Sei sulla pagina 1di 24

REVIEW

Tissue Engineering www.advhealthmat.de

Tissue Engineering at the Blood-Contacting Surface:


A Review of Challenges and Strategies in Vascular Graft
Development
Daniel Radke, Wenkai Jia, Dhavan Sharma, Kemin Fena, Guifang Wang,
Jeremy Goldman, and Feng Zhao*

can cause stenosis or occlusion of blood


Tissue engineered vascular grafts (TEVGs) are beginning to achieve clinical vessels and is commonly associated with
success and hold promise as a source of grafting material when donor coronary artery disease, cerebrovascular
grafts are unsuitable or unavailable. Significant technological advances have disease, deep vein thrombosis, atheroscle-
rosis, and other serious complications.[3]
generated small-diameter TEVGs that are mechanically stable and promote
Treatment methods of CVD include
functional remodeling by regenerating host cells. However, developing a lifestyle modification, pharmaceuticals, or
biocompatible blood-contacting surface remains a major challenge. The TEVG surgery. For situations requiring surgery,
luminal surface must avoid negative inflammatory responses and thrombo- autologous grafts are the current gold
genesis immediately upon implantation and promote endothelialization. The standard treatment method. However,
although autologous grafts are preferred
surface has therefore become a primary focus for research and development
over synthetic grafts, acceptable donor
efforts. The current state of TEVGs is herein reviewed with an emphasis on sites are not always available. Synthetic
the blood-contacting surface. General vascular physiology and developmental grafts are a useful alternative for large
challenges and strategies are briefly described, followed by an overview of the diameter vessels but not ideal for small-
materials currently employed in TEVGs. The use of biodegradable materials diameter vessels (diameters less than
and stem cells requires careful control of graft composition, degradation 6 mm) due to poor patency rates. Tissue
engineered vascular grafts (TEVGs) hold
behavior, and cell recruitment ability to ensure that a physiologically relevant
promise to overcome the limitations seen
vessel structure is ultimately achieved. The establishment of a stable mono­ with synthetic grafts and serve as alterna-
layer of endothelial cells and the quiescence of smooth muscle cells are tive replacement vessels for clinical appli-
critical to the maintenance of patency. Several strategies to modify blood- cations. However, engineering materials
contacting surfaces to resist thrombosis and control cellular recruitment are to mimic the properties of native tissues
is challenging due to the complex struc-
reviewed, including coatings of biomimetic peptides and heparin.
ture and composition. In order to develop
a functional small-diameter vascular graft,
it is essential to understand the blood-
1. Introduction contacting surface and antithrombogenic mechanisms. The
engineering strategies for TEVGs have been extensively
Cardiovascular disease (CVD) is currently the leading cause of reviewed by Pashneh-Tala et al.[3] This review paper focuses on
death in the United States and the world. ≈800 000 CVD deaths the blood-contracting surface of TEVGs.
occur annually in the United States as reported in 2017 by the
American Heart Association.[1] Additionally, the World Health
Organization estimated that 17.5 million people died world- 1.1. Blood-Contacting Surface and Antithrombogenicity
wide from cardiovascular related diseases in the year 2013, and
this number is expected to grow to 23.6 million by 2030.[2] CVD The blood-contacting surface of a vessel’s lumen is covered
with a confluent monolayer of endothelial cells (ECs). A healthy
D. Radke, W. Jia, D. Sharma, K. Fena, Dr. G. Wang, Dr. J. Goldman, endothelium plays a critical role in hemostasis as it directly
Dr. F. Zhao contacts and interacts with substances within circulating
Department of Biomedical Engineering blood. The ECs are anchored to an underlying basement mem-
Michigan Technological University brane, which consists of a thin mat of laminin, collagen, and
1400 Townsend Drive, Houghton, MI 49931, USA
assorted extracellular matrix (ECM) biomacromolecules. This
E-mail: fengzhao@mtu.edu
matrix often contains basic fibroblast growth factor (bFGF) and
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adhm.201701461. transforming growth factor (TGF)-β1, which activates SMCs
to synthesize elastin and other components of ECM.[3,4] The
DOI: 10.1002/adhm.201701461 morphology and function of ECs are critical for maintaining

Adv. Healthcare Mater. 2018, 1701461 1701461  (1 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

vascular health. Endothelial abnormalities are well known


for their significant role in cardiovascular diseases, especially Daniel Radke is an
atherosclerosis.[5,6] Accelerated Master’s student
Hemostasis and thrombus formation are regulated by several at Michigan Technological
interconnected systems and biological factors, including the University where he com-
coagulation and complement cascades, biomechanical forces, pleted his bachelor of science
and circulating cells and blood borne molecules. ECs synthe- in biomedical engineering
size and control many key factors such as heparans, throm- with a minor in psychology.
bomodulin, tissue plasminogen activator (tPA), plasminogen Mr. Radke’s research project
activator inhibitor-1 (PAI-1), adhesion proteins, and platelet is focused on vascular bio-
activating factor (PAF) to regulate thrombus formation through mechanics and vascular graft
interactions with secondary molecules. These EC-secreted mol- engineering.
ecules include thrombin, antithrombin III (AT III), protein C,
endothelial protein coupled receptor (EPCR), platelet activating
factor receptor (PAFR), plasmin, and plasminogen. Figure 1 Jeremy Goldman is a pro-
depicts the ECs’ key role in regulating the antithrombogenic fessor in the Department
properties of blood vessels. of Biomedical Engineering
at Michigan Technological
University, where he has been
1.1.1. Platelet Adhesion, Coagulation Cascade, and Regulation on the faculty since 2004.
of Thrombus Formation Dr. Goldman’s research
interests are focused mainly
The coagulation cascade is divided into an intrinsic and extrinsic in the area of vascular
pathway. The endothelium plays a central role in coagulation engineering. He is developing
cascades due to its unique antithrombogenic, fibrinolytic, and vascular biomaterials,
anticoagulant properties. Because collagen and other ECM pro- including metallic and animal-
teins are highly thrombogenic, trauma or disease that damages derived processed extracellular matrix, for bioabsorbable
the endothelium and exposes underlying thrombogenic mate- arterial stents and vascular grafts, respectively.
rials can trigger thrombosis. Platelets initiate the coagulation
cascade by immediately binding to exposed ECM components Feng Zhao is an associate
such as collagen, fibronectin, laminin, and thrombospondin professor in the Department
at the site of vascular damage. Binding to these molecules of Biomedical Engineering
activates platelets, which release prothrombotic molecules, at Michigan Technological
including adenosine diphosphate (ADP) and thromboxane, University. Dr. Zhao’s research
leading to further activation and aggregation of platelets.[7,8] interests are focused mainly
Platelet activation leads toward synthesis of additional adhesive on stem cell engineering and
molecules, which further increase the platelet adhesion rate. cardiovascular tissue engi-
For example, the de novo expression of vascular cell adhesion neering. She also develops
molecule (VCAM-1), also known as CD106, facilitates the adhe- synthetic and naturally derived
sion of leukocytes to ECs.[6,9] von Willebrand factor (vWF) is biomaterials for tissue engi-
another platelet adhesive protein that is synthesized by ECs and neering applications.
links platelets to collagen. vWF is present in all ECs but is not
secreted unless activated by the coagulation cascade.[7,10] CD31
(PECAM1), CD54, (ICAM-1), P-selectin, and E-selectin are cel-
lular adhesion proteins present on ECs that can bind with plate- coagulation cascade and is released from damaged subendothe-
lets and leukocytes.[6] In order to stabilize the blood clot formed lial tissues. TF is regulated by TFPI, which is a serine protease
by platelet aggregation, components of the intrinsic pathway inhibitor secreted by healthy vascular ECs. Undamaged ECs
activate thrombin, which converts fibrinogen into fibrin poly- also express thrombomodulin, an integral membrane protein
mers to facilitate thrombus cross-linking. The presence of a that binds thrombin and stimulates the activation of protein C,
fibrin clot further increases platelet incorporation.[11] Avoiding another anticoagulant. In addition, it is crucial to disintegrate
activation of these adhesive proteins can reduce blood coagula- and clear the thrombus after damage is repaired in order to
tion and thrombus formation after TEVG implantation. maintain homeostasis. Tissue plasminogen activator (tPA) pro-
Prothrombin, fibrinogen, tissue factor (TF), and kallikrein motes fibrinolysis by catalyzing the breakdown of plasminogen
are key procoagulation factors. These thrombus-forming com- to plasmin and is inhibited by plasminogen activator inhibitor
ponents are controlled by several anticoagulation factors, (PAI-1). tPA and PAI-1 are secreted by ECs to regulate fibrinol-
including tissue factor pathway inhibitor (TFPI), thrombomod- ysis of thrombi.[5,6,8–10]
ulin, protein C, and tissue plasminogen activator. Thrombo- The natural anticoagulation mechanism involves several
modulin and EPCR stimulate protein C, which contributes to factors that can be used or mimicked to improve patency of
anticoagulation.[6,8,10] TF initiates the extrinsic pathway of the TEVGs. AT III and TFPI are upregulated by the presence

Adv. Healthcare Mater. 2018, 1701461 1701461  (2 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 1.  The role of endothelial cells (ECs) in the regulation of thrombogenesis: Heparans, thrombomodulin, tissue plasminogen activator (tPA),
plasminogen activator inhibitor-1 (PAI-1), adhesion proteins, and platelet activating factor (PAF) play key roles in regulating coagulation. These impor-
tant factors interact with other molecules such as thrombin, antithrombin III (AT III), protein C, endothelial protein coupled receptor (EPCR), platelet
activating factor receptor (PAFR), plasmin, and plasminogen to promote or prevent thrombus formation. Thrombi are formed when damaged endothe-
lium expose extracellular matrix (ECM) proteins such as collagen, which then recruit platelets, von Willebrand factor (vWF), and red blood cells (RBCs).

of an intact endothelial layer, and they prevent activation of endothelial activation can help prevent the accumulation of
thrombus promoting factors in the coagulation cascade. Other atherosclerotic plaque, which could otherwise lead to vascular
anticoagulation factors present on ECs include heparans, occlusion.[9,16]
thrombomodulin, and ADPase. ECs synthesize heparans
(or heparin-like glycosaminoglycans (GAG)), which bind to
AT III and greatly decrease thrombogenesis by deactivating 1.1.3. Influence of Mechanical Force on Vascular Tone Maintenance
coagulation factors.[5,7,12] Several studies have demonstrated
the antithrombogenic properties of heparin-coated vascular Pulsatile flow and mechanical stresses in the lumen of blood
grafts.[13] vessels also influence vascular patency. Blood flow produces
shear stress on ECs and causes hyperpolarization of the cell
membrane via potassium channels. This activation stimulates
1.1.2. Inflammation and Complement Cascade Activation the release of nitric oxide (NO) by transcriptional upregula-
tion of endothelial nitric oxide synthase (eNOS) and generates
The complement cascade is an important system to terminate prostacyclin (PGI2), leading to vasodilation. The lipid molecule
foreign cells. There are more than 20 types of plasma proteins PGI2 acts as an antagonist to thromboxane A2, inhibits platelet
involved in the complement system, which function as either aggregation, and promotes vasodilation.[6,17]
binding proteins or catalysts in both the classical and alterna- Relaxing factors are important for regulating vascular tone
tive pathway.[14] Several components of the complement cascade and contribute to the antithrombotic properties of the endothe-
are controlled by ECs. Complement factors, including C1s, C3, lium.[5] Endothelium-derived relaxing factor (EDRF) relaxes
factor B, and the terminal complex components are secreted smooth muscle cells (SMCs) and inhibits thrombus formation.
from ECs upon activation and initiate the cascade. Several other EDRF was identified as NO by Luis Ignarro and his colleagues,
EC-secreted factors inhibit the complement cascade, including for which they won a Nobel Prize in 1998.[18] PGI2 and NO
C1 inhibitor, factor H, and factor I. The complement inhibitors inhibit platelet aggregation by stimulating increased levels of
CD46, CD55, and CD59 are bound to the surface of ECs.[6,15] cAMP and cGMP. Endothelium-derived hyperpolarizing factor
Endothelial inflammation plays a key role in atherosclerosis (EDHF) also contributes to vasodilation by hyperpolarization
and vascular occlusion. Endothelial inflammation is a complex of the cell membrane through K+ channels, which causes the
response triggered by either an acute endothelial response or vascular smooth muscle layer to relax.[5,19] In recent literature,
a phenotypic alteration, which can stimulate a prolonged reac- NO release was found to decrease thrombus formation and
tion. Monocyte chemoattractant protein (MCP)-1, secreted by improve vascular patency, demonstrating that eNOS and NO
actively inflamed ECs, contributes to the inflammatory and are important factors in regulating the antithrombogenic prop-
immune response by recruiting monocytes (macrophages) to erties of ECs.[20]
an injury site. Agonists (such as endotoxin, interleukin-1, and The endothelium acts as a mechanoreceptor, sensing
tumor necrosis factor (TNF)), oxidized lipoproteins, advanced changes in blood flow and pressure, which triggers the release
glycation end products, and mechanical stimulation mediate of vasodilation or vasoconstriction signaling molecules to the
endothelial activation and secretion of MCP-1. These factors vascular smooth muscle cells. To counter vasodilation, ECs pro-
regulate gene expression within ECs by activating the pleio- duce endothelin-1 if activated by shear stress in the presence of
tropic transcription factor, nuclear factor-κB. Prevention of thrombin or under hypoxic conditions, which stimulates SMCs

Adv. Healthcare Mater. 2018, 1701461 1701461  (3 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

to contract, leading to vasoconstriction.[6,21] Studies investi- 2. Blood Interface of Vascular Grafts


gating the effects of biomechanical stimulation on ECs have
shown that laminar shear stress plays an important role in Vascular graft implantation triggers protein absorption, com-
maintaining natural endothelial morphology. Cultured ECs plement activation, platelet adhesion, and foreign reaction.
under laminar shear stress upregulated transcription factors Recapitulating a hemocompatible blood-contacting interface on
such as KLF-2 (Kruppel-like factor), KLF-4, and nuclear factor vascular grafts is a major goal of bioengineering replacement
erythroid 2-related factor-2 (NRF-2). This biomechanical stimu- vascular tissues. The character of the blood–material interface
lation promotes an antithrombogenic phenotype and normal is determined by material properties, blood flow effects, and
cell shape, alignment, and organization. It also promotes glyco- the biological environment.[25] Material selection and under-
calyx formation, a glycosaminoglycan-rich layer on the surface standing their effect on blood and blood borne cells are critical
of ECs, which triggers the expression of eNOS and subsequent for TEVG engineering.
release of NO.[9,22] In contrast, ECs under disturbed flow (i.e.,
nondirected, as found in branched or curved arteries) express
the pleiotropic transcription factor nuclear factor-κB and mimic 2.1. Naturally Derived Materials
the abnormal phenotype of ECs found in patients with athero-
sclerotic cardiovascular disease.[9,23] A major benefit of naturally derived materials is that their chem-
ical structure and biological effects are similar to their human
counterparts. Most of the natural materials are biodegradable and
1.1.4. The Influence of Endothelial Cell Activation on can be remodeled and replaced by native cells and ECM. Conse-
Thrombus Formation quently, grafts fabricated from naturally derived materials can be
integrated and attain a close approximation to a native vessel.[26]
Activation of ECs can stimulate phenotypic changes that influ-
ence the antithrombogenic properties of blood vessels. ECs
can be activated by vascular endothelial growth factor (VEGF), 2.1.1. Elastin
cellular adhesion, several cytokines, mechanical stimulation,
or endotoxin. An understanding of endothelial activation and Elastin, composed of tropoelastin and fibrillin, is an important
thrombotic endothelial diseases can help design TEVGs with ECM constituent in vertebrate tissues that require elastic recoil.[27]
improved antithrombogenic properties.[5,9] As an essential constituent of blood vessels, it plays a vital role
Surgical implantation of a TEVG damages neighboring tis- in vessel elasticity and SMC phenotype regulation.[28] Tropoe-
sues and can disrupt endothelial continuity. It is particularly lastin can be secreted by SMCs, ECs, and fibroblasts. Elastin has
important to avoid surgical trauma induced intimal hyperplasia become an important component for constructing TEVGs, as it
at the interface between the implant and native vasculature. exhibits antithrombogenic and anti-inflammatory properties in
This disease promoting tissue has mismatching mechanical addition to imparting material properties of elasticity. Its degra-
properties and can protrude into the vascular lumen, which can dation products, bioactive elastin-derived peptides (EDPs), retain
disrupt laminar blood flow and predispose toward thrombus the property of decreasing platelet aggregation and thrombosis.[29]
formation. Vascular trauma induces platelet adhesion, migra- Elastin directly isolated from vasculature was found to cause only
tion of proinflammatory cells especially monocytes/mac- minor platelet adhesion.[30] A study by Simionescu et al. obtained
rophages, and promotes excessive cellular proliferation and pure elastin from porcine aortas and tested its potential as a TEVG
activation, which can result in atherosclerotic plaque develop- scaffold. In vitro results revealed reduced platelet adhesion and
ment.[5,9,24] Activation of ECs or damage to the endothelial layer aggregation compared to collagen scaffolds derived from porcine
compromises the antithrombogenic properties of blood vessels. aortas. The research demonstrated that elastin scaffolds promote
Therefore, maintaining a healthy and undamaged endothelial EC proliferation, viability, and endothelial layer formation.[31]
layer is the first and most important consideration to enhance Another study applied purified porcine arterial elastin combined
the patency of cellularized TEVGs. Various strategies have been with fibrin-bonded layers from acellular small intestinal mucosa
developed to address this issue that are reviewed in the present to improve the mechanical properties of elastin.[32] The researchers
contribution. These strategies include novel in vitro antithrom- tested acute thrombogenicity of the construct using a porcine
bogenic cell seeding, EC maturation by provision of physiolog- carotid artery interposition model. The elastin-containing construct
ical pulsatile flow, as well as promoting efficient in vitro and/ remained patent significantly longer than commercially available
or in situ endothelialization by modifying the blood-contacting PTFE. While thrombus in the elastin construct was restricted to
surface via attachment of biomimetic peptides, antibodies, and the suture sites, thrombosis was elicited along the entire length
growth factors. of the PTFE graft. Incorporating elastin into TEVGs is therefore
When developing vascular grafts for CVD patients, a broad a promising approach to achieve a stable blood-contacting surface
range of disease characteristics that affect the confluence and based on its antithrombogenic and natural properties.
functionality of the endothelium need to be considered. Symp-
toms of vascular dysfunction include generalized or localized
vascular spasms, abnormal thrombosis, atherosclerosis, and 2.1.2. Silk Fibroin
restenosis.[5] Understanding mechanisms of endothelial dys-
function and treatment methods are critical in TEVG develop- Fibroin is the main component of silk fibers, composed of
ment to prevent coagulation and improve patency results. antiparallel beta sheets. Silk fibroin is a biodegradable and

Adv. Healthcare Mater. 2018, 1701461 1701461  (4 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

biocompatible scaffold material with high mechanical strength tissues which contain a relatively organized ECM structure,
and a low degradation rate. It has been shown to be nonthrom- such as human greater saphenous vein and intestine, have been
bogenic, as it avoided activation of platelets.[33] The antithrom- investigated for use as vascular scaffolds.[42] Following decel-
botic property of fibroin was also observed by Enomoto et al., in lularization processing, decellularized tissues have been found
a long-term implantation of a vascular graft fabricated with silk to maintain a high compositional similarity to native vascula-
fibroin thread by plaiting.[34] The fibroin graft maintained high ture with respect to collagen and elastin content. Furthermore,
patency after a one year implantation. Implantation of grafts the mechanical strength of decellularized scaffold, including
by Soffer that were fabricated into tubular silk fibroin scaffolds suture retention and compliance, are comparable to native
by electrospinning demonstrated sufficient structural integ- vasculature.[43] In vitro testing revealed that the decellularized
rity to allow handling and to maintain open conduits when human greater saphenous vein exhibits similar burst strength and
hydrated.[35] The scaffolds possess a promising elastic mod- suture-holding strength as fresh veins (fresh: 2480 ± 460 mm Hg,
ulus (average 2.45 MPa) and ultimate tensile strength (average 185 ± 30 g, decellularized: 2380 ± 620 mm Hg, 178 ± 66 g).
2.42 MPa) for potential use as vascular grafts, including the However, tissue engineering approaches that rely upon xeno-
ability to withstand arterial pressures and to behave mechani- graft ECM must surmount pathogenic concerns, while allograft
cally similar to native vessels. These systems also support EC and homograft ECM is in limited supply. To solve this problem,
and SMC proliferation,[35,36] suggesting their future potential in one unique strategy developed tubular ECM by body foreign
TEVG applications. reaction over three weeks of subcutaneous implantation of
polymer rods.[44] The tubular fibrocellular tissue capsule can be
applied as a TEVG material. The graft can also be decellularized
2.1.3. Collagen and repopulated with specialized cell types for improved perfor-
mance as a TEVG. Widespread experience demonstrates that the
Collagen is a main protein component of arteries and veins, ECM supports cell attachment, proliferation, and infiltration,
made from amino acid-packed triple helices, providing mechan- which permits their use as potential TEVG candidates.[45] Never-
ical support along the protein longitudinal axis. Although pre- theless, based on the complex composition of ECM, a bulk ECM
sent in each layer of vasculature, larger bundles of collagen implanted into the vascular system will promote thrombosis.[46]
are concentrated in the vessel adventitia. TEVG scaffolds with Antithrombogenic strategies need to be incorporated into these
high collagen content exhibit high mechanical strength, bio- ECM constructs to improve the TEVG patency.
compatibility, degradability, and accessibility to native cells. Besides decellularization of natural tissues, cell-derived ECM
The collagen fibrils contain integrin binding sites, which allow has been developed to circumvent pathogenic concerns associ-
cells to attach and migrate.[37] However, at the same time, col- ated with xenografts.[47,48] Our group has derived highly aligned
lagen can bind vWF and blood coagulation proteins, which ECM nanofibers by decellularizing aligned human dermal fibro-
promote platelet adhesion on the TEVG surface.[38] Similarly, blasts sheets. The nanofibers are around 80 nm in diameter, sim-
activated platelets also express adhesion proteins including ilar to the physiological size of collagen nanofibers.[49] Recently,
Gp1b, GpIIb-IIIa, CD40L, P-selectin, GPIbα,[46] and ICAM-2, we assembled a completely biological and anisotropic vascular
which induce platelet aggregation and vessel wall invasion of graft by combining the aligned ECM nanofibrous sheet and
activated leucocytes.[39] Therefore, when constructing TEVGs hMSCs in a rotating wall bioreactor system.[47] The TEVG demo­­
from collagen-containing matrices, antithrombogenic materials nstrated increased tensile strength and anisotropy compared
or anticoagulation reagents should be incorporated into the with its static culture counterpart. Dr. Tranquillo’s group has cre-
scaffold, especially on the blood-contacting surface, to reduce ated a TEVG by growing ovine dermal fibroblasts in a sacrificial
the thrombogenic effects of collagen.[40] Huynh et al. developed tubular fibrin gel mold and then decellularizing the resulting
an acellular collagen-based TEVG by depositing bovine type I tubular cellular assembly. The four weeks implantation of this
collagen on the submucosa of swine small intestines, which TEVG in sheep femoral arteries demonstrated graft patency and
is itself a highly collagenous material.[41] After treatment with mechanical anisotropy along with extensive recellularization.[50] A
an antithrombosis reagent and crosslinking, the graft was similar approach was applied to create an “off-the-shelf” decellu-
implanted into a rabbit model. Histological analysis identified larized vascular graft, which was implanted into young lambs.[51]
SMCs infiltrating within the graft and an endothelial layer at After 50 weeks, explanted grafts displayed physiological stiff-
90 d. The grafts were responsive to various vasoactive agents, ness and complete lumen endothelialization without any signs
indicating successful functional remodeling. of calcification, aneurysm, or restenosis.[51] Most recently, this
group has developed decellularized arteriovenous grafts (AVGs)
by seeding neonatal human dermal fibroblasts in the sacrificial
2.1.4. Decellularized ECM fibrin mold.[52] These decellularized constructs were implanted
into baboons and tested as hemodialysis access points. AVGs
ECM is the collection of biomacromolecules secreted and organ- were extensively recellularized, showed an 83 and 60% patency
ized by cells, which contains complex components including rate at 3 and 5 months, respectively, without calcifications, loss
collagen, laminin, hyaluronic acid, and fibronectin. The ECM of burst strength, or outflow stenosis.[52] Although significant
provides an ideal environment for cells to proliferate and carry progress has been achieved using this strategy, anticoagulation
out their functions. It can mechanically support cells to form an drugs were required for the entire duration of the study, and thus
integrated and functional tissue, as well as provide biomolecules the study focused on long term graft function and biointegration
to maintain cell motility and viability. Decellularized tubular without considering the blood-contacting interface.

Adv. Healthcare Mater. 2018, 1701461 1701461  (5 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

2.2. Synthetic Materials biomaterial that has been extensively investigated. It degrades by
the hydrolysis of ester linkages, and giant cells and macrophages
Synthetic materials provide a reproducible, controllable, and can digest the fragments. Unlike PGA or PLA, the degradation
inexpensive source for TEVG engineering. Synthetic mate- of PCL is significantly slower (longer than 24 month), making
rials usually have higher mechanical strength than naturally it more compatible with the slow rate of tissue healing.[59] One
derived materials, which make them promising in blood pres- study compared small-diameter PCL grafts with ePTFE grafts at
sure-bearing applications. One major obstacle of constructing 24 weeks in rats in terms of endothelial coverage, intima for-
TEVGs from synthetic materials is their lack of cell communi- mation, and cell infiltration after implantation.[60] The authors
cation signals and integrin-binding sites, which might hinder found faster endothelialization and intima formation in the PCL
cell attachment and infiltration into the graft. Moreover, a grafts relative to ePTFE, with better cell infiltration and ECM
broad mechanical compliance mismatch further undermines deposition, indicating that PCL is a more appropriate candidate
the long-term patency of these grafts. To improve the proper- than ePTFE for TEVGs. Varied combinations of polyesters have
ties of synthetic materials, innovative modifications have been been used to synthesize biodegradable materials with different
developed. mechanical properties and degradation rates. A PGA nonwoven
fabric sheet was combined with P(CL/LA) (50:50) by pouring
P(CL/LA) solution onto the sheet followed by freeze-drying.
2.2.1. Non-Biodegradable Synthetic Materials Tubular grafts were constructed with the material to a diameter
of 8 mm. The grafts were implanted in the inferior vena cava of
Biostable polymers, such as polyethylene terephthalate (aka canines for a long-term performance test at low pressure.[61] The
PET, Dacron) and expanded polytetrafluoroethylene (ePTFE), goal was to achieve a biodegradable scaffold that was capable
have been utilized as prosthetic large vascular grafts of promoting regeneration through optimized mechanics and
(ID > 6 mm).[53] When applied as large diameter vessels, the degradation time without the need for cell seeding. After two
higher rate of blood flow and larger flow area together with the years of implantation the graft exhibited well-formed vascula-
relatively inert property of PET and ePTFE decreases the possi- ture with high similarity to native vessels without observations
bility of TEVG occlusion. They also provide strong mechanical of serious thrombus in the inner layer of the graft. However, due
support to tolerate blood pressure and pulsatile blood flow.[54] to the hydrolysis of the graft, the loss of mechanical strength at
However, when applied in small diameter vessels (ID < 6 mm) early stages of implantation resulted in stenosis. Reinforcement
that have a much lower blood flow rate, the vascular grafts to increase mechanical strength and endothelialization of the
tend to fail by occlusive thrombus. Moreover, biostable mate- graft is needed to improve the TEVG performance. Polydiox-
rials are prone to chronic foreign body responses that form anone ((C4H6O3)n, PDO) is a biocompatible and biodegradable
fibrotic capsules around the graft. The mechanical mismatch polymer that has been widely used as wound closure sutures.
between material and host tissue may also compromise vessel Similar to Dacron and ePTFE, PDO provides strong mechanical
functionality, promoting cell hyperplasia near the graft–tissue retention when applied as a TEVG scaffold.[62] Its degradation
interface. In order to improve the blood-contacting surface of rate is lower than PLGA and PGA,[63] but faster than PCL. Sell
nonbiodegradable polymeric TEVGs, ECs have been seeded et al. cospun PDO with elastin at a 1:1 ratio, using PDO as a
into the vascular lumen. Nevertheless, the generated endothe- backbone to provide mechanical support while elastin provided
lium fails to completely cover the neointima, which contributes bioactive signals to prevent platelet adhesion.[64] The mechanical
to thrombus formation.[55] Thus, the biostable polymers require strength of the construct was similar to that of native vessels
surface modification and bioactive molecular incorporation to and the incorporation of elastin promoted cell infiltration. A
improve EC confluence on the material surface.[56] similar study by Smith et al. employed an electrospun PDO-
elastin tube reinforced by cross-linking to withstand suturing.
The burst strength of the tube was significantly elevated by the
2.2.2. Biodegradable Synthetic Materials processing.[65]
Overall, natural materials are more biocompatible since they
Linear aliphatic polyesters such as poly(caprolactone) (PCL), are remodelable by host cells and their degradation products
poly(lactic acid) (PLA), poly(glycolic acid) (PGA), and their copol- can be assimilated or metabolized and excreted by the body.
ymers poly(lactic acid-co-glycolic acid) (PLGA) are biodegrad- However, using natural or naturally derived materials often
able and biocompatible polymers that have achieved approval requires complicated and time-consuming purification or iso-
from the U.S. Food and Drug Administration (FDA) for clinical lation processes, and reproducibility may be challenged due
use.[57] The first preliminary study evaluating a PGA tube as a to natural variations. The low mechanical strength of natural
vascular graft was conducted in a rat model by Lauritzen in 1983 materials often necessitates additional processing for reinforce-
utilizing grafts with an internal diameter of 1 mm.[58] This ini- ment. Synthetic materials are more reproducible and easily
tial experiment discovered that half the venous PGA grafts were processed but require incorporation of bioactive molecules
completely replaced by regenerated vascular tissue with com- and careful design to obtain suitable degradation rates and
plete endothelium and developed subendothelial layers over the mechanical strength. More importantly, a functional TEVG
duration of the study, suggesting that PGA tubes are promising design must be antithrombotic. Incorporation of antithrom-
constructs for vascular graft engineering. It should be noted that botic materials such as elastin and designing synthetic mate-
the failure mode for the other half of the PGA grafts was due to rials with nonthrombogenic surfaces provide new directions for
damage caused to the intima during surgery. PCL is a versatile engineering TEVGs.

Adv. Healthcare Mater. 2018, 1701461 1701461  (6 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

2.3. Influence of Surface Roughness, Topography, and cell orientation and alignment on aligned patterns ridges larger
Mechanical Properties on TEVG Hemocompatibility than 800 nm.[74] Moreover, all of the cell types excluding human
aortic ECs showed a significantly increased rate of cell migra-
In addition to considerations related to TEVG material selec- tion on aligned patterns having ridges larger than 1200 nm.[74]
tion, the architecture and topography of the blood-contacting The mechanical properties of the blood-contacting surface are
surface are also important factors affecting thrombus forma- also important regulators of platelet adhesion, spreading and
tion. The aortic endothelium is rough at the sub-micrometer activation.[75–77] A fibrinogen-immobilized polyacrylamide gel
scale, with ridges and grooves that are generally aligned in the with varied stiffness (0.25–100 kPa) revealed that stiffer surfaces
blood flow direction.[66,67] These ridges have an ≈500 nm width (5–50 kPa) significantly enhance platelet adhesion and spreading
and ≈100 nm height.[66,68] These natural topographical features through activation of integrin αIIbβ2, which binds fibrinogen.[76]
may provide increased cellular attachement as well as align- They found that platelets respond to surface stiffness via Rac1
ment in the direction of blood flow. In recent years, various and actomyosin activity by secreting more alpha granules on
studies have focused on elucidating the impact of biophysical stiffer substrates.[76] Another study with a collagen-conjugated
stimulation on the behavior of platelets and ECs. These studies polyacrylamide gel with varying stiffness from 0.25 to 5 kPa
can help in designing appropariate blood-contacting surfaces confirmed this result, as the platelet spreading area increased
for TEVGs that can provide better hemocompatibility. (50–60 µm2) on gels stiffer than 5 kPa, compared to a spreading
Platelet adhesion and activation are significantly affected by area of 30–40 of µm2 on softer gels with a stiffness between
material surface nanotopography gradients. A rough surface has 0.25 and 2.5 kPa.[75] This study found that collagen mediated the
been created by coating gold nanoparticles (36 and 56 nm of diam- stiffness effects by regulating extracellular Ca+2 levels and acto-
eter) onto a smooth gold substrate, which was further coated with myosin pathways rather than through Rho-associated protein
fibrinogen cell adhesion molecules.[69] Results showed that plate- kinase pathways.[75] Although the influence of a TEVG’s blood-
lets were more adherent and readily activated on a smooth surface contacting surface topography and mechanical properties on
relative to a rough one. Others have also found that increased sur- platelet activation/adhesion is a relatively new research direction,
face roughness at a micrometer-level increases thrombogenicity, this needs to be carefully considered for successful TEVG design.
possibly due to the increased surface area available for platelet
adhesion and activation.[70] Surfaces with roughness larger than a
platelet dimension (≈2 µm) have higher thrombogenic potential. 3. Strategies to Improve the Blood Interface
Interestingly, roughness from a 36 nm nanoparticle coating does
of Vascular Grafts
not affect platelet adhesion and activation, suggesting platelets
may have a minimum roughness detection limit of ≈36 nm for Whether off-the-shelf grafts made from biomaterials or cellular
stimulating thrombogenic effects.[71] Variations in surface topog- grafts fabricated by allogeneic or autologous cells, TEVGs all
raphy also affect platelet activity. In contrast to roughness, surface require a functional blood-contacting surface to reduce throm-
topography with structured ridges or grooves (50 nm to 2 µm) bogenicity and maintain patency. Different strategies, such as
significantly reduce platelet adhesion area, as platelet attach- including antithrombotic cells in the graft lumen, stimulating
ment is generally restricted to the groove top. Thus, structured in vivo endothelialization, utilizing thrombosis-resistant graft
surfaces can be highly thrombo-resistant. Similarly, electrospun biomaterials, and chemical modification of the graft surface,
fibers having diameter less than 1 µm effectively avoid platelet have been employed to produce successful blood-contacting
adhesion and blood coagulation, while a significantly increased surfaces in TEVGs (Figure 2).
platelet adhesion has been observed on fibers with diameters of
2–3 µm.[72] Therefore, a well-designed TEVG surface with appro-
priate roughness and topography could significantly improve the 3.1. Incorporating Antithrombotic Luminal Cells to Improve
hemocompatibility of the TEVG blood-contacting surface.[69] TEBV Blood Interface
The native endothelial vascular basement membrane has
nano to microscale topographical features, which guide EC ori- In most TEVGs, thrombosis occurs due to the lack of an EC
entation.[67] These surface topographical features can be used layer or the failure to properly re-endothelialize after implan-
to modulate EC behavior in order to enhance TEVG hemocom- tation. Preimplant luminal cell seeding to provide a func-
patibility. For example, a study conducted to explore rat aortic tional endothelium or antithrombotic cell layer would provide
endothelial cell (RAEC) behavior on a patterned titanium sur- an effective blood-contacting surface. Seeding cells before
face having periodic arrays of grooves with width and pitch implantation, if they are accepted by the host immune system,
ranging from 750 nm to 100 µm demonstrated that a smaller shortens the duration of TEVG regeneration postimplantation.
feature size elicits enhanced cellular effects.[73] The patterned An ideal cell source is one that is easy to obtain and expands
surface with a feature size less than 10 µm promoted increased quickly to large quantities in vitro, retains native function, and
cell density, elongation, and proliferation compared to smooth is nonimmunogenic in the intended recipient.
or randomly pattered surfaces.[73] A similar study was con-
ducted on patterns with ridge and groove sizes ranging from
200 to 2000 nm and at a fixed 300 nm groove depth. Four dif- 3.1.1. Cell Types
ferent vascular cell types including human umbilical vein ECs,
human dermal microvascular ECs, human aortic ECs, and An ideal TEVG should have biomimetic components and struc-
human saphenous vein ECs exhibited significantly increased ture. It should be able to withstand the natural mechanical

Adv. Healthcare Mater. 2018, 1701461 1701461  (7 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 2.  Modifying blood-contacting surface of TEVG: (1) In vitro endothelial cell seeding on the graft lumen provides a biomimetic environment and
prevents direct contact of blood with the graft surface. (2) In situ endothelialization can be achieved by coating the graft lumen with ECM proteins,
growth factors, EC/EPC specific cell binding peptides or antibodies. (3) Fabrication of TEVG with antithrombogenic natural or synthetic materials
maintains graft patency by preventing platelet adhesion and activation. (4) Coating of various antithrombogenic molecules such as heparin, silver
nanoparticles, argatroban, and thrombomodulin actively maintains graft patency by preventing platelet adhesion and activation.

forces from blood flow without rupture and should be function- the graft revealed an absence of thrombus due to the presence
ally antithrombogenic, a property in which the vascular cells of a uniform endothelial layer similar to native vasculature.[78]
play a vital role. Including vascular cells in the design of TEVGs Including ECs in the inner layer of TEVGs or applying growth
would help to reconstruct functional vascular tissue. Currently, factors to stimulate EC recruitment onto TEVGs contributes to
ECs and SMCs are widely applied in TEVGs. Compared to the formation of a stable endothelium and reduces thrombosis
traditional vascular cell types, stem cells expand the range of after implantation. SMCs are distributed in the vascular media
options when designing TEVGs, since the process to obtain vas- layer, sandwiched between layers of elastin. Although these
cular cells is invasive and the vitality and proliferative capability cells do not directly contact the blood, they indirectly regulate
of adult cells are limited. Furthermore, the multidifferentiation the function of ECs through secretion, maturation, and organi-
capability of stem cells may decrease the number of required zation of ECM,[106] in addition to their mechanical role. SMC
cell types involved in the whole procedure, which could shorten and EC interactions through paracrine factors influence vessel
the time required to fabricate TEVGs. Table 1 provides exam- wall assembly and vessel maturation.[107] The presence of SMCs
ples of typical cell types that have been incorporated in TEVG in EC culture decreases EC apoptosis and stimulates EC qui-
fabrication to improve the TEVG–blood interface. escence via Tie2 signaling, which increases vascular stability
Vascular Cells: A monolayer of ECs line the vasculature, and EC survival.[108] Research by Neff et al. compared TEVG
which plays a vital role in avoiding thrombus formation. functionality in which ECs were seeded on the inner surface
The permeability of the vessel, fibrinolysis, and inflamma- of decellularized porcine arteries either with or without SMCs
tion are also associated with a healthy EC layer that secretes on the outer layer.[91] The group with SMCs not only exhibited
an assortment of molecules to regulate the function of SMCs higher tensile strength and more rapid medial healing but also
and blood borne cells. The expression level of these molecules displayed a greater EC physiologic response to receptor-medi-
is controlled by interactions between circulating blood, ECs ated agonists compared with TEVGs seeded without SMCs.
and other vascular cells.[6] Early stage studies recognized the Unfortunately, although an engineered endothelial layer is the
importance of ECs in the TEVG blood-contacting layer to avoid most straightforward and biomimetic analogue of the natural
thrombosis post implantation. An endothelialized Dacron vas- endothelium, ECs have limited proliferative and regenera-
cular graft was implanted in a patient for nine months before tive ability. Moreover, the sudden exposure to blood flow and
requiring a resection, and upon explant the inner surface of physiological conditions could reduce the attachment of EC on

Adv. Healthcare Mater. 2018, 1701461 1701461  (8 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Table 1.  TEVG with incorporated cells (EC: endothelial cell; ECFC: endothelial colony forming cell; MSC: mesenchymal stem cell; BM-MNC: bone
marrow mononuclear cell; PCL: poly(ε-caprolactone); CS: chitosan; PLLA: poly(l-lactide); PTFE: polytetrafluoroethylene; PLGA: poly(lactic-glycolic
acid); PU: polyurethane)

ECs Nondegradable scaffolds: Human vein,[78] human ECs EC seeding significantly ECs participated in
Dacron,[78,79] Dacron- artery,[79,81,86,88] canine improved TEVG patency. re-endothelialization.
collagen,[80] PTFE;[81] Degra- artery,[83,84] porcine artery[87] Dynamic seeding increased Detached ECs circulated
dable scaffolds: PCL,[82] endothelium confluence in peripheral blood. EC
P(LLA/CL),[83] P(CS/CL),[84] after implantation. attachment depended upon
collagen,[85] decellularized EC source, seeding strategy,
vessels,[86,87] fibroblast cell and scaffold material.
sheets[88]
ECFCs Degradable scaffolds: decel- Sheep artery,[89,91] Canine ECFCs, ECFCs-derived ECs ECFCs promoted and ECFCs differentiated
lularized vessels,[89–91] decel- artery[90,93] directly integrated into the into ECs and integrated
lularized small intestine regenerating endothelium. into the reformed
submucosa,[92]collagen [93] Cell implantation improved endothelium. ECFCs
TEVG patency. recruited circulating blood
progenitor cells to promote
re-endothelialization.
MSCs Degradable scaffolds: Rat artery,[95,98] ovine MSCs, MSCs-derived ECs MSCs possess anti- MSCs recruited endo-
P(CL/LA),[94] PLLA,[95] PCL- artery,[97] canine artery[99] thrombogenic properties, thelial lineage cells and
gelatin,[96] decellularized increasing TEVG patency. stabilized EC colonization
vessels,[97,98] PLGA-PU[99] MSC-derived ECs involved through paracrine fac-
in re-endothelialization. tors. MSC-derived ECs
directly participated in
re-endothelialization.
BM-MNCs Degradable scaffolds: P(CL/ Canine artery,[100] lamb BM-MNCs, BM-MNC BM-MNCs enabled TEVG to BM-MNCs differentiated
LA),[100–102] decellularized vein,[101] canine artery,[103] derived ECs regenerate SMCs and endo- into endothelial lineage
vessels,[103,104] PGLA-P human vein,[104,105] mice thelium. BM-MNCs reduced cells, lining the endothe-
(CL/LA)[105] artery[102] platelet activation and lium. The MSC subpopula-
inflammation, improving tion within the BM-MNCs
TEVG patency. further stabilized the
endothelium.

TEVGs.[109] In order to improve endothelialization as well as model.[89] The graft exhibited vascular function similar to native
reduce processing complexity, different cell sources and strate- arteries, including contraction and relaxation responses. The
gies have been explored. results suggest that ECFCs could be used as an alternative
Endothelial Progenitor Cells (EPCs): EPCs are defined as cells source of ECs in TEVG biofabrication.
that can differentiate into ECs and contribute to new blood Mesenchymal Stem Cells (MSCs): MSCs are a promising cell
vessel formation.[110] By using cell culture technology, two phe- type for vascular engineering. They are widely available in the
notypes of EPCs have been produced: myeloid angiogenic cells body and can be derived from various tissue sources, including
(MACs) and endothelial colony forming cells (ECFCs). MACs adipose tissue, bone marrow, and umbilical vein blood. They
do not have the ability to differentiate into ECs but can secrete can be easily expanded through at least 40 population dou-
paracrine factors to promote angiogenesis. ECFCs give rise to blings in vitro.[113] MSCs are immunoregulatory and nonau-
ECs and directly participate in vasculature formation.[111] ECFCs tologous sources do not cause severe immune responses when
are positive for CD31, CD146, VEGFR-2, and vWF, but nega- implanted into foreign recipients.[114] More importantly, they
tive for CD45 and CD14, reflecting their normal morphology are antithrombogenic because of heparan sulfate proteoglycan
and function for EC differentiation.[24] ECFCs are recruited expression on their surface.[95] They can also recruit ECFCs and
on-site where there is injured endothelium and differentiate ECs on site,[95] and be transdifferentiated into vascular cell types,
into ECs to repair vascular damage. Compared to ECs, ECFCs including ECs and SMCs.[115] These properties make MSCs
have a higher proliferation ability and are therefore easier to especially attractive as a cell source for TEVG engineering,
expand in vitro,[112] which provides an alternative source of ECs. as they could increase graft patency.[116] A completely biologi­
ECFCs from dogs were isolated and expanded in vitro, and then ­cal TEVG was synthesized by MSC sheets by Zhao et al.[117]
prelined on a collagen mesh that was wrapped with segmented The TEVG integrated with native vasculature and formed an
polyurethane. The graft exhibited high patency postimplanta- endothelial inner layer after four weeks implantation, which
tion at three months, as 11 of 12 of the grafts were patent and maintained the patency of the TEVG. Zhou et al. developed a
covered confluently with cells expressing factor VIIIb-related completely biological TEVG by combining MSC-derived ECs
antigen, indicating that the prelined ECFCs had formed into a and SMCs from transdifferentiation.[118] MSC-derived SMCs
functional vascular graft with a confluent EC layer.[93] ECFCs were seeded on a PCL-gelatin mesh to help SMCs maintain
derived from sheep peripheral blood were seeded on decel- a tubular shape and then the scaffold was sacrificed, leaving
lularized porcine iliac vessels and then implanted in a sheep behind a SMC-filled layer. MSC-derived ECs were then seeded

Adv. Healthcare Mater. 2018, 1701461 1701461  (9 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

in the lumen under pulsatile stimulation. This method pro- by PGLA.[129] The conduits were implanted into patients with a
duced a TEVG with similar structure as native vessels and com- median age of 5.5 years. In a follow-up with a median duration
parable mechanical properties including elastic modulus (12.7 of 16.7 months postoperation, there were no complications due
± 1.19 MPa), suture retention (1.62 ± 0.1 N), and burst pressure to thrombosis or obstruction. The maximum follow-up time of
(1.72 ± 0.14 MPa) to the human saphenous vein. The study 32 months demonstrated the long-term feasibility of TEVGs
demonstrated that the multidifferentiation property of MSC constructed from BM-MNCs with an effective blood-contacting
makes it a viable cell source for TEVGs. luminal surface.
Induced Pluripotent Stem Cells (iPSCs): iPSCs are pluripotent The establishment of an EC lining or a complete layer of
stem cells that are derived by reprogramming adult cells.[119] non-EC antithrombotic cells on the lumen of vascular grafts
iPSCs can avoid the ethical issues involved in the use of embry- offers the most effective method to avoid thrombus forma-
onic stem cells (ESCs) while retaining the pluripotent ability to tion and maintain patency after implantation. The cell seeding
differentiate into all cell types. In use, iPSCs are always induced strategy is as important as the cell type selection to ensure
to differentiate into specific lineages before seeding since their a confluent and functional blood-contacting surface in the
pluripotent property might otherwise produce a teratoma. TEVG lumen.
iPSCs provide a novel source for obtaining vascular cells. The
SMCs derived from iPSCs have been shown to maintain phe-
notype stability and perform better than vascular-derived SMCs 3.1.2. Strategies for Preventing Immune Rejection
in terms of SMC-specific proteins and gene expression.[120]
Since iPSCs can be induced into an EC type with phenotypic Vascular grafts seeded with autologous ECs normally do not
plasticity,[121,122] their use as blood contacting cells in a TEVG evoke host immune responses. However, obtaining autolo-
may reduce platelet activation and thrombogenesis postimplan- gous cells requires a biopsy, which might be distressing for
tation. Studies utilizing iPSC-derived ECs in engineering vas- patients. Moreover, this approach suffers from various limita-
culature in 3D tissues[121,123] demonstrated the proangiogenic tions including a long culture period for host cell expansion
capability of the ECs. Research by Nakayama et al. engineered and decreased cellular health depending upon the patient’s age
a bilayered vascular graft by incorporating iPSC-derived SMCs and disease status. Allogenic ECs are readily available from
and ECs into aligned fibrillary collagen. The aligned EC layer donors but increase the risk of immune rejection. In addition
significantly reduced the inflammatory response of monocytes to humeral responses, host T cells can detect allogenic anti-
as compared to randomly oriented ECs.[124] The study demon- gens by allorestricted (direct) recognition and/or self-restricted
strated the feasibility of applying iPSC-derived ECs in building (indirect) recognition, which might result in massive cell death
TEVGs. The iPSCs provide a promising cell source for engi- and graft rejection.[130] Monoclonal antibody treatment is an
neering TEVGs, overcoming the limitations of ESCs while pos- efficient way to modulate host immune responses. Antibodies
sessing pluripotent differentiation capability. targeted to immune cell receptors can block chronic graft rejec-
Bone Marrow Mononuclear Cells (BM-MNCs): BM-MNCs tion. Treatment with an anti-VCAM-1 monoclonal antibody
include EPCs, ECs, MSCs, immune-related cells, and hemat- after allogenic vascular graft implantation resulted in pro-
opoietic stem cells. By combining their features, BM-MNCs longed graft survival.[131] This treatment reduced macrophage
may provide an antithrombogenic property when incorpo- and T cell infiltration at the implant site. Moreover, infiltrated
rated into TEVGs. Harvesting BM-MNCs from bone marrow T cells appeared to be quiescent after antibody treatment.[131]
is also less invasive than collecting vascular cells from blood Very late antigen-4 (VLA-4) can bind with VCAM-1 and induce
vessels.[125] The BM-MNCs can be differentiated into ECs a costimulatory immune reaction. Monoclonal antibody treat-
and SMCs, prior to seeding them in a scaffold,[126] or seeded ment against VLA-4 and VCAM-1 prevents active graft rejection
while undifferentiated.[127] In a study by Cho et al., BM-MNCs and provides immunoosupression to cardiac allografts.[132] In
were differentiated to SMC and EC phenotypes and seeded another study, a rat anti-CD2 monoclonal antibody was tested
on decellularized canine carotid arteries.[103] The resulting in a preclinical model of pig-to-baboon xenograft transplanta-
TEVGs were evaluated in a canine model. They found a sig- tion.[133] This treatment prevented direct cytotoxicity of baboon
nificantly improved patency of the BM-MNC graft compared immune cells toward pig aortic ECs by depleting all peripheral
with unseeded groups after eight weeks. The structure of the CD2 positive cells.[133] A similar study was performed to deter-
TEVG was similar to native tissue and the BM-MN-derived mine the influence of an anti-CD4 monoclonal antibody and
ECs and SMCs were detected in the construct, demonstrating gallium nitrate for murine cardiac allograft acceptance. During
the long-term self-renewal capacity of the BM-MNCs. Fuku- this treatment, immune processes similar to the acute graft
nishi et al. combined BM-MNCs with a nanofibrous scaffold rejection were observed. Moreover, transplanted allografts did
employed as an infrarenal inferior vena cava graft. The patency not undergo long-term tissue remodeling.[134]
of the conduits with BM-MNCs was significantly increased Although current immunosuppression regimens are ade-
(from 1/10 to 9/10) relative to the nonseeded group after a six quate for preventing acute allograft rejection, these treatments
month implantation.[128] Histological results depicted concen- pose a risk of malignancy, infection, hypertension, diabetes,
tric laminated SMCs and a confluent EC layer, demonstrating and hypercholesterolemia.[135] Interestingly, ECFC express all
the differentiation ability of BM-MNCs and their applicability EC markers but are negative for CD14, CD115, and CD45,[136]
in TEVGs. A clinical study was also conducted employing BM- which might reduce the risk of immune rejection. ECFCs can be
MNCs in vascular tissue engineering. BM-MNCs were seeded cultured in endothelial differentiation medium to induce their
on a polymer tube composed of PLA and PCL and reinforced differentiated into ECs. In order to compare immunogenicity of

Adv. Healthcare Mater. 2018, 1701461 1701461  (10 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

allogenic ECFC-derived ECs with allogenic aortic ECs (not dif- poor seeding efficiency, longer culture periods (typically 5–9 d),
ferentiated from ECFC), Ladhoff et al., seeded both of these cell and the risk of contamination.[142,171] In addition, coating the
types into an acellular aortic graft, which was implanted into lumen with adhesion molecules increases the risk of platelet
an allogeneic mismatch rat model.[137] Their results indicated adhesion.[172] If the EC layer is damaged during implantation,
that ECFC-derived ECs moderately upregulate MHC-II and the underlying thrombogenic adhesion molecules can promote
are protected against allogenic humoral as well as cytotoxic serious complications including graft occlusion. Thus, with cel-
T cell mediated immune responses. In addition, allogenic lularized grafts, it is crucial to protect the surface with a con-
ECFC-derived ECs showed excellent endothelialization and fluent layer of antithrombotic cells.
very mild inflammatory responses without any signs of graft Dynamic Cell Seeding: Dynamic cell seeding uses centrifugal
rejection compared to allogenic aortic ECs.[137] Besides ECFCs, force, vacuum pressure, and fluid sheer stress to enhance
MSCs can also be used to modulate host immune responses. seeding efficiency. Graft porosity plays a crucial role for suc-
Allogenic MSCs can alter the cytokine secretion profile of cessful cell seeding under dynamic conditions, especially in
major immune cells, including dendritic cells (DCs), natural vacuum seeding.[144] However, synthetic grafts such as PTFE
killer (NK) cells, and effector T cells.[115] It has been shown that and Dacron need to be preclotted after cell seeding in order to
allogenic hMSCs can reduce secretion of TNF-α, interferon-γ close pores present in the constructs. There are several chal-
and increase secretion of IL-4, IL-10 from DCs, T helper cells, lenges associated with preclotting procedures, as reviewed by
and macrophages.[115] These immune-modulatory properties of Burkel.[173] For example, the preclotted surface is rough and can
MSCs make them an appropriate candidate for development promote platelet adhesion or activation, which eventually leads
of vascular grafts. Another study demonstrated that IL-1β acti- to the induction of neointimal hyperplasia. Thus, patients are
vated macrophages can induce MSC differentiation into α-actin required to take systemic heparin treatments to prevent platelet
positive SMC like cells, which can provide contractility to vas- activation after implantation.[174]
cular grafts.[138] Most recently, a novel approach was developed Tissue engineered scaffolds can be used to mitigate these
to reduce immunogenicity of ECs by silencing the human leu- problems. It is possible to create vascular scaffolds with
kocyte antigen class I (HLA I) using lentiviral vector mediated desired pore size and mechanical properties similar to native
RNA interference (RNAi), which could silence HLA-I expres- arteries.[175] Tissue-engineering approaches eliminate the
sion up to 67%.[139] Importantly, HLA I silenced ECs were posi- need for preclotting the scaffolds with a patient’s blood. Park
tive for CD31 and vascular endothelial (VE)-cadherin, which et al. used a dynamic perfusion reactor for seeding cells into
are critical for successful tight junction formation. In addition, elastic scaffolds to form small diameter vascular grafts.[143]
HLA I silenced ECs were able to synthesize vWF and NO.[139] They seeded a collagen-SMC mixture into poly (l-lactide-co-
This approach can possibly reduce immunogenicity of autolo- -caprolactone) (PLCL) scaffolds under vacuum and cultured
gous and allogenic ECs for successful TEVG construction. the constructs under dynamic strain in a perfusion bioreactor
It is crucial to design efficient approaches to modulate host to promote ECM synthesis and uniform SMC distribution. ECs
immune responses in order to prevent TEVG rejection. These were seeded on the lumen of the graft under the same dynamic
approaches might include (but are not limited to) the use of culture conditions to complete the TEVG. The engineered
autologous cell types, systemic immune suppression treat- grafts showed a confluent EC layer on the lumen along with
ment, inclusion of immunosuppressive antibodies into graft secreted elastin content similar to native arteries.[143] Various
design, and use of immunomodulatory stem cells for allogenic types of bioreactors are being used to generate negative pres-
cellular TEVGs. sure for enhanced cell seeding along with dynamic fluid flow
for uniform cell distribution and matrix deposition.
With advances in bioreactor design and technology, various
3.1.2. Cell Seeding Strategies physiological conditions can be generated during cell culture.
For example, physiological pulse dynamics can be mimicked
Various strategies have been developed for cell seeding in using perfusion bioreactors to improve endothelialization in
small diameter vascular grafts. In vitro cell seeding techniques small diameter TEVGs. Decellularized umbilical vein scaffolds
include static/gravitational cell seeding, dynamic cell seeding, promote efficient cell seeding and development of uniform
magnetic cell seeding, and electrostatic cell seeding.[140] These EC monolayers on the graft lumen under axial rotation.[147]
techniques are briefly summarized in Figure 3. Moreover, use of perfusion circuits (flow-ramping paradigm)
Static Cell Seeding: Static or gravitational cell seeding is the to generate physiological fluid sheer stress and pressure of
simplest technique, in which cells are seeded directly on the 1.45 Pa with pulse frequencies at 80 pulses min−1 significantly
lumen of vascular scaffolds. Static cell seeding usually requires improves expression of a nonactivated EC phenotype, which
coating the graft lumen with cell adhesion molecules, such inhibits the induction of neointimal hyperplasia.[147]
as ECM proteins. The cell adhesion molecule fibronectin has The physiological pulsatile flow generated by a perfusion
been used to coat small-caliber PTFE and Dacron vascular bioreactor creates temporal and spatial variations in shear
grafts in order to promote EC attachment and growth.[142] Simi- stress experienced at the graft lumen, which directly affects the
larly, other ECM proteins such as collagen, gelatin, fibrin, and function and phenotype of luminally seeded ECs. ECs respond
laminin, alone or in various combinations, promotes in vitro to differences in fluid flow conditions, including laminar,
EC adhesion and proliferation on the lumen of PTFE and sinusoidal, and physiological pulsatile flow. These different
Dacron grafts.[141] However, static cell seeding is associated flow conditions affect EC morphology, retention rate, align-
with several notable problems including nonuniform seeding, ment, and apoptosis.[145] Among these various flow conditions,

Adv. Healthcare Mater. 2018, 1701461 1701461  (11 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 3.  Cell-seeding strategies for TEVG endothelialization: 1) Static cell seeding: ECs/EPCs can be directly seeded on the vascular graft lumen. A graft
lumen can be coated with individual or combinations of various ECM proteins for enhanced cell attachment and retention.[141,142] 2) Dynamic cell seeding:
A) Vacuum pressure and centrifugal force applied toward graft lumen increases cell seeding efficiency and reduces cell culture period.[143,144] B) Bioreac-
tors can provide a physiological pulsatile flow environment, which can be used to promote a mature, nonactivated EC phenotype.[145–147] 3) Magnetic cell
seeding: An external magnetic field can efficiently guide and regulate seeding of paramagnetic/superparamagnetic particle coated EC/EPCs on a graft
lumen.[148–150] 4) Electrostatic cell seeding: attachment of e-PTFE graft with capacitor generates temporary positive charge on graft lumen. Cell surface
is usually negatively charged; thus, this temporary positive charge on graft lumen can be used for successful adhesion of ECs/EPCs.[151] 5) Biological
approaches for cell seeding: A graft lumen can be coated with biomimetic cell adhesion peptides,[152–161] specific antibodies to capture ECs/EPCs,[162–165]
and various growth factors to promote in vitro/in situ migration and maturation of ECs/EPCs during cell culture and/or after implantation.[166–170]

physiological pulsatile fluid flow with a pulse frequency of Magnetic Cell Seeding: One of the major obstacles in the devel-
1.25 Hz and a time-average pressure of 100 mmHg produces opment of TEVGs is long cell culture periods and the main-
excellent cell retention with unidirectional EC alignment and tenance of complicated dynamic physiological conditions for
actin bundles oriented in the same direction. In addition, physi- proper cell seeding into vascular scaffolds. A novel strategy for
ological pulsatile fluid flow significantly reduces the rate of faster and more efficient seeding of vascular cells involves the
apoptosis compared to laminar flow and sinusoidal flow condi- use of magnetic beads to label desired cells and application of
tions.[145] Novel advances in pulsatile perfusion bioreactors are an external magnetic field to regulate cellular distribution. Car-
being made by optimizing culture conditions for continuous boxydextran-coated superparamagnetic nanoparticles (Resovist,
maintenance of O2 and CO2 concentrations along with constant Schering, Berlin, Germany) with a diameter of 50 nm can be
delivery of nutrients and removal of waste products.[146] This used to label human umbilical vein endothelial cells (HUVECs),
approach eliminates the conventional need to replace culture which is a typical and widely used EC model.[148] Concentra-
medium. tions of superparamagnetic nanoparticles of 10–200 µg mL−1

Adv. Healthcare Mater. 2018, 1701461 1701461  (12 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

do not affect cellular viability or eNOS expression. An external capacitor. When the graft is removed from the capacitor, the
magnetic field can be used to seed labeled HUVECs on the negative charge returns to the e-PTFE surface. After implan-
lumen of a PTFE graft. In addition, cell seeding can be observed tation, this negatively charged surface prevents platelet adhe-
noninvasively using magnetic resonance imaging (MRI). This sion and contributes to graft patency if the EC layer becomes
method produces a complete and homogeneous monolayer of damaged.
HUVECs on the graft lumen. However, long-term cell retention In summary, small diameter vascular grafts are especially vul-
and potential side effects from the beads need to be evaluated nerable to occlusion. They must be engineered with favorable
to validate the usefulness of this technology. surface properties to avoid thrombogenesis.[178] Endotheliali-
Internalization of magnetic nanoparticles may negatively zation and chemical stability have been shown to be the most
affect cellular physiology and function. Thus, noninvasive cell important factors controlling the thrombosis property.[179] As
coating with magnetic beads may provide a better solution. ECs form the interface between blood and tissues, the pres-
Superparamagnetic polymer particles known as Dynabeads ence of a confluent EC monolayer in a vascular graft can inhibit
(DynaBiotec, Oslo, Norway), have been used to coat the cell sur- the bioactive substances responsible for SMC migration and
face. Similarly, an external magnetic field is used to uniformly proliferation and their production of ECM, and thus improve
seed cells on graft lumens. However, the concentration of thromboresistance and reduce intimal hyperplasia. However,
beads on a single cell surface must be optimized. Tiwari et al. innovative approaches are needed to improve the limited
used CD31 coated Dynabeads to attach them on the surface of capacity of ECs to re-endothelialize. In the absence of a con-
HUVECs (4, 10, and 50 beads per cell) and seeded these cells fluent endothelium, surface treatments have the potential to
on the lumen of PTFE TEVGs using an external magnetic improve thrombosis resistance of small diameter grafts.[180] For
field.[150] Their results indicate that a higher number of beads instance, elastic laminae and modified elastin coatings, or hep-
per cell produced detrimental effects on cell proliferation and arin conjugates can control platelet distribution and decrease
metabolism over time. Although this is an effective method for the degree of platelet activation.[181]
cell seeding, the internalization of the nanoparticles and their
side effects were not discussed.
In addition to distributing cells on synthetic grafts, such as 3.2. Biological Approaches for Enhancing Cell Recruitment
PTFE and Dacron, Dynabeads can be used to cover tissue-engi- and Retention on TEVGs
neered scaffolds with different vascular cell types. For example,
Perea et al. labeled SMCs and HUVECs with CD44 and CD31 Various biological approaches are being developed for suc-
coated Dynabeads, respectively.[149] They seeded SMCs into cessful in vitro and/or in situ recruitment of EPCs and ECs.
the lumen of a collagen membrane under a magnetic field for These include attachment of EPC/EC specific adhesion pep-
40 min, followed by HUVEC seeding for another 20 min under tides, growth factors, and EPC/EC capturing antibodies for
an external magnetic force of 2 pN on each magnetic bead, enhanced endothelialization of the graft lumen. This section
with 90% seeding efficiency. Magnetic cell seeding can create will explore the advantages and limitations associated with var-
uniform and monolayer cell coatings on the lumen of both ious biological cell adhesion approaches that have been devel-
synthetic and natural scaffolds, accurately and within short cul- oped for TEVGs.
ture times. However, the long-term cell retention, phenotypic
changes, and other side effects after in vivo implantation need
to be addressed. 3.2.1. Biomimetic Peptides
Electrostatic Cell Seeding: The material properties of vascular
grafts also play a crucial role in successful endothelialization Coating TEVGs with the Arg-Gly-Asp (RGD) peptide is an
processing. For example, synthetic grafts such as e-PTFE are effective approach to promote EC adhesion and migration.[182]
highly negatively charged.[176] The surfaces of ECs and plate- Several research groups have used variants of RGD peptides
lets are also negatively charged.[177] Thus, ECs must overcome to enhance endothelialization, including Gly-Arg-Gly-Asp-
the repulsive force present between the material and cell inter- Ser-Pro (GRGDSP),[159] RGD combined with the Pro-His-Ser-
face for successful cell attachment. To overcome this challenge, Arg-Asn (PHSRN) peptide (a fibronectin associated binding
various research groups have evaluated the effects of a range sequence), and RGD combined with the Tyr-Ile-Gly-Ser-Arg
of ECM protein coatings on the graft lumen for successful cell (YIGSR) peptide (part of the laminin β1 chain).[153] How-
adhesion.[141,160] However, these coatings create a risk of platelet ever, coating the graft lumen with chemically synthesized low
adhesion and activation if the EC layer becomes damaged molecular weight RGD peptides has some limitations. For
during or after implantation. instance, instead of increasing EC adhesion, the RGD peptide
Electrostatic cell seeding temporarily alters the electrical can detach from the graft substrate after becoming attached
charge of the graft lumen to enhance cell attachment. For to a cell’s surface via integrin receptors. In order to increase
example, e-PTFE is dielectric. When the graft is attached to a the molecular weight of RGD peptides and their retention
capacitor, negatively charged nuclei (electrons) of dielectric capability on the graft lumen, a novel recombinant RGD-con-
(graft) material are attracted toward the surface of the capacitor, taining fusion protein was created by connecting the cellulose-
temporarily imposing a positively charged lumen surface.[151] binding domain (CBD) to RGD peptides. This fusion protein
This temporary positive charge facilitates EC attachment. It is was coated on an inner gelatin layer of polyurethane grafts[155]
important to remember that the graft is made up of insulating and significantly increased EC retention and reduced platelet
material, so there is no electric current passing from graft to adhesion/activation .

Adv. Healthcare Mater. 2018, 1701461 1701461  (13 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Other biomimetic substances, such as the mussel-inspired 3.2.2. Growth Factors


adhesive polydopamine, are being investigated to determine
their potential for enhancing endothelialization. A study con- VEGF and bFGF are known regulators of angiogenesis, vas-
ducted by Lee et al. found that a polydopamine coating in com- cular growth, and blood vessel maturation. VEGF in particular
bination with RGD and YIGSR on decellularized vein matrix is a potent mitogen and chemoattractant for ECs.[184] Thus, it
promotes EPC differentiation into ECs with enhanced focal is a good promoter for TEVG endothelialization.[185] Decellular-
adhesions.[158] However, platelet adhesion/activation was not ized carotid arteries can be coated with VEGF and heparin.[170]
investigated in this study. In order to promote in situ recruit- This modified graft can sustainably release VEGF up to 20 d
ment of EPCs and ECs, a microporous PCL graft coated with and promotes complete endothelialization within six months
fusion proteins containing the mussel adhesive protein (MAP) of implantation. Moreover, they showed significantly reduced
and RGD peptide was evaluated over four weeks in the rabbit neointimal hyperplasia and increased patency compared to
carotid artery.[156] Results showed that the MAP-RGD pro- unmodified grafts following in vivo implantation.[170] In addi-
moted in situ EPC/EC recruitment with a 66% patency rate. For tion to decellularized scaffolds, nanofibrous scaffolds coated
unknown reasons, the graft endothelialized prior to implanta- with VEGF and heparin exhibited anticoagulation properties
tion was less effective at maintaining patency than the graft that and enhanced EPC growth in vitro.[166]
was endothelialized in situ.[156] PDGF plays a critical role in the recruitment of pericytes and
Various studies suggest that stromal cell–derived factor-1α smooth muscle cells, which stabilizes blood vessels.[169] Multi-
(SDF-1α) plays a major role in the transportation of hematopoi- layer electrospun nanofibrous scaffolds with an inner layer of
etic stem cells from bone marrow to the peripheral blood. It also poly(ethylene glycol)-b-poly(l-lactide-co-ε-caprolactone) (PELCL)
promotes EPC recruitment into ischemic microenvironments along with gelatin and VEGF, a middle layer containing PLGA
and regulates neoangiogenesis.[152,160] Thus, SDF-1α can be along with gelatin and PDGF and an outer layer containing
used to promote TEVG endothelialization. Nanofibrous vascular PCL enhances recruitment of vascular ECs and SMCs. Release
scaffolds prepared from a PLLA and PCL polymer blend, with of VEGF and PDGF increases endothelialization and inhibits
the luminal surface coated with heparin and SDF-1α, reduces SMC hyperproliferation, with enhanced patency over eight
platelet adhesion and activation. Moreover, the combination of weeks.[167] The cross-linking process to conjugate various
heparin and SDF-1α improved long-term graft patency as well growth factors depends on the type of scaffold (i.e., synthetic
as in situ recruitment of EPCs and smooth muscle progenitor grafts, decellularized scaffolds, and electrospun scaffolds), and
cells (SMPCs). The SMPCs differentiated into SMCs, which sig- details can be found in articles cited for each graft type.
nificantly improved the elastic modulus of the grafts.[161] In addition to growth factor coatings, plasmids encoding
Currently, peptides/proteins that bind selectively to ECs or genes of specific growth factors can be incorporated into the
EPCs are being developed to allow highly specific cell recruit- graft lumen. Lahtinen et al. developed this novel approach
ment for endothelialization with minimal risk of platelet adhe- for vascular graft endothelialization via incorporation of plas-
sion and activation side effects. The basement membrane that mids encoding the human VEGF165 and FGF-2 genes into the
separates ECs and SMCs in arteries contains a high collagen lumen of ePTFE and preclotted polyester grafts.[168] They found
type IV content. Trimeric peptides found with high redundancy that the VEGF165 plasmid improves graft endothelialization
in human collagen type IV were screened using peptide array while combined FGF-2 and VEGF165 plasmids reduce graft
based cell adhesion effects.[183] By comparing results between endothelialization. They also found that VEGF165 plasmid
EC and SMC adhesion using cysteine-alanine-glycine (CAG) incorporation into preclotted polyester grafts increases endothe-
peptides, it was found that CAG has a high affinity for ECs lialization and patency relative to ePTFE grafts.[168]
with minimal cross-reactivity for SMCs.[157] Small caliber PCL
grafts containing the CAG peptide sequence, incorporated by
blending the CAG peptide with the polymer during electrospin- 3.2.3. Antibody Coating
ning, experience a significantly increased endothelialization
and eNOS expression after implantation compared to grafts Antibodies can specifically target and recruit ECs/EPCs on
without the CAG peptide.[157] Hao et al. studied the cell recruit- the graft lumen by engaging with specific surface antigens
ment potential for the LXW7 ligand, which specifically binds or receptors. For example, several studies have reported that
to the αvβ3 integrin present on the EC/EPC surface. LXW7 coating cardiovascular stents with anti-CD34 antibodies
is a disulfide cyclic octa-peptide with unnatural amino acids enhances in situ recruitment, adhesion, and proliferation of
flanking both sides of the main functional motif. Due to the ECs and circulating EPCs.[186] Similarly, ePTFE graft lumens
presence of unnatural amino acids, it is more resistant to pro- can be coated with anti-CD34 antibodies using peptide link-
teolysis in vivo compared to linear and natural amino acid con- ages for faster endothelialization and enhanced patency.[165]
taining peptides..[154] They found that the LXW7 ligand exhibits After 28 d of implantation into pig models, 85% of the graft
very strong binding affinity toward ECs/EPCs and weaker surface was covered with an EC layer compared to 32% EC
binding to platelets compared to the conventional GRGD coverage in noncoated grafts. However, despite a promising
ligand for αvβ3 integrin. Moreover, it promotes cellular prolif- endothelialization, anti-CD34 antibody coated grafts evoke an
eration, possibly due to activation of VEGFR2 and the MAPK excessive intimal hyperplasia at the anastomoses.[165] Similarly,
pathway.[154] These EPC/EC specific proteins/peptide sequences another study showed that anti-CD34-coating significantly
can significantly improve the rate of endothelialization in vitro increases platelet adhesion along with protein adsorption,
and/or after graft implantation. compared to noncoated grafts.[164]

Adv. Healthcare Mater. 2018, 1701461 1701461  (14 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Although an anti-CD34 coating improves endothelializa- elastin. It has been shown that the elastin-rich arterial elastic
tion, this approach risks platelet adhesion and induction of laminae is able to suppress inflammatory responses, in part by
neointimal hyperplasia and thus alternative solutions need to inhibiting leukocyte adhesion via activation of Src homology
be investigated. In situ recruitment of EPCs can be achieved 2 domain-containing protein-tyrosine phosphatase (SHP)-1
by coating antibodies targeted toward EPC specific cell recep- through signal regulatory protein (SIRP) α.[192] Elastin con-
tors. CD133 is an EPC-specific surface receptor that can be tains ≈75% hydrophobic amino acids and is therefore highly
used to enhance EPC recruitment on the graft lumen using an insoluble.[193] Novel approaches to synthetically mimic elastin
anti-CD133 coating.[163] ePTFE grafts coated with multilayers have produced materials with a similar inhibition of platelet
of anti-CD133 along with heparin and collagen showed accel- adhesion and activation.[191,194–196] Polyurethane surfaces modi-
erated endothelialization and patency at 7 d after implantation fied with peptides modeled from the cross-linking domains
in a pig model.[162] However, long-term patency has yet to be of elastin have been used to cross-link elastin-like peptides to
investigated. Although antibody coatings accelerate cell recruit- their surfaces, resulting in decreased platelet adhesion and
ment with high cell type specificity, increased manufacturing activation, increased endothelialization, and increased eNOS
costs compromise their commercial application for TEVG expression relative to uncoated controls in vitro.[191,194] Inspired
production. by the ability of elastin to self-aggregate and self-organize,[196]
researchers were able to genetically engineer elastin mimics
capable of both chemical and physical cross-linking.[195] The
3.3. Modification of Blood-Contacting Surface elastin mimic LysB10, a triblock amphiphilic copolymer with
hydrophobic endblocks and a hydrophilic midblock, was able to
Synthetic, naturally derived, and biodegradable TEVGs maintain complete patency for a two week in vivo study when
require blood-contacting surface processing to reduce throm- used as the blood-contacting surface for an acellular vascular
bogenicity and maintain patency. Systemic anticoagulant drug graft.[197] Histological evaluation identified a stable neointima
regimens are commonly used to maintain graft patency in positive for vWF, indicating the acellular graft had successfully
clinical applications, particularly when working with synthetic re-endothelialized.
materials. Modification of the blood-contacting surface can Tropoelastin: The elastin precursor, tropoelastin, rapidly
reduce or eliminate the need for such treatments, decreasing and spontaneously self-aggregates under physiological condi-
the corresponding risk of bleeding by restricting anticoagu- tions to be cross-linked into elastin fibers by the recruitment
lation effects locally to the TEVG. This section will examine of the enzyme lysyl oxidase.[198] This property made isolation
methods currently employed to produce successful blood- and extraction difficult and inefficient prior to the development
contacting TEVG surfaces. of a synthetic elastin gene in E. coli.[199] Tropoelastin has since
been covalently coated on metal stents[200] and polymer vas-
cular grafts[201] where it has successfully imparted antithrom-
3.3.1. Elastic Lamina and Modified Elastin as Blood-Contacting bogenicity, reduced SMC proliferation, and enhanced the devel-
Surface opment of a stable endothelium to improve biocompatibility.
Another study was able to determine that the first 10 N-ter-
Elastin is a key regulator within the vasculature. In addition minal domains (N10) and first 18 N-terminal domains (N18)
to providing the physical elasticity and resilience necessary of tropoelastin facilitate EC attachment and proliferation.[202]
for the conduction of pulsatile blood flow, elastin regulates the These domains retained the antithrombogenicity of full-length
migration and proliferation of SMCs by inducing a quiescent, tropoelastin when plasma coated onto stainless steel and evalu-
contractile phenotype via a nonintegrin-dependent G-protein ated in vitro.
coupled signaling pathway.[187] Elastin exhibits antithrombic Elastin provides a promising biocompatible material for the
and inflammation-resistant properties in vivo, resisting the construction of vascular grafts, able to closely recreate the nat-
adhesion and activation of leukocytes and platelets, suggesting ural structure and mechanics of native tissue while providing
a lack of integrin-binding sites on these cell types.[188] a hemocompatible surface for immediate biocompatibility
These properties make elastin an attractive choice as a blood- and being able to regulate EC and SMC proliferation for long-
contacting material. Elastin has been successfully employed as term stability.[203] A close recreation of native tissue mechanics
a blood-contacting surface in a small diameter vascular graft may be more important than previously thought. While many
by McCarthy et al.[189] The elastin-rich elastic laminae of donor researchers attempt to approximate native compliance and elas-
vessels was decellularized and mechanically reinforced with ticity as closely as is necessary to facilitate undisturbed flow
biodegradable poly(ether urethane) (PEU). This study utilized without risk of aneurysm or burst, recent work has implicated
naturally sourced elastin. However, elastin can also be used to alterations in the mechanical properties of blood vessels as risk
coat synthetic materials through use of its precursor, tropoe- factors for atherosclerosis.[204]
lastin, and peptide mimics, as will be discussed later.[190]
Elastin Mimicry: Synthetic materials can be fabricated with
elastin-mimicking surfaces or coated with elastin-mimicking 3.4. Development of Scaffolds Incorporating
peptides to activate the receptor-mediated signaling mecha- Anticoagulation Molecules
nisms that inhibit platelet and inflammation activation.[191]
These mechanisms are incompletely understood, but are A variety of methods have been developed to actively maintain
thought to be due to the lack of integrin-binding sites for TEVG patency. Figure 4 illustrates the mechanisms of some of

Adv. Healthcare Mater. 2018, 1701461 1701461  (15 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 4.  Inclusion of anticoagulation molecules in TEVGs. 1) Thrombomodulin (TM) is a transmembrane protein present on ECs that binds thrombin.
The thrombin–TM complex activates protein C (PC, APC), which then forms a membrane-bound complex with protein S (S) that inactivates factors Va
and VIIIa. Recombinant TM can be covalently bound to synthetic materials.[206] 2) Argatroban locally and directly inhibits thrombin by noncovalently
interacting with Asp-189 in the S1 binding pocket of thrombin to block its enzymatic activity. Truncated forms were developed which retained the
guanidyl group for ionic interaction and featured a spacer arm on the piperazinyl amide moiety for surface grafting.[207] 3) Nitric oxide (NO) activates
soluble guanylate cyclase (sGC) to increase cyclic guanosine monophosphate (cGMP) production by binding to the sixth position of the heme ring,
breaking the bond to His-105. cGMP then decreases expression of thromboxane A2 and the platelet surface adhesion protein P-selectin. 4) Silver
nanoparticles (SNP) inhibit platelet aggregation by blocking biochemical pathways associated with membrane restructuring, preventing the expression
of surface adhesion proteins.

these methods. Many of these methods, such as heparin, inhibit Thrombomodulin: Since thrombomodulin not only prevents
coagulation by targeting either thrombin or platelets. Heparin thrombin from serving as a procoagulation factor but also
is perhaps the most widely utilized antithrombic agent for repurposes it for anticoagulation, engineering active thrombo-
coating blood-contacting TEVG surfaces and vascular devices. modulin onto blood contacting surfaces would locally generate
This well-characterized molecule has been reproduced syn- protein C to inhibit thrombin activation. One study aimed at
thetically and is therefore inexpensive and readily available. But optimizing surface activity by controlling the random orien-
alternatives, such as argatroban are available if necessary.[205] tation associated with traditional covalent linkages utilized a
truncated form of thrombomodulin containing the binding
sites for thrombin and protein C. The truncated thrombo-
3.4.1. Thrombin Regulation modulin was selectively bound to the surface of ePTFE grafts
through Staudinger ligation.[206] For evaluation purposes, an
The enzyme thrombin plays a pivotal role in coagulation. It is ex vivo femoral arteriovenous shunt model was used in which
responsible for the activation of platelets, the conversion of the treated ePTFE grafts were placed between a collagen-coated
fibrinogen into a fibrin network during clotting, and providing ePTFE graft, as a thrombogenic source, and an expansion
feedback amplification for coagulation. However, thrombin is chamber. The authors found a near 50% reduction in platelet
not exclusively a procoagulant. Thrombomodulin, a membrane deposition in the expansion chamber compared to ePTFE grafts
protein present on ECs, has the ability to bind thrombin, which without the truncated thrombomodulin.[206]
activates protein C and exerts an inhibitory effect on the coagula- Argatroban: Argatroban interacts with Asp-189 of thrombin,
tion system.[208] Thus, a variety of approaches aimed at imparting also known as the specificity pocket S1 binding site, to directly
anticoagulation properties to biomaterials target thrombin. and locally inhibit thrombin. Argatroban is one of only two

Adv. Healthcare Mater. 2018, 1701461 1701461  (16 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

synthetic thrombin inhibitors currently approved for med- NO as a negative feedback mechanism against aggregation, and
ical use by the FDA.[207,209] Argatroban-loaded small diameter its short half-life makes NO an ideal agent to achieve local, bio-
(1–3 mm) vascular grafts fabricated by subcutaneous implan- compatible inhibition of thrombosis.[220,221] Recently, Gao et al.
tation and maturation of silicone rods exhibited patency for developed a small diameter (2 mm) vascular graft from elec-
up to 12 weeks in 3/4 samples implanted into rabbit carotid trospun PCL with a surface treatment of organoselenium-
arteries, as compared to 0/4 patency by two weeks for untreated modified polyethyleneimine (SePEI) to catalyze the production
grafts.[210] These grafts were composed primarily of col- of NO in situ.[222] The grafts displayed a steady generation of
lagen and fibroblasts, known to trigger severe thrombosis.[211] NO in vitro that could be controlled by adjusting the number of
Argatroban has also been used as an antithrombic coating for SePEI layers, allowing it to promote rapid adhesion and prolif-
balloon catheters and stents.[212] Nakayama et al. developed eration of ECs while inhibiting the adhesion and activation of
water-soluble argatroban for improved adsorption on biological SMCs and macrophages. The use of an NO generating catalyst
materials by utilizing ionization of the carboxyl group to dis- rather than NO storage avoids the limitations of NO depletion
solve it in an alkaline solution, followed by freeze-drying to pro- and cytotoxic complications from a burst release profile.
duce a water-soluble argatroban sodium salt.[213] Small diameter Heparin: Heparin is a GAG composed of alternating chains
vascular grafts (1.5 mm) and heart tri-leaflet valves formed via of d-glucosamine and uronic acid residues.[223,224] It functions
subcutaneous implantation of silicone molds treated with the as an anticoagulant by binding with AT III, enacting a confor-
water-soluble argatroban avoided any signs of thrombus or mational change that enhances the ability of AT III to inacti-
neointimal hyperplasia for up to 12 weeks in a canine model, vate thrombin by forming a ternary complex with heparin and
although they were covered by native tissue.[213] thrombin.[225] However, the unique pentasaccharide responsible
for facilitating this binding is only present in about a third of
heparin molecules, making its effectiveness variable.[226] Hep-
3.4.2. Platelet Regulation arin is heterogenous in molecular size, with molecular weight
ranging from 5000 to 30 000 with a mean molecular weight of
The activation of a platelet negatively charges its surface by 15 000, or ≈50 monosaccharide chains.[223] Heparin molecules
activating scramblase, which transports negatively charged shorter than 18 chains are unable to simultaneously bind
phospholipids from its inner to outer membrane and creates thrombin and AT III, but retain anticoagulation properties by
an optimal surface for binding the tenase and prothrombic their ability to catalyze AT III to inhibit factor Xa.[227] Heparin is
complex.[214] Regulating the activation or aggregation of also able to catalyze the inhibition of thrombin through heparin
platelets thus offers another target for controlling coagulation. cofactor II. This cofactor does not require the AT III-binding
Silver Nanoparticles (SNPs): While investigating the potential pentasaccharide, is specific for thrombin, but requires much
for spherical SNPs (10–15 nm in diameter) as cytotoxic antimi- higher doses of heparin for comparative effectiveness to AT
crobials, it was found that they inhibited platelet aggregation III catalysis.[228] Investigation into this alternative mechanism
in a concentration-dependent manner.[215] Further characteriza- led to the development of low molecular weight (LMW) heparin,
tion of this phenomenon determined that platelets incubated however a detailed distinction will not be drawn here due to their
with SNP retained significantly reduced aggregation poten- similar clinical effectiveness. Barrowcliffe’s History of Heparin
tial after removal of the nanoparticles. Thrombin stimulation provides a comprehensive review.[229] More recently, heparin
increased release of ATP/ADP from platelets. Pretreatment has found application in the biomedical field as a means to
with SNP significantly reduced ATP/ADP release in aggregated improve hemocompatibility of biomaterials by reducing platelet
platelets, indicating the SNP interfered with an integrin-medi- adhesion, inhibiting coagulation kinetics, and supporting re-
ated mechanism since platelet aggregation involves ligation of endothelialization even on synthetic materials.[230] Clinical trials
surface integrins.[215] More recent studies have suggested that have determined that heparin immobilized to the surface of
SNPs reversibly inhibit platelet aggregation by blocking bio- ePTFE vascular grafts via covalent bonding remain bioactive
chemical pathways associated with platelet membrane restruc- for up to 12 weeks after implantation, producing results com-
turing during activation.[216] This activity is critically dependent parable to autologous grafting and preferable when the saphe-
upon their size, as SNPs larger than 24 nm have been found nous vein is absent or unsuitable for transplantation, as may be
to promote platelet activation and the coagulation cascade.[217] the case in systemic CVD.[231] Materials may also be engineered
SNPs have been synthesized and immobilize onto a porous to release heparin by utilizing responsive polymers to break the
PCL scaffold through the reaction of PEG and silver nitrate conjugate bonds.[232]
to inhibit platelet adhesion and aggregation in vitro while Of particular interest is the application of heparin to vas-
retaining antimicrobial properties.[216,218] The aggregation of cular scaffolds, where thrombosis poses a serious challenge to
SNP in the medium indicates the protective PEG coating is lost the use of synthetic materials and the ideal vascular replace-
as SNP leach out of the scaffold, but will not cause any cytotoxic ment is one that promotes native tissue regrowth. Several
effects as long as the initial concentration is set optimally low. long-term clinical studies showed that heparin significantly
Nitric Oxide (NO): NO primarily serves as a vasodilator in the improves the patency of synthetic vascular grafts, concur-
regulation of blood pressure and as a cytotoxic agent for nonspe- rent with the results of animal studies, but host integration
cific immune responses. NO also inhibits platelet adhesion and has remained elusive.[233–235] Stable binding of heparin to
aggregation via the activation of soluble guanylate cyclase and the luminal surface of ePTFE grafts significantly improved
consequent increase in the concentration of cyclic guanosine patency compared to untreated controls in the canine model
monophosphate (GMP).[219,220] Platelets themselves produce and remained active for up to 12 weeks.[235] A one year clinical

Adv. Healthcare Mater. 2018, 1701461 1701461  (17 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

trial comparing heparin-bonded PTFE to untreated PTFE grafts through an access graft that had at least one previous failure.
in femoral artery bypass found patency rates favoring the Three grafts failed within the first three months, one patient
heparin-bonded grafts (86.4 to 79.9% primary patency, 88 to was withdrawn because of severe gastrointestinal bleeding,
81% secondary patency) with an overall 37% reduction in risk one died of unrelated cause, and five grafts functioned for 6–20
of graft failure.[236] Another five year clinical study comparing months. Although an endothelium was included in the lumen,
heparin-bonded Dacron to untreated PTFE in femoral artery the graft failure at mid- to long-term was associated with the
bypass found similar results (46 to 35% primary patency, 47 to formation of thrombosis and aneurysm. A major limitation of
36% secondary patency).[234] this approach is the long time required to build the completely
Heparin holds potential to further enhance vascular engi- autologous vascular graft.[243] To address this problem, the group
neering through its ability to promote endothelialization and reported in 2011 the first implantation of a frozen, devitalized,
regulate SMCs.[237] Heparin contributes to the hemocompat- completely autologous Lifeline 4.8 mm vascular graft derived
ibility of synthetic scaffolds, especially in the case of acellular entirely from a fibroblast layer. Living autologous ECs were
biodegradable scaffolds where they endow an immediate, off- seeded in the lumen 5 d before implantation. The graft func-
the-shelf potential.[238] Novel approaches are being developed to tioned effectively within eight weeks in a patient who required
exploit heparin-mediated tissue regrowth activity. Smith et al. a semipermanent dialysis catheter.[244] Grafts produced by this
demonstrated that a heparin-bound poly-l-lysine (PLL) sur- method can be stored “off-the-shelf.” The grafts were implanted
face could be used to immobilize VEGF for the capture of ECs into three hemodialysis patients and no degradation or dilation
under shear with high selectivity (37 ± 2 cells mm−2 at shear were observed up to 11 months.[245] However, the time required
of 15 dyne cm−2 compared to 10 ± 4 cells mm−2 for heparin to prepare the grafts and the lack of mature elastin may limit the
controls without immobilized VEGF).[239] This highlights the application of the technology. While vascular grafts produced by
potential use of heparin’s growth factor binding properties to self-assembly approaches have proceeded to more clinical trials,
accelerate endothelialization and graft integration under in vivo they cannot be conveniently commercialized. Although self-
flow. A similar method was employed to recruit a confluent assembly grafts can utilize devitalization to achieve long-term
and functional endothelium on an acellular TEVG fabricated storage similar to scaffold-based designs, doing so still requires
from small intestinal submucosa (SIS), within one month in a preemptive harvest to collect autologous cells.
a sheep model.[240] The graft possessed functional host SMCs To solve the problems associated with “off-the-shelf” grafts,
at one month, and both endothelium and SMCs were aligned Dahl et al. from Humacyte Inc. constructed a completely
with flow direction and vessel circumference, respectively, after biological tubular small-diameter vascular graft (3–6 mm in
three months. In another study, the sequential surface modi- diameter) using cadaver SMC secreted ECM.[246] These decel-
fication with thrombomodulin followed by heparin imparted lularized grafts can be stored for at least 12 months and provide
a strong local anticoagulation effect via generation of activated “off-the-shelf” grafts without using antithrombotic drugs for
protein C (APC). This effect was demonstrated in an arte- clinical use.[247] The clinical trials for the 6 mm acellular grafts
riovenous (AV) shunt constructed from decellularized porcine have been registered both in Poland and the United States.
SIS, using a baboon model.[241] Covalent attachment of heparin The grafts were implanted as arteriovenous (AV) conduits in
to a decellularized rat pancreas scaffold significantly improved 40 patients with end-stage renal disease, in Poland starting from
the proliferation and growth of seeded HUVECs.[242] These suc- December 2012 (ClinicalTrials.gov, NCT01744418), followed by
cesses demonstrate that heparinized TEVG surfaces can effec- a U.S. clinical trial in 20 patients with end-stage renal disease,
tively inhibit thrombogenesis and stimulate endothelialization. beginning in May 2013 (ClinicalTrials.gov, NCT01840956). The
study reported adequate blood flow through the vessels, no
dilation and no postcannulation bleeding after 16 months.[248]
4. Conclusion Remarks and Translational The clinical trials are still ongoing. Clinical trials of completely
biological “off-the-shelf” small-diameter vascular grafts have
Challenges
achieved promising initial results, but their long-term efficacy
Although considerable effort has been invested to develop still needs to be assessed.
small-diameter TEVGs, presently there is no commercial- An ideal small-diameter vascular graft should be “off-the-
ized vascular graft for small diameter vessels, including coro- shelf,” antithrombotic, immune-compatible, compliant, and
nary arteries. The failures of small diameter vascular grafts eventually completely integrated into native vessels. Biodegrad-
are generally associated with thrombosis, intimal hyperplasia, able materials hold the greatest promise for engineering “off-
or inflammation, all linked to the blood-contacting TEVG sur- the-shelf” vascular grafts. However, their mechanical proper-
face. The blood interface has thus become an important focus ties, composition, degradation behavior, and cell recruitment
for translational applications. Some clinical results are encour- ability need to be precisely controlled to realize a physiologically
aging and present attractive directions for future development. relevant vessel structure with a stable endothelium to main-
The early clinical trials for small vascular grafts produced by tain long-term graft patency. Most recently, a 3D printing tech-
cell self-assembly were undertaken for hemodialysis access. The nology has been developed to print vascular grafts.[249] Although
L’Heureux’s group clinically tested the durability of a 4.8 mm the technology itself is encouraging for biomanufacturing and
diameter TEVG with autologous fibroblasts and ECs obtained scaling-up biological tissues, developmental challenges for vas-
from small skin and vein biopsies. The grafts were grown in cular grafts are still related to materials, cells, and the blood-
culture supplemented with bovine serum and implanted as contracting surface. Improved strategies are in high demand
arteriovenous shunts in ten patients receiving hemodialysis for engineering functional small-diameter TEVGs.

Adv. Healthcare Mater. 2018, 1701461 1701461  (18 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Acknowledgements P. J. Bendick, G. B. Zelenock, C. J. Shanley, J. Vasc. Surg. 2004, 40,


803; b) S. Chen, J. An, L. Weng, Y. Li, H. Xu, Y. Wang, D. Ding,
This study was supported by the National Institutes of Health D. Kong, S. Wang, Mater. Sci. Eng., C 2014, 45, 491; c) S. H. Lim,
(1R15CA202656 and 1R15HL115521-01A1) and the National Science S.-W. Cho, J.-C. Park, O. Jeon, J. M. Lim, S.-S. Kim, B.-S. Kim,
Foundation (1703570). J. Biomed. Mater. Res., Part B 2008, 85B, 537; d) P. Ponnuswamy,
A. Schröttle, E. Ostermeier, S. Grüner, P. L. Huang, G. Ertl,
U. Hoffmann, B. Nieswandt, P. J. Kuhlencordt, PLoS One 2012, 7,
Conflict of Interest e30193.
[21] L. R. Stow, M. E. Jacobs, C. S. Wingo, B. D. Cain, FASEB J. 2011,
The authors declare no conflict of interest. 25, 16.
[22] a) A. Koo, C. F. Dewey, G. García-Cardeña, Am. J. Physiol. 2013,
304, C137; b) P. F. Davies, Nat. Clin. Pract. Cardiovasc. Med. 2009,
6, 16.
Keywords [23] a) S. Dimmeler, I. Fleming, B. Fisslthaler, C. Hermann, R. Busse,
blood-contacting surfaces, endothelialization, surface modification, A. M. Zeiher, Nature 1999, 399, 601; b) D. Nam, C.-W. Ni,
vascular engineering, vascular grafts A. Rezvan, J. Suo, K. Budzyn, A. Llanos, D. Harrison, D. Giddens,
H. Jo, Ame. J. Physiol. 2009, 297, H1535.
Received: December 15, 2017 [24] D. B. Cines, E. S. Pollak, C. A. Buck, J. Loscalzo, G. A. Zimmerman,
Revised: February 9, 2018 R. P. McEver, J. S. Pober, T. M. Wick, B. A. Konkle, B. S. Schwartz,
Published online: E. S. Barnathan, K. R. McCrae, B. A. Hug, A.-M. Schmidt,
D. M. Stern, Blood 1998, 91, 3527.
[25] S. L. Cooper, N. A. Peppas, A. S. Hoffman, B. D. Ratner, Biomate-
rials: Interfacial Phenomena and Applications, American Chemical
[1] J. Xu, S. L. Murphy, K. D. Kochanek, B. A. Bastian, Natl. Vital Stat. Society 1982.
Rep. 2016, 64, 39. [26] Y. Naito, T. Shinoka, D. Duncan, N. Hibino, D. Solomon,
[2] S. C. Smith Jr., A. Collins, R. Ferrari, D. R. Holmes Jr., S. Logstrup, M. Cleary, A. Rathore, C. Fein, S. Church, C. Breuer, Adv. Drug
D. V. McGhie, J. Ralston, R. L. Sacco, H. Stam, K. Taubert, Delivery Rev. 2011, 63, 312.
D. A. Wood, W. A. Zoghbi, Global Heart 2012, 7, 297. [27] K. A. Woodhouse, P. Klement, V. Chen, M. B. Gorbet, F. W. Keeley,
[3] S. Pashneh-Tala, S. MacNeil, F. Claeyssens, Reviews 2016, 22, 68. R. Stahl, J. D. Fromstein, C. M. Bellingham, Biomaterials 2004, 25,
[4] S. Lin, M. Sandig, K. Mequanint, Tissue Eng., Part A 2011, 17, 1561. 4543.
[5] G. M. Rubanyi, J. Cardiovasc. Pharmacol. 1993, 22, S1. [28] A. Patel, B. Fine, M. Sandig, K. Mequanint, Cardiovasc. Res. 2006,
[6] A. P. McGuigan, M. V. Sefton, Biomaterials 2007, 28, 2547. 71, 40.
[7] S. Sarkar, K. M. Sales, G. Hamilton, A. M. Seifalian, J. Biomed. [29] a) A. Waterhouse, S. G. Wise, M. K. Ng, A. S. Weiss, Tissue Eng.,
Mater. Res., Part B 2007, 82B, 100. Part B 2011, 17, 93; b) A. Scandolera, L. Odoul, S. Salesse,
[8] K. C. Dee, D. A. Puleo, R. Bizios, An Introduction to Tissue-Biomaterial A. Guillot, S. Blaise, C. Kawecki, P. Maurice, H. El Btaouri,
Interfaces, Wiley, Hoboken, NJ 2002. B. Romier-Crouzet, L. Martiny, Front. Pharmacol. 2016, 7, 32;
[9] M. A. Gimbrone, G. García-Cardeña, Circ. Res. 2016, 118, 620. c) C. Kawecki, N. Hézard, O. Bocquet, G. Poitevin, F. Rabenoelina,
[10] E. W. Davie, K. Fujikawa, W. Kisiel, Biochemistry 1991, 30, 10363. A. Kauskot, L. Duca, S. Blaise, B. Romier, L. Martiny, Arterioscler.,
[11] S. Beumer, M. IJsseldijk, P. G. de Groot, J. J. Sixma, Blood 1994, Thromb., Vasc. Biol. 2014, 34, 2570.
84, 3724. [30] M. Barnes, D. Maclntyre, Pathophysiol. Haemostasis Thromb. 1979,
[12] W. Li, D. J. D. Johnson, C. T. Esmon, J. A. Huntington, Nat. Struct. 8, 158.
Mol. Biol. 2004, 11, 857. [31] D. T. Simionescu, Q. Lu, Y. Song, J. Lee, T. N. Rosenbalm,
[13] a) Z. Wang, B. Sun, M. Zhang, L. Ou, Y. Che, J. Zhang, D. Kong, C. Kelley, N. R. Vyavahare, Biomaterials 2006, 27, 702.
J. Bioact. Compat. Polym. 2012, 28, 154; b) A. J. Melchiorri, [32] M. T. Hinds, R. C. Rowe, Z. Ren, J. Teach, P. C. Wu, S. J. Kirkpatrick,
N. Hibino, T. Yi, Y. U. Lee, T. Sugiura, S. Tara, T. Shinoka, C. Breuer, K. D. Breneman, K. W. Gregory, D. W. Courtman, J. Biomed. Mater.
J. P. Fisher, Biomacromolecules 2015, 16, 437; c) M. A. Nugent, Res., Part A 2006, 77, 458.
H. M. Nugent, R. V. Iozzo, K. Sanchack, E. R. Edelman, Proc. Natl. [33] M. Lovett, G. Eng, J. Kluge, C. Cannizzaro, G. Vunjak-Novakovic,
Acad. Sci. USA 2000, 97, 6722; d) P. H. Lin, C. Chen, R. L. Bush, D. L. Kaplan, Organogenesis 2010, 6, 217.
Q. Yao, A. B. Lumsden, S. R. Hanson, J. Vasc. Surg. 2004, 39, 1322; [34] S. Enomoto, M. Sumi, K. Kajimoto, Y. Nakazawa, R. Takahashi,
e) M. S. Lord, W. Yu, B. Cheng, A. Simmons, L. Poole-Warren, C. Takabayashi, T. Asakura, M. Sata, J. Vasc. Surg. 2010, 51, 155.
J. M. Whitelock, Biomaterials 2009, 30, 4898. [35] L. Soffer, X. Wang, X. Zhang, J. Kluge, L. Dorfmann, D. L. Kaplan,
[14] M. B. Gorbet, M. V. Sefton, Biomaterials 2004, 25, 5681. G. Leisk, J. Biomater. Sci., Polym. Ed. 2008, 19, 653.
[15] M. Bevilacqua, M. Gimbrone Jr., presented at Leukocyte Adhesion [36] H. Liu, X. Li, G. Zhou, H. Fan, Y. Fan, Biomaterials 2011, 32, 3784.
Molecules: Proc. First Int. Conf. Structure, Function and Regulation [37] J. Heino, Matrix Biol. 2000, 19, 319.
of Molecules Involved in Leukocyte Adhesion, Titisee, West Germany, [38] a) L. Badimon, J. J. Badimon, V. T. Turitto, S. Vallabhajosula,
September–October 1988. V. Fuster, Circulation 1988, 78, 1431; b) D. Wu, K. Vanhoorelbeke,
[16] A. Csiszar, M. Wang, E. G. Lakatta, Z. Ungvari, J. Appl. Physiol. N. Cauwenberghs, M. Meiring, H. Depraetere, H. F. Kotze,
2008, 105, 1333. H. Deckmyn, Blood 2002, 99, 3623.
[17] a) J. Frölich, J. Hypertens. 1990, 8, S73; b) S. Moncada, [39] a) M. G. Davies, P. O. Hagen, Br. J. Surg. 1994, 81, 1254;
Br. J. Pharmacol. 1982, 76, 3; c) Y. Tanaka, F. Yamaki, K. Koike, b) V. Henn, J. R. Slupsky, M. Grafe, I. Anagnostopoulos, R. Forster,
L. Toro, Curr. Med. Chem.: Cardiovasc. Hematol. Agents 2004, 2, 257. G. Muller-Berghaus, R. A. Kroczek, Nature 1998, 391, 591;
[18] J. Loscalzo, Circ. Res. 2013, 113, 100. c) T. G. Diacovo, S. J. Roth, J. M. Buccola, D. F. Bainton,
[19] S.-P. Olesen, D. Claphamt, P. Davies, Nature 1988, 331, 168. T. A. Springer, Blood 1996, 88, 146; d) D. I. Simon, Z. Chen, H. Xu,
[20] a) P. S. Fleser, V. K. Nuthakki, L. E. Malinzak, R. E. Callahan, C. Q. Li, J. Dong, L. V. McIntire, C. M. Ballantyne, L. Zhang,
M. L. Seymour, M. M. Reynolds, S. I. Merz, M. E. Meyerhoff, M. I. Furman, M. C. Berndt, J. A. Lopez, J. Exp. Med. 2000, 192, 193.

Adv. Healthcare Mater. 2018, 1701461 1701461  (19 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[40] B. Furie, B. C. Furie, Cell 1988, 53, 505. [69] M. Hulander, A. Lundgren, L. Faxalv, T. L. Lindahl, A. Palmquist,
[41] T. Huynh, G. Abraham, J. Murray, K. Brockbank, P.-O. Hagen, M. Berglin, H. Elwing, Colloids Surf., B 2013, 110, 261.
S. Sullivan, Nat. Biotechnol. 1999, 17, 1083. [70] a) Y. Ikada, Polymers in Medicine, Springer, Berlin 1984, p. 103;
[42] P. J. Schaner, N. D. Martin, T. N. Tulenko, I. M. Shapiro, b) J. F. Hecker, R. O. Edwards, J. Biomed. Mater. Res. 1981, 15, 1.
N. A. Tarola, R. F. Leichter, R. A. Carabasi, P. J. DiMuzio, J. Vasc. [71] a) T. Sun, H. Tan, D. Han, Q. Fu, L. Jiang, Small 2005, 1, 959;
Surg. 2004, 40, 146. b) L. Chen, D. Han, L. Jiang, Colloids Surf., B 2011, 85, 2.
[43] B. Conklin, E. Richter, K. Kreutziger, D.-S. Zhong, C. Chen, [72] V. Milleret, T. Hefti, H. Hall, V. Vogel, D. Eberli, Acta Biomater.
Med. Eng. Phys. 2002, 24, 173. 2012, 8, 4349.
[44] a) T. C. Rothuizen, F. F. Damanik, T. Lavrijsen, M. J. Visser, [73] J. Lu, M. P. Rao, N. C. MacDonald, D. Khang, T. J. Webster,
J. F. Hamming, R. A. Lalai, J. M. Duijs, A. J. van Zonneveld, Acta Biomater. 2008, 4, 192.
I. E. Hoefer, C. A. van Blitterswijk, Biomaterials 2016, 75, 82; [74] S. J. Liliensiek, J. A. Wood, J. Yong, R. Auerbach, P. F. Nealey,
b) T. C. Rothuizen, F. F. Damanik, J. M. Anderson, T. Lavrijsen, C. J. Murphy, Biomaterials 2010, 31, 5418.
M. A. Cox, T. J. Rabelink, L. Moroni, J. I. Rotmans, Tissue Eng., Part [75] M. F. Kee, D. R. Myers, Y. Sakurai, W. A. Lam, Y. Qiu, PLoS One
C 2014, 21, 436. 2015, 10, e0126624.
[45] L. Gui, A. Muto, S. A. Chan, C. K. Breuer, L. E. Niklason, Tissue [76] Y. Qiu, A. C. Brown, D. R. Myers, Y. Sakurai, R. G. Mannino,
Eng., Part A 2009, 15, 2665. R. Tran, B. Ahn, E. T. Hardy, M. F. Kee, S. Kumar, G. Bao,
[46] Y. Wang, R. C. Gallant, H. Ni, Curr. Opin. Hematol. 2016, 23, 280. T. H. Barker, W. A. Lam, Proc. Natl. Acad. Sci. USA 2014, 111, 14430.
[47] Q. Xing, Z. Qian, M. Tahtinen, A. H. Yap, K. Yates, F. Zhao, [77] Y. Qiu, J. Ciciliano, D. R. Myers, R. Tran, W. A. Lam, Blood Rev.
Adv. Healthcare Mater. 2017, 6, 1601333. 2015, 29, 377.
[48] C. Quint, M. Arief, A. Muto, A. Dardik, L. E. Niklason, J. Vasc. Surg. [78] P. K. Park, B. E. Jarrell, S. K. Williams, T. L. Carter, D. G. Rose,
2012, 55, 790. A. Martinez-Hernandez, R. A. Carabasi, J. Vasc. Surg. 1990, 11, 468.
[49] a) Q. Xing, K. Yates, M. Tahtinen, E. Shearier, Z. Qian, F. Zhao, [79] a) M. Herring, A. Gardner, J. Glover, J. Vasc. Surg. 1984, 1, 279;
Tissue Eng., Part C 2015, 21, 77; b) Q. Xing, C. Vogt, K. W. Leong, b) P. Örtenwall, H. Wadenvik, J. Kutti, B. Risberg, J. Vasc. Surg.
F. Zhao, Adv. Funct. Mater. 2014, 24, 3027. 1990, 11, 403.
[50] Z. H. Syedain, L. A. Meier, M. T. Lahti, S. L. Johnson, [80] C. B. Weinberg, E. Bell, Science 1986, 231, 397.
R. T. Tranquillo, Tissue Eng., Part A 2014, 20, 1726. [81] P. Örtenwall, H. Wadenvik, B. Risberg, J. Vasc. Surg. 1989, 10, 374.
[51] Z. Syedain, J. Reimer, M. Lahti, J. Berry, S. Johnson, R. Bianco, [82] S. H. Ku, C. B. Park, Biomaterials 2010, 31, 9431.
R. T. Tranquillo, Nat. Commun. 2016, 7, 12951. [83] a) W. Zhai, L. J. Qiu, X. M. Mo, S. Wang, Y. F. Xu, B. Peng, M. Liu,
[52] Z. H. Syedain, M. L. Graham, T. B. Dunn, T. O’Brien, S. L. Johnson, J. H. Huang, G. C. Wang, J. H. Zheng, J. Biomed. Mater. Res., Part
R. J. Schumacher, R. T. Tranquillo, Sci. Transl. Med. 2017, 9, B 2013; b) S. Wang, X. M. Mo, B. J. Jiang, C. J. Gao, H. S. Wang,
pii: eaan4209. Y. G. Zhuang, L. J. Qiu, Int. J. Nanomed. 2013, 8, 2131.
[53] B. H. Walpoth, G. L. Bowlin, Expert Rev. Med. Devices 2005, 2, 647. [84] M. Zhou, W. Qiao, Z. Liu, T. Shang, T. Qiao, C. Mao, C. Liu, Tissue
[54] H. V. Damme, M. Deprez, E. Creemers, R. Limet, Acta Chir. Belg. Eng., Part A 2013, 20, 79.
2005, 105, 249. [85] J. Carnagey, D. Hern-Anderson, J. Ranieri, C. E. Schmidt, J. Biomed.
[55] a) A. W. Clowes, A. Gown, S. Hanson, M. Reidy, Am. J. Pathol. Mater. Res., Part B 2003, 65, 171.
1985, 118, 43; b) S. R. Hanson, H. F. Kotze, B. Savage, [86] P. Lamm, G. Juchem, S. Milz, M. Schuffenhauer, B. Reichart,
L. A. Harker, Arterioscler., Thromb., Vasc. Biol. 1985, 5, 595. Circulation 2001, 104, I.
[56] a) M. Gabriel, H. Weiler, U. Mehlhorn, C.-F. Vahl, Thorac. [87] N. Dahan, U. Sarig, T. Bronshtein, L. Baruch, T. Karram,
Cardiovasc. Surg. 2014, 62, OP53; b) M. Gabriel, K. Niederer, A. Hoffman, M. Machluf, Tissue Eng., Part A 2017, 23, 69.
M. Becker, C. M. Raynaud, C.-F. Vahl, H. Frey, Bioconjugate Chem. [88] N. L’Heureux, T. N. McAllister, L. M. de la Fuente, N. Engl. J. Med.
2016, 27, 1216. 2007, 357, 1451.
[57] I. Armentano, M. Dottori, E. Fortunati, S. Mattioli, J. M. Kenny, [89] S. Kaushal, G. E. Amiel, K. J. Guleserian, O. M. Shapira, T. Perry,
Polym. Degrad. Stab. 2010, 95, 2126. F. W. Sutherland, E. Rabkin, A. M. Moran, F. J. Schoen, A. Atala,
[58] C. Lauritzen, Scand. J. Plast. Reconstr. Surg. 1983, 17, 133. Nat. Med. 2001, 7, 1035.
[59] a) C. G. Pitt, T. Marks, A. Schindler, Biodegradable Drug Delivery [90] M. Zhou, Z. Liu, C. Liu, X. Jiang, Z. Wei, W. Qiao, F. Ran, W. Wang,
Systems Based on Aliphatic Polyesters: Application to Contra- T. Qiao, C. Liu, J. Biomed. Mater. Res., Part B 2012, 100, 111.
ceptives and Narcotic Antagonists, Academic, New York 1980; [91] L. P. Neff, B. W. Tillman, S. K. Yazdani, M. A. Machingal, J. J. Yoo,
b) K. Van de Velde, P. Kiekens, Polym. Test. 2002, 21, 433. S. Soker, B. W. Bernish, R. L. Geary, G. J. Christ, J. Vasc. Surg. 2011,
[60] E. Pektok, B. Nottelet, J.-C. Tille, R. Gurny, A. Kalangos, 53, 426.
M. Moeller, B. H. Walpoth, Circulation 2008, 118, 2563. [92] J.-J. Rong, H.-F. Sang, A.-M. Qian, Q.-Y. Meng, T.-J. Zhao, X.-Q. Li,
[61] G. Matsumura, N. Nitta, S. Matsuda, Y. Sakamoto, N. Isayama, Int. J. Clin. Exp. Pathol. 2015, 8, 1282.
K. Yamazaki, Y. Ikada, PLoS One 2012, 7, e35760. [93] H. He, T. Shirota, H. Yasui, T. Matsuda, J. Thorac. Cardiovasc. Surg.
[62] E. D. Boland, B. D. Coleman, C. P. Barnes, D. G. Simpson, 2003, 126, 455.
G. E. Wnek, G. L. Bowlin, Acta Biomater. 2005, 1, 115. [94] Y.-Q. Gu, C.-M. Li, C.-R. Wang, Z.-G. Feng, R.-X. Qiu, B. Chen,
[63] N. Goonoo, R. Jeetah, A. Bhaw-Luximon, D. Jhurry, Eur. J. Pharm. J.-X. Li, S.-W. Zhang, Z.-G. Wang, J. Zhang, Acta Pharmacol. Sin.
Biopharm. 2015, 97, 371. 2009, 30, 530.
[64] S. Sell, M. J. McClure, C. P. Barnes, D. C. Knapp, B. H. Walpoth, [95] C. K. Hashi, Y. Zhu, G.-Y. Yang, W. L. Young, B. S. Hsiao, K. Wang,
D. G. Simpson, G. L. Bowlin, Biomed.Mater. 2006, 1, 72. B. Chu, S. Li, Proc. Natl. Acad. Sci. USA 2007, 104, 11915.
[65] M. J. Smith, M. J. McClure, S. A. Sell, C. P. Barnes, B. H. Walpoth, [96] R. Zhou, L. Zhu, S. Fu, Y. Qian, D. Wang, C. Wang, Sci. Rep. 2016,
D. G. Simpson, G. L. Bowlin, Acta Biomater. 2008, 4, 58. 6, 35422.
[66] Y. Mao, Q. Sun, X. Wang, Q. Ouyang, L. Han, L. Jiang, D. Han, [97] Y. Zhao, S. Zhang, J. Zhou, J. Wang, M. Zhen, Y. Liu, J. Chen, Z. Qi,
Appl. Phys. Lett. 2009, 95, 013704. Biomaterials 2010, 31, 296.
[67] D. Klee, H. Hocker, Biomed. Appl.: Polym. Blends 1999, 149, 1. [98] A. Mirza, J.-M. Hyvelin, G. Y. Rochefort, P. Lermusiaux, D. Antier,
[68] H. Fan, P. Chen, R. Qi, J. Zhai, J. Wang, L. Chen, L. Chen, Q. Sun, B. Awede, P. Bonnet, J. Domenech, V. Eder, J. Vasc. Surg. 2008, 47,
Y. Song, D. Han, L. Jiang, Small 2009, 5, 2144. 1313.

Adv. Healthcare Mater. 2018, 1701461 1701461  (20 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[99] L. Zhang, J. Zhou, Q. Lu, Y. Wei, S. Hu, Biotechnol. Bioeng. 2008, [126] S. Shintani, T. Murohara, H. Ikeda, T. Ueno, T. Honma, A. Katoh,
99, 1007. K.-I. Sasaki, T. Shimada, Y. Oike, T. Imaizumi, Circulation 2001,
[100] G. Matsumura, S. Miyagawa-Tomita, T. Shin’oka, Y. Ikada, 103, 2776.
H. Kurosawa, Circulation 2003, 108, 1729. [127] J. T. Krawiec, D. A. Vorp, Biomaterials 2012, 33, 3388.
[101] M. P. Brennan, A. Dardik, N. Hibino, J. D. Roh, G. N. Nelson, [128] T. Fukunishi, C. A. Best, C. S. Ong, T. Groehl, J. Reinhardt, T. Yi,
X. Papademitris, T. Shinoka, C. K. Breuer, Ann. Surg. 2008, 248, H. Miyachi, H. Zhang, T. Shinoka, C. K. Breuer, J. Johnson,
370. N. Hibino, Tissue Eng., Part A 2018, 24, 135.
[102] T. Fukunishi, C. A. Best, C. S. Ong, T. Groehl, J. Reinhardt, T. Yi, [129] T. Shin’oka, G. Matsumura, N. Hibino, Y. Naito, M. Watanabe,
H. Miyachi, H. Zhang, T. Shinoka, C. K. Breuer, Tissue Eng., Part A T. Konuma, T. Sakamoto, M. Nagatsu, H. Kurosawa, J. Thorac.
2018, 24, 135. Cardiovasc. Surg. 2005, 129, 1330.
[103] S.-W. Cho, S. H. Lim, I.-K. Kim, Y. S. Hong, S.-S. Kim, K. J. Yoo, [130] M. H. Sayegh, B. Watschinger, C. B. Carpenter, Transplantation
H.-Y. Park, Y. Jang, B. C. Chang, C. Y. Choi, Ann. Surg. 2005, 241, 1994, 57, 1295.
506. [131] C. G. Orosz, R. G. Ohye, R. P. Pelletier, A. M. Van Buskirk,
[104] M. Olausson, P. B. Patil, V. K. Kuna, P. Chougule, N. Hernandez, E. Huang, C. Morgan, P. W. Kincade, R. M. Ferguson, Transplantation
K. Methe, C. Kullberg-Lindh, H. Borg, H. Ejnell, S. Sumitran- 1993, 56, 453.
Holgersson, Lancet 2012, 380, 230. [132] M. Isobe, J. Suzuki, H. Yagita, K. Okumura, S. Yamazaki,
[105] N. Hibino, E. McGillicuddy, G. Matsumura, Y. Ichihara, Y. Naito, R. Nagai, Y. Yazaki, M. Sekiguchi, J. Immunol. 1994, 153, 5810.
C. Breuer, T. Shinoka, J. Thorac. Cardiovasc. Surg. 2010, 139, [133] J. P. Dehoux, B. de la Parra, D. Latinne, H. Bazin, P. Gianello,
431. Xenotransplantation 2001, 8, 193.
[106] K.-W. Lee, D. B. Stolz, Y. Wang, Proc. Natl. Acad. Sci. USA 2011, [134] C. G. Orosz, E. Wakely, D. D. Sedmak, S. D. Bergese,
108, 2705. A. M. VanBuskirk, Transplantation 1997, 63, 1109.
[107] T. Korff, S. Kimmina, G. Martiny-baron, H. G. Augustin, FASEB J. [135] H. Valantine, J. Heart Lung Transplant. 2004, 23, S187.
2001, 15, 447. [136] M. C. Yoder, L. E. Mead, D. Prater, T. R. Krier, K. N. Mroueh, F. Li,
[108] B. Lilly, Physiology 2014, 29, 234. R. Krasich, C. J. Temm, J. T. Prchal, D. A. Ingram, Blood 2007, 109,
[109] S. Baiguera, D. Ribatti, Angiogenesis 2013, 16, 1. 1801.
[110] D. P. Basile, M. C. Yoder, J. Cell. Physiol. 2014, 229, 10. [137] J. Ladhoff, B. Fleischer, Y. Hara, H. D. Volk, M. Seifert, Cardiovasc.
[111] a) A. Zampetaki, J. P. Kirton, Q. Xu, Cardiovasc. Res. 2008, 78, Res. 2010, 88, 121.
413; b) R. J. Medina, C. L. Barber, F. Sabatier, F. Dignat-George, [138] M. J. Lee, M. Y. Kim, S. C. Heo, Y. W. Kwon, Y. M. Kim, E. K. Do,
J. M. Melero-Martin, K. Khosrotehrani, O. Ohneda, A. M. Randi, J. H. Park, J. S. Lee, J. Han, J. H. Kim, Arterioscler., Thromb., Vasc.
J. K. Chan, T. Yamaguchi, Stem Cells Transl. Med. 2017, 6, 1316. Biol. 2012, 32, 2733.
[112] Y. Lin, D. J. Weisdorf, A. Solovey, R. P. Hebbel, J. Clin. Invest. 2000, [139] S. Lau, D. Eicke, M. C. Oliveira, B. Wiegmann, C. Schrimpf,
105, 71. A. Haverich, R. Blasczyk, M. Wilhelmi, C. Figueiredo, U. Boer,
[113] N. B. Nardi, L. da Silva Meirelles, Stem Cells, Springer, Berlin 2008, Tissue Eng., Part A 2017.
p. 249. [140] a) G. A. Villalona, B. Udelsman, D. R. Duncan, E. McGillicuddy,
[114] S. Aggarwal, M. F. Pittenger, Blood 2005, 105, 1815. R. F. Sawh-Martinez, N. Hibino, C. Painter, T. Mirensky,
[115] a) J. Oswald, S. Boxberger, B. Jørgensen, S. Feldmann, B. Erickson, T. Shinoka, C. K. Breuer, Tissue Eng., Part B 2010, 16,
G. Ehninger, M. Bornhäuser, C. Werner, Stem Cells 2004, 22, 377; 341; b) K. J. Pawlowski, S. E. Rittgers, S. P. Schmidt, G. L. Bowlin,
b) T. Wang, Z. Xu, W. Jiang, A. Ma, Int. J. Cardiol. 2006, 109, 74. Front. Biosci. 2004, 9, 1412.
[116] A. Bronckaers, P. Hilkens, W. Martens, P. Gervois, J. Ratajczak, [141] J. S. Anderson, T. M. Price, S. R. Hanson, L. A. Harker, Surgery
T. Struys, I. Lambrichts, Pharmacol. Ther. 2014, 143, 181. 1987, 101, 577.
[117] J. Zhao, L. Liu, J. Wei, D. Ma, W. Geng, X. Yan, J. Zhu, H. Du, [142] T. L. Foxall, K. R. Auger, A. D. Callow, P. Libby, J. Surg. Res. 1986,
Y. Liu, L. Li, Artif. Organs 2012, 36, 93. 41, 158.
[118] R. Zhou, L. Zhu, S. Fu, Y. Qian, D. Wang, C. Wang, Sci. Rep. [143] I. S. Park, Y. H. Kim, Y. Jung, S. H. Kim, S. H. Kim, J. Biomater. Sci.,
2016, 6. Polym. Ed. 2012, 23, 1807.
[119] D. Zhang, W. Jiang, BioScience 2015, 65, 468. [144] P. B. van Wachem, J. W. Stronck, R. Koers-Zuideveld, F. Dijk,
[120] a) Y. Wang, J. Hu, J. Jiao, Z. Liu, Z. Zhou, C. Zhao, L.-J. Chang, C. R. Wildevuur, Biomaterials 1990, 11, 602.
Y. E. Chen, P. X. Ma, B. Yang, Biomaterials 2014, 35, 8960; b) J. Luo, [145] X. Gong, H. Liu, X. Ding, M. Liu, X. Li, L. Zheng, X. Jia,
L. Qin, M. H. Kural, J. Schwan, X. Li, O. Bartulos, X.-Q. Cong, G. Zhou, Y. Zou, J. Li, X. Huang, Y. Fan, Biomaterials 2014, 35,
Y. Ren, L. Gui, G. Li, Biomaterials 2017, 147, 116; c) S. Sundaram, 4782.
J. Siewert, S. Teodosescu, L. Zhao, S. Dimitrievska, H. Qian, [146] L. Song, Q. Zhou, P. Duan, P. Guo, D. Li, Y. Xu, S. Li, F. Luo,
A. H. Huang, L. Niklason, Stem Cells Transl. Med. 2014, 3, Z. Zhang, PLoS One 2012, 7, e42569.
1535. [147] J. S. Uzarski, J. Cores, P. S. McFetridge, Tissue Eng., Part C 2015,
[121] D. G. Belair, J. A. Whisler, J. Valdez, J. Velazquez, J. A. Molenda, 21, 1125.
V. Vickerman, R. Lewis, C. Daigh, T. D. Hansen, D. A. Mann, Stem [148] H. Perea, J. Aigner, J. T. Heverhagen, U. Hopfner, E. Wintermantel,
Cell Rev. Rep. 2015, 11, 511. J. Tissue Eng. Regener. Med. 2007, 1, 318.
[122] W. J. Adams, Y. Zhang, J. Cloutier, P. Kuchimanchi, G. Newton, [149] H. Perea, J. Aigner, U. Hopfner, E. Wintermantel, Cells, Tissues,
S. Sehrawat, W. C. Aird, T. N. Mayadas, F. W. Luscinskas, Organs 2006, 183, 156.
G. García-Cardeña, Stem Cell Rep. 2013, 1, 105. [150] A. Tiwari, G. Punshon, A. Kidane, G. Hamilton, A. M. Seifalian,
[123] H. E. Abaci, Z. Guo, A. Coffman, B. Gillette, W. h. Lee, S. K. Sia, Cell Biol. Toxicol. 2003, 19, 265.
A. M. Christiano, Adv. Healthcare Mater. 2016, 5, 1800. [151] G. L. Bowlin, S. E. Rittgers, Cell Transplant. 1997, 6, 631.
[124] K. H. Nakayama, P. A. Joshi, E. S. Lai, P. Gujar, L.-M. Joubert, [152] E. De Falco, D. Porcelli, A. R. Torella, S. Straino, M. G. Iachininoto,
B. Chen, N. F. Huang, Regener. Med. 2015, 10, 745. A. Orlandi, S. Truffa, P. Biglioli, M. Napolitano, M. C. Capogrossi,
[125] B. Wang, A. Borazjani, M. Tahai, A. L. de Jongh Curry, M. Pesce, Blood 2004, 104, 3472.
D. T. Simionescu, J. Guan, F. To, S. H. Elder, J. Liao, J. Biomed. [153] M. H. Fittkau, P. Zilla, D. Bezuidenhout, M. P. Lutolf, P. Human,
Mater. Res., Part A 2010, 94, 1100. J. A. Hubbell, N. Davies, Biomaterials 2005, 26, 167.

Adv. Healthcare Mater. 2018, 1701461 1701461  (21 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[154] D. Hao, W. Xiao, R. Liu, P. Kumar, Y. Li, P. Zhou, F. Guo, D. L. Farmer, D. H. Qian, Z. X. Qin, Y. Lei, X. Y. Hou, C. Wen, J. Mater.
K. S. Lam, F. Wang, A. Wang, ACS Chem. Biol. 2017, 12, 1075. Chem. B 2017, 5, 1053; g) K. O. Cardinal, G. T. Bonnema,
[155] S. H. Hsu, S. H. Sun, D. C. Chen, Artif. Organs 2003, 27, 1068. H. Hofer, J. K. Barton, S. K. Williams, Tissue Eng. 2006, 12, 3431;
[156] T. Y. Kang, J. H. Lee, B. J. Kim, J. A. Kang, J. M. Hong, B. S. Kim, h) J. L. Cezeaux, C. E. Romoser, R. S. Benson, C. K. Buck,
H. J. Cha, J. W. Rhie, D. W. Cho, Biofabrication 2015, 7, 015007. J. E. Sackman, Nucl. Instrum. Methods Phys. Res., Sect. B 1998, 141,
[157] F. Kuwabara, Y. Narita, A. Yamawaki-Ogata, K. Kanie, R. Kato, 193.
M. Satake, H. Kaneko, H. Oshima, A. Usui, Y. Ueda, Ann. Thorac. [180] a) A. W. Tulloch, Y. Chun, D. S. Levi, K. P. Mohanchandra,
Surg. 2012, 93, 156. G. P. Carman, P. F. Lawrence, D. A. Rigberg, J. Surg. Res. 2011,
[158] J. S. Lee, K. Lee, S. H. Moon, H. M. Chung, J. H. Lee, S. H. Um, 171, 317; b) B. Heublein, C. Ozbek, K. Pethig, J. Endovasc. Surg.
D. I. Kim, S. W. Cho, Macromol. Biosci. 2014, 14, 1181. 1998, 5, 32; c) A. Mayer, S. Lee, A. Lendlein, F. Jung, B. Hiebl,
[159] J. Li, M. Ding, Q. Fu, H. Tan, X. Xie, Y. Zhong, J. Mater. Sci.: Mater. Clin. Hemorheol. Microcirc. 2011, 48, 57; d) G. B. Fields, J. L. Lauer,
Med. 2008, 19, 2595. Y. Dori, P. Forns, Y. C. Yu, M. Tirrell, Biopolymers 1998, 47, 143.
[160] J. Yamaguchi, K. F. Kusano, O. Masuo, A. Kawamoto, M. Silver, [181] a) F. M. Zhang, G. C. Li, P. Yang, W. Qin, C. H. Li, N. Huang,
S. Murasawa, M. Bosch-Marce, H. Masuda, D. W. Losordo, Colloids Surf., B 2013, 102, 457; b) G. W. Bos, N. M. Scharenborg,
J. M. Isner, T. Asahara, Circulation 2003, 107, 1322. A. A. Poot, G. H. M. Engbers, J. G. A. Terlingen, T. Beugeling,
[161] J. Yu, A. Wang, Z. Tang, J. Henry, B. Lee, Y. Zhu, F. Yuan, F. Huang, W. G. Van Aken, J. Feijen, Tissue Eng. 1998, 4, 267.
S. Li, Biomaterials 2012, 33, 8062. [182] a) L. V. Antonova, A. M. Seifalian, A. G. Kutikhin,
[162] S. Lu, P. Zhang, X. Sun, F. Gong, S. Yang, L. Shen, Z. Huang, V. V. Sevostyanova, E. O. Krivkina, A. V. Mironov, A. Y. Burago,
C. Wang, ACS Appl. Mater. Interfaces 2013, 5, 7360. E. A. Velikanova, V. G. Matveeva, T. V. Glushkova, E. A. Sergeeva,
[163] E. Miller-Kasprzak, P. P. Jagodzinski, Arch. Immunol. Ther. Exp. G. Y. Vasyukov, Y. A. Kudryavtseva, O. L. Barbarash, L. S. Barbarash,
2007, 55, 247. Front. Pharmacol. 2016, 7, 136; b) W. Zheng, Z. Wang, L. Song,
[164] W. Mrówczyński, A. Rungatscher, F. Buchegger, J.-C. Tille, Q. Zhao, J. Zhang, D. Li, S. Wang, J. Han, X. L. Zheng, Z. Yang,
S. Namy, O. Ratib, M. Kutryk, B. H. Walpoth, Pol. J. Cardio-Thorac. D. Kong, Biomaterials 2012, 33, 2880.
Surg. 2014, 11, 182. [183] R. Kato, C. Kaga, M. Kunimatsu, T. Kobayashi, H. Honda, J. Biosci.
[165] J. I. Rotmans, J. M. Heyligers, H. J. Verhagen, E. Velema, Bioeng. 2006, 101, 485.
M. M. Nagtegaal, D. P. de Kleijn, F. G. de Groot, E. S. Stroes, [184] N. Ferrara, H. P. Gerber, J. LeCouter, Nat. Med. 2003, 9, 669.
G. Pasterkamp, Circulation 2005, 112, 12. [185] a) P. Davoudi, S. Assadpour, M. A. Derakhshan, J. Ai, A. Solouk,
[166] X. Chen, J. Wang, Q. An, D. Li, P. Liu, W. Zhu, X. Mo, Colloids Surf., H. Ghanbari, Mater. Sci. Eng., C 2017, 80, 213; b) L. V. Antonova,
B 2015, 128, 106. A. M. Seifalian, A. G. Kutikhin, V. V. Sevostyanova, V. G. Matveeva,
[167] F. Han, X. Jia, D. Dai, X. Yang, J. Zhao, Y. Zhao, Y. Fan, X. Yuan, E. A. Velikanova, A. V. Mironov, A. R. Shabaev, T. V. Glushkova,
Biomaterials 2013, 34, 7302. E. A. Senokosova, G. Y. Vasyukov, E. O. Krivkina, A. Y. Burago,
[168] M. Lahtinen, P. Blomberg, G. Baliulis, F. Carlsson, H. Khamis, Y. A. Kudryavtseva, O. L. Barbarash, L. S. Barbarash, Int. J. Mol.
V. Zemgulis, Eur. J. Cardio-thorac. Surg. 2007, 31, 383. Sci. 2016, 17, 1920.
[169] Q. Sun, E. A. Silva, A. Wang, J. C. Fritton, D. J. Mooney, [186] a) M. Yin, Y. Yuan, C. Liu, J. Wang, J. Mater. Sci.: Mater. Med.
M. B. Schaffler, P. M. Grossman, S. Rajagopalan, Pharm. Res. 2009, 20, 1513; b) Q. Lin, X. Ding, F. Qiu, X. Song, G. Fu, J. Ji,
2010, 27, 264. Biomaterials 2010, 31, 4017; c) C. L. Song, Q. Li, J. C. Zhang,
[170] M. Zhou, Z. Liu, Z. Wei, C. Liu, T. Qiao, F. Ran, Y. Bai, X. Jiang, J. P. Wang, X. Xue, G. Wang, Y. F. Shi, H. Y. Diao, B. Liu, Eur. Rev.
Y. Ding, Artif. Organs 2009, 33, 230. Med. Pharmacol. Sci. 2016, 20, 311.
[171] J. D. Roh, G. N. Nelson, B. V. Udelsman, M. P. Brennan, [187] S. K. Karnik, B. S. Brooke, A. Bayes-Genis, L. Sorensen,
B. Lockhart, P. M. Fong, R. I. Lopez-Soler, W. M. Saltzman, J. D. Wythe, R. S. Schwartz, M. T. Keating, D. Y. Li, Development
C. K. Breuer, Tissue Eng. 2007, 13, 2743. 2003, 130, 411.
[172] M. Mazzucato, P. Spessotto, A. Masotti, L. De Appollonia, [188] S. Q. Liu, C. Tieche, P. K. Alkema, Biomaterials 2004, 25, 1869.
M. R. Cozzi, A. Yoshioka, R. Perris, A. Colombatti, L. De Marco, [189] C. W. McCarthy, D. C. Ahrens, D. Joda, T. E. Curtis, P. K. Bowen,
J. Biol. Chem. 1999, 274, 3033. R. J. Guillory, S. Q. Liu, F. Zhao, M. C. Frost, J. Goldman, ACS
[173] W. E. Burkel, Med. Prog. Technol. 1988, 14, 165. Appl. Mater. Interfaces 2015, 7, 16202.
[174] S. G. Yates, A. A. Barros D’Sa, K. Berger, L. G. Fernandez, [190] W. F. Daamen, J. Veerkamp, J. Van Hest, T. Van Kuppevelt,
S. J. Wood, E. A. Rittenhouse, C. C. Davis, P. B. Mansfield, Biomaterials 2007, 28, 4378.
L. R. Sauvage, Ann. Surg. 1978, 188, 611. [191] P. H. Blit, W. G. McClung, J. L. Brash, K. A. Woodhouse,
[175] A. Abruzzo, C. Fiorica, V. D. Palumbo, R. Altomare, G. Damiano, J. P. Santerre, Biomaterials 2011, 32, 5790.
M. C. Gioviale, G. Tomasello, M. Licciardi, F. S. Palumbo, [192] S. Q. Liu, P. K. Alkema, C. Tieché, B. J. Tefft, D. Z. Liu, Y. C. Li,
G. Giammona, A. I. Lo Monte, Int. J. Polym. Sci. 2014, 2014, 9. B. E. Sumpio, J. A. Caprini, M. Paniagua, J. Biol. Chem. 2005, 280,
[176] S. Srinivasan, P. N. Sawyer, J. Colloid Interface Sci. 1970, 32, 456. 39294.
[177] a) P. N. Sawyer, S. Sprinivasan, Am. J. Surg. 1967, 114, 42; [193] S. Ayad, R. Boot-Handford, M. Humphries, K. Kadler,
b) H. A. Abramson, J. Exp. Med. 1928, 47, 677. A. Shuttleworth, The Extracellular Matrix Factsbook, Academic,
[178] a) D. M. Scharn, W. F. Daamen, T. H. van Kuppevelt, J. A. van der Vliet, San Diego, CA 1998.
Eur. J. Vasc. Endovasc. Surg. 2012, 43, 66; b) A. A. Owida, H. Do, [194] P. H. Blit, K. G. Battiston, K. A. Woodhouse, J. P. Santerre,
Y. S. Morsi, Comput. Methods Prog. Biol. 2012, 108, 689. J. Biomed. Mater. Res., Part A 2011, 96, 648.
[179] a) T. Pennel, G. Fercana, D. Bezuidenhout, A. Simionescu, [195] R. E. Sallach, W. Cui, J. Wen, A. Martinez, V. P. Conticello,
T. H. Chuang, P. Zilla, D. Simionescu, Biomaterials 2014, 35, 6311; E. L. Chaikof, Biomaterials 2009, 30, 409.
b) D. Liao, X. W. Wang, P. H. Lin, Q. Z. Yao, C. Y. Chen, J. Cell. Mol. [196] a) F. W. Keeley, C. M. Bellingham, K. A. Woodhouse, Philos.
Med. 2009, 13, 2736; c) M. G. Jeschke, V. Hermanutz, S. E. Wolf, Trans. R. Soc., B 2002, 357, 185; b) C. Bellingham, K. Woodhouse,
G. B. Koveker, J. Vasc. Surg. 1999, 29, 168; d) Q. K. Lin, Y. Hou, P. Robson, S. Rothstein, F. Keeley, Biochim. Biophys. Acta, Protein
K. F. Ren, J. Ji, Thin Solid Films 2012, 520, 4971; e) M. S. Baguneid, Struct. Mol. Enzymol. 2001, 1550, 6.
A. M. Seifalian, H. J. Salacinski, D. Murray, G. Hamilton, [197] V. A. Kumar, J. M. Caves, C. A. Haller, E. Dai, L. Liu, S. Grainger,
M. G. Walker, Br. J. Surg. 2006, 93, 282; f) W. W. Dai, H. F. Guo, E. L. Chaikof, Acta Biomater. 2013, 9, 8067.

Adv. Healthcare Mater. 2018, 1701461 1701461  (22 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[198] G. C. Yeo, F. W. Keeley, A. S. Weiss, Adv. Colloid Interface Sci. 2011, 6454; c) V. Ellis, M. F. Scully, V. V. Kakkar, Biochem. J. 1986, 238, 329;
167, 94. d) U. Lindahl, L. Thunberg, G. Bäckström, J. Riesenfeld,
[199] S. G. Wise, G. C. Yeo, M. A. Hiob, J. Rnjak-Kovacina, D. L. Kaplan, K. Nordling, I. Björk, J. Biol. Chem. 1984, 259, 12368; e) J. Choay,
M. K. Ng, A. S. Weiss, Acta Biomater. 2014, 10, 1532. M. Petitou, J. Lormeau, P. Sinay, B. Casu, G. Gatti, Biochem.
[200] Y. Yin, S. G. Wise, N. J. Nosworthy, A. Waterhouse, D. V. Bax, Biophys. Res. Commun. 1983, 116, 492; f) J. Choay, J. C. Lormeau,
H. Youssef, M. J. Byrom, M. M. Bilek, D. R. McKenzie, A. S. Weiss, M. Petitou, P. Sinay, J. Fareed, Ann. N. Y. Acad. Sci. 1981, 370,
Biomaterials 2009, 30, 1675. 644; g) B. Casu, P. Oreste, G. Torri, G. Zoppetti, J. Choay,
[201] S. G. Wise, H. Liu, A. Kondyurin, M. J. Byrom, P. G. Bannon, J. Lormeau, M. Petitou, P. Sinac, Biochem. J. 1981, 197, 599;
G. A. Edwards, A. S. Weiss, S. Bao, M. M. Bilek, ACS Biomater. h) R. D. Rosenberg, L. Lam, Proc. Natl. Acad. Sci. 1979, 76, 1218;
Sci. Eng. 2016, 2, 1286. i) U. Lindahl, G. Bäckström, M. Höök, L. Thunberg, L.-A. Fransson,
[202] M. A. Hiob, S. G. Wise, A. Kondyurin, A. Waterhouse, M. M. Bilek, A. Linker, Proc. Natl. Acad. Sci. USA 1979, 76, 3198; j) M. Höök,
M. K. C. Ng, A. S. Weiss, Biomaterials 2013, 34, 7584. I. Björk, J. Hopwood, U. Lindahl, FEBS Lett. 1976, 66, 90.
[203] a) U. Yokoyama, Y. Tonooka, R. Koretake, T. Akimoto, Y. Gonda, [226] a) P. Brill-Edwards, J. S. Ginsberg, M. Johnston, J. Hirsh,
J. Saito, M. Umemura, T. Fujita, S. Sakuma, F. Arai, Sci. Rep. Ann. Intern. Med. 1993, 119, 104; b) J. Harenberg, B. Casu,
2017, 7, 140; b) Q. Xing, Z. Qian, M. Tahtinen, A. H. Yap, K. Yates, M. Guerrini, R. Malsch, A. Naggi, L. Piazolo, G. Torri, Semin.
F. Zhao, Adv. Healthcare Mater. 2017, 6, 1601333. Thromb. Hemost. 2002, 28, 343; c) L.-O. Andersson, T. Barrowcliffe,
[204] C. Palombo, M. Kozakova, Vasc. Pharmacol. 2016, 77, 1. E. Holmer, E. Johnson, G. Söderström, Thromb. Res. 1979, 15, 531;
[205] Z. Qu, S. Muthukrishnan, M. K. Urlam, C. A. Haller, S. W. Jordan, d) E. A. Johnson, B. Mulloy, Carbohydr. Res. 1976, 51, 119.
V. A. Kumar, U. M. Marzec, Y. Elkasabi, J. Lahann, S. R. Hanson, [227] a) A. Danielsson, E. Raub, U. Lindahl, I. Björk, J. Biol. Chem.
E. L. Chaikof, Adv. Funct. Mater. 2011, 21, 4736. 1986, 261, 15467; b) D. Lane, J. Denton, A. Flynn, L. Thunberg,
[206] C. Salvagnini, S. Gharbi, T. Boxus, J. Marchand-Brynaert, U. Lindahl, Biochem. J. 1984, 218, 725; c) D. Tollefsen,
Eur. J. Med. Chem. 2007, 42, 37. D. W. Majerus, M. K. Blank, J. Biol. Chem. 1982, 257, 2162;
[207] L. K. von Segesser, X. Mueller, B. Marty, J. Horisberger, A. Corno, d) R. E. Jordan, G. Oosta, W. Gardner, R. Rosenberg, J. Biol.
Perfusion 2001, 16, 411. Chem. 1980, 255, 10081; e) E. Holmer, U. Lindahl, G. Bäckström,
[208] B. Dahlbäck, Lancet 2000, 355, 1627. L. Thunberg, H. Sandberg, G. Söderström, L.-O. Andersson,
[209] C. J. Lee, J. E. Ansell, Br. J. Clin. Pharmacol. 2011, 72, 581. Thromb. Res. 1980, 18, 861.
[210] T. Watanabe, K. Kanda, H. Ishibashi-Ueda, H. Yaku, Y. Nakayama, [228] a) P. Sié, M. Petitou, J.-C. Lormeau, D. Dupouy, B. Boneu, J. Choay,
J. Biomed. Mater. Res., Part B 2010, 92, 236. Biochim. Biophys. Acta, Gen. Subj. 1988, 966, 188; b) M. Petitou,
[211] a) Y. Nakayama, H. Ishibashi-Ueda, K. Takamizawa, Cell Transplant. J. Lormeau, B. Perly, P. Berthault, V. Bossennec, P. Sie, J. Choay,
2004, 13, 439; b) R. Kikumoto, Y. Tamao, T. Tezuka, S. Tonomura, J. Biol. Chem. 1988, 263, 8685; c) M. M. Maimone, D. M. Tollefsen,
H. Hara, K. Ninomiya, A. Hijikata, S. Okamoto, Biochemistry 1984, Biochem. Biophys. Res. Commun. 1988, 152, 1056; d) R. Hurst,
23, 85; c) T. Kumada, Y. Abiko, Thromb. Res. 1981, 24, 285. M.-C. Poon, M. Griffith, J. Clin. Invest. 1983, 72, 1042.
[212] a) S. Nishi, Y. Nakayama, H. Ishibashi-Ueda, Y. Okamoto, [229] T. Barrowcliffe, Heparin - A Century of Progress, Springer, Berlin
Y. Kinoshita, J. Artif. Organs 2009, 12, 35; b) T. Richey, H. Iwata, 2012, p. 3.
H. Oowaki, E. Uchida, S. Matsuda, Y. Ikada, Biomaterials 2000, [230] R. A. Hoshi, R. Van Lith, M. C. Jen, J. B. Allen, K. A. Lapidos,
21, 1057; c) T. Imanishi, M. Arita, M. Hamada, Y. Tomobuchi, G. Ameer, Biomaterials 2013, 34, 30.
T. Hano, I. Nishio, Jpn. Circ. J. 1997, 61, 256. [231] a) W. Dorigo, R. Pulli, P. Castelli, V. Dorrucci, F. Ferilli,
[213] Y. Nakayama, S. Yamaoka, M. Yamanami, M. Fujiwara, M. Uechi, G. De Blasis, V. Monaca, E. Vecchiati, C. Pratesi, P. I. R. Group,
K. Takamizawa, H. Ishibashi-Ueda, M. Nakamichi, K. Uchida, J. Vasc. Surg. 2011, 54, 1332; b) R. Pulli, W. Dorigo, P. Castelli,
T. Watanabe, K. Kanda, H. Yaku, J. Biomed. Mater. Res., Part B V. Dorrucci, F. Ferilli, G. De Blasis, V. Monaca, E. Vecchiati,
2011, 99B, 420. C. Pratesi, P. I. R. Group, J. Vasc. Surg. 2010, 51, 1167.
[214] R. F. Zwaal, P. Comfurius, E. M. Bevers, Biochim. Biophys. Acta, [232] a) Y. Li, K. Neoh, E. Kang, J. Biomed. Mater. Res., Part A 2005,
Rev. Biomembr. 1998, 1376, 433. 73, 171; b) A. Gutowska, Y. H. Bae, H. Jacobs, F. Mohammad,
[215] S. Shrivastava, T. Bera, S. K. Singh, G. Singh, P. Ramachandrarao, D. Mix, J. Feijen, S. W. Kim, J. Biomed. Mater. Res., Part A 1995, 29,
D. Dash, ACS Nano 2009, 3, 1357. 811.
[216] V. Ragaseema, S. Unnikrishnan, V. K. Krishnan, L. K. Krishnan, [233] a) J. S. Lindholt, B. Gottschalksen, N. Johannesen, D. Dueholm,
Biomaterials 2012, 33, 3083. H. Ravn, E. D. Christensen, B. Viddal, T. Flørenes, G. Pedersen,
[217] a) E. Fröhlich, Curr. Med. Chem. 2016, 23, 408; b) K. A. Tomaszewski, M. Rasmussen, Eur. J. Vasc. Endovasc. Surg. 2011, 41, 668;
M. W. Radomski, M. J. Santos-Martinez, Nanomedicine 2015, 10, b) M. Vicaretti, W. Hawthorne, P. Ao, J. Fletcher, Cardiovasc.
1451. Surg. 1998, 6, 268; c) J. K. Drury, T. R. Ashton, J. D. Cunningham,
[218] R. V. Madhavan, M. J. Rosemary, M. A. Nandkumar, K. V. Krishnan, R. Maini, J. G. Pollock, Ann. Vasc. Surg. 1987, 1, 542.
L. K. Krishnan, Tissue Eng., Part A 2010, 17, 439. [234] C. Devine, C. McCollum, J. Vasc. Surg. 2004, 40, 924.
[219] Q. Xing, L. Zhang, T. Redman, S. Qi, F. Zhao, J. Biomed. Mater. [235] P. Begovac, R. Thomson, J. Fisher, A. Hughson, A. Gällhagen,
Res., Part A 2015, 103, 3807. Eur. J. Vasc. Endovasc. Surg. 2003, 25, 432.
[220] S. Moncada, A. Higgs, N. Engl. J. Med. 1993, 329, 2002. [236] J. S. Lindholt, B. Gottschalksen, N. Johannesen, D. Dueholm,
[221] L. J. Ignarro, Nitric Oxide: Biology and Pathobiology, Academic, H. Ravn, E. D. Christensen, B. Viddal, T. Florenes, G. Pedersen,
San Diego, CA 2000. M. Rasmussen, M. Carstensen, N. Grondal, H. Fasting,
[222] J. Gao, Y. Wang, S. Chen, D. Tang, L. Jiang, D. Kong, S. Wang, Eur. J. Vasc. Endovasc. Surg. 2011, 41, 668.
RSC Adv. 2017, 7, 18775. [237] a) J. A. Beamish, L. C. Geyer, N. A. Haq-Siddiqi, K. Kottke-Marchant,
[223] J. Hirsh, R. Raschke, T. E. Warkentin, J. E. Dalen, D. Deykin, R. E. Marchant, Biomaterials 2009, 30, 6286; b) Z. Gu,
L. Poller, Chest J. 1995, 108, 258S. B. E. Rolfe, Z. P. Xu, A. C. Thomas, J. H. Campbell, G. Q. Lu,
[224] J. Choay, M. Petitou, Med. J. Aust. 1986, 144, HS7. Biomaterials 2010, 31, 5455.
[225] a) R. Rosenberg, Hemostasis and Thrombosis, Basic Principles and [238] a) M. Shafiq, Y. Jung, S. H. Kim, J. Biomed. Mater. Res., Part A
Clinical Practice, Lippincott Williams & Wilkins, Philadelphia 1994, 2016, 104, 1352; b) W. Gong, D. Lei, S. Li, P. Huang, Q. Qi, Y. Sun,
p. 837; b) D. Atha, J. Lormeau, M. Petitous, Biochemistry 1987, 26, Y. Zhang, Z. Wang, Z. You, X. Ye, Q. Zhao, Biomaterials 2016, 76, 359.

Adv. Healthcare Mater. 2018, 1701461 1701461  (23 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[239] R. J. Smith Jr., M. T. Koobatian, A. Shahini, D. D. Swartz, [245] W. Wystrychowski, T. N. McAllister, K. Zagalski, N. Dusserre,
S. T. Andreadis, Biomaterials 2015, 51, 303. L. Cierpka, N. L’Heureux, J. Vasc. Surg. 2014, 60, 1353.
[240] M. T. Koobatian, S. Row, R. J. Smith Jr., C. Koenigsknecht, [246] S. L. Dahl, A. P. Kypson, J. H. Lawson, J. L. Blum, J. T. Strader,
S. T. Andreadis, D. D. Swartz, Biomaterials 2016, 76, 344. Y. Li, R. J. Manson, W. E. Tente, L. DiBernardo, M. T. Hensley,
[241] J. J. Glynn, M. T. Hinds, Adv. Healthcare Mater. 2016, 5, R. Carter, T. P. Williams, H. L. Prichard, M. S. Dey, K. G. Begelman,
1439. L. E. Niklason, Sci. Transl. Med. 2011, 3, 68ra9.
[242] L. Xu, Y. Guo, Y. Huang, Y. Xiong, Y. Xu, X. Li, J. Lu, L. Wang, [247] S. L. Dahl, J. L. Blum, L. E. Niklason, Trends Cardiovasc. Med. 2011,
Y. Wang, Y. Lu, J. Biomater. Appl. 2018. 21, 83.
[243] a) T. N. McAllister, M. Maruszewski, S. A. Garrido, [248] J. H. Lawson, M. H. Glickman, M. Ilzecki, T. Jakimowicz,
W. Wystrychowski, N. Dusserre, A. Marini, K. Zagalski, A. Fiorillo, A. Jaroszynski, E. K. Peden, A. J. Pilgrim, H. L. Prichard,
H. Avila, X. Manglano, J. Antonelli, A. Kocher, M. Zembala, M. Guziewicz, S. Przywara, J. Szmidt, J. Turek, W. Witkiewicz,
L. Cierpka, L. M. de la Fuente, N. L’Heureux, Lancet 2009, 373, N. Zapotoczny, T. Zubilewicz, L. E. Niklason, Lancet 2016, 387, 2026.
1440; b) N. L’Heureux, T. N. McAllister, L. M. de la Fuente, [249] a) A. J. Melchiorri, N. Hibino, C. A. Best, T. Yi, Y. U. Lee,
N. Engl. J. Med. 2007, 357, 1451. C. A. Kraynak, L. K. Kimerer, A. Krieger, P. Kim, C. K. Breuer,
[244] W. Wystrychowski, L. Cierpka, K. Zagalski, S. Garrido, N. Dusserre, J. P. Fisher, Adv. Healthcare Mater. 2016, 5, 319; b) T. Mirabella,
S. Radochonski, T. N. McAllister, N. L’Heureux, J. Vasc. Access J. W. MacArthur, D. Cheng, C. K. Ozaki, Y. J. Woo, M. T. Yang,
2011, 12, 67. C. S. Chen, Nat. Biomed. Eng. 2017, 1, 0083.

Adv. Healthcare Mater. 2018, 1701461 1701461  (24 of 24) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

Potrebbero piacerti anche