You are on page 1of 6

REVIEW ARTICLE

Artificial blood vessel: The Holy Grail of


peripheral vascular surgery
John D. Kakisis,a,b Christos D. Liapis,b Christopher Breuer,a and Bauer E. Sumpio,a New Haven, Conn;
and Athens, Greece

Artificial blood vessels composed of viable tissue represent the ideal vascular graft. Compliance, lack of thrombogenicity,
and resistance to infections as well as the ability to heal, remodel, contract, and secrete normal blood vessel products are
theoretical advantages of such grafts. Three basic elements are generally required for the construction of an artificial
vessel: a structural scaffold, made either of collagen or a biodegradable polymer; vascular cells, and a nurturing
environment. Mechanical properties of the artificial vessels are enhanced by bioreactors that mimic the in vivo
environment of the vascular cells by producing pulsatile flow. Alternative approaches include the production of
fibrocollagenous tubes within the recipient’s own body (subcutaneous tissue or peritoneal cavity) and the construction of
an artificial vessel from acellular native tissues, such as decellularized small intestine submucosa, ureter, and allogeneic or
xenogeneic arteries. This review details the most recent developments on vascular tissue engineering, summarizes the
results of initial experiments on animals and humans, and outlines the current status and the challenges for the future.
( J Vasc Surg 2005;41:349-54.)

There is a tremendous impetus in the vascular commu- of the environment, and even have the ability to grow when
nity to develop synthetic small-diameter grafts that have placed in children. Compliance, lack of thrombogenicity,
high long-term patency rates. “Off the shelf” availability in and resistance to infections are theoretical advantages of
various diameters and lengths, uncomplicated storage or such grafts, and burst strength and “off the shelf” availabil-
preparation requirements, and ease of handling are the ity are additional desirable goals.4
main advantages of such grafts; their inherent thromboge- This review summarizes recent developments in vascu-
nicity and compliance mismatch are the main drawbacks. lar tissue engineering and the initial in vivo studies. A
Synthetic grafts have been associated with excellent long- Medline search of the English-language literature was per-
term results in treating the pathology of large-diameter formed using the key words artificial vessels, tissue engineer-
arteries where the flow is high and the resistance is low. The ing, and vascular grafts. Reference lists from all relevant
patency rates have been disappointing when they are used articles were reviewed to identify additional studies.
to replace small-diameter (⬍6 mm) arteries such as the Construction of an artificial vessel. Three basic ele-
coronary and the infragenicular vessels.1-3 ments are generally required for the construction of an
One strategy to overcome these limitations applies artificial vessel: a structural scaffold, cells, and a nurturing
tissue engineering techniques to construct biologic substi- environment (Table).5-13 The scaffold provides a tempo-
tutes of diseased native vessels. These artificial blood vessels rary skeleton to support the growing tissue and provides the
should ideally be composed of viable tissue, able to contract desired shape until the cells produce their own extracellular
in response to hemodynamic forces and chemical stimuli, matrix (ECM). Most scaffolds are based on collagen matri-
and secrete normal blood vessel products. Artificial blood ces and biodegradable polymers. Another strategy is to use
vessels should also allow complete healing without any a tubular mandrel as a supporting device in vitro and
immunologic reaction, remodeling according to the needs remove it prior to implantation of the graft into the host.14
Autologous or allogeneic smooth muscle cells (SMCs),
From the Department of Vascular Surgery,a Yale University School of fibroblasts, and endothelial cells (ECs) can then be seeded
Medicine, and the 2nd Department of Propedeutic Surgery,b Athens on the scaffold and cultured until a cellularized vessel with
University Medical School, Laiko Hospital. optimal mechanical properties is created. Bioreactors that
Competition of interest: none.
Additional material for this article may be found online at
mimic the in vivo environment of the vascular cells by
www.mosby.com/jvs. producing pulsatile flow are being evaluated and may be
Reprint requests: Bauer E. Sumpio, MD, PhD, Chief, Vascular Surgery, Yale useful in this process.12,15,16
University School of Medicine, 333 Cedar Street, New Haven, CT Artificial vessels based on collagen scaffolds.
06510.
0741-5214/$30.00
Weinberg and Bell5 were the first to report the in vitro
Copyright © 2005 by The Society for Vascular Surgery. construction of a vessel; its multilayer structure resembled
doi:10.1016/j.jvs.2004.12.026 that of an artery. Collagen and bovine aortic SMCs were
349
JOURNAL OF VASCULAR SURGERY
350 Kakisis et al February 2005

Methods for preparing an artificial vessel and initial results

Mechanical
Author Scaffold Cells properties Implantation site Patency

Natural (collagen)
Weinberg and Bell,5 1986 Collagen Bovine aortic SMC, Poor, necessitating a Not tested
EC and Dacron mesh
fibroblasts
L’Heureux et al,6 1993 Type I and III Human umbilical Poor Not tested
collagen vein SMC and
EC
Human skin
fibroblasts
Hirai et al,7,8 1994 Type I collagen Canine jugular Poor, necessitating a Canine posterior vena 65% at 6
SMC and EC Dacron mesh cava months
Synthetic
(biodegradable
polymer)
Shum-Tim et al,9 1999 PGA-PHA Ovine carotid Good Ovine infrarenal aorta 100% at 5
SMC, EC and months
fibroblasts
Niklason et al,10 1999 PGA Bovine aortic SMC Good Swine saphenous artery 100% at 4
and EC weeks
11
Watanabe et al, 2001 PGA-CL/LA Canine femoral vein Moderate Canine inferior vena 100% at 13
SMC and cava months
fibroblasts
Hoerstrup et al,12 2001 PGA-P4HB Ovine carotid Good Not tested
fibroblasts and
EC
Hoerstrup et al,13 2002 PGA-P4HB Human umbilical Good Not tested
cord cells
None
L’Heureux et al,14 1998 Human umbilical Good Canine femoral artery 50% at 7 days
vein SMC and
EC
Human skin
fibroblasts
SMC, Smooth muscle cells; EC, endothelial cells; PGA, polyglycolic acid; PHA, polyhydroxyalkanoate; CL, caprolactone; LA, lactic acid; P4HB, poly-4-
hydroxybutyrate.

casted together in culture medium to create an annular with jugular ECs and the resulting graft was implanted in
mold (Fig 1, online only). The inner surface of the graft was canine vena cava. Despite the low venous pressures, the
seeded with bovine ECs and the outer with bovine adven- graft ruptured within a few days of implantation. Subse-
titial fibroblasts. A Dacron mesh was embedded into the quent grafts were wrapped with a Dacron mesh to prevent
wall to enhance the mechanical strength of the model. tearing, and the patency rate of the wrapped grafts was 64%
Models made without the mesh dilated and ruptured at at 6 months. Complete remodeling could be documented
very low intraluminal pressures (⬍10 mm Hg). A model by a monolayer of longitudinally oriented ECs on the
with one mesh had a burst strength of 40 to 70 mm Hg, luminal surface, circumferentially oriented SMCs of the
whereas three layers of collagen lattice alternating with two contractile phenotype at the subendothelial layer, ingrown
meshes provided a burst strength of 120 to 180 mm Hg. fibroblasts throughout the vessel wall, meshes of collagen
The absence of elastin, the longitudinal (instead of fibrils in the intercellular space, and sheet-like elastic lamel-
circular) orientation of the SMCs, and the low densities of lae in the subendothelial layer.
the SMCs and collagen accounted for the poor mechanical The poor mechanical strength of collagen-based artifi-
properties of the model. However, well differentiated cial vessels has been partially attributed to the longitudinal
SMCs and functional endothelium, which could produce orientation of the SMCs before implantation. Several strat-
von Willebrand’s factor and prostacyclin, made this blood egies have been used to stimulate reorientation of the
vessel model attractive and sparked further research into the SMCs in a circumferential direction. L’Heureux et al6
tissue engineering of blood vessels. hypothesized that SMC orientation is directly influenced
A similar technique used by Hirai et al7,8 yielded similar by tensions that oppose cell contraction. In this context,
results. Hybrid medial tissue was prepared by pouring a circumferential forces induced during the maturation of the
cold solution of canine jugular SMCs and type I collagen artificial vessel are favorable, whereas longitudinal forces are
into a tubular glass mold. The luminal surface was seeded not. To minimize longitudinal forces, the adherence of
JOURNAL OF VASCULAR SURGERY
Volume 41, Number 2 Kakisis et al 351

media to the glass mandrel was repeatedly broken up by animals. It is expected that as the support sleeve will de-
tapping the tube on a hard surface at least once a day. This grade, the cellular layer will gradually strengthen and even-
allowed free longitudinal contraction of the media. The tually become the load-bearing component of the graft.
radial forces remained, stimulating reorientation of the The in vivo performance of the graft remains to be tested,
SMCs in a circumferential fashion. however.
A collagen gel containing human dermal fibroblasts Artificial vessels based on biodegradable scaffolds.
was added around the media-like structure to simulate an Several biocompatible and biodegradable polymers have
adventitia; an endothelium was established by injecting a been used as scaffolds for the construction of artificial
solution of human umbilical vein ECs onto the inner lining vessels. Polyglycolic acid (PGA)10,20,21 is most commonly
of the vessel. The structure of the complete model resem- used. Its high porosity permits the diffusion of nutrients
bled that of an artery, but it still was not able to withstand upon implantation and subsequent neovascularization, and
intraluminal pressure at a physiological level. it is easily handled and fabricated into different shapes.19
An innovative technique was introduced by Kanda Unfortunately, PGA meshes are rapidly bioabsorbed
et al16 to induce longitudinal orientation of the ECs, which (within 6 to 8 weeks) and unable to withstand systemic
enhances their stability under flow shear stress, and circum- pressures. To improve the physical properties of PGA,
ferential orientation of the SMCs, which is expected to several copolymers have been produced by combining PGA
increase the mechanical strength of the artificial vessel and with polyhydroxyalkanoate (PHA),9 poly(lactic acid-co-
improve its vasomotor reactivity. The investigators hypoth- lysine),11 poly-4-hydroxybutyrate (P4HB),12,13 poly-L-
esized that by applying pulsatile flow on a collagen-based lactic acid (PLLA),21,22 or polyethylene glycol (PEG).23
artificial vessel, the ECs would orient parallel to the direc- These combinations not only define the mechanical char-
tion of flow and the SMCs would align parallel to the acteristics of the scaffold but also regulate the phenotype of
direction of stretch (i.e., circumferentially). They utilized a the cells grown on them through cellular interactions with
nondegradable polyurethane graft with a collagen gel em- the scaffold material.21
bedding canine SMCs. Autologous ECs were then seeded Shum-Tim et al9 have seeded a PGA-PHA co-polymer
onto the media equivalent to create the framework of an directly with a mixed population of ovine carotid ECs,
intima, and the graft was subjected to pulsatile flow of SMCs and fibroblasts. The PGA inner layer was used to
culture medium. After 10 days of stress loading, the ECs promote cell attachment and tissue formation, whereas the
aligned parallel to the longitudinal axis of the graft and the outer PHA layer provided the necessary mechanical
SMCs were oriented in a circumferential direction. strength with a longer degradation period of ⬎52 weeks.
In the initial experiments, the application of direct, The tissue-engineered grafts were used to replace infrarenal
pulsatile flow on an immature artificial vessel necessitated aortic segments in seven lambs and all remained patent for
the use of a polyurethane scaffold. To avoid the use of a up to 5 months. Collagen and DNA content, as well as the
scaffold, the investigators17 utilized an elastomeric silicon mechanical strain-stress curve, approached those of the
tube in the bioreactor. A hybrid media tissue composed of native aorta over time. Histology revealed elastic fibers in
type I collagen gel and bovine aortic SMCs was constructed the medial layer and endothelial-specific von Willebrand
around this tube, which was subsequently subjected to factor on the luminal surface.
periodic cyclic inflation. After 5 days of culture under these Watanabe et al11 created a hybrid polymer scaffolding
conditions, both the SMCs and collagen fibers were aligned composed of a PGA sheet and a co-polymer consisting of
circumferentially. L-lactides and ⑀-caprolactone. The scaffold was seeded with
Another strategy for influencing reorientation of SMCs a mixed cell population obtained from the femoral veins of
within artificial grafts was developed by Tranquillo et al.18 mongrel dogs and, after 7 days of culture, implanted as an
A strong magnetic field was applied during collagen fibril- interposition graft into the inferior vena cava of the same
logenesis, when the media equivalent was first created, dog. The graft remained patent for up to 13 months, with
resulting in circumferential orientation of the collagen no evidence of stenosis or dilatation. The scaffold was
fibrils within the collagen scaffold. The entrapped SMCs completely degraded by 3 months, and the overall gross
were subsequently aligned along the circumferentially ori- appearance of the artificial vein was similar to that of the
entated fibrils through cell contact guidance. The tech- native inferior vena cava.
nique appears to be simple, reproducible, and effective, Niklason et al10 used a PGA scaffold and a biomimetic
resulting in media equivalents that are stiffer and more system similar to that described by Kanda et al.17 However,
viscous than the nonmagnetically orientated controls. instead of pulsatile flow at 60 beats per minute, the fre-
Berglund et al19 recently presented the concept of a quency of cyclic stretch was set at 165 to mimic the fetal
construct-sleeve hybrid graft composed of an outer colla- environment in large mammals and, presumably, facilitate
gen layer containing neonatal human dermal fibroblasts in vitro vasculogenesis. Bovine aortic SMCs were seeded on
and an inner support sleeve fabricated from type I collagen tubular biodegradable PGA scaffolds and placed around
gel and cross-linked with glutaraldehyde, ultraviolet, or highly distensible silicon tubes. After 8 weeks of pulsatile
dehydrothermal treatment. Although inferior to native ar- stretch in the bioreactor, the silicon tubing was removed,
teries, the hybrid grafts approached the mechanical prop- and the luminal surfaces of the grafts were seeded with
erties that are considered necessary for implantation in bovine aortic ECs. Continuous perfusion was instituted for
JOURNAL OF VASCULAR SURGERY
352 Kakisis et al February 2005

another 3 days, imposing shear stress to the endothelialized would be reasonable to expect that decellularized alloge-
vessel lumen. The resulting graft had a wall thickness and neic or xenogeneic arteries would give the best biome-
collagen content comparable to native arteries and a rup- chanical profile for vascular tissue engineering. Neverthe-
ture strength of 2150 ⫾ 709 mm Hg, which is greater than less, decellularized canine arteries did not appear to
the 1680 ⫾ 307 mm Hg reported for native human saphe- perform well when implanted as interposition carotid or
nous veins. coronary artery bypass grafts in dogs, with an early throm-
Engineered vessels showed measurable contractions to bosis rate of 20% to 56%.26,27
pharmacologic agents such as serotonin, endothelin-1 and The thrombogenicity of decellularized xenogeneic ves-
prostaglandin F2a and contained well-differentiated SMCs, sels is mainly due to the lack of EC lining of the luminal
as displayed by the expression of calponin and myosin heavy surface of the vessel. Bader et al28 have tried to overcome
chains. The initial experience with two of these grafts this problem by reseeding decellularized porcine aortas
implanted into the saphenous artery of miniature swine was with pre-expanded human ECs and myofibroblasts har-
promising. They were patent for up to 4 weeks, in contrast vested from saphenous vein biopsy specimens. Apart from
to two nonpulsed grafts that thrombosed. demonstrating the feasibility of reseeding decellularized
Hoerstrup et al12 applied direct flow of culture medium vessels with vascular cells, these experiments also revealed
with a bioreactor through the vascular lumen, thereby that the in vivo immune response was greatly reduced after
generating significant shear on the luminal surface and such processing. Ex vivo reseeding of decellularized vascu-
pulsatile stretch to the vessel wall. A co-polymer of PGA- lar matrix scaffolds could therefore be the answer to throm-
P4HB was used as the scaffold material because its strength, bogenicity, immunologic rejection, and lack of healing,
pliability, and thermoplastic characteristics allowed fabrica- which are the three main problems of cryopreserved or
tion into almost any shape using a heat-welding technique. glutaraldehyde-fixed grafts.
Grafts were sequentially seeded with ovine vascular myofi- Artificial vessels without the use of a scaffold.
broblasts and ECs and placed in the pulse duplicator system L’Heureux et al14 have investigated a novel technique for the
for up to 4 weeks. production of artificial vessels, based exclusively on cultured
Increased cell mass and collagen content, well orga- human cells, without the use of a scaffold (Fig 2, online only).
nized in a layered fashion, was achieved in the pulsed Human umbilical vein SMCs and skin fibroblasts were cul-
vascular grafts, resulting in supraphysiologic burst pressures tured for ⬃30 days, at which time both cell types had formed
and suture retention strength appropriate for surgical im- cohesive cellular sheets composed of cells and ECM, which
plantation within 3 weeks of culture. The burst pressure could be manually peeled from the culture flasks.
(262 ⫾ 26 mm Hg), however, was still significantly lower The SMC sheet was then wrapped around a tubular
than that obtained by Niklason et al10 at 8 weeks of culture. support to produce the media of the vessel. After 1 week of
In addition to ovine vascular cells, Hoerstrup et al13 also media maturation, the fibroblast sheet was rolled around
demonstrated the feasibility of seeding the grafts with human the media to provide the adventitia. After a 7-week matu-
umbilical cord cells. These cells exhibit myofibroblast-like ration period, the mandrel was removed and human um-
characteristics and have the advantage of being readily avail- bilical vein ECs were seeded in the lumen and allowed to
able and easy to obtain, without necessitating the sacrifice of grow for another week.
intact donor vessels. In addition, the authors suggest that The end product was a well-organized, three-layer wall
individual cell pools could be created from a person’s own vessel resembling a human artery. The tissue-engineered
umbilical cord for potential future use. This technique would graft had burst strength of 2594 ⫾ 501 mm Hg and
eliminate the need to harvest autologous cells and the prob- demonstrated good handling and suturability characteris-
lem of immunogenicity of heterogenic cells. tics. It showed no signs of degradation, tearing, or dilata-
The first clinical application of an artificial vessel based on tion when implanted as an interposition femoral artery graft
a biodegradable scaffold was reported by Shin’oka et al in in mongrel dogs. On the other hand, intramural blood
2001.24 Cells isolated from a peripheral vein were seeded on a infiltration was observed between the vessel wall layers and
polycaprolactone-polylactic acid co-polymer tube reinforced the patency rate was only 50% at 7 days of implantation.
with woven PGA. The graft was used for the reconstruction of Current status and future perspectives. The ability
an occluded pulmonary artery in a 4-year-old girl. Seven to apply tissue engineering techniques to construct autol-
months after implantation, the patient was doing well, with no ogous vascular grafts for surgical reconstruction is now a
evidence of graft occlusion or aneurysm formation. clinical reality. Shin’oka’s group24,29,30 have reported their
An alternative approach to the use of synthetic biode- clinical experience using tissue-engineered vessels as venous
gradable scaffolds is to implant an acellular native tissue and conduits for reoperative reconstructive cardiovascular sur-
let the body act as a bioreactor and cell source to seed the gical procedures in ⬎40 children with varying forms of
acellular matrix. Several tissues, including skin, fascia, peri- congenital heart disease. They report ⬎95% patency at 1
toneum, muscle, small intestine submucosa, and ureter, year, without evidence of stenosis or aneurysm formation
have been used for this purpose with various outcomes.4 It despite the small caliber of these vessels in the setting of a
has been well-established, however, that the mechanical re-do surgical environment.
properties of uncross-linked, cell-extracted tissues are re- Although significant advances have already been made,
markably similar to those of fresh tissue.25 Therefore, it the construction of a synthetic graft with mechanical prop-
JOURNAL OF VASCULAR SURGERY
Volume 41, Number 2 Kakisis et al 353

erties identical to those of native arteries remains an elusive SMCs with the cyclic guanosine monophosphate-dependent
goal. It should be emphasized, however, that a major protein kinase gene may enhance the contractile characteris-
advantage of an artificial vessel is that it does not need to tics of the cells and, consequently, of the graft.38
have mechanical properties identical to those of a native In this context, Matsuda39 harvested fibroblasts from
artery at implantation.31 Being composed of viable tissue skin tissue and transfected them with a triple-gene-encod-
with the potential to remodel, repair, and grow, an artificial ing adenovirus to express a tissue factor pathway inhibitor,
vessel is theoretically able to completely adapt to local C-type natriuretic peptide, and vascular endothelial growth
hemodynamic conditions and acquire the structural and factor. The transfected fibroblasts were incorporated into a
mechanical characteristics of the vessel it replaces, whether collagen gel wrapped with a polyurethane film, and the
this is an artery or a vein. construct was implanted into canine carotid arteries. The
Although some authors are optimistic enough to be- grafts exhibited 100% patency at 1 month and 70% patency
lieve that we are only a decade away from United States at 3 months, whereas all of the nontransfected fibroblast-
Food and Drug Administration (FDA) approval of a tissue- inoculated grafts occluded ⱕ2 days after implantation.
engineered blood vessel substitute,32 certain problems Genetic and molecular engineering can also be applied to
need to be addressed: scaffold technology to improve its mechanical and biologic
properties. Modification of chain length and location of cross-
1. Grafts currently being investigated require an extended
link junctions in the polymer allow for the preparation of
period of preparation, usually 1 to 3 months, so they
materials with well-defined mechanical characteristics.40 The
cannot be used in emergency situations.
application of nanofibrous technology to create scaffolds that
2. The prolonged duration of culture increases the risk of
mimic the natural extracellular matrix enhances the ability to
infection and raises the cost in terms of manpower,
achieve ingrowth of vascular cells,41 whereas incorporation of
equipment, and materials needed.
adhesion peptides or growth factors will improve the capacity
3. Most of the biodegradable polymers currently in use as
of the scaffold to support the attachment of vascular cells and
scaffolds for the construction of artificial vessels already
their production of ECM.42-45
have FDA approval. This could actually be a step back-
It should be noted, however, that the incorporation of
wards. We need a biopolymer appropriate for use as a
cell adhesion peptides in polymer scaffolds is a double-edge
vascular conduit, not one that is off the shelf.
sword. Several peptides, such as the RGDS (Arg-Gly-Asp-
Alternative types of cells are already being used in Ser), KQAGDV (Lys-Gln-Ala-Gly-Asp-Val) and VAPG
animal models to eliminate the need for surgical harvest of (Val-Ala-Pro-Gly), have been shown to promote cell
ECs from large veins or tissue microvessels of patients with attachment to collagen-based, PEG, or poly(lactic acid-co-
atherosclerosis.33 Endothelial progenitor cells (EPCs) pro- lysine) scaffolds and prevent cell wash-out after exposure to
vide a valuable cell source for cardiovascular tissue engi- shear.42-44 On the other hand, increased cell adhesion
neering, because they are present in peripheral blood and results in decreased ECM protein synthesis by the cells.43
can differentiate into ECs with comparable antithrombo- This may lead to graft failure because the mechanical integ-
genic potential. Shirota et al34 have shown that the produc- rity of the tissue construct will depend entirely on the ECM
tion rate of endothelial-type nitric oxide synthase and 6- density after the degradation of the scaffold.
keto-prostaglandin-F1-a by EPCs are approximately one This problem may be overcome by tethering growth
third and one half of mature ECs, respectively, whereas the factors to the scaffold. For example, as reported by Mann et
tissue-type plasminogen activator production rate of EPCs al,45 transforming growth factor-␤1 attached to a PGA scaf-
is almost the same as that of ECs. Moreover, EPC-seeded fold increases the production of ECM proteins by vascular
decellularized porcine iliac vessels implanted in sheep ex- SMCs, thus counteracting the effects of adhesive ligands on
hibited high patency, rapid vascular wall regeneration, con- ECM production, but it does not increase SMC proliferation.
tractile activity, and nitric oxide-induced vasodilation sim- This is particularly important in artificial vessels, as prolifera-
ilar to those of native carotid arteries.35 tion of SMCs could lead to narrowing of the vessel lumen.
Apart from EPCs, smooth muscle progenitor cells (SPCs) Drug-eluting scaffolds are another revolutionary devel-
have also been identified in peripheral blood.36 Ex vivo expan- opment in the fabrication of artificial vessels. Yoon et al46
sion of these cells has been achieved and may be exploited for have incorporated dexamethasone, a steroidal anti-inflam-
the construction of an artificial vessel in the lab. Even more matory drug, into the polymer phase of a PLGA scaffold.
exciting is that in vivo recruitment of SPCs to an implanted Dexamethasone was slowly released in a controlled manner
construct seems feasible, because these cells show increased for over 30 days, leading to a drastic suppression in the
integrin ␣5␤1 expression, which allows them to attach to proliferation of lymphocytes and SMCs in vitro. Although not
fibronectin or other ␣5␤1 adhesive matrices. yet tested in vivo, such scaffolds show promise for the preven-
Significant advancements in the properties of artificial tion of intimal hyperplasia.
vessels can be expected in the foreseeable future with the In conclusion, initial results show that the construction
combination of tissue engineering and gene therapy. For of an artificial vessel that combines desirable biologic and
instance, the incorporation in the vessel wall of SMCs trans- mechanical characteristics is feasible. The incorporation of
fected with the tropoelastin gene may result in increased recent advances in molecular and genetic engineering to
elastin synthesis and deposition,37 whereas transfection of the already developed techniques is expected to aid in the
JOURNAL OF VASCULAR SURGERY
354 Kakisis et al February 2005

solution of the problems that have emerged. The clinical 23. Wake MC, Gupta PK, Mikos AG. Fabrication of pliable biodegradable
imperative for better vascular grafts, the great scientific polymer foams to engineer soft tissues. Cell Transplant 1996;5:465-73.
24. Shin’oka T, Imai Y, Ikada Y. Transplantation of a tissue-engineered
challenge, and the large commercial opportunity for such a pulmonary artery. N Engl J Med 2001;344:532-3.
graft guarantee that significant developments are to be 25. Schmidt CE, Baier JM. Acellular vascular tissues: natural biomaterials
anticipated in this field in the near future. for tissue repair and tissue engineering. Biomaterials 2000;21:2215-31.
26. Malone JM, Brendel K, Duhamel RC, Reinert RL. Detergent-extracted
small-diameter vascular prostheses. J Vasc Surg 1984;1:181-91.
REFERENCES 27. Wilson GJ, Courtman DW, Klement P, Lee JM, Yeger H. Acellular
matrix: a biomaterials approach for coronary artery bypass and heart
1. Steinthorsson G, Sumpio B. Clinical and biological relevance of vein valve replacement. Ann Thorac Surg 1995;60:S353-8.
cuff anastomosis. Acta Chir Belg 1999;99:282-8. 28. Bader A, Steinhoff G, Strobl K, Schilling T, Brandes G, Mertsching H,
2. Sapsford RN, Oakley GD, Talbot S. Early and late patency of expanded et al. Engineering of human vascular aortic tissue based on a xenogeneic
polytetrafluoroethylene vascular grafts in aorta-coronary bypass. J Tho- starter matrix. Transplantation 2000;70:7-14.
rac Cardiovasc Surg 1981;81:860-4. 29. Matsumura G, Hibino N, Ikada Y, Kurosawa H, Shin’oka T. Successful
3. Veith FJ, Gupta SK, Ascer E, White-Flores S, Samson RH, Scher LA, et application of tissue engineered vascular autografts: clinical experience.
al. Six-year prospective multicenter randomized comparison of autolo- Biomaterials 2003;24:2303-8.
gous saphenous vein and expanded polytetrafluoroethylene grafts in 30. Shin’oka T. Clinical experience with tissue engineered venous conduits.
infrainguinal arterial reconstructions. J Vasc Surg 1986;3:104-14. Tissue Engineering Society International. Orlando, FL 2003, abstract.
4. Teebken OE, Haverich A. Tissue engineering of small diameter vascular 31. Edelman ER. Vascular tissue engineering: designer arteries. Circ Res
grafts. Eur J Vasc Endovasc Surg 2002;23:475-85. 1999;85:1115-7.
5. Weinberg CB, Bell E. A blood vessel model constructed from collagen 32. Nerem RM. Tissue engineering a blood vessel substitute: the role of
and cultured vascular cells. Science 1986;231:397-400. biomechanics. Yonsei Med J 2000;41: 735-9.
6. L’Heureux N, Germain L, Labbe R, Auger FA. In vitro construction of 33. Langer RS, Vacanti JP. Tissue engineering: the challenges ahead. Sci
a human blood vessel from cultured vascular cells: a morphologic study. Am 1999;280:86-9.
J Vasc Surg 1993;17:499-509. 34. Shirota T, He H, Yasui H, Matsuda T. Human endothelial progenitor
7. Hirai J, Kanda K, Oka T, Matsuda T. Highly oriented, tubular hybrid cell-seeded hybrid graft: proliferative and antithrombogenic potentials
vascular tissue for a low pressure circulatory system. ASAIO J 1994;40: in vitro and fabrication processing. Tissue Eng 2003;9:127-36.
M383-8. 35. Kaushal S, Amiel GE, Guleserian KJ, Shapira OM, Perry T, Sutherland
8. Hirai J, Matsuda T. Venous reconstruction using hybrid vascular tissue FW, et al. Functional small-diameter neovessels created using endothe-
composed of vascular cells and collagen: tissue regeneration process. lial progenitor cells expanded ex vivo. Nat Med 2001;7:1035-40.
Cell Transplant 1996;5:93-105. 36. Simper D, Stalboerger PG, Panetta CJ, Wang S, Caplice NM. Smooth
9. Shum-Tim D, Stock U, Hrkach J, Shinoka T, Lien J, Moses MA, et al. muscle progenitor cells in human blood. Circulation 2002;106:1199-204.
Tissue engineering of autologous aorta using a new biodegradable 37. Robb BW, Wachi H, Schaub T, Mecham RP, Davis EC. Characteriza-
polymer. Ann Thorac Surg 1999;68:2298-305. tion of an in vitro model of elastic fiber assembly. Mol Biol Cell
10. Niklason LE, Gao J, Abbott WM, Hirschi KK, Houser S, Marini R, et al. 1999;10:3595-605.
Functional arteries grown in vitro. Science 1999;284:489-93. 38. Boerth NJ, Dey NB, Cornwell TL, Lincoln TM. Cyclic GMP-depen-
11. Watanabe M, Shin’oka T, Tohyama S, Hibino N, Konuma T, Mat- dent protein kinase regulates vascular smooth muscle cell phenotype. J
sumura G, et al. Tissue-engineered vascular autograft: inferior vena cava Vasc Res 1997;34:245-59.
replacement in a dog model. Tissue Eng 2001;7:429-39. 39. Matsuda T. Recent progress of vascular graft engineering in Japan. Artif
12. Hoerstrup SP, Zund G, Sodian R, Schnell AM, Grunenfelder J, Turina Organs 2004;28:64-71.
MI. Tissue engineering of small caliber vascular grafts. Eur J Cardio- 40. Welsh ER, Tirrell DA. Engineering the extracellular matrix: a novel
thorac Surg 2001;20:164-9. approach to polymeric biomaterials. I. Control of the physical proper-
13. Hoerstrup SP, Kadner A, Breymann C, Maurus CF, Guenter CI, Sodian ties of artificial protein matrices designed to support adhesion of vascu-
R, et al. Living, autologous pulmonary artery conduits tissue engineered lar endothelial cells. Biomacromolecules 2000;1:23-30.
from human umbilical cord cells. Ann Thorac Surg 2002;74:46-52. 41. Xu C, Inai R, Kotaki M, Ramakrishna S. Electrospun nanofiber fabri-
14. L’Heureux N, Paquet S, Labbe R, Germain L, Auger FA. A completely cation as synthetic extracellular matrix and its potential for vascular
biological tissue-engineered human blood vessel. FASEB J 1998;12:47-56. tissue engineering. Tissue Eng 2004;10:1160-8.
15. Sumpio BE, editor. Hemodynamic forces and vascular cell biology. 42. Myles JL, Burgess BT, Dickinson RB. Modification of the adhesive
Austin, TX: RG Landes Publishers; 1993; p.1-134. properties of collagen by covalent grafting with RGD peptides. J Bio-
16. Kanda K, Matsuda T, Oka T. In vitro reconstruction of hybrid vascular mater Sci Polym Ed 2000;11:69-86.
tissue. Hierarchic and oriented cell layers. ASAIO J 1993;39:M561-5. 43. Mann BK, West JL. Cell adhesion peptides alter smooth muscle cell
17. Kanda K, Matsuda T. In vitro reconstruction of hybrid arterial media with adhesion, proliferation, migration, and matrix protein synthesis on
molecular and cellular orientations. Cell Transplant 1994;3:537-45. modified surfaces and in polymer scaffolds. J Biomed Mater Res 2002;
18. Tranquillo RT, Girton TS, Bromberek BA, Triebes TG, Mooradian DL. 60:86-93.
Magnetically orientated tissue-equivalent tubes: application to a circumfer- 44. Barrera DA, Zylstra E, Lansbury PT, Langer R. Synthesis and RGD
entially orientated media-equivalent. Biomaterials 1996;17:349-57. peptide modification of a new biodegradable copolymer: poly(lactic
19. Berglund JD, Mohseni MM, Nerem RM, Sambanis A. A biological acid-co-lysine). J Am Chem Soc 1993;115:11010-1.
hybrid model for collagen-based tissue engineered vascular constructs. 45. Mann BK, Schmedlen RH, West JL. Tethered-TGF-beta increases
Biomaterials 2003;24:1241-54. extracellular matrix production of vascular smooth muscle cells. Bioma-
20. Zund G, Hoerstrup SP, Schoeberlein A, Lachat M, Uhlschmid G, Vogt terials 2001;22:439-44.
PR, et al. Tissue engineering: a new approach in cardiovascular surgery: 46. Yoon JJ, Kim JH, Park TG. Dexamethasone-releasing biodegradable
Seeding of human fibroblasts followed by human endothelial cells on polymer scaffolds fabricated by a gas-foaming/salt-leaching method.
resorbable mesh. Eur J Cardiothorac Surg 1998;13:160-4. Biomaterials 2003;24:2323-9.
21. Kim BS, Nikolovski J, Bonadio J, Smiley E, Mooney DJ. Engineered
smooth muscle tissues: regulating cell phenotype with the scaffold. Exp
Submitted Oct 12, 2004; accepted Dec 6, 2004.
Cell Res 1999;251:318-28.
22. Mooney DJ, Mazzoni CL, Breuer C, McNamara K, Hern D, Vacanti
JP, et al. Stabilized polyglycolic acid fibre-based tubes for tissue engi- Additional material for this article may be found online
neering. Biomaterials 1996;17:115-24. at www.mosby.com/jvs.

You might also like