducing environmen-tal cues to cells seeded within or on the matrix. Inessence, the matrix acts as the translator between thelocal (e.g., mechanical) environment (either in vitro or invivo) and the developing tissue, aiding in the develop-ment of biologically viable functional tissue. The matrixmust support cell attachment, spreading, growth anddifferentiation. In most instances it is advantageous ifthe matrix degrades into biocompatible fragments ormonomers capable of being metabolized by host cells;however, the rate of degradation must match or be lessthan the rate of tissue ingrowth and development. This

balance assures appropriate mechanical and physiologi-cal compatibility during integration of the host and/orimplanted tissue in vivo. A mismatch in these rates canlead to premature failure of the tissue. Silk fibroin offersversatility in matrix scaffold design for a number oftissue engineering needs (Table 8) in which mechanicalperformance and biological interactions are majorfactors for success, including bone, ligaments, tendons,blood vessels and cartilage. Silk fibroin can be processedinto foams, films, fibers and meshes.

Inouye et al. [59] demonstrated the utility of a silkfibroin film in culturing animal cells (SE1116, humancolon adenocarcinoma; KB, human mouth epidermoidcarcinoma; Colo201, human colon adenocarcinoma;QG56, human lung carcinoma) in comparison to acollagen matrix. Films of both fibroin and collagensupported equivalent cell growth after 5 days in cellculture conditions. Minoura et al. [26] compared theability of fibroin and sericin films, as well as a control,collagen (cast bovine acid soluble) film, in supportingattachment, spreading and growth of the L-929 fibro-blast cell line. The results indicated that fibroin andcollagen films were equivalent in their ability to supportcell attachment, physiological morphology and growthwhen compared to the sericin film. These results supportthe concept that extracted fibroin free of sericin is asuitable matrix for cell and tissue culture.

In our own studies, fibroin films induced bone tissuegrowth in vitro when seeded with osteoblasts [60]. Whenthe films were chemically decorated with the peptideRGD to promote integrin interactions for adhesion, theinduction of bone formation in vitro was significantlyenhanced. The response was determined based onincreased alkaline phosphatase levels, upregulation ofbone-specific transcripts, and calcification levels over 4weeks. Similar responses were not observed whenparathyroid hormone was immobilized and also assayedwith osteoblasts. The utility of a protein matrix offering

Table 8

Examples of silk’s utility as a matrix material in tissue engineering

Silk form Supported cell type in vitro Comments Authors

Fibroin film L-929 mouse fibroblast Comparable growth rates to collagen films Minoura et al. [26]

Fibroin film SE1116 (human colon adenocarcinoma); KB

(human mouth epidermoid carcinoma);

Colo201 (human colon adenocarcinoma);

QG56 (human lung carcinoma)

Comparable growth rates to collagen films as

well as rates of protein production of

carcinoembryonic antigen (CEA)

Inouye et al. [59]

Fibroin film Saos-2 (human osteoblast-like cells) Bone formation was evident on fibroin films,

but was enhanced on RGD-coupled matrices

Sofia et al. [60]

Fibroin film hBMSC (human bone marrow stromal cells) Supports bone nodule formation from adult

stem cells

(Unpublished data)

Fibroin fibers hBMSC; human adult anterior cruciate

ligament fibroblasts

Supports ligament specific development

in vitro

Altman et al. [61]

G.H. Altman et al. / Biomaterials 24 (2003) 401–416410

Page 11: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

a diversity of sites (e.g., amino acids) for selectivechemical couplings for tissue engineering is a benefit ofthe silk system. Bone tissue formation was a logicaltarget tissue for silk due to the unique mechanicalproperties of the protein in fiber and film forms.

Recent research with silk has focused on the devel-opment of a wire rope matrix for the development ofautologous tissue engineered anterior cruciate ligaments(ACL) using a patient’s own adult stem cells [61].Biologically compatible and mechanically robust bio-materials are critical based on the stringent requirementsof ligaments and the dynamic and demanding mechan-ical environment of the knee. Human bone marrowstromal cells (BMSCs) cultured in collagen gels weregrown in a mechanically dynamic environment relevantto that present in vivo leading to the formation ofligament fibroblasts (from the BMSCs) and maturetissue development in vitro [62]. However, the poorintegrity of the collagen gels and the demanding anddynamic intra-articular mechanical and biochemicalenvironment of the knee have prompted renewedinterest in silk as a long term absorbable material withgood mechanical integrity and biocompatibility.

Collagen fibers have been used as a matrix biomater-ial to support de novo ligament formation in vivo withlimited success. In the early 1990s Dunn et al. [63]reported initial work on the development of a ligamentprosthesis involving a collagenous ACL prosthesis [63].Results showed inconsistent neoligament formation andsignificant weakening of the prosthesis in a rabbitmodel. Enhanced scaffolds were examined including acollagen fiber-PLA composite to further maintainmechanical integrity allowing for neoligament tissueingrowth [64]. In both studies, only half of the structuresremained intact 4 weeks post-reconstruction suggestingthat this composition (collagen and PLA) was inade-quate for the rigorous in vivo environment of the ACL.

Silk’s unique mechanical properties, coupled with theability to weave the fibers into a wire-rope geometryprovides control over the matrix’s final mechanicalproperties (i.e., matrix stiffness can be altered byadjusting the pitch angle of a wire rope) to mimic themechanical properties of the native ACL and supporthost tissue ingrowth offering new options in ACL tissueengineering. Pilot scale manufacturing equipment hasbeen developed for the fabrication of ACL wire-rope

Fig. 1. (A) Pilot-scale manufacturing equipment for the fabrication of silk wire-rope matrices showing (i) two computer controlled twisting

machines, (ii) a 160 l stainless steel water bath with immersion heaters, and (iii) a custom designed fiber extraction rack holding 94 individual groups

of fibers to be extracted free of sericin; (B) a close up of the extraction rack containing 94 individually anchored groups of fibers; (C) spring-loaded

clamps used to keep fibers or groups of fibers in equal tension during extraction, rinsing and drying.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 411

Page 12: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

fibroin matrices (Fig. 1). A 160 l stainless steel waterbath (Fig. 1A) and custom designed fiber extractionracks to keep individual fibers or groups of fibers inequal tension were developed. The extraction rack wasdesigned to anchor up to 94 individual fibers or groupsof fibers (Fig. 1B) with spring-loaded clamps (Fig. 1C).The water bath (214 cm� 61 cm� 15 cm) combined withtwo immersion heaters and an external pump allowrecirculating flow of the extraction broth at 901Cover the rack containing the silk. Following sericinextraction, two computer controlled twisting machines(Figs. 1A and 2) were developed to fabricate wire-ropematrices with desired geometries (e.g., material proper-ties). One of many fabricated matrices is shown inFig. 3. Confirming silks utility as a biocompatiblematerial for in vitro ligament tissue engineering, BMSCswere shown to attach, spread and proliferate on the silkfiber matrices [61]. Scanning electron microscopy (SEM)images of virgin silk and extracted silk (Fig. 4) as well asBMSC seeded fibroin wire-rope matrices are shown(Fig. 5). BMSC are able to attach within 5 min(Fig. 5A), initially spread at 1 h (Fig. 5B), and formconfluent cell sheets with possible extracellular matrixformation 7 days and 14 days post-seeding (Figs. 5Cand D).

It is generally accepted in the literature that silk willslowly degrade in vivo as a function of proteolyticattack. However, in vitro in immune system deficienttissue culture conditions, sericin-extracted fibroin silkfibers retained their initial tensile integrity over 21 days

[61]. In vitro studies of the extracted silk fibroinexhibited a negligible response from macrophages asassessed by cytokine release in vitro (unpublished data).As a result, appropriate, directed and rigorous mechan-ical stimuli can be communicated to adhered stem cellsin vitro without concern for premature graft mechanicalfailure; however, the effects of mechanical fatigue ongraft integrity will need to be characterized for eachculture condition and variant matrix geometry.

We have shown that the application of mechanicalstimuli will induce BMSC ligament specific differentia-tion and matrix formation in vitro [62]. The utility ofthis tissue engineering approach in combination with amechanical robust silk matrix is the potential inhibitionof a FBR following implantation in vivo due to the

Fig. 2. A close-up view of the twisting machines showing the motor controlled spring-loaded clamps. The clamps can anchor from 2 to 6 fibers or

groups of fibers for twisting.

Fig. 3. One of many silk cords manufactured by the twisting

equipment. The silk cord shown contains 5 levels of twisting hierarchy

and 540 individual fibers twisted to approximate the stiffness of the

human ACL.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416412

Page 13: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

Fig. 4. (A) SEM of raw virgin B. mori silk fibers prior to extraction showing the gum-like sericin proteins coating the core fibroin and (B) following

extraction at 901C for 60 min in conditions previously described [59] showing the individual smooth fibroin filaments following the removal of the

covering sericin.

Fig. 5. (A) Initially attached BMSCs on the fibroin cord shown in Fig. 3, 5 min following seeding of the fibers with 2 million cells/ml (1 ml total per

3 cm of silk cord). (B) Initial cell spreading on the fibroin 1 h after seeding. (C) Seven days post-seeding with a cell and ECM sheet evident. (D)

Fourteen days following seeding, thick encapsulation of the matrix by cells and ECM.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 413

Page 14: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

presentation of autologous ligament tissue (derived fromthe BMSC) to the host. This approach should allowboth infiltration of host tissue and the in vitro developedligament tissue incorporated into and surrounding thematrix to form mature functional, biologically viableautologous ligament tissue. Studies are currently on-going to identify an optimal matrix geometry andstiffness to best support tissue ingrowth both in vitroand in vivo. Furthermore, silk’s high ultimate tensilestress (N/mm2) provides significant void volume in vivooccupying only B12% of the space of a human ACL(given a human ACL 27 mm in length and 8 mm indiameter). We suspect, following angiogenesis andrevascularization of the tissue engineered ligamentwithin 8–12 weeks in vivo, the core original silk matrixwill be proteolytically degraded and metabolized in vivo.The effects of the rigorous mechanical knee jointenvironment on matrix fatigue life over the long termremain to be determined but will clearly guarantee theeventual failure of the silk matrix in vivo, a desired goalto ensure biologically viable, host ligamentous tissueeventually sustains the functional roles of the ACL overthe life of the patient.

In terms of matrices for tissue engineering, the novelmechanical properties of silk in film, fiber (Table 1) orsponge forms, coupled with the facile chemical decora-tion, the potential to form complex rope designs tomatch functional requirements for specific tissues, andthe slow rate of degradation in vivo to provide adequaterobust support, suggest that silks offer many benefitswhen compared to other types of natural or syntheticdegradable fibers, films and foams. For example, in thecase of bone, biomaterials that can provide robustsupport during compression would be advantageous,and silks are known to function well under compressionwith little evidence for cracking or crazing underconditions in which synthetic high performance fibersfail [13]. Furthermore, in the case of ligaments, the hightensile strength of silks provides a strong advantage inmatching ligament function to immediately restore kneefunction, a feature difficult to achieve with collagen [61].

Silks fibers and films are often prepared fromresolubilized protein after dissolution in high concentra-tions of lithium salts or calcium nitrate. Once soluble,the proteins can be dialyzed, lyophilized and resolubi-lized in organic solvents such as hexafluoroisopropanolor in water for limited periods of time depending onconcentration. In the case of fiber formation, silks[65,66] and collagens [67] have been electrospun to formnanometer-scale diameter fibers. Thus, modulation infiber diameters from the native 10’s of microns in thecase of B. mori to submicron by electrospinning offers arange of morphologies for cell interactions. It is alsointeresting to note that unrestrained dragline silk fromN. clavipes will supercontract. However, when em-bedded in a material or restrained, this unusual feature

is lost. Silkworm silk fibers from B. mori do notsupercontract.

7. Conclusions

B. mori silk fibers are composed primarily of twotypes of proteins: (1) sericin, the antigenic gum-likeprotein surrounding the fibers and (2) fibroin, the corefilaments of silk comprised of highly organized b-sheetcrystal regions and semi-crystalline regions responsiblefor silk’s elasticity compared to fibers of similar tensileintegrity. Silk has been used in biomedical applicationsfor centuries primarily for the ligation of wounds. Virginsilk suture (containing sericin) induces hypersensitivityin patients, causing a Type I allergic reaction. Exposureto silk debris (e.g., broken virgin silk fibers used inbedding and fabrics) may sensitize patients to silkcausing adverse allergic reactions when silk is used asa suture material. Sericin, identified as the antigenicagent of silk, is removed and replaced with a wax orsilicone coating in commercial black braided silksutures. As a result, T-cell mediated hypersensitivityhas not been observed in response to black braided silkin the literature. Unfortunately, poor documentation ofthe exact type of silk suture used when describing hostresponses to silk in the literature has led to confusionregarding the utility of silk in biomedical applications.As a result, the use of silk over the past 20 years hasdeclined since the arrival of commonly used absorbableand non-degradable synthetics such as VicrylTM andEthibondTM.

Another misconception about silk revolves around itsability to degrade in vivo. Defined as non-degradable bythe USP because it retains the majority of its tensilestrength beyond 60 days in vivo, silk is commonlythought of as a permanent suture once implanted intothe body. As a protein, silk is susceptible to proteolyticdegradation, but over a longer time periods. Based onthe literature, it is apparent that silk fibroin will lose themajority of its tensile strength within 1 year in vivo. Insome cases, the silk or the features associated withinflammation cannot be found at the implantation sitewithin months following surgery. Therefore, silk is along-term absorbable suture.

Silk fibroin elicits a foreign body response followingimplantation in vivo. Yet the response is comparable tothe most popular synthetic materials in use today asbiomaterials, and is dependent upon the implantationsite and model used for investigation. In rare cases agranuloma may form as a result of an abandonedphagocytic response to silk by macrophages and giantbody cells. Again, the response is dependent on theimplantation site of the silk. It is known that withinblood, silk is highly thrombic. Clearly, this results froma unique feature of the silk fibroin protein to bind

G.H. Altman et al. / Biomaterials 24 (2003) 401–416414

Page 15: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

components of the clotting cascade such as fibrin andfibrinogen. However, in most cases, the response ismoderate and subsides with time.

To examine the material properties of silk fibroin inrelation to other common degradable and non-degrad-able biomaterials in vivo, comparisons should be madeby equating surface area of the materials underinvestigation. Individual silk fibroin filaments, due totheir small diameter (B5 mm) increase the surface areato volume ratio of silk sutures when compared to otherbiomaterials where the equating factor is typically finalsuture diameter. As a result, in cases that demonstrate agreater foreign body response to silk in vivo, thecomparison may not be equitable to the inherentmaterial properties of silk fibroin. Therefore, silk fibroinwhen utilized as films, foams and fibers, may offer a‘new’ alternative biomaterial for use as matrices in tissueengineering where mechanically robust, long-term de-gradable materials are needed.

Acknowledgements

We thank the NIH (R01 DE13405-01 and R01AR46563-01), the NSF (DMR-0090384) and TissueRegeneration, Inc., for support of this research.

References

[1] Kaplan DL, Mello SM, Arcidiacono S, Fossey S, Senecal K,

Muller W. Silk. In: McGrath K, Kaplan DL, editors. Protein

based materials. Boston: Birkhauser, 1998. p. 103–31.

[2] Kaplan DL, Adams WW, Farmer B, Viney C. Silk: biology,

structure, properties and genetics. In: Kaplan DL, Adams WW,

Farmer B, Viney C, editors. Silk polymers: materials science and

biotechnology. ACS Symp Ser 1994;544:2–16.

[3] Kaplan DL, Fossey S, Viney C, Muller W. Self-organization

(assembly) in biosynthesis of silk fibers—a hierarchical problem.

In: Aksay IA, Baer E, Sarikaya M, Tirrell DA, editors.

Hierarchically structured materials. Materials Res Symp Proc

1992;255:19–29.

[4] Guerette PA, Ginzinger DG, Weber BHF, Gosline JM. Silk

properties determined by gland-specific expression of a spider

fibroin gene family. Science 1996;272:112–5.

[5] Cappello J. The biological production of protein polymers and

their use. Trends Biotechnol 1990;8:309–11.

[6] Xu M, Lewis RV. Structure of a protein superfiber: spider

dragline silk. Proc Natl Acad Sci USA 1990;87:7120–4.

[7] Gosline JM, Denny MW, DeMont ME. Spider silk as rubber.

Nature 1984;309:551–2.

[8] Hayashi CY, Lewis RV. Molecular architecture and evolution of

a modular spider silk protein gene. Science 2000;287:1477–9.

[9] Arcidiacono S, Mello C, Kaplan DL, Cheley S, Bayley H.

Purification and characterization of recombinant spider silk

expressed in Escherichia coli. Appl Microbiol Biotechnol

1998;49:31–8.

[10] Hayashi CY, Lewis RV. Evidence from flagelliform silk cDNA

for the structural basis of elasticity and modular nature of spider

silks. J Mol Biol 1998;275:773–84.

[11] Hinman MB, Lewis RV. Isolation of a clone encoding a second

dragline silk fibroin. J Biol Chem 1992;267:19320–4.

[12] Zhou CZ, Confalonieri F, Medina N, Zivanovic Y, Esnault C,

Yang T, Jacquet M, Janin J, Duguet M, Perasso R, Li ZG. Fine

organization of Bombyx mori fibroin heavy chain gene. Nucleic

Acids Res 2000;28:2413–9.

[13] Cunniff PM, Fossey SA, Auerbach MA, Song JW, Kaplan DL,

Adams WW, Eby RK, Mahoney D, Vezie DL. Mechanical and

thermal properties of dragline silk from the spider, Nephila

clavipes. Polym Adv Technol 1994;5:401–10.

[14] Gosline JM, DeMont ME, Denny MW. The structure and

properties of spider silk. Endeavour 1986;10:37–43.

[15] Lazaris A, Arcidiacono S, Huang Y, Zhou JF, Duguay F,

Chretien N, Welsh EA, Soares JW, Karatzas CN. Spider silk

fibers spun from soluble recombinant silk produced in mamma-

lian cells. Science 2002;295:472–6.

[16] Hood EE, Jilka JM. Plant-based production of xenogenic

proteins. Curr Opin Biotechnol 2000;10:382–6.

[17] Wilson D, Valluzzi R, Kaplan DL. Conformational transitions in

model silk peptides. Biophys J 2000;78:2690–701.

[18] Szela S, Avtges P, Valluzzi R, Winkler S, Wilson D, Kirschner D,

Kaplan DL. Reduction-oxidation control of b-sheet assembly

in genetically engineered silk. Biomacromolecules 2000;1:

534–42.

[19] Winkler S, Wilson D, Kaplan DL. Controlling b-sheet assembly

in genetically engineered silk by enzymatic phosphorylation/

dephosphorylation. Biochemistry 2000;39:12739–46.

[20] Valluzzi R, Szela S, Avtges P, Kirschner D, Kaplan DL.

Methionine redox controlled crystallization of biosynthetic silk

spidroin. J Phys Chem B 1999;103:11382–92.

[21] Winkler S, Szela S, Avtges P, Valluzzi R, Kirschner DA, Kaplan

DL. Designing recombinant spider silk proteins to control

assembly. Int J Biol Macro 1999;24:265–70.

[22] Winkler S, Kaplan DL. Molecular biology of spider silk. Rev Mol

Biotech 2000;74:85–93.

[23] Heslot H. Artificial fibrous proteins: a review. Biochimie

1998;80:19–31.

[24] Kadler KE. Fibril forming collagens. In: Sheterline P, editor.

Protein profiles: extracellular matrix, vol. 1. London, UK:

Academic Press, 1995.

[25] Moy RL, Lee A, Zalka A. Commonly used suture materials in

skin surgery. Am Fam Physician 1991;44:2123–8.

[26] Minoura N, Aiba S, Gotoh Y, Tsukada M, Imai Y. Attachment

and growth of cultured fibroblast cells on silk protein matrices.

J Biomed Mater Res 1995;29:1215–21.

[27] Moynihan BGA. The ritual of a surgical operation. Br J Surg

1920;8:27–35.

[28] Hollander DH. Interstitial cystitis and silk allergy. Med Hypoth-

eses 1994;43:155–6.

[29] Wen CM, Ye ST, Zhou LX, Yu Y. Silk-induced asthma in

children: a report of 64 cases. Ann Allergy 1990;65:375–8.

[30] Kurosaki S, Otsuka H, Kunitomo M, Koyama M, Pawankar R,

Matumoto K. Fibroin allergy: IgE mediated hypersensitivity

to silk suture materials. Nippon Ika Daigaku Zasshi 1999;66:

41–4.

[31] Rossitch Jr. E, Bullard DE, Oakes WJ. Delayed foreign-body

reaction to silk sutures in pediatric neurosurgical patients. Childs

Nerv Syst 1987;3:375–8.

[32] Dewair M, Baur X, Ziegler K. Use of immunoblot technique for

detection of human IgE and IgG antibodies to individual silk

proteins. J Allergy Clin Immunol 1985;76:537–42.

[33] Zaoming W, Codina R, Fernandez-Caldas E, Lockey RF. Partial

characterization of the silk allergens in mulberry silk extract. J Inv

Allergy Clin Immunol 1996;6:237–41.

[34] Soong HK, Kenyon KR. Adverse reactions to virgin silk sutures

in cataract surgery. Ophthalmology 1984;91:479–83.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416 415

Page 16: Silk-based biomaterials - Biomedical Engineeringorion.bme.columbia.edu/lulab/pdf/altman_silk-based-biomaterials_2003.pdfSilk-based biomaterials Gregory H. Altman,Frank Diaz,Caroline

[35] Postlethwait RW. Long-term comparative study of nonabsorb-

able sutures. Ann Surg 1970;171:892–8.

[36] Postlethwait RW. Tissue reaction to surgical sutures. In: Dumphy

JE, Van Winkle W, editors. Repair and regeneration. New York:

McGraw-Hill, 1969. p. 263–85.

[37] Postlethwait RW, Dillon MD, Reeves JW. Experimental study of

silk suture. Arch Surg 1962;84:698–702.

[38] Morrow FA, Kogan SJ, Freed SZ, Laufman H. In vivo

comparison of polyglycolic acid, chromic catgut and silk in tissue

of the genitourinary tract: an experimental study of tissue retrieval

and calculogenesis. J Urol 1974;112:655–8.

[39] Nebel L, Rosenberg G, Tobias B, Nathan H. Autograft suture in

peripheral nerves. Eur Surg Res 1977;9:224–34.

[40] Peleg H, Rao UN, Emrich LJ. An experimental comparison of

suture materials for tracheal and bronchial anastomoses. J Thorac

Cardiovasc Surg 1986;34:384–8.

[41] Lam KH, Nijenhuis AJ, Bartels H, Postema AR, Jonkman MF,

Pennings AJ, Nieuwenhuis P. Reinforced poly(l-Lactic Acid)

fibers as suture material. J Appl Biomater 1995;6:191–7.

[42] Anderson JM. Inflammation, wound healing, and the foreign

body response. In: Ratner BD, Hoffman AS, Schoen FJ, Lemons

JE, editors. Biomaterials science: an introduction to materials in

medicine. Boston: Academic Press, 1996. p. 165–72.

[43] Foschi D, Corsi F, Cellerino P, Rizzi A, Morandi E, Trabucchi E.

Angiogenic effects of suture biomaterials. An experimental study

in rats. Eur Surg Res 2001;33:16–20.

[44] Setzen G, Williams EF. Tissue response to suture materials

implanted subcutaneously in a rabbit model. Plast Reconstr Surg

1997;100:1788–95.

[45] Bucknall TE, Teare L, Ellis H. The choice of a suture to close

abdominal incisions. Eur Surg Res 1983;15:59–66.

[46] Wong PC, Crain EJ, Nguan O, Watson CA, Racanelli A.

Antithrombotic actions of selective inhibitors of blood coagula-

tion factor Xa in rat models of thrombosis. Thromb Res

1996;83:117–26.

[47] Dahlke H, Dociu N, Thurau K. Thrombogenicity of different

suture materials as revealed by scanning electron microscopy.

J Biomed Mater Res 1980;14:251–68.

[48] Lee KY, Kong SJ, Park WH, Ha WS, Kwon IC. Effect of surface

properties on the antithrombogenicity of silk fibroin/S-carbox-

ymethyl kerateine blend films. J Biomater Sci Polym Ed

1998;9:905–14.

[49] Sakabe H, Ito H, Miyamoto T, Noishiki Y, Ha WS. In vivo blood

compatibility of regenerated silk fibroin. Sen-I Gakkaishi

1989;45:487–90.

[50] Santin M, Motta A, Freddi G, Cannas M. In vitro evaluation of

the inflammatory potential of the silk fibroin. J Biomed Mater

Res 1999;46:382–9.

[51] Uff CR, Scott AD, Pockley AG, Phillips RK. Influence of soluble

suture factors on in vitro macrophage function. Biomaterials

1995;16:355–60.

[52] Salthouse TN, Matlaga BF, Wykoff MH. Comparative tissue

response to six suture materials in rabbit cornea, sclera, and

ocular muscle. Am J Ophthalmol 1977;84:224–33.

[53] Ethicon, Inc., Wound closure manual. The suture: specific

suturing materials. Non-absorbable sutures. Somerville, NJ:

Ethicon, Inc. [copyright 2000, Chapter 2].

[54] Minoura N, Tsukada M, Nagura M. Physico-chemical properties

of silk fibroin membrane as a biomaterial. Biomaterials

1990;11:430–4.

[55] Asakura T, Demura M, Date T, Miyashita N, Ogawa K,

Williamson MP. NMR study of silk I structure of Bombyx mori

silk fibroin with N-15- and C-13-NMR chemical shift contour

plots. Biopolymers 1997;41:193–203.

[56] Kurioka A, Yamazaki M, Hirano H. Primary structure and

possible functions of a trypsin inhibitor of Bombyx mori. Eur J

Biochem 1999;259:120–6.

[57] Greenwald D, Shumway S, Albear P, Gottlieb L. Mechanical

comparison of 10 suture materials before and after in vivo

incubation. J Surg Res 1994;56:372–7.

[58] Kourie JI, Shorthouse AA. Properties of cytotoxic peptide-

formed ion channels. Am J Physiol Cell Physiol 2000;278:

C1063–87.

[59] Inouye K, Kurokawa M, Nishikawa S, Tsukada M. Use of

Bombyx mori silk fibroin as a substratum for cultivation of animal

cells. J Biochem Biophys Meth 1998;37:159–64.

[60] Sofia S, McCarthy MB, Gronowicz G, Kaplan DL. Functiona-

lized silk-based biomaterials for bone formation. J Biomed Mater

Res 2001;54:139–48.

[61] Altman GH, Horan RL, Lu H, Moreau J, Martin I, Richmond

JC, Kaplan DL. Silk matrix for tissue engineered anterior cruciate

ligaments. Biomaterials 2002;23:4131–41.

[62] Altman GH, Horan RL, Martin I, Farhadi J, Stark PRH, Volloch

V, Richmond JC, Vunjak-Novakovic, Kaplan DL. Cell differ-

entiation by mechanical stress. FASEB J 2001;16:270–2 (print)

and 10:1096 (electronic).

[63] Dunn MG, Tria AJ, Kato P, Bechler JR, Ochner RS, Zawadski

JP, Silver FH. Anterior cruciate ligament reconstruction using a

composite collagenous prosthesis. Am J Sports Med 1992;20:

507–15.

[64] Dunn MG, Bellincampi LD, Tria AJ, Zawadski JP. Preliminary

development of a collagen-PLA composite for ACL reconstruc-

tion. J Appl Polym Sci 1997;63:1423–8.

[65] Zarkoob S, Reneker DH, Eby RK, Hudson SD, Ertley D, Adams

WW. Generation of synthetic elastin-mimetic small diameter

fibers and fiber networks. Polym Preprints Am Chem Soc

1998;39:244–5.

[66] Jin H-J, Fridrikh SV, Rutledge GC, Kaplan DL. Electrospinning

Bombyx mori silk with poly(ethylene oxide). Biomacromolecules,

in press.

[67] Matthews JA, Wnek GE, Simpson DG, Bowlin GL. Electrospin-

ning of collagen nanofibers. Biomacromolecules 2002;3:232–8.

[68] Perez-Rigueiro J, Viney C, Llorca J, Elices M. Mechanical

properties of single-brin silkworm silk. J Appl Polym Sci

2000;75:1270–7.

[69] Pins GD, Christiansen DL, Patel R, Silver FH. Self-assembly of

collagen fibers: influence of fibrillar alignment and decorin on

mechanical properties. Biophys J 1997;73:2164–72.

[70] Engelberg I, Kohn J. Physicomechanical properties of degradable

polymers used in medical applications: a comparative study.

Biomaterials 1991;12:292–304.

[71] Gosline JM, Guerette PA, Ortlepp CS, Savage KN. The

mechanical design of spider silks: from fibroin sequence to

mechanical function. J Exp Biol 1999;202(Pt 23):3295–303.

G.H. Altman et al. / Biomaterials 24 (2003) 401–416416