ArticlePDF Available

Endovascular Gene Delivery from a Stent Platform: Gene- Eluting Stents

Authors:

Abstract

A synergistic impact of research in the fields of post-angioplasty restenosis, drug-eluting stents and vascular gene therapy over the past 15 years has shaped the concept of gene-eluting stents. Gene-eluting stents hold promise of overcoming some biological and technical problems inherent to drug-eluting stent technology. As the field of gene-eluting stents matures it becomes evident that all three main design modules of a gene-eluting stent: a therapeutic transgene, a vector and a delivery system are equally important for accomplishing sustained inhibition of neointimal formation in arteries treated with gene delivery stents. This review summarizes prior work on stent-based gene delivery and discusses the main optimization strategies required to move the field of gene-eluting stents to clinical translation.
Research Article Open Access
Fishbein et al., Angiol 2013, 1:2
http://dx.doi.org/10.4172/2329-9495.1000109
Review Article Open Access
Angiology: Open Access
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
Keywords: Endovascular stents; Restenosis; Vascular gene delivery
Abbreviations: Ad: Adenovirus; AAV: Adeno-Associated
Virus; BMS: Bare Metal Stents; DES: Drug-Eluting Stents; ECM:
Extracellular Matrix; E-NTPDase: Ectonucleoside Triphosphate
Diphosphohydrolase; GFP: Green Fluorescent Protein; FGF-2:
Fibroblasts Growth Factor-2; HC: Hydrolyzable Cross-Linker;
IGF: Insulin-Like Growth Factor; IL-1β: Interleukin-1β; IL-6:
Interleukin-6; IL-8: Interleukin-8; ISR: In-stent Restenosis; LMW: Low
Molecular Weight; LST: Late Stent rombosis; MCP-1: Monocyte
Chemoattractant Protein-1; MNP: Magnetic Nanoparticles; NFκB:
Nuclear factor κB; NO: Nitric Oxide; NOS: Nitric Oxide Synthase;
ODN: Oligodeoxynucleotides; PAB: Polyallylamine Bisphosphonate;
PABT: Polyallylamine Bisphosphonate with Latent iol Groups; PC:
Poly-Phosphorylcholine; PCI: Percutaneous Coronary Interventions;
PDGF: Platelet-Derived Growth Factor; PEI (PDT): Polyethyleneimine
Modied with Pyridyldithio Groups; PFU: Particle Forming Units;
PLGA: Poly [lactic-co-glycolic acid]); PVA: Poly Vinyl Alcohol; ROS:
Reactive Oxygen Species; SMC: Smooth Muscle Cells; TIMP-3: Tissue
Inhibitor of Metalloproteinases-3; TNF: Tumor Necrosis Factor;
VEGF: Vascular Endothelial Growth Factor
DES for the Prevention of In-Stent Restenosis
A percutaneous vascular intervention (PCI) is a standard
therapeutic approach for treating patients with symptomatic coronary
disease. PCI are carried out annually in more than one million patients
in USA alone [1]. An overwhelming majority of PCI cases involve stent
angioplasty performed with either bare metal stents (BMS) or drug-
eluting stents (DES). Compared to BMS, DES achieve better outcomes
related to early and mid-term arterial patency and are associated with a
reduced need for target vessel revascularization [2]. However, in some
studies DES have been linked to a higher rate of late stent thrombosis
(LST) [3]. Aborted healing response, lack of re-endothelialization and
unresolved inammation have been suggested as the major factors
contributing to this complication [3]. LST is a grave medical condition
carrying a high risk of sudden cardiac death. To minimize the risk
of thrombosis, patients implanted with DES are routinely prescribed
prolonged aggressive dual anti-platelet therapy that causes bleeding
complications in a signicant fraction of post-PCI patients [4]. e
incidence of ISR and LST varies widely among dierent groups of
patients and lesion characteristics. Most registries based on unselected
patient populations report 20-30% and 8-10% ISR, and 0.8-1% and 1.5-
2% LST in the rst year following stent deployment, for the patients
receiving BMS and DES, respectively [5,6].
Moreover, the anti-restenotic eect of DES is less pronounced in
patients with peripheral artery disease [7], as well as coronary disease
patients aicted by diabetes or renal failure [8]. Furthermore, the
presence of ostial lesions, pre-existing ISR, smaller arterial diameter
and longer lesion length are also associated with poor outcomes [8].
e binary rate of restenosis in patients with these and some other
anatomical and clinical circumstances is currently in double digits
even with the use of the 2nd and 3rd generation DES devices [9]. It is
also noteworthy that ISR developed within DES is especially resistant
to further interventional therapeutic approaches and, thus, is oen an
indication for bypass surgery.
Vascular Gene erapy and the Concept of Gene-
Eluting Stents (GES)
e use of specialized catheters to deliver and lodge gene vectors
in the vessel wall preceded stent-based technologies for vascular gene
therapy by 15 years [10]. ese initial studies provided important
insights in transfectability/transducibility of adventitia, media and
intima (neointima) in various experimental species and established
multiple potential molecular targets for restenosis prevention [11].
However, the extent of vascular tissue transduction even with
sophisticated catheter devices (Dispatch®, Inltrator®) remained
inadequate for clinical translation of vascular gene therapy mostly
due to the short intraluminal retention time of the delivered gene
therapeutics [10].
Immobilization of gene vectors on stents embraces the best of both
DES and catheter-based methods of vascular gene therapy, by providing
the means to overcome the limitations of DES. First, gene therapy can
*Corresponding author: Ilia Fishbein, Dept of Pediatrics, Division of Cardiology,
The Children’s Hospital of Philadelphia, USA, Tel: (215) 590-8740; Fax: (215) 590-
5454; E-mail: FISHBEIN@email.chop.edu
Received June 27, 2013; Accepted July 25, 2013; Published July 27, 2013
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013)
Endovascular Gene Delivery from a Stent Platform: Gene- Eluting Stents. Angiol 1:
109. doi: 10.4172/2329-9495.1000109
Copyright: © 2013 Fishbein I, et al. This is an open-access article distributed under
the terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and
source are credited.
Abstract
A synergistic impact of research in the elds of post-angioplasty restenosis, drug-eluting stents and vascular
gene therapy over the past 15 years has shaped the concept of gene-eluting stents. Gene-eluting stents hold
promise of overcoming some biological and technical problems inherent to drug-eluting stent technology. As the
eld of gene-eluting stents matures it becomes evident that all three main design modules of a gene-eluting stent: a
therapeutic transgene, a vector and a delivery system are equally important for accomplishing sustained inhibition of
neointimal formation in arteries treated with gene delivery stents. This review summarizes prior work on stent-based
gene delivery and discusses the main optimization strategies required to move the eld of gene-eluting stents to
clinical translation.
Endovascular Gene Delivery from a Stent Platform: Gene- Eluting Stents
Ilia Fishbein1,2*, Michael Chorny1,2, Richard F Adamo1, Scott P Forbes1, Ricardo A Corrales1, Ivan S Alferiev1,2 and Robert J Levy1,2
1Dept of Pediatrics, Division of Cardiology, The Children’s Hospital of Philadelphia, USA
2The University of Pennsylvania, USA
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 2 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
attain a longer lasting therapeutic modication of vascular substrate;
second, gene interventions allow for selective inhibition of SMC
proliferation and migration while maintaining and even enhancing
endothelial re-growth; third, the underlying atherosclerotic process
can be addressed; and fourth, a modulation of the biological activity of
gene-eluting stents can be achieved with systemically administered low
molecular weight compounds.
Furthermore, compared with the scaold-less vascular gene
delivery as accomplished with the use of delivery catheters, stents
present an advantageous physical platform for local arterial gene
transfer. Indeed, higher arterial concentrations of gene vectors can be
achieved with the immobilization on stent surface when compared to
non-immobilized vectors following administration of a smaller vector
input dose. Superior vascular wall retention is a combined result of
physical vector association with a permanently implanted scaolding
device, and a shielding eect of the vector deposited on the adluminal
surface of the stent (i.e. at a stent/tissue interface) from the shearing
eect of blood ow. As a consequence of protracted vascular residence,
immobilization on the stent minimizes the distal spread of gene vector,
reducing inadvertent inoculation of non-target tissues. Moreover,
stent-based delivery of gene therapeutics is feasible in diseased
arterial segments with side branches. Human arteries susceptible to
atherosclerosis, with the exception of the common carotid artery, are
extensively branched and therefore scaold-less delivery of uid phase
vector formulations leads to immediate run-o of the vector to the
circulation through the side branches. Furthermore, since stent-based
gene transfer does not require isolation of the treated vascular segment,
procedure-related ischemic complications can be minimized. Finally,
stent-based gene delivery provides an optimal spatial conguration
to prevent cellular proliferation and migration in the setting of ISR,
since cell activation aer stent deployment is typically observed in the
vicinity of stent struts [12]. Physical association of gene vector with a
stent establishes a transgene expression gradient that overlaps with the
cell activation gradient, thus approximating vectors to the projected
site of the action.
us, the use of gene delivery stents for atherosclerotic vascular
disease oers the appealing possibility of site specic delivery of
gene therapy with long lasting and controllable transgene expression
that can prevent in-stent restenosis, and also provide therapy for the
underlying vascular disease.
Minimal requirements for GES function and performance
e design requirements for GES share several characteristics with
DES platforms. First, deliverability of both types of devices should not
be compromised by the accommodation of a therapeutic agent and its
matrix on the stent struts. Second, the matrix associated with DES and
GES struts has to withstand the mechanical stretch associated with a
change of the stent diameter during deployment. Cracking, peeling
and delamination of the matrix during stent implantation should be
prevented since these post-deployment defects lead to irregularity in
the release rate of a therapeutic agent and can cause distal embolization.
ird, the matrix has to be highly biocompatible, causing no more than
a minimal thrombogenic and inammatory response.
ere are several important distinctions, however, in the design
characteristics of DES and GES. While DES have been devised to be able
to provide sustained release and vessel wall delivery of low molecular
weight therapeutic agents for as long as 3 months, this may not be
necessary for the GES, since transfected/transduced cell populations in
the stented artery are able to produce and secrete a transgene product
for prolonged period of time. A minimal required duration of stent-
associated vector perseverance in its active transduction competent
form is not entirely clear. Since the medial and neointimal SMC
proliferation starts no earlier than 1 week aer stent deployment in
human and higher animal models [13], the target cell population is not
yet present at the time of vascular injury. is fact argues for at least
2-3 week-long release period of a transduction competent gene vector
from the stent, posing additional requirements to GES technology
related to extended vector stability in vivo. Unlike LMW drugs, which
are oen stabilized by matrix immobilization, gene vectors (both non-
viral and especially virus-based) are vulnerable and lose their activity if
not properly protected. Elicitation of immune response to a therapeutic
moiety is another specic problem of GES since LMW drugs used
in DES are rarely strong immunogenes unless bound to proteins.
erefore, preventing distal vector spread to regional lymph nodes and
spleen, as well as physical shielding of the vector from direct contact
with pre-formed antibodies and eector T-cells are of paramount
importance for sustaining therapeutic levels of transgene expression
following GES use. Finally, the 1-2 orders of magnitude size dierence
between LMW drugs and gene vectors dictates use of dierent matrices
to enable timely release of a therapeutic agent from a stent platform,
aecting the predominant mechanism of the agent release (diusion vs
matrix degradation).
Conceptual Technologies in GES Design
Research concerning gene delivery stents over the past 15 years
illustrates a number of principles and mechanisms involved in stent-
facilitated delivery of gene therapy vectors to the arterial wall. Several
dierent approaches have been exploited to achieve labile association
of gene vectors with stent struts (Table 1).
Bulk immobilization (polymer coatings)
Initial investigations concerning gene delivery stents utilized
polymer coatings on the surface of metallic stents in which gene vectors
were dispersed or pseudo-dissolved. ese coatings (typically 50-250
µm thick) were deposited on a primed stent surface either by a multiple
dip technique or by aerosol deposition of a polymer solution containing
a suspension of the vector. Aer solvent evaporation and polymer
precipitation on the stent surface, the gene vector stays contained
between the polymer bers. A subsequent release of vector particles is
then controlled by diusion though the polymer matrix (determined by
the relative hydrophilicity/hydrophobicity of the matrix and the pore
size), matrix degradation and/or dissolution of matrix in blood and
tissue uid. Depending on the polymer type, these three mechanisms
have varying contributions to the overall release kinetics of the vector
from the stent coating. A variety of synthetic polymers (PLGA [14],
poly(beta-aminoester) [15-18], polyurethane [19,20], PVA [21], poly
[phosphorylcholine-lauryl methacrylate] [22-28]), semi-synthetic
polymers (dopamine-modied hyaluronic acid [29,30], cationized
gelatin [31], styrene-modied gelatin [32], cationized pullulan [33] or
Immobilization method Advantages Disadvantages
Bulk coating High loading of gene
vectors, ease of scale-up
Inadequate release
kinetics, polymer-induced
inammation
Surface tethering Controllable release rate,
no inammation
Low loading of gene
vectors
In-situ stent loading using
magnetic targeting
No vector loss on route to
delivery site, possibility of
repeated loading
A limited choice of stent
materials, MNP safety
issues
Table 1: Comparison of gene vector immobilization methods on stent surfaces.
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 3 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
naturally occurring macromolecules (collagen [34], denatured collagen
[35], gelatin [17,18]) have been employed for the bulk incorporation of
genetic material on stent wires.
An obvious advantage of the bulk immobilization is the ability to
integrate substantial amounts of the vector in a thick coating. Plasmid
DNA quantities as high as 4 mg [20] or Ad doses up to 5×1010 particles
[34] have been reported. ere are several important disadvantages of
the bulk immobilization as well. Typically, 80-90% of the vector load
is released within the rst 24 hours [17,19,21,23,24,33,36], especially
in the case of non-viral systems. Part of this eect is due to the “burst
release” of a fraction of the vector payload, which is either deposited
in the outermost layer of the polymer coating or has been adsorbed
onto the polymer layer aer going out of solution during the solvent
evaporation step. A partial solution for preventing this undesired
rapid release of the vector is provided by a super-coating of a vector/
polymer composite with additional layers of vector-free polymer
[14,17,18]. Another potential method to modulate the release rate of
gene therapeutics from bulk polymer coatings uses controlled cross-
linking of a polymer as exemplied by Nakayama [32] who developed a
photoactivatable coating system consisting of styrene-modied gelatin,
a polymerization initiator, carboxylated camphoroquinone and a
reporter Ad-LacZ adenovirus vector. Upon irradiation of dipped stents
with a broad spectrum halogen lamp, gelatin chains become cross-
linked forming a hydrogel that entraps Ad.
Another important disadvantage of the bulk polymer coatings is
an inammatory reaction triggered by native and especially synthetic
polymers in the arterial segments placed in contact with the polymers
[37]. us, unless potent agents, such as the anti-cancer drugs commonly
used in drug-eluting stents are utilized, the inammatory response to
the polymer coating negates the therapeutic eect of a transgene. One
important exception to the uniformly pro-inammatory character of
polymer coatings is an almost complete lack of inammatory inltrates
and generally high biocompatibility of poly [phosphorylcholine-
lauryl methacrylate] co-polymer utilized in the BiodivYsio stent [23-
28] approved by FDA for clinical use in 2000. A hydrophilic poly-
phosphorylcholine (PC) moiety of this macromolecule mimics an
outer phospholipid layer of cell membranes, thereby reducing platelet
activation on the metal surface. Platelet passivation makes this stent
a device of choice in patients with absolute contraindications to dual
anti-platelet therapy aer stenting. A positive surface charge on the
BiodivYsio stent imparted by choline residues renders the PC coating
useful for electrostatic binding of negatively charged DNA plasmids,
as well as Ad and AAV vectors. e latest generation of BiodivYsio
stents feature extremely thin coatings (50 nm) precluding signicant
incorporation of the vector in the bulk of the polymer. erefore, this
system constitutes a paradigm that includes both elements of bulk
immobilization and the coatless surface immobilization systems as
described below.
Surface immobilization of gene vectors on coatless metal
substrate
Surface immobilization of gene vectors on the surface of
endovascular stents represents a particular case of substrate mediated
gene delivery, a concept put forward by the work of Shea [38,39]
primarily to increase biocompatibility of bioprosthetic scaolds.
e main premise of substrate-mediated gene transfer is to use the
tissue-facing surface of an implanted device to append (rather than
bulk-incorporate) therapeutic gene vectors in an arranged pattern
to facilitate transduction of tissue elements on the interface with the
bioprosthesis, thereby improving functional integration of the device.
Protein anity adaptors for vector immobilization: Studies by
our group further developed the idea of substrate mediated delivery for
use in conjunction with endovascular stents. Driven by the established
data on detrimental pro-inammatory and pro-restenotic eects of
stent coatings we designed and implemented a chemical strategy for
reversible attachment of both viral and non-viral gene vectors that
does not rely on surface coating. Our method of vector immobilization
on non-coated metal surface is based on surface modication with
poly(allylamine)-bisphoshonate (PAB) compounds bridging the metal
surface and gene vectors [40]. Bare metal gene delivery using PAB
involves rst forming a polymer monolayer on the metal surface of the
stent that is established due to the formation of strong coordination
bonds between pendant bisphosphonic groups of the polymer and
Fe, Ni and Cr atoms and their oxides on the steel surface. is ultra-
thin (less than 5 nm) permanently attached polymer lm is rapidly
formed upon exposure of metal samples to an aqueous PAB solution.
e molecular monolayer of PAB can be further covalently modied
in order to permit a covalent attachment of vector binding agents
using well established amine conjugation chemical strategies targeting
multiple primary amines of the allylamine backbone [40]. High anity
vector binding proteins such as anti-Ad knob [40], anti–DNA [41]
antibodies or a recombinant D1 domain of Coxsackie-Adenovirus
Receptor [40] can be used as binding adaptors enabling high-anity
binding of gene vectors to stents. Stent loading with gene vectors is
then achieved by simple immersion of a stent in a vector suspension.
We further demonstrated that agents with dierent binding anities
can be exploited to adjust a vector capacity of the gene-eluting stent
and to modulate the release rate of gene therapeutics both in vitro and
in vivo [40]. However, limited control over the vector release kinetics
remains a disadvantage of the protein anity binding immobilization
technology. Although we have demonstrated the vector presence at
the interface between stent struts and the arterial wall 24 hours post-
deployment and associated reporter (GFP) activity in all 3 arterial
layers 7 days post-stenting [40], the duration of the vector binding to
a stent in vivo may be far from optimal. It is challenging to prolong
vector association with the stent surface using this technique, since
the vector association with stents is determined by the antigen/
antibody or receptor/ligand anity, local pH, and the protein
content of blood and tissue uid; the factors that cannot be changed
deliberately. Furthermore, the protein anity binding technology
cannot be immediately adapted to any given vector, since it implies
availability of a vector-binding molecule with Kd in the range of 10-8-
10-9 M. Moreover, vector-binding strategies based on protein anity
interactions are dicult to adapt to manufacturing scale because of
protein stability and species specicity issues.
Viral vector tethering to bare metal stents via hydrolyzable
cross-linkers: Motivated by the limitations of anity immobilization
we developed an alternative methodology for the reversible binding
of recombinant replication-defective adenoviruses to metal surfaces
that completely avoids using protein adaptors. is method is based
on hydrolyzable cross-linker (HC) molecules that directly append
vectors to PAB-activated steel (Figure 1). e subsequent release
of the vectors is governed by the kinetics of cross-linker hydrolysis
(Figure 1) and can be modulated by the usage of HC with variable
hydrolysis rates. A new conjugation strategy necessitated a change of
PAB chemical design with the introduction of latent thiol groups in
the side chains of the polymer molecule. is novel compound, PABT,
was successfully synthesized and characterized by our group [42]. e
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 4 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
devised linking strategy by itself was sucient to achieve signicant
binding of thiol-reactive Ad particles to the surface of model steel
samples. However, we choose to expand the amount of available thiol
group on the metal surface using additional exposure of thiol-activated
metal samples to an aqueous solution of pyridyldithio (PDT)-engraed
polyethyleneimine, PEI (PDT) followed by reduction of PDT to thiols
with dithiothreitol. We demonstrated that the “amplication” protocol
resulted in a more eective Ad tethering when compared with the basic
“no-amplication” protocol [42].
Synthetic, biodegradable HC are particularly promising tools for
achieving a site-specic tunable release of gene therapy vectors since
they can be synthesized to have a broad range of hydrolysis rates and
thus the vector release rate from the surface of a stent can programmed
per the formulation parameters [43].
Magnetic stent targeting
Both bulk immobilization and surface tethering approaches make
use of stent-based gene delivery systems that are assembled prior
to stent implantation in the artery. Introduction of a stent into the
vasculature through the hemostatic valve of a vascular sheath and
routing it to a deployment site through the atherosclerotic and oen
calcied arterial conduit always involves some physical damage to the
stent coating and vector depot. As an alternative approach, stent loading
can be accomplished in situ aer the stent has already been implanted.
Since the deployed stent is freely accessible to blood ow, its surface
can be actively targeted with therapeutic agents delivered to systemic
or regional circulation, provided the targeting forces are strong enough
to capture and retain a therapeutic agent on the stent surface.
is concept of in situ stent loading with gene vectors was recently
implemented by our group using a magnetic targeting paradigm
[44]. Stents made of magnetic alloys, when placed in a uniform
magnetic eld, such as that created within an MRI coil or induced
by paired electromagnets generate strong highly localized magnetic
A B
Catheter
Magnet
Magnetic
nanoparticles
Figure 2: A targeted delivery of MNP co-formulated with Ad-Luc vectors to a deployed stent mediated by the uniform eld induced magnetization
effect. The uniform eld generated by paired electromagnets (A) both induces high gradients on the stent and magnetizes Ad-loaded MNP, thus
creating a magnetic force driving MNP to the stent struts and adjacent arterial tissue (B). (Adapted from [45] with permission).
A
5 nm
H2O
O
O
O
O
O
O
O
O
O
O
O
O
O
P
S
S
P
P
P
NH
NH NH
NH
NS
S
S
S
S
S
N
N
NH
n
H
N
NH NH
HL
HL
HN
OH
Z2
Z1
Z1Z2
Z1
O
O
HO
CZ
2
-HN
O
OO
O
O
O
O
O N
O
SO2Na
CH CH2CH CH2CH CH2
PO3H2
C3H7
PO3H2
NH2NH NH
n-k-m k m
O
Fe Ni Cr Fe Ni Cr Fe Ni Fe Ni Cr Fe Ni Cr Fe Ni
O
H
N
SS
S
S
S
S
PEI(PDT)
PEI(PDT)
amplification
layer
PABT
coordination
layer
PABT
=
O
O
O
O
O
O
O
O
O
O
O
O
P
P
P
P
Figure 1: A scheme illustrating specic chemical interactions to enable adenovirus binding to a stent surface. Ad vectors were modied by
reacting lysine residues of capsid proteins with a bifunctional amine/thiol-reactive Hydrolyzable Cross-Linker (HL) possessing a hydrolyzable
ester bond separating fragment Z1 and Z2. Stainless steel stents were consecutively exposed to a solution of Polyallylamine Bisphosphonate
Comprising Latent Thiol Groups (PABT) and a reducing agent, TCEP, to activate thiol groups on the surface. To expand the amount of available
thiol functions, a subsequent treatment with polyethyleneimine modied with pyridyldithio groups, PEI (PDT), and DTT was used. Finally,
HL-modied Ad vectors were reacted with thiolated stent surfaces to achieve covalent tethering of Ad. The subsequent release of covalently
immobilized Ad is driven by hydrolysis of the ester bond in the cross-linker’s backbone. (Adapted from [42] with permission).
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 5 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
forces due to steep eld gradients between stent struts [44-47]. ese
magnetic forces enable the targeted capture of systemically or locally
administered gene vectors formulated in MNP [44-47] (Figure 2).
e same external magnetic source can be used to magnetize both
stents and MNP. In our study [44], aer implanting stents made of
magnetically responsive 304-grade stainless steel in the common
carotid arteries of rats, suspension of MNP incorporating 2.5×109
Ad-Luc was slowly injected through the catheter into the aortic arch.
e delivered vector was then transported en masse with antegrade
blood ow to carotid circulation across the stented segment. During
and immediately aer MNP/Ad-Luc delivery a uniform magnetic eld
of 0.1 Tesla was established across the neck area of the rats. While
no direct quantication of the amount of Ad vector attached to stent
struts following magnetic targeting was pursued, the vector uptake was
signicant enough to cause a localized transduction of stented carotid
segment, greatly exceeding that achievable with free Ad, as determined
by bioluminescence imaging [44].
Magnetic reloading of depleted GES: Prolonged arterial expression
of a transgene with an established anti-restenotic activity following a
single intervention is the ultimate goal of stent-based vascular gene
therapy. Despite ongoing research in the eld for the past decade,
it is still too early to conclude whether this goal is realistic. Indeed,
even with protracted release kinetics of gene vectors from stents,
such as one achieved with hydrolyzable cross-linker immobilization,
the duration of the transgene expression and hence the durability of
the therapeutic eect is a concern. In most mammalian tissues, aer
reaching peak level, transgene expression tends to decrease over time
due to both promoter attenuation and mounting immune response
to the vector and the transgene product. Previous reports [48,49]
have documented that suppressed neointimal growth can recur if the
inhibiting modality is withdrawn early. e vulnerability period for
restenosis recurrence following premature treatment withdrawal in
human arteries is unknown, but appears to be longer for peripheral
compared to coronary vasculature. erefore, the potential to
rejuvenate therapeutic transgene expression by reloading stents with
additional payloads of the same or other gene vectors is of paramount
importance in designing translatable vascular gene therapy approaches
to prevent restenosis, especially in peripheral vasculature. Uniform
eld high-gradient magnetic targeting provides a potential way of
addressing reloading of depleted GES at delayed time points. In order
to reload a stent in situ, a gene vector needs to be physically partitioned
at or near the depot for sucient time to saturate binding sites in the
depot matrix with the re-delivered vectors. Magnetic eld mediated
targeting provides a unique physical opportunity to achieve this task.
Reporter Studies and Pharmacokinetics of Transgene
Expression
Reporter studies are instrumental for optimizing stent-based
gene delivery systems in regards to transgene expression strength
and durability. GFP [14,16,20,34-36,40,42,50,51], β-galactosidase
[20,21,23,26,27,32] and luciferase [20,36,40,42,43] have been
extensively employed to map temporal and spatial expression patterns
of stent-delivered transgenes. It is noteworthy that the information
provided by dierent reporter systems is not redundant. For example,
while luciferase is typically more sensitive than the other two reporter
systems, it does not readily provide a tissue level resolution. Use of
GFP and β-galactosidase, on the other hand, is compatible with direct
histological examination of transduced tissue, providing valuable
information about transduced cell populations and their distribution
in distinct compartments of the vessel wall. Native GFP uorescence is
labile and extremely sensitive to tissue xation methodology. Moreover,
GFP emits light in the same part of spectrum as elastin, making
unambiguous detection of GFP positive cells dicult, especially in the
injured tissue, where elastin is partially disintegrated. A recent advent
of high quality commercial anti-GFP antibodies makes this task easier
by streamlining immunouorescence and immunohistochemical
approaches for the identication of transduced cells.
A direct comparison of the extent and duration of reporter arterial
expression among the published studies is challenging because of
dissimilarities between animal models, reporter systems and reporter
detection methods employed by dierent groups. Nevertheless, several
common traits of arterial tissue transduction from a stent platform
are preserved throughout most of these studies regardless of the
specic experimental setup. First, gene transfer is highly site-specic,
leading to more eective transduction of cells underlying stent struts
compared to cells located between the stent wires [20,21,26,27,40,42].
Second, despite neointimal growth not occurring until 4-14 days aer
a stent deployment, a nascent neointima is more eectively transduced
than media and adventitia [20,26,36,40,42]. ird, duration of gene
expression may exceed one month following implantation of GES
[26,27]. Fourth, no massive distal spread of a transgene to liver, spleen
and lungs is apparent [14,20,34,40,42]. Fih, dierent vector systems
may have predilection for transducing specic cell populations in the
arterial wall [20,26].
Disease-related Targets and erapeutic Transgenes
In-stent restenosis is a multifactorial disease [52,53]. e stenting
related factors with proven pathogenic bearing for ISR include
endothelial denudation, direct trauma to medial SMC resulting in
altered SMC apoptosis, proliferation and ECM synthesis programs,
parietal thrombus formation, unresolved inammation and foreign
body reaction to the stent and its coating. Accordingly, the number
of molecular targets relevant to inhibition or stimulation of signaling
pathways altered during ISR development is extremely high [52,53].
Comprehensive reviews [11,54,55] focused on gene therapy of
restenosis and related vasculoproliferative conditions list more than
150 dierent genes implicated in regulation of cell proliferation,
migration, ECM turnover, thrombosis and inammation that were
targeted with gene therapy interventions to mitigate restenosis. Only a
small fraction of these therapeutic gene constructs was investigated in
conjunction with GES.
GES targeting re-endothelialization
Since the seminal study by Asahara et al. [56] describing inhibition
of restenosis in a rat model with locally administered vascular
endothelial growth factor (VEGF) was published in 1995, strategies
aimed at attenuating restenosis via enhanced re-endothelialization
have come into focus. In line with this concept, Walter et al. [28] have
used poly[phosphorylcholine-lauryl methacrylate]-coated stents to
electrostatically adsorb 200 µg plasmid DNA encoding human VEGF-
2. When deployed in Fogarty balloon-denuded external iliac arteries
of hypercholesterolemic rabbits, VEGF-2 plasmid-eluting stents
enhanced endothelial recovery in the stented arteries in comparison
with the animals receiving control stents and resulted in a 60%
reduction of cross-sectional arterial narrowing of the stented region 3
months aer the procedure [28].
GES targeting SMC proliferation, migration and ECM
remodeling
Serine/threonine kinase, Akt1, also known as protein kinase B
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 6 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
(PKB) is an important signaling hub integrating input from several pro-
proliferative pathways, in particular from PI3K [57]. Akt1 activation
during neointimal tissue maturation [58] as well as the anti-restenotic
eectiveness of Akt1 inhibition [59] are well established. Recently
Che et al. [29] applied coordination chemistry to achieve stable
attachment of dopamine-conjugated hyaluronic acid (HA) to stents via
bonds formed between dopamine residues and metal ions on the stent
surface. e HA-coated stents were further modied with polyplexes
made of siRNA to Akt1 and disulde cross-linked PEI. A sustained
release of siRNA from the stent surface was observed in vitro for at
least 72 hours. In in vivo study, an undisclosed amount of Akt1 siRNA
polyplexes was deposited on HA-coated stents. Akt1 siRNA stents
implanted in rabbit iliac arteries decreased the extent of ISR by 50% at
4 weeks aer deployment compared to BMS [29].
Platelet-derived growth factor (PDGF) existing in two
homodimeric forms (PDGF-AA, PDGF-BB) and as a heterdimer
(PDGF-AB) is a well established SMC mitogen [60] and pro-migratory
chemoattractant [61]. All three isoforms were shown to promote SMC
accumulation in vitro and in vivo, while demonstrating relatively low
activity toward endothelial cells [62]. erefore, LMW inhibitors of
PDGF receptor tyrosine kinase activity [63], as well as gene therapy
modalities of inhibiting the PDGF-triggered signaling cascades [64,65]
are appealing directions to selectively inhibit SMC growth without
aecting endothelial cell regeneration. To this end, Li et al. [66]
synthesized phosphorothioate antisense oligodeoxynucleotides (ODN)
to the 15 base-long conserved coding sequence of PDGF-A gene and
complexed them with PEI to make a polyplex formulation that was
absorbed on hydrogel coated stents. When implanted in the coronary
arteries of healthy young pigs, the anti-sense-eluting stents resulted in
a 50% reduction of neointimal thickening in comparison with non-
sense ODN-eluting and plain hydrogel stents [66]. Importantly, the
endothelial lining was complete 28 days aer stenting in the arterial
sections from the animals treated with the antisense PDGF-A –eluting
stents, while the endothelialization was patchy at the 28 days time point
in the arteries of animals receiving commercial sirolimus-eluting stents
[66]. Likewise, complete endothelialization of stented pig coronaries
and a 40% reduction of restenosis were observed with a six-ring
phosphorothioate morpholino backbone anti-sense construct eluted
from a PC-coated stent and targeting expression of a cell cycle check
point transcription factor c-myc [67].
In-stent neointima is primarily formed by the progeny of medial
SMC and adventitial myobroblasts that populate the intimal space
of an injured artery. Matrix remodeling triggered by vascular trauma
makes possible unhindered migration of these cell populations along
the gradients of specic chemoattractants [68]. us, stabilization
of ECM was proposed for limiting SMC migration toward (neo)
intima. In accordance with this idea, Johnson and colleagues [23]
bulk-immobilized a suspension of Ad vector (4×108 pfu) encoding
synthesis of the tissue inhibitor of metalloprotease-3 (TIMP3) on PC-
coated stainless steel stents. At 28 days aer stent implantation in pig
carotid arteries neointimal volume was reduced by 40% and 50% in the
animals treated with Ad-TIMP3-decorated stents in comparison with
pigs receiving BMS or Ad-LacZ immobilized stents, respectively [23].
GES targeting inammatory cell recruitment
Acute inammatory response in the form of platelet and
brin deposition and massive neutrophil recruitment to the site
of vascular trauma via P-selectin and CD11b/CD18 engagement is
triggered immediately upon stent deployment [52,69]. Days later, an
escalating production of IL-6, IL-8 and MCP-1 by the injured SMC
and ingressing neutrophils attracts monocytes that transmigrate into
the tissue becoming resident macrophages thus denoting a stage of
chronic inammation. Macrophages are the key cell type bridging
inammation to neointimal formation, since macrophage-produced
and secreted chemokines (TNF, IL-1β) and growth factors (PDGF,
FGF-2, IGF) stimulate luminal migration and proliferation of medial
SMC and adventitial myobroblasts that collectively form neointima
[52,69,70]. erefore, eectively curbing the inammatory response
represents a well-recognized anti-restenotic strategy [71,72].
In keeping with this therapeutic approach, Egashira and co-
workers [21] evaluated stents formulated with plasmid DNA encoding
dominant negative variant of MCP1. e stents deployed in the iliac
arteries of hypercholesterolemic rabbits and cynomolgus monkeys
resulted in approximately a 20% reduction of macrophage inltration
at the stent implantation site and a 20-30% restenosis attenuation in
comparison with BMS [21].
NFkB is a master transcription factor involved in the regulation
of multiple inammatory mediators. Ohtani et al. [19] used NFkB
decoy ODN (500 µg/stent) incorporated in a polyurethane coating.
Stents deployed in femoral arteries of hypercholesterolemic rabbits
demonstrated approximately a 30% decrease of neointimal thickening
in comparison with BMS and polyurethane coated stents at 4 weeks
post-stenting and a signicant decrease of inammatory marker
expression at 3 and 10 days post-implantation [19].
GES targeting thrombus formation in stented arteries
Platelets are the rst cellular elements to contact the surface of
newly deployed stents. Since both human patients and experimental
animals undergoing stent placement are pretreated with anti-platelet
and anti-coagulant drugs, acute blockage of a treated artery is seldom
an issue. However, a non-occluding parietal thrombus is routinely
formed at the angioplasty/stenting site. If the parietal thrombus
persists, it gets populated with the migrating SMC, thus facilitating fast
neointimal growth.
In an attempt to minimize platelet activation at the site of stent
implantation, Takemoto et al. [31] formulated gene-eluting stents
incorporating plasmid DNA encoding E-NTPDase, a crucial regulator
of platelet thrombus formation. is molecule is a membrane bound
enzyme that locally hydrolyses ATP, thus opposing platelet aggregation.
e plasmid DNA (78 ± 5 µg) was absorbed into cationized gelatin
hydrogel coating on the stent struts. Implantation of E-NTPDase-
eluting stents in a rabbit model of repeated femoral injury, which
is characterized by a high rate of thrombus formation completely,
prevented the occurrence of intravascular thrombi, while 25% and 50%
of femoral arteries treated with control βGal plasmid-eluting stents
had compromised patency at 3 and 7 days post-stenting. Moreover,
since thrombus serves as a nidus for SMC colonization, thus enhancing
neointimal formation, the extent of early restenosis (at day 7) was
signicantly reduced in the vessels treated with E-NTPDase stents [31].
Stents targeting nitric oxide production
Nitric oxide (NO), a by-product of enzymatic conversion
of arginine to citrulline has recently emerged as a key signaling
molecule in vascular homeostasis [73]. Increased NO production and
availability were shown to prevent platelet aggregation [74], inhibit
SMC proliferation [75,76], and migration [77], promote endothelial
growth [78], and decrease expression of cell adhesion molecules
by endothelium, inhibiting ingress of activated leukocytes into the
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 7 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
arterial intima [79]. Additionally NO promotes viability of endothelial
progenitor cells and enhances their capacity to reconstitute damaged
endothelium [80]. e pleiotropic nature of its benecial actions makes
NO-related interventions a plausible strategy for the prevention of ISR.
Both endothelial [81,82] and inducible [83,84] forms of NO
synthase (eNOS and iNOS, respectively) were investigated as genes
with potential anti-restenotic activity in animal models of arterial
injury/local delivery generally showing strong inhibiting eects on
neointimal formation. Driven by the wide scope of anti-restenotic
mechanisms bestowed by NO, we [40,42] and others [17,25,26,51] have
pursued the idea of the stent-based delivery of NOS-encoding vectors.
e studies by Sharif and others [25] employed PC coated stents
congured with 5x109 pfu of Ad-eNOS to address the hypothesis
that enhanced reendothelialization and reduced restenosis can be
achieved in a rabbit model with GES-induced eNOS overexpression.
e authors demonstrated signicantly enhanced restoration of the
endothelial layer and reduced neointimal thickness in both normo-
and hypercholesterolemic rabbits treated with Ad-eNOS stents in
comparison with animals that received Ad-βGal-eluting stents or
blank PC-coated stents. In an attempt to formulate a gene-eluting
stent devoid of immunogenic and pro-inammatory adenoviral
proteins, thus facilitating clinical translation of stent-based eNOS
overexpression, the same group recently reported [26] on a Lipofectin-
based lipoplex formulation containing human eNOS plasmid. Ten µg
of the lipoplex formulation were uniformly deposited on PC-coated
stents. e non-viral gene-eluting stents deployed in iliac arteries of
hypercholesterolemic rabbits achieved levels of transgene expression
comparable to Ad-containing GES. Moreover re-endothelialization
at 28 days was similarly enhanced by eNOS lipoplex-eluting stents.
However, no signicant reduction in restenosis indices was observed
in this study. e authors explain this discrepancy by the fact that
liposomal formulation primarily targets the macrophage population
in the diseased artery and not SMC, thus missing the vital cell type
aecting restenosis.
In contrast, using a similar rabbit model for therapeutic assessment
of eNOS-lipoplex-eluting stents Brito [17] demonstrated a 30%
reduction of restenosis accompanied by a 50% decrease of proliferation
in the eNOS lipoplex-treated rabbit arteries in comparison with an
empty vector control. e dierence in the vector dose (10 µg vs 25 µg)
and the delivery system used (adsorption of lipoplexes on a PC-coated
stent vs stent coating with lipoplexes dispersed in a gelatin/mannitol
formulation with additional lipoplex-free gelatin/PLGA supercoating)
between the former and the latter studies may account for the dierent
outcomes, highlighting the importance of proper pharmaceutical
adjustments for revealing full therapeutic potential of GES.
In our studies, Ad-iNOS delivered in a rat model from PAB- or
PABT-modied bare metal stents using anity adaptors [40] or
hydrolyzable cross-linkers [42] for vector tethering, respectively,
resulted in a 50%-60% reduction of neointimal thickening in
comparison with the BMS-implanted arteries. ese eects were
achieved using 109-1010 Ad-iNOS particles (107-108 pfu) appended to
a stent. Our recent studies assessing NO synthesis and ROS production
in cultured SMC and endothelium transduced with Ad-iNOS tethered
to stainless steel disks have determined that the super-physiological
levels of iNOS expression driven by the immobilized vector can lead to
NOS decoupling due to deciency of arginine and tetrahydrobiopterin
[85]. Decoupled NOS is unable to generate NO, producing superoxide
instead [86]. If not eectively removed by superoxide dismutases,
superoxide triggers a sequence of redox reactions that result in the
formation of multiple additional ROS, which can cause extensive
tissue damage [86]. erefore, pharmacological supplementation with
arginine and tetrahydrobiopterin may be required to modulate the
function of NOS overexpressed in the arterial wall using stent based
delivery of a respective vector [85].
Which of three known isoforms of NOS is the best choice for the
use in conjunction with GES is not immediately obvious. A study
by Cooney [82] compared the antirestenotic eectiveness of Ad-
iNOS and Ad-eNOS in a model of intraluminal dwell delivery of the
vectors to temporarily isolated segment of balloon-injured rabbit
carotid arteries. While a 20-min incubation of both vectors (109 pfu)
resulted in an identical extent of neointimal inhibition in comparison
with Ad-βGal-treated counterparts, this was allegedly achieved by
dissimilar mechanisms, since eNOS overexpression promoted and
iNOS overexpression inhibited endothelial regeneration in the treated
arterial segments [82]. Additional in-depth studies are required to
elucidate the actual mechanisms involved in the antirestenotic eects of
dierent NOS isoforms and their utility in the setting of GES-induced
overexpression.
Conclusions
Gene-eluting stents represent an interesting alternative to drug-
eluting stents in clinical circumstances where current DES formulations
oen fail to provide satisfactory results. Research related to GES has
now come to a phase when the perspectives of clinical translation
should always be taken into consideration. However, further work
related to optimization of all three main design components of GES:
a therapeutic transgene, a vector and a delivery system has to be done
before stent based vascular gene therapy comes to clinical fruition.
Acknowledgement
Studies carried out as part of the experimental work discussed in this review
were supported by American Heart Association Scientist Development grants
(I.F. 0735110N and M.C. 10SDG4020003), U.S. National Heart, Lung, and Blood
Institute grant HL 72108 (R.J.L.), U.S. National Heart, Lung, and Blood Institute
grant HL 111118 (M.C.) and U.S. National Heart, Lung, and Blood Institute
T32 007915 (S.P.F. and R.A.C.). The authors wish to acknowledge secretarial
assistance of Ms. Susan Kerns.
References
1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, et al. (2013) Heart
disease and stroke statistics--2013 update: a report from the American Heart
Association. Circulation 127: e6-6e245.
2. Wallace EL, Abdel-Latif A, Charnigo R, Moliterno DJ, Brodie B, et al. (2012)
Meta-analysis of long-term outcomes for drug-eluting stents versus bare-
metal stents in primary percutaneous coronary interventions for ST-segment
elevation myocardial infarction. Am J Cardiol 109: 932-940.
3. Joner M, Finn AV, Farb A, Mont EK, Kolodgie FD, et al. (2006) Pathology of
drug-eluting stents in humans: delayed healing and late thrombotic risk. J Am
Coll Cardiol 48: 193-202.
4. Valgimigli M, Park SJ, Kim HS, Park KW, Park DW, et al. (2013) Benets
and risks of long-term duration of dual antiplatelet therapy after drug-eluting
stenting: A meta-analysis of randomized trials. Int J Cardiol.
5. Doyle B, Rihal CS, O’Sullivan CJ, Lennon RJ, Wiste HJ, et al. (2007) Outcomes
of stent thrombosis and restenosis during extended follow-up of patients treated
with bare-metal coronary stents. Circulation 116: 2391-2398.
6. King SB 3rd (2007) Applying drug-eluting stents in clinical practice. Am J
Cardiol 100: 25K-31K.
7. Duda SH, Pusich B, Richter G, Landwehr P, Oliva VL, et al. (2002) Sirolimus-
eluting stents for the treatment of obstructive supercial femoral artery disease:
six-month results. Circulation 106: 1505-1509.
8. Rathore S, Terashima M, Katoh O, Matsuo H, Tanaka N, et al. (2009) Predictors
of angiographic restenosis after drug eluting stents in the coronary arteries:
contemporary practice in real world patients. EuroIntervention 5: 349-354
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 8 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
9. De Luca G, Dirksen MT, Spaulding C, Kelbaek H, Schalij M, et al. (2013)
Impact of diabetes on long-term outcome after primary angioplasty: insights
from the DESERT cooperation. Diabetes Care 36: 1020-1025.
10. Sharif F, Daly K, Crowley J, O’Brien T (2004) Current status of catheter- and
stent-based gene therapy. Cardiovasc Res 64: 208-216.
11. Appleby CE, Kingston PA (2004) Gene therapy for restenosis--what now, what
next? Curr Gene Ther 4: 153-182.
12. Yutani C, Ishibashi-Ueda H, Suzuki T, Kojima A (1999) Histologic evidence of
foreign body granulation tissue and de novo lesions in patients with coronary
stent restenosis. Cardiology 92: 171-177.
13. Schwartz RS (1998) Pathophysiology of restenosis: interaction of thrombosis,
hyperplasia, and/or remodeling. Am J Cardiol 81: 14E-17E.
14. Klugherz BD, Jones PL, Cui X, Chen W, Meneveau NF, et al. (2000) Gene
delivery from a DNA controlled-release stent in porcine coronary arteries. Nat
Biotechnol 18: 1181-1184.
15. Jewell CM, Zhang J, Fredin NJ, Wolff MR, Hacker TA, et al. (2006) Release
of plasmid DNA from intravascular stents coated with ultrathin multilayered
polyelectrolyte lms. Biomacromolecules 7: 2483-2491.
16. Saurer EM, Jewell CM, Roenneburg DA, Bechler SL, Torrealba JR, et al.
(2013) Polyelectrolyte multilayers promote stent-mediated delivery of DNA to
vascular tissue. Biomacromolecules 14: 1696-1704.
17. Brito LA, Chandrasekhar S, Little SR, Amiji MM (2010) Non-viral eNOS gene
delivery and transfection with stents for the treatment of restenosis. Biomed
Eng Online 9: 56.
18. Brito LA, Chandrasekhar S, Little SR, Amiji MM (2010) In vitro and in vivo
studies of local arterial gene delivery and transfection using lipopolyplexes-
embedded stents. J Biomed Mater Res A 93: 325-336.
19. Ohtani K, Egashira K, Nakano K, Zhao G, Funakoshi K, et al. (2006) Stent-
based local delivery of nuclear factor-kappaB decoy attenuates in-stent
restenosis in hypercholesterolemic rabbits. Circulation 114: 2773-2779.
20. Takahashi A, Palmer-Opolski M, Smith RC, Walsh K (2003) Transgene delivery
of plasmid DNA to smooth muscle cells and macrophages from a biostable
polymer-coated stent. Gene Ther 10: 1471-1478.
21. Egashira K, Nakano K, Ohtani K, Funakoshi K, Zhao G, et al. (2007) Local
delivery of anti-monocyte chemoattractant protein-1 by gene-eluting stents
attenuates in-stent stenosis in rabbits and monkeys. Arterioscler Thromb Vasc
Biol 27: 2563-2568.
22. Ganly S, Hynes SO, Sharif F, Aied A, Barron V, et al. (2013) Liposomal surface
coatings of metal stents for efcient non-viral gene delivery to the injured
vasculature. J Control Release 167: 109-119.
23. Johnson TW, Wu YX, Herdeg C, Baumbach A, Newby AC, et al. (2005) Stent-
based delivery of tissue inhibitor of metalloproteinase-3 adenovirus inhibits
neointimal formation in porcine coronary arteries. Arterioscler Thromb Vasc
Biol 25: 754-759.
24. Radke PW, Griesenbach U, Kivela A, Vick T, Judd D, et al. (2005) Vascular
oligonucleotide transfer facilitated by a polymer-coated stent. Hum Gene Ther
16: 734-740.
25. Sharif F, Hynes SO, Cooney R, Howard L, McMahon J, et al. (2008) Gene-
eluting stents: adenovirus-mediated delivery of eNOS to the blood vessel wall
accelerates re-endothelialization and inhibits restenosis. Mol Ther 16: 1674-1680.
26. Sharif F, Hynes SO, McCullagh KJ, Ganley S, Greiser U, et al. (2012) Gene-
eluting stents: non-viral, liposome-based gene delivery of eNOS to the blood
vessel wall in vivo results in enhanced endothelialization but does not reduce
restenosis in a hypercholesterolemic model. Gene Ther 19: 321-328.
27. Sharif F, Hynes SO, McMahon J, Cooney R, Conroy S, et al. (2006) Gene-
eluting stents: comparison of adenoviral and adeno- associated viral gene
delivery to the blood vessel wall in vivo. Hum Gene Ther 17: 741-750.
28. Walter DH, Cejna M, Diaz-Sandoval L, Willis S, Kirkwood L, et al. (2004) Local
gene transfer of phVEGF-2 plasmid by gene-eluting stents: an alternative
strategy for inhibition of restenosis. Circulation 110: 36-45.
29. Che HL, Bae IH, Lim KS, Song IT, Lee H, et al. (2012) Suppression of post-
angioplasty restenosis with an Akt1 siRNA-embedded coronary stent in a rabbit
model. Biomaterials 33: 8548-8556.
30. Kim TG, Lee Y, Park TG (2010) Controlled gene-eluting metal stent fabricated
by bio-inspired surface modication with hyaluronic acid and deposition of
DNA/PEI polyplexes. Int J Pharm 384: 181-188.
31. Takemoto Y, Kawata H, Soeda T, Imagawa K, Somekawa S, et al. (2009)
Human placental ectonucleoside triphosphate diphosphohydrolase gene
transfer via gelatin-coated stents prevents in-stent thrombosis. Arterioscler
Thromb Vasc Biol 29: 857-862.
32. Nakayama Y, Ji-Youn K, Nishi S, Ueno H, Matsuda T (2001) Development
of high-performance stent: gelatinous photogel-coated stent that permits drug
delivery and gene transfer. J Biomed Mater Res 57: 559-566.
33. San Juan A, Bala M, Hlawaty H, Portes P, Vranckx R, et al. (2009) Development
of a functionalized polymer for stent coating in the arterial delivery of small
interfering RNA. Biomacromolecules 10: 3074-3080.
34. Klugherz BD, Song C, DeFelice S, Cui X, Lu Z, et al. (2002) Gene delivery to
pig coronary arteries from stents carrying antibody-tethered adenovirus. Hum
Gene Ther 13: 443-454.
35. Perlstein I, Connolly JM, Cui X, Song C, Li Q, et al. (2003) DNA delivery from
an intravascular stent with a denatured collagen-polylactic-polyglycolic acid-
controlled release coating: mechanisms of enhanced transfection. Gene Ther
10: 1420-1428.
36. Zhu D, Jin X, Leng X, Wang H, Bao J, et al. (2010) Local gene delivery
via endovascular stents coated with dodecylated chitosan-plasmid DNA
nanoparticles. Int J Nanomedicine 5: 1095-1102.
37. van der Giessen WJ, Lincoff AM, Schwartz RS, van Beusekom HM,
Serruys PW, et al. (1996) Marked inammatory sequelae to implantation of
biodegradable and nonbiodegradable polymers in porcine coronary arteries.
Circulation 94: 1690-1697.
38. Bengali Z, Shea LD (2005) Gene Delivery by Immobilization to Cell-Adhesive
Substrates. MRS Bull 30: 659-662.
39. Segura T, Volk MJ, Shea LD (2003) Substrate-mediated DNA delivery: role of
the cationic polymer structure and extent of modication. J Control Release
93: 69-84.
40. Fishbein I, Alferiev IS, Nyanguile O, Gaster R, Vohs JM, et al. (2006)
Bisphosphonate-mediated gene vector delivery from the metal surfaces of
stents. Proc Natl Acad Sci U S A 103: 159-164.
41. Ma G, Wang Y, Fishbein I, Yu M, Zhang L, et al. (2013) Anchoring of self-
assembled plasmid DNA/anti-DNA antibody/cationic lipid micelles on
bisphosphonate-modied stent for cardiovascular gene delivery. Int J
Nanomedicine 8: 1029-1035.
42. Fishbein I, Alferiev I, Bakay M, Stachelek SJ, Sobolewski P, et al. (2008) Local
delivery of gene vectors from bare-metal stents by use of a biodegradable
synthetic complex inhibits in-stent restenosis in rat carotid arteries. Circulation
117: 2096-2103.
43. Fishbein I, Forbes SP, Chorny M, Connolly JM, Adamo RF, et al. (2013)
Adenoviral vector tethering to metal surfaces via hydrolyzable cross-linkers for
the modulation of vector release and transduction. Biomaterials 34: 6938-6948.
44. Chorny M, Fishbein I, Tengood JE, Adamo RF, Alferiev IS, et al. (2013) Site-
specic gene delivery to stented arteries using magnetically guided zinc oleate-
based nanoparticles loaded with adenoviral vectors. FASEB J 27: 2198-2206.
45. Chorny M, Fishbein I, Yellen BB, Alferiev IS, Bakay M, et al. (2010) Targeting
stents with local delivery of paclitaxel-loaded magnetic nanoparticles using
uniform elds. Proc Natl Acad Sci U S A 107: 8346-8351.
46. Polyak B, Fishbein I, Chorny M, Alferiev I, Williams D, et al. (2008) High eld
gradient targeting of magnetic nanoparticle-loaded endothelial cells to the
surfaces of steel stents. Proc Natl Acad Sci U S A 105: 698-703.
47. Polyak B, Friedman G (2009) Magnetic targeting for site-specic drug delivery:
applications and clinical potential. Expert Opin Drug Deliv 6: 53-70.
48. Carter AJ, Aggarwal M, Kopia GA, Tio F, Tsao PS, et al. (2004) Long-term
effects of polymer-based, slow-release, sirolimus-eluting stents in a porcine
coronary model. Cardiovasc Res 63: 617-624.
49. Sirois MG, Simons M, Kuter DJ, Rosenberg RD, Edelman ER (1997) Rat
arterial wall retains myointimal hyperplastic potential long after arterial injury.
Circulation 96: 1291-1298.
50. Jin X, Mei L, Song C, Liu L, Leng X, et al. (2008) Immobilization of plasmid DNA
on an anti-DNA antibody modied coronary stent for intravascular site-specic
gene therapy. J Gene Med 10: 421-429.
Citation: Fishbein I, Chorny M, Adamo RF, Forbes SP, Corrales RA, et al. (2013) Endovascular Gene Delivery from a Stent Platform: Gene- Eluting
Stents. Angiol 1: 109. doi: 10.4172/2329-9495.1000109
Page 9 of 9
Volume 1 • Issue 2 • 1000109
Angiol
ISSN: 2329-9495 AOA, an open access journal
51. Zhang LH, Luo T, Zhang C, Luo P, Jin X, et al. (2011) Anti-DNA antibody
modied coronary stent for plasmid gene delivery: results obtained from a
porcine coronary stent model. J Gene Med 13: 37-45.
52. Chaabane C, Otsuka F, Virmani R, Bochaton-Piallat ML (2013) Biological
responses in stented arteries. Cardiovasc Res 99: 353-363.
53. Curcio A, Torella D, Indol C (2011) Mechanisms of smooth muscle cell
proliferation and endothelial regeneration after vascular injury and stenting:
approach to therapy. Circ J 75: 1287-1296.
54. Gaffney MM, Hynes SO, Barry F, O’Brien T (2007) Cardiovascular gene
therapy: current status and therapeutic potential. Br J Pharmacol 152: 175-188.
55. Rissanen TT, Ylä-Herttuala S (2007) Current status of cardiovascular gene
therapy. Mol Ther 15: 1233-1247.
56. Asahara T, Bauters C, Pastore C, Kearney M, Rossow S, et al. (1995) Local
delivery of vascular endothelial growth factor accelerates reendothelialization
and attenuates intimal hyperplasia in balloon-injured rat carotid artery.
Circulation 91: 2793-2801.
57. Hers I, Vincent EE, Tavaré JM (2011) Akt signalling in health and disease. Cell
Signal 23: 1515-1527.
58. Zhou RH, Lee TS, Tsou TC, Rannou F, Li YS, et al. (2003) Stent implantation
activates Akt in the vessel wall: role of mechanical stretch in vascular smooth
muscle cells. Arterioscler Thromb Vasc Biol 23: 2015-2020.
59. Stabile E, Zhou YF, Saji M, Castagna M, Shou M, et al. (2003) Akt controls
vascular smooth muscle cell proliferation in vitro and in vivo by delaying G1/S
exit. Circ Res 93: 1059-1065.
60. Fager G, Hansson GK, Ottosson P, Dahllöf B, Bondjers (1988) Human arterial
smooth muscle cells in culture. Effects of platelet-derived growth factor and
heparin on growth in vitro. Exp Cell Res 176: 319-335.
61. Grotendorst GR, Chang T, Seppä HE, Kleinman HK, Martin GR (1982) Platelet-
derived growth factor is a chemoattractant for vascular smooth muscle cells. J
Cell Physiol 113: 261-266.
62. Kazlauskas A, DiCorleto PE (1985) Cultured endothelial cells do not respond
to a platelet-derived growth-factor-like protein in an autocrine manner. Biochim
Biophys Acta 846: 405-412.
63. Fishbein I, Waltenberger J, Banai S, Rabinovich L, Chorny M, et al. (2000)
Local delivery of platelet-derived growth factor receptor-specic tyrphostin
inhibits neointimal formation in rats. Arterioscler Thromb Vasc Biol 20: 667-676.
64. Deguchi J, Namba T, Hamada H, Nakaoka T, Abe J, et al. (1999) Targeting
endogenous platelet-derived growth factor B-chain by adenovirus-mediated
gene transfer potently inhibits in vivo smooth muscle proliferation after arterial
injury. Gene Ther 6: 956-965.
65. Lin ZH, Fukuda N, Suzuki R, Takagi H, Ikeda Y, et al. (2004) Adenovirus-
encoded hammerhead ribozyme to PDGF A-chain mRNA inhibits neointima
formation after arterial injury. J Vasc Res 41: 305-313.
66. Li Y, Fukuda N, Kunimoto S, Yokoyama S, Hagikura K, et al. (2006) Stent-
based delivery of antisense oligodeoxynucleotides targeted to the PDGF
A-chain decreases in-stent restenosis of the coronary artery. J Cardiovasc
Pharmacol 48: 184-190.
67. Kipshidze NN, Iversen P, Kim HS, Yiazdi H, Dangas G, et al. (2004)
Advanced c-myc antisense (AVI-4126)-eluting phosphorylcholine-coated
stent implantation is associated with complete vascular healing and reduced
neointimal formation in the porcine coronary restenosis model. Catheter
Cardiovasc Interv 61: 518-527.
68. Johnson C, Galis ZS (2004) Matrix metalloproteinase-2 and -9 differentially
regulate smooth muscle cell migration and cell-mediated collagen organization.
Arterioscler Thromb Vasc Biol 24: 54-60.
69. Welt FG, Rogers C (2002) Inammation and restenosis in the stent era.
Arterioscler Thromb Vasc Biol 22: 1769-1776.
70. Sarkar K, Sharma SK, Sachdeva R, Romeo F, Garza L, et al. (2006) Coronary
artery restenosis: vascular biology and emerging therapeutic strategies. Expert
Rev Cardiovasc Ther 4: 543-556.
Submit your next manuscript and get advantages of OMICS
Group submissions
Unique features:
• Userfriendly/feasiblewebsite-translationofyourpaperto50world’sleadinglanguages
• AudioVersionofpublishedpaper
• Digitalarticlestoshareandexplore
Special features:
• 250OpenAccessJournals
• 20,000editorialteam
• 21daysrapidreviewprocess
• Qualityandquickeditorial,reviewandpublicationprocessing
• IndexingatPubMed(partial),Scopus,DOAJ,EBSCO,IndexCopernicusandGoogleScholaretc
• SharingOption:SocialNetworkingEnabled
• Authors,ReviewersandEditorsrewardedwithonlineScienticCredits
• Betterdiscountforyoursubsequentarticles
Submityourmanuscriptat:http://www.omicsonline.org/submission/
Citation: Yamasaki T, Koizumi T, Tamaki T, Sakamoto A, Kikutani T, et al. (2013)
Differing Behavior of Plasma Pentraxin3 and High-Sensitive CRP at the Very
Onset of Myocardial Infarction with ST-Segment Elevation. Angiol 1: 108. doi:
10.4172/2329-9495.1000108
71. Inoue T, Croce K, Morooka T, Sakuma M, Node K, et al. (2011) Vascular
inammation and repair: implications for re-endothelialization, restenosis, and
stent thrombosis. JACC Cardiovasc Interv 4: 1057-1066.
72. Danenberg HD, Fishbein I, Gao J, Mönkkönen J, Reich R, et al. (2002)
Macrophage depletion by clodronate-containing liposomes reduces neointimal
formation after balloon injury in rats and rabbits. Circulation 106: 599-605.
73. Ignarro LJ (2002) Wei Lun Visiting Professorial Lecture: Nitric oxide in the
regulation of vascular function: an historical overview. J Card Surg 17: 301-306.
74. Nong Z, Hoylaerts M, Van Pelt N, Collen D, Janssens S (1997) Nitric oxide
inhalation inhibits platelet aggregation and platelet-mediated pulmonary
thrombosis in rats. Circ Res 81: 865-869.
75. Garg UC, Hassid A (1989) Nitric oxide-generating vasodilators and 8-bromo-
cyclic guanosine monophosphate inhibit mitogenesis and proliferation of
cultured rat vascular smooth muscle cells. J Clin Invest 83: 1774-1777.
76. Groves PH, Banning AP, Penny WJ, Newby AC, Cheadle HA, et al. (1995) The
effects of exogenous nitric oxide on smooth muscle cell proliferation following
porcine carotid angioplasty. Cardiovasc Res 30: 87-96.
77. Sarkar R, Meinberg EG, Stanley JC, Gordon D, Webb RC (1996) Nitric oxide
reversibly inhibits the migration of cultured vascular smooth muscle cells. Circ
Res 78: 225-230.
78. Papapetropoulos A, García-Cardeña G, Madri JA, Sessa WC (1997) Nitric
oxide production contributes to the angiogenic properties of vascular endothelial
growth factor in human endothelial cells. J Clin Invest 100: 3131-3139.
79. De Caterina R, Libby P, Peng HB, Thannickal VJ, Rajavashisth TB, et al.
(1995) Nitric oxide decreases cytokine-induced endothelial activation. Nitric
oxide selectively reduces endothelial expression of adhesion molecules and
proinammatory cytokines. J Clin Invest 96: 60-68.
80. Kong D, Melo LG, Mangi AA, Zhang L, Lopez-Ilasaca M, et al. (2004) Enhanced
inhibition of neointimal hyperplasia by genetically engineered endothelial
progenitor cells. Circulation 109: 1769-1775.
81. Chen L, Daum G, Forough R, Clowes M, Walter U, et al. (1998) Overexpression
of human endothelial nitric oxide synthase in rat vascular smooth muscle cells
and in balloon-injured carotid artery. Circ Res 82: 862-870.
82. Cooney R, Hynes SO, Sharif F, Howard L, O’Brien T (2007) Effect of gene
delivery of NOS isoforms on intimal hyperplasia and endothelial regeneration
after balloon injury. Gene Ther 14: 396-404.
83. Muhs A, Heublein B, Schletter J, Herrmann A, Rüdiger M, et al. (2003)
Preclinical evaluation of inducible nitric oxide synthase lipoplex gene therapy
for inhibition of stent-induced vascular neointimal lesion formation. Hum Gene
Ther 14: 375-383.
84. Wang K, Kessler PD, Zhou Z, Penn MS, Forudi F, et al. (2003) Local adenoviral-
mediated inducible nitric oxide synthase gene transfer inhibits neointimal
formation in the porcine coronary stented model. Mol Ther 7: 597-603.
85. Forbes SP, Alferiev IS, Chorny M, Adamo RF, Levy RJ, et al. (2013)
Modulation of NO and ROS production by AdiNOS transduced vascular cells
through supplementation with L-Arg and BH4: Implications for gene therapy of
restenosis. Atherosclerosis 230: 23-32.
86. Forstermann U (2008) Oxidative stress in vascular disease: causes, defense
mechanisms and potential therapies. Nat Clin Pract Cardiovasc Med 5: 338-349.
... As autonomous implantable devices, DES possess a limited loading capacity that is often inadequate to provide therapeutic concentrations of drug for the entire period of post-angioplasty vessel remodeling. Conversely, stents furnished with gene delivery vectors immobilized on the surface of the struts, i.e., gene delivery stents (GDS), may overcome this intrinsic DES deficiency by delivering the gene vector into the stented vessel wall and making the vascular tissue a permanent production site of the encoded therapeutic protein 43 . We 44 and others 45 showed sustained transgene expression in mammalian vasculature upon transduc- Advantages of apoA1(4WF) over apoA1(WT) in cholesterol efflux under conditions of oxidative stress. ...
Article
Full-text available
Abstract In-stent restenosis (ISR) complicates revascularization in the coronary and peripheral arteries. Apolipoprotein A1 (apoA1), the principal protein component of HDL possesses inherent anti-atherosclerotic and anti-restenotic properties. These beneficial traits are lost when wild type apoA1(WT) is subjected to oxidative modifications. We investigated whether local delivery of adeno-associated viral (AAV) vectors expressing oxidation-resistant apoA1(4WF) preserves apoA1 functionality. The efflux of 3H-cholesterol from macrophages to the media conditioned by endogenously produced apoA1(4WF) was 2.1-fold higher than for apoA1(WT) conditioned media in the presence of hypochlorous acid emulating conditions of oxidative stress. The proliferation of apoA1(WT)- and apoA1(4FW)-transduced rat aortic smooth muscle cells (SMC) was inhibited by 66% ± 10% and 65% ± 11%, respectively, in comparison with non-transduced SMC (p
... GES may hold promise for contributing new ideas to the stent-based prevention of ISR through genetic interventions by capitalizing on a wide variety of molecular targets. Several GES studies have been performed in recent years targeting signalling pathways and different specific cell types, thus allowing for the selective inhibition of smooth muscle cell proliferation and migration.55 9 | GES TRIALS FOR ACCELERATION OF RE-ENDOTHELIALIZATIONA study by Paul et al.56 investigated a nanobiohybrid hydrogelbased endovascular stent device carrying endosomolytic Tat peptide/ DNA nanoparticles or nanoparticles hybridized to polyacrylic acid conjugated to carbon nanotubes in adult beagle dogs. ...
... The nature of therapeutic genes capable of inhibiting deleterious remodeling of the stented arteries has been investigated in many studies 16,17 . We 7, 8 and others 5,11 have previously demonstrated that augmented nitric oxide production with overexpression of endothelial and inducible isoforms of nitric oxide synthase encoded by gene vectors delivered from the stents inhibits restenosis though multiple mechanisms related to reduced SMC proliferation 18 and migration 19 , enhanced endothelial re-growth 20 , and decreased ingress of inflammatory cell types into the vascular wall 21 . ...
Article
Full-text available
In-stent restenosis remains an important clinical problem in the era of drug eluting stents. Development of clinical gene therapy protocols for the prevention and treatment of in-stent restenosis is hampered by the lack of adequate local delivery systems. Herein we describe a novel stent-based gene delivery platform capable of providing local arterial gene transfer with adeno-associated viral (AAV) vectors. This system exploits the natural affinity of protein G (PrG) to bind to the Fc region of mammalian IgG, making PrG a universal adaptor for surface immobilization of vector-capturing antibodies (Ab). Our results: (1) demonstrate the feasibility of reversible immobilization of AAV2 vectors using vector tethering by AAV2-specific Ab appended to the stent surface through covalently attached PrG, (2) show sustained release kinetics of PrG/Ab-immobilized AAV2 vector particles into simulated physiological medium in vitro and site-specific transduction of cultured cells, (3) provide evidence of long-term (12 weeks) arterial expression of luciferase with PrG/Ab-tethered AAV2Luc, and (4) show anti-proliferative activity and anti-restenotic efficacy of stent-immobilized AAV2iNOS in the rat carotid artery model of stent angioplasty.Gene Therapy accepted article preview online, 23 August 2017. doi:10.1038/gt.2017.82.
... Altogether, the superior gene expression profiles combined with the lower cytotoxicity with respect to the gold standard 25 kDa bPEI make PEI2-PrA1 a very promising candidate for the transfection of VSMCs. These transfection results are extremely important because they were obtained in the presence of serum and upon centrifugation of the polyplexes over the cells, mimicking the high concentration at the cell surface that could be found in cardiovascular devices such as gene-eluting stents [35] or catheter balloons [36,37]. ...
Article
Full-text available
Gene therapy targeted to vascular cells represents a promising approach for prevention and treatment of pathological conditions such as intimal hyperplasia, in-stent and post-angioplasty restenosis. In this context, polymeric non-viral gene delivery systems are a safe alternative to viral vectors but a further improvement in efficiency and cytocompatibility is needed to improve their clinical success. Herein, a library of 24 branched polyethylenimine (bPEI) derivatives modified with hydrophobic moieties was synthesised, characterised and tested in vitro on primary vascular cells, aiming to identify delivery agents with superior transfection efficiency and low cytotoxicity. Low molecular weight PEIs (0.6, 1.2 and 2 kDa) were grafted with long (C18) and short (C3) aliphatic chains, featuring different unsaturation degrees and degrees of substitution. 0.6 kDa bPEI-based derivatives were generally ineffective in transfection on vascular smooth muscle cells (VSMCs), while among the other derivatives some promising vectors were identified. Forcing polyplexes on the cell surface by means of centrifugation invariably boosted transfection levels but increased cytotoxicity as well. Of note, a propionyl-substituted derivative (PEI2-PrA1, C3:0) was the most effective on both VSMCs and endothelial cells (ECs), with higher and more sustained gene expression in combination with markedly lower cytotoxicity with respect to the gold standard 25 kDa bPEI. In addition, a linoleoyl-substituted derivative (PEI1.2-LA6, C18:2) owing to its high efficiency in VSMCs and relative inefficacy in ECs, combined with tolerable cytotoxicity was proposed as a vector for specific VSMCs targeting.
... GES may hold promise for contributing new ideas to the stent-based prevention of ISR through genetic interventions by capitalizing on a wide variety of molecular targets. Several GES studies have been performed in recent years targeting signalling pathways and different specific cell types, thus allowing for the selective inhibition of smooth muscle cell proliferation and migration.55 9 | GES TRIALS FOR ACCELERATION OF RE-ENDOTHELIALIZATIONA study by Paul et al.56 investigated a nanobiohybrid hydrogelbased endovascular stent device carrying endosomolytic Tat peptide/ DNA nanoparticles or nanoparticles hybridized to polyacrylic acid conjugated to carbon nanotubes in adult beagle dogs. ...
Article
Full-text available
Aim: This article concisely recapitulates the different existing modes of stent mediated gene/drug delivery, their considerable advancements in clinical trials and a rationale of other merging new technologies such as nanotechnology and microRNA based therapeutics, furthermore addressing the breach in each of these perpetual stent platforms. Discussion: Over the past decade stent mediated gene/drug delivery have materialized as a hopeful alternative for cardiovascular disease and cancer in contrast to the routine conventional treatment modalities. Regardless of the phenomenal recent developments figured out with coronary interventions and cancer therapy by use of gene and drug eluting stents, practical hurdles still remain a challenge. The article moreover, highlights the limitations that each of the existing stent based gene/drug delivery system encompasses and therefore a vision for the future of discovering an ideal stent therapeutic platform that would circumvent all the practical hurdles witnessed with the existing technology. Conclusion: Further delve into improvisation of next generation DES to an extent has helped in overcoming the issue of restenosis. However, current stent formulations fall short of anticipated clinically meaningful outcomes and there is an explicit need for more randomized trials to further evaluate stent platforms in favour of enhanced safety and clinical value. GES may hold promise in contributing new ideas for stent based prevention of in-stent restenosis through genetic interventions by capitalizing wide variety of molecular targets. Therefore, the heart of the matter directs us to foresee in finding an ideal stent therapeutic platform that would tackle all gaps in the existing technology.
... One promising solution for gene therapy in-stent is the use of endovascular stents, which can act both as the gene carrier and the scaffold for localized and prolonged delivery of therapeutic genes into the diseased blood vessel wall. [9][10][11] Several reports in the literature have described gene therapy approaches using naked plasmids, viral vectors, and non-viral nano-delivery systems. [12][13][14] However, several recurring issues have led researchers to reconsider their use in human clinical trials. ...
Article
Full-text available
Background and objectives: MicroRNA 145 is known to be responsible for cellular proliferation, and its enhanced expression reportedly inhibits the retardation of vascular smooth muscle cell growth specifically. In this study, we developed a microRNA 145 nanoparticle immobilized, hyaluronic acid (HA)-coated stent. Materials and methods: For the gene therapy, we used disulfide cross-linked low molecular polyethylenimine as the carrier. The microRNA 145 was labeled with YOYO-1 and the fluorescent microscopy images were obtained. The release of microRNA 145 from the stent was measured with an ultra violet spectrophotometer. The downstream targeting of the c-Myc protein and green fluorescent protein was determined by Western blotting. Finally, we deployed microRNA 145/ssPEI nanoparticles immobilized on HA-coated stents in the balloon-injured external iliac artery in a rabbit restenosis model. Results: Cellular viability of the nanoparticle-immobilized surface tested using A10 vascular smooth muscle cells showed that MSN exhibited negligible cytotoxicity. In addition, microRNA 145 and downstream signaling proteins were identified by western blots with smooth muscle cell (SMC) lysates from the transfected A10 cell, as the molecular mechanism for decreased SMC proliferation that results in the inhibition of in-stent restenosis. MicroRNA 145 released from the stent suppressed the growth of the smooth muscle at the peri-stent implantation area, resulting in the prevention of restenosis at the post-implantation. We investigated the qualitative analyses of in-stent restenosis in the rabbit model using micro-computed tomography imaging and histological staining. Conclusion: MicroRNA 145-eluting stent mitigated in-stent restenosis efficiently with no side effects and can be considered a successful substitute to the current drug-eluting stent.
Article
Vascular stent is tubular shaped medical device designed to treat occluded circulatory or digestive systems by opening the blocked vascular sites. Originally stents were made of bare metals, such as stainless steel and cobalt–chromium alloys, due to their inherent corrosion resistance and good mechanical properties. Damage of intimal layer during expansion of stent, however, have caused neointimal hyperplasia resulting in in-stent restenosis which is one of main complications of vascular stent treatment. Drug-eluting stents (DESs) were introduced to alleviate this restenosis by local delivery of anti-proliferative drugs to the implanted or inserted site. The DES systems have been developed over the years by making changes in its main component, namely, drug, polymeric coating layer and expandable scaffold. Nevertheless, late thrombosis caused by drugs themselves and polymeric coating layer still remains as a serious problem of DES systems. Numerous studies, therefore, consistently focus on more advanced DES systems to solve the current issues. In this mini review article, we cast an opinion on how to improve the potential of DES system by means of (i) summarizing the history of DES system developed so far and (ii) updating current advances for the future generation of DES system.
Article
Full-text available
Gene therapy with viral vectors encoding for NOS enzymes has been recognized as a potential therapeutic approach for the prevention of restenosis. Optimal activity of iNOS is dependent on the intracellular availability of L-Arg and BH4 via prevention of NOS decoupling and subsequent ROS formation. Herein, we investigated the effects of separate and combined L-Arg and BH4 supplementation on the production of NO and ROS in cultured rat arterial smooth muscle and endothelial cells transduced with AdiNOS, and their impact on the antirestenotic effectiveness of AdiNOS delivery to balloon-injured rat carotid arteries. Supplementation of AdiNOS transduced endothelial and vascular smooth muscle cells with L-Arg (3.0 mM), BH4 (10 μM) and especially their combination resulted in a significant increase in NO production as measured by nitrite formation in media. Formation of ROS was dose-dependently increased following transduction with increasing MOIs of AdiNOS. Exposure of RASMC to AdiNOS tethered to meshes via a hydrolyzable cross-linker, modeling viral delivery from stents, resulted in increased ROS production, which was decreased by supplementation with BH4 but not L-Arg or L-Arg/BH4. Enhanced cell death, caused by AdiNOS transduction, was also preventable with BH4 supplementation. In the rat carotid model of balloon injury, intraluminal delivery of AdiNOS in BH4-, L-Arg-, and especially in BH4 and L-Arg supplemented animals was found to significantly enhance the antirestenotic effects of AdiNOS-mediated gene therapy. Fine-tuning of iNOS function by L-Arg and BH4 supplementation in the transduced vasculature augments the therapeutic potential of gene therapy with iNOS for the prevention of restenosis.
Article
Full-text available
Endothelial cell loss is a critical event in the pathological repair of the injured blood vessel. Impaired endothelial function results in reduced production of key vascular mediators such as nitric oxide (NO) within the vessel wall leading to enhanced smooth muscle cell proliferation and migration and ultimately intimal hyperplasia. The aim of the present study was to directly compare the effects of adenoviral-mediated gene delivery of two nitric oxide synthase (NOS) isoforms, eNOS and iNOS on endothelial regeneration and intimal hyperplasia following endothelial injury in the rabbit carotid artery.Methods: The right carotid arteries of male New Zealand white rabbits were denuded by passing a 3French Fogarty balloon catheter along the artery three times. 1×109 pfu of AdeNOS, AdiNOS or AdβGal was then delivered intraluminally and allowed to dwell for twenty minutes. Transgene expression was sought after 3 days by immunohistochemistry. The effect on intimal hyperplasia was sought using histological staining after 14 days. Evans blue staining was used to determine the effect on endothelial regeneration.Results: eNOS and iNOS expression was detected in transduced arteries. Neointima/media ratios were significantly reduced in eNOS (0.07±0.033) and iNOS (0.12±0.1) transduced arteries compared with βGal (0.39±0.14) transduced arteries. eNOS (3.61±2.025% de-endothlialized) had no effect on endothelial regeneration compared to βGal (8.48±5.32), however iNOS (28.78±5.8) inhibited endothelial regeneration in the injured rabbit carotid artery.Conclusion: These results highlight the potential of NOS gene therapy, in particular eNOS gene therapy as a potential therapeutic strategy for the prevention of restenosis after angioplasty procedures.
Article
Although pentraxin3 (PTX3) has been reported as marker of more directly reflect the vascular inflammatory status than short pentraxin including high-sensitive CRP (hs-CRP), detailed difference in blood levels between PTX3 and hs-CRP at the onset of ST-segment elevation myocardial infarction (STEMI) are not fully investigated. Blood levels of pentraxins (PTX3 and hs-CRP) in 20 patients with early arrival of STEMI (2.9 ± 2.2 hours after onset) were measured at baseline, 24, 48, 72 and 120 hours after primary percutaneous coronary intervention (PCI). Also, the blood levels in infarct-related artery (IRA) were measured by thrombus aspiration during PCI. Samples of control (not myocardial infarction) with normal coronary artery (n=10) were drawn from both coronary and peripheral arteries during diagnostic coronary angiography. At baseline, the levels of PTX3 in both femoral and coronary artery in STEMI were significantly higher than those in control, but the hs-CRP did not different between STEMI and control. The level of both PTX3 and hs-CRP did not different between femoral artery and IRA in STEMI patients at baseline. Systemic level of PTX3 peaked 24 hours (p=0.01) followed by the hs-CRP that peaked 48 hours (p<0.01) after the PCI. PTX3 had appeared earlier than hs-CRP in the systemic circulation in the STEMI patients, but they may not be locally released from the IRA.
Article
Background— Stent implantation for obstructive femoropopliteal artery disease has been associated with poor long-term outcomes. This study evaluated the effectiveness of shape memory alloy recoverable technology (SMART) nitinol self-expanding stents coated with a polymer impregnated with sirolimus (rapamycin) versus uncoated SMART stents in superficial femoral artery obstructions. Methods and Results— Thirty-six patients were recruited for this double-blind, randomized, prospective trial. All patients had chronic limb ischemia and femoral artery occlusions (57%) or stenoses (average lesion length, 85±57 mm). Patients were eligible for randomization after successful guidewire passage across the lesion. Eighteen patients received sirolimus-eluting SMART stents and 18 patients received uncoated SMART stents. The primary end point of the study was the in-stent mean percent diameter stenosis, as measured by quantitative angiography at 6 months. The in-stent mean percent diameter stenosis was 22.6% in the sirolimus-eluting stent group versus 30.9% in the uncoated stent group ( P =0.294). The in-stent mean lumen diameter was significantly larger in the sirolimus-eluting stent group (4.95 mm versus 4.31 mm in the uncoated stent group; P =0.047). No serious adverse events (death or prolonged hospitalization) were reported. Conclusions— The use of sirolimus-eluting SMART stents for superficial femoral artery occlusion is feasible, with a trend toward reducing late loss compared with uncoated stents. The coated stent also proved to be safe and was not associated with any serious adverse events. Received March 29, 2002; revision received June 27, 2002; accepted June 27, 2002.