You are on page 1of 8

Brief Reviews

Applications of Nanotechnology to Atherosclerosis,


Thrombosis, and Vascular Biology
Samuel A. Wickline, Anne M. Neubauer, Patrick Winter, Shelton Caruthers, Gregory Lanza
AbstractThe role of nanotechnology in cardiovascular diagnosis is expanding rapidly. The goal of this brief review is
to illustrate selected examples of nanosystems that have been applied to the arenas of atherosclerosis, thrombosis, and
vascular biology. The technologies for producing targeted nanosystems are multifarious and reflect end uses in many
cases. The results to date indicate rapid growth of interest and capability in the field. The future of cardiovascular
diagnosis already is being impacted by nanosystems that can both diagnose pathology and treat it with targeted delivery
systems. (Arterioscler Thromb Vasc Biol. 2006;26:435-441.)
Key Words: nanotechnology contrast agents imaging drug therapy

dvances in cellular and molecular biology are extending


the horizons of medical imaging from gross anatomic
description toward delineation of cellular and biochemical
signaling processes. The emerging fields of cellular and
molecular imaging are converging to allow noninvasive
detection of the molecular components of pathological processes, such as image-based identification of specific molecules associated with inflammation or angiogenesis. Techniques have been developed recently to achieve molecular
and cellular imaging with most imaging modalities, including
nuclear,1 optical,2 ultrasound,3,4 and magnetic resonance imaging (MRI).5,6 In general, these methods might be considered the nondestructive in vivo analog of traditional immunocytochemistry.7 This brief review focuses on both
advanced imaging methods and on new targeted nanoparticle
contrast agents for early characterization of atherosclerosis
and cardiovascular pathology at the cellular and molecular
levels that might represent the next frontier for combining
imaging and rational drug delivery to facilitate personalized
medicine.5
The rapid growth of nanotechnology and nanoscience
could greatly expand the clinical opportunities for molecular
imaging.6,8 Nanotechnology seeks to develop and combine
new materials by precisely engineering atoms and molecules
to yield new molecular assemblies on the scale of individual
cells, organelles, or even smaller components, generally in
the range of 5 to 500 nm. The specific organization of such
nanoscale materials is anticipated to confer unique chemical
and biological properties on the basis of interactions that
occur at their surfaces. Synthesis of such materials may occur
from a top down approach by miniaturizing existing microscopic materials, or from a bottom up approach involv-

ing self assembly of molecules into reproducible and


well-defined nanoscale constructs.

Nanoparticle Classes
Liposomes, 50- to 700-nm uni- or multilammelar vesicles
comprising lipid bilayer membranes surrounding an aqueous
interior, have been approved for enhancing the efficacy and
safety of drugs such as doxorubicin (eg, Doxil, ALZA
Corporation, Tibotec Therapeutics). Applications of liposomal technology as molecular imaging agents have been
reported for both ultrasound and MRI.9,10
Emulsions, which are chemically distinct from liposomes,
are oil-in-water type mixtures that are stabilized with surfactants to maintain size and shape. Perfluorocarbon core emulsions (200 to 400 nm) have been used for molecular imaging
with MRI, ultrasound, fluorescence, nuclear and computed
tomography imaging.4 6,11 For example, by incorporating
vast numbers of paramagnetic gadolinium complexes
(50 000) onto emulsion particles, the signal enhancement
possible for each binding site is magnified dramatically by a
factor of 106 over conventional paramagnetic extracellular
contrast agents.12,30 Modified micellar particles such as highdensity lipoprotein (HDL) or low-density lipoprotein particles have been used as molecular imaging agents for
MRI.14,15
Polymers (40 to 200 nm) offer a wide variety of flexible
designer approaches to construction of molecular imaging
agents and therapeutic delivery devices.16 Size and shape can
be tightly controlled, and functionalization of their surface
permits binding of myriad targeting and therapeutic moieties
for imaging, as well as drug and gene delivery. Polymers
made from poly hydroxy acids such as the copolymer of poly

Original received October 26, 2005; final version accepted December 8, 2005.
From Washington University, Departments of Medicine (S.A.W., P.W., G.L.) and Biomedical Engineering (S.A.W., A.M.N., G.L.), St Louis, Mo, and
Philips Medical Systems (S.C.), Best, the Netherlands.
Correspondence to Samuel A. Wickline, Washington University School of Medicine, Campus Box 8086, 660 South Euclid Ave, St Louis, MO 63110.
E-mail wicklines@aol.com
2006 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org

DOI: 10.1161/01.ATV.0000201069.47550.8b

435
Downloaded from http://atvb.ahajournals.org/
by guest on January 8, 2015

436

Arterioscler Thromb Vasc Biol.

March 2006

(lactic acid) and poly (D,L-lactide-co-glycolide) have been


investigated for localized drug and gene delivery. Dendrimers, or cascade polymers, are highly branched polymeric
structures that are globular in configuration. Paramagnetic
polyamidoamine and diaminobutane dendrimers have been
reported for MRI applications.17,18 The multivalent surface
comprises a number of functional sites that can undergo
reactions to add drugs, imaging agents, and targeting ligands.
Metallic particles such as iron oxide nanoparticles (15 to
60 nm) generally comprise a class of superparamagnetic
agents that can be coated with dextran, phospholipids, or
other compounds to inhibit aggregation and enhance stability
for use as passive or active targeting agents. The iron in
monocrystaline iron oxide nanoparticles, small particles of
iron oxide (50 to 500 nm), or ultrasmall particles of iron oxide
(USPIOs, 10 to 50 nm) produces strong local disruptions in
the magnetic field of MRI scanners, which leads increased
T2* relaxation, causing a decrease in image intensity in areas
with iron particle accumulation (termed susceptibility effects). These particles exhibit a very long circulating half-life
(24 hours) and have been used for passive targeted imaging
of pathological inflammatory processes such as unstable
atherosclerotic plaques by MRI.19 Alternatively, similar types
of particles (eg, cross-linked iron oxide particles complexed
with retroviral tat protein ligands) have been used for
localization and transcellular deposition.20
Other metal-based agents such as gold shell nanoparticles
(120 nm) have been used for both imaging and therapy.21,22
Carbon nanotubes and fullerenes (4 nm) have been used as
particulate systems whose surfaces also can be functionalized
for tissue binding,23 Native fluorescent properties have been
reported.24 Quantum dots (2 to 8 nm) are constructed from
semiconductor materials (eg, cadmium selenide) that manifest stable (nonquenching) fluorescent properties at various
wavelengths depending on exact composition.2527 For use in
vivo, they must be coated with materials (polymers) that
allow solubilization while also preventing leaching of the
toxic heavy metals.

Specific Examples of Cardiovascular Imaging


Atherosclerotic Plaque
A sine qua non of the disrupted plaque is fibrin deposition.
Not only is fibrin deposition one of the earliest signs of
plaque rupture or erosion, but along with intraplaque hemorrhage, it also forms a considerable part of the core of growing
lesions. The diagnosis of disrupted plaque by detection of
small deposits of fibrin in erosions or microfractures could
allow characterization of a potential culprit lesion before a
high-grade stenosis has been formed that is detectable by
cardiac catheterization.
The possibility of nanoparticle-targeted fibrin imaging
with either ultrasound or paramagnetic MR contrast agents
was first demonstrated by Lanza et al3,28 as early as 1996. In
this case, the ligand comprised an antibody fragment highly
specific for certain cross-linked fibrin peptide domains,
which can be complexed to the particle either through
avidin-biotin linkages or covalently to the functionalized
nanoparticle as has been shown for tissue factor targeting.29,30

For ultrasound imaging, thrombi formed in situ in canine


carotid arteries were detectable within 30 minutes with
commercially available 7.5-MHz linear array imaging
transducers.28
Tissue factor is a prothrombotic transmembrane glycoprotein expressed within plaques that is upregulated after vascular injury or stent placement and that contributes as a mitogen
to restenosis.31 Tissue factor imaging has been demonstrated
in vivo for molecular imaging with ultrasound (Figure 1) and
in vitro with MRI.3,32 The ability to image tissue factortargeted paramagnetic nanoparticles bound to smooth muscle
cell monolayers in cell culture at 1.5T attests to the potency
of nanoparticles agents that carry 50 000 or more gadolinium
chelates.
Echogenic liposomes, in contrast to nanoparticles or emulsions, are composed of alternating layers of aqueous fluid and
lipid bilayers that are formulated to produce an ultrasound
signal.9 Hamilton et al33,34 used these liposomes to target
thrombi and various vascular signatures associated with
atheroma development in injured vessels of miniswine for
intravascular ultrasound imaging (Figure 1). By targeting
intercellular adhesion molecule-1, vascular cell adhesion
molecule-1, fibrin, fibrinogen, and tissue factor, they were
able to produce targeted enhancement in the vessel walls 5
minutes after intravenous administration of the liposomes.
MR imaging of vascular cell adhesion molecule also has been
reported recently with the use of peptide-targeted superparamagnetic nanoparticles in aortas of apolipoprotein E null
mice by Kelly et al.35
For MRI, perfluorocarbon particles loaded with 50 to
90 000 gadolinium atoms per particle yielded a substantial
amplification of signal from fibrin clots at 1.5T both in vitro
and in vivo (Figure 2).6,36 Furthermore, the detection of
disrupted plaque was illustrated in actual human carotid
endarterectomy specimens obtained from patients symptomatic with transient ischemic attacks, stroke, or bruits (Figure
2).12 Epix Pharmaceuticals more recently has used phage
display methods to produce a peptide ligand specific for
fibrin (EP-2104R), which may be useful for imaging thrombi
in various body locations such as the left atrium, pulmonary
arteries, or coronary arteries in experimental preparations.37,38

Angiogenesis

The v3-integrin is a general marker of angiogenesis and


plays an important role in a wide variety of disease states,
including atherosclerosis.39 The v3-integrin is a wellcharacterized heterodimeric adhesion molecule that is widely
expressed by endothelial cells, monocytes, fibroblasts, and
vascular smooth muscle cells, and it plays a critical part in
smooth muscle cell migration and cellular adhesion,40,41 both
of which are required for the formation of new blood vessels.
The v3-integrin is expressed on the luminal surface of
activated endothelial cells but not on mature quiescent cells.42
The utility of v3-integrintargeted nanoparticles has been
shown for the detection and characterization of angiogenesis
associated with growth factor expression,43 tumor growth,44
and atherosclerosis.45
Angiogenesis plays a critical role in plaque growth and
rupture.46,47 In regions of atherosclerotic lesions, angiogenic

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

Wickline et al

Applications of Nanotechnology

437

Figure 1. Ultrasound molecular imaging with nanoparticles. Left, top panels, Tissue factor (TF)-targeted nanoparticles binding to tissue
factor constitutively expressed on porcine endothelial cells in vitro. Bottom panels, Imaging the porcine carotid artery with 30 MHz
intravascular ultrasound (IVUS) catheter after balloon injury. Note contrast enhancement heterogeneously distributed throughout media
of vessel representing binding of TF-targeted nanoparticles to medial smooth muscle cells expressing tissue factor epitopes (left), and
no enhancement in control injured segments treated with untargeted nanoparticles. Reprinted with permission from Lanza et al and
Lippincott Williams & Wilkins.29,36 Right, top panels, Four-chamber view of dog heart with experimental thrombus before (top left) and
after (top right) intravenous delivery and binding of echogenic liposomes targeted to fibrin/fibrinogen molecules. Bottom panels,
parasternal view of clot before (left) and after (right). LV indicates left ventricle. M indicates myocardium. Reprinted with permission
from Hamilton et al and Lippincott Williams & Wilkins.33

vessels proliferate from the vasa vasorum to meet the high


metabolic demands of plaque growth.48,49 Molecular imaging
of expanded vasa vasorum in atherosclerotic lesions in
cholesterol-fed rabbits was first demonstrated for MRI by
Winter et al45 with the use of paramagnetic nanoparticles
targeted to v3-integrin expressing endothelial cells (Figure
3). Animals on a control diet exhibited no increased signal
and background was minimal. Expression of v3-integrins in
the adventitial layer and beyond was confirmed by colocalized histological staining of v3-integrin and platelet endothelial cell adhesion molecule, which is a general endothelial
marker.

Other Plaque Components


Macrophage imaging with the use of nontargeted USPIOs
was reported first by Schmitz et al19 in Watanabe rabbits and

by Ruehm et al50 in cholesterol-fed atherosclerotic rabbits.


Because macrophages are abundant in plaques throughout the
vascular tree and are well known to ingest particulate matter,
the use of superparamagnetic agents to delineate macrophages and foam cells has been pursued in both animal
models and in clinical trials.51 The demonstration of macrophage targeting in vivo in rabbits required a waiting period of
1 to 3 days to allow for both passive uptake of sufficient
numbers of particles and for blood stream clearance of the
long circulating particles. In general, the susceptibility artifacts produced extended beyond the confines of the plaque
macrophages and appeared as heterogeneously distributed
signal voids up and down the aorta.
In similar clinical trials of patients undergoing carotid
endarterectomy by Kooi et al52 and by Trivedi et al,53 USPIO
particles accumulated in the macrophages in plaques and

Figure 2. MRI of thrombi with paramagnetic nanoparticles targeted to fibrin.


Left, Thrombus formed in canine jugular
vein imaged with T1 weighted pulse
sequences at 1.5T. Right, Disrupted
carotid endarterectomy specimens incubated with fibrin-targeted nanoparticles
binding to small amounts of fibrin at
shoulders of ruptured plaque cap imaged
in vitro at 1.5T. Reprinted with permission from Lippincott Williams & Wilkins.12

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

438

Arterioscler Thromb Vasc Biol.

March 2006
Figure 3. Left: Angiogenesis induced in
cholesterol-fed rabbits. Aortic cross sections imaged at 1.5T with v3-integrin
targeted nanoparticles. Note heterogeneous distribution in aortic cross sections
(false colored contrast enhancement), but
little enhancement in nontargeted rabbits
(v3-) or rabbits on a standard diet
(Chol-). Upper right, MRI signal modeling
illustrates that picomolar (100 pM) intravoxel concentrations of perfluorocarbonbased paramagnetic nanoparticles are
required to achieve a diagnostic contrastto-noise (CNR) ratio of 5 (blue dashed
line). Lower right, Immunochemical staining
for v3-integrin at the media-adventitia
border of aorta segments. Note abundant
red-brown vascular segments. Reprinted
with permission from Lippincott Williams &
Wilkins.45

were optimally imaged as signal reductions at 24 hours after


injection. Kooi et al also noted that more contrast change was
observed for ruptured plaques than for stable plaques:
USPIO-labeled macrophages have been imaged and localized
to unstable and ruptured plaques (75% demonstrating uptake)
but not in stable lesions (only 7% showing USPIO uptake).52
Recently, Frias et al14 reported the development of recombinant paramagnetic HDL-like particles that have been shown
to enhance atherosclerotic regions in apolipoprotein
E-deficient mice. These particles are formed through the
delipidation of normal isolated human HDL particles, followed by reconstitution with phospholipids and addition of a
phospholipid-based conjugate of Gd-DTPA (15 to 20 molecules of gadolinium included in each 9 nm particle) for signal
enhancement. Nonselective accumulation in atherosclerosis
has been demonstrated.
Fayad et al54 have also demonstrated the use of other
nontargeted agents such as gadofluorine, a lipophilic chelate
of gadolinium that forms 5-nm micelles in aqueous solution,
to preferentially label the fatty cores of plaques. The small
size and lipophilic nature of this contrast agent allows it to
accumulate in lipid rich areas of plaque in cholesterol-fed
rabbits.

Stem Cell Imaging


Stem cell imaging with MRI is another emerging area that
might fit under the rubric of molecular imaging with targeted

nanoparticle contrast agents. Cells can be treated with superparamagnetic nanoparticles in vitro and then engrafted into
the selected location by local injection. The stem cells ingest
nanoparticles through endocytosis by various strategies, including coating the particles with dendrimers, transfection
agents, or antibodies/peptides,5557 which results in the intracellular accumulation of significant amounts of intact nanoparticles that then can exert a local susceptibility effect for
detection in vivo. These particles appear to be well tolerated
by cells over the long term, although the signal ultimately
dissipates as the cells divide and distribute the material or as
the particles are catabolized naturally. Along these lines,
Frank et al, 56 Bulte et al, 58 and others have demonstrated the
utility of stem cell tracking after in vitro preparation with
superparamagnetic nanoparticles. More recently, Kraitchman
et al59 demonstrated the ability to detect and track mesenchymal stem cells injected into necrotic regions of a pig heart at
1.5 T (Figure 4). Recent data indicating clinical feasibility for
MRI stem cell tracking was reported for labeled dendritic
cells injected into lymph nodes in patients with melanoma.60
Alternatively, the fluorine component of perfluorocarbonbased nanoparticles might be used to advantage for cell
imaging after particle ingestion. Our group originally demonstrated the concept of targeted nanoparticle fluorine imaging at 1.5T or 4.7T for detection of experimental thrombi or
small fibrin deposits in disrupted human carotid arteries using
nanoparticles made with perfluoroctylbromide, crown ether,

Figure 4. Stem cell labeling and imaging


with iron oxide nanoparticles. Left, Stem
cells incubated with nontargeted iron
oxide nanoparticles that undergo endocytosis are injected near the apex in a
porcine heart and imaged at 1.5T. Note
dark spot due to susceptibility effect
(arrow). Right, Iron stain of heart tissue
illustrating stem cells containing abundant nanoparticles in cytoplasmic compartment. Reprinted with permission
from Kraitchman et al59 and Lippincott
Williams & Wilkins.

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

Wickline et al

Applications of Nanotechnology

439

Figure 5. Fluorine imaging with fibrin targeted perfluorocarbon-based nanoparticles. A, Optical image of an excised human carotid endarterectomy
sample shows asymmetrical plaque distribution, with areas of fat deposition (yellow). B, Using nuclear magnetic resonance spectroscopy, the fluorine image can be converted into a false color map of the nanoparticle binding and coregistered and overlaid on the proton image (in gray scale),
corresponding to the intravoxel concentration of nanoparticles bound to fibrin epitopes. Note quantitative false color mapping scale at right illustrating quantification of signal enhancement as a nanoparticle concentration expressed in nanomoles per voxel.

or other perfluorocarbon core materials (Figure 5).7,30 Subsequently, Ahrens et al61 used a crown ether preparation of
nanoparticles to load dendritic immune cells with no loss of
viability and illustrated the use of fluorine imaging at high
field strengths (11.7 T) for tracking cells after local and
systemic injection. Again, the advantage to this approach is
that no background signal exists because there is no appreciable amount of fluorine in the body to confound the signal
from the targeted cells.

Delivering and Monitoring Therapy


The potential dual use of nanoparticles for both imaging and
site-targeted delivery of therapeutic agents to cardiovascular
disease offers great promise for individualizing therapeutics.
Image-based therapeutics with site-selective agents should
enable conclusive assurance that the drug is reaching the
intended target and a molecular effect is occurring. As an
example of this new paradigm for drug delivery, Lanza et al30
treated smooth muscle cells in culture with tissue factortargeted nanoparticles that were loaded with paclitaxel. The
smooth muscle cells were harvested from pig aortae and
constitutively expressed tissue factor epitopes in vitro. Binding of the drug-free nanoparticles to the cells yielded no
alterations in growth characteristics of the cultured cells.
When paclitaxel-loaded nanoparticles were applied to the
cells, however, specific binding elicited a substantial reduction in smooth muscle cell proliferation. Nontargeted
paclitaxel-loaded particles applied to the cells (ie, no binding
of nanoparticles to cells occurred) resulted in normal cell
proliferation, indicating that selective targeting may be a
requirement for effective drug delivery for these emulsions.
Similar behavior has been demonstrated for doxorubicincontaining particles.30 Recent reports indicate that intravenous delivery of fumagillin-loaded nanoparticles (an antiangiogenic agent) targeted to v3-integrin epitopes on vasa
vasorum in growing plaques results in marked inhibition of
plaque angiogenesis in cholesterol fed rabbits.62 Kolodgie et
al63 also used taxol-containing nontargeted albumin nanoparticles for limitation of the restenotic response after angioplasty and stent placement in experimental animals.
The unique mechanism of drug delivery for highly lipophilic agents such as paclitaxel contained within emulsions
depends on close apposition between the nanoparticle carrier

and the targeted cell membrane and has been described as


contact facilitated drug delivery.30 In contrast to liposomal
drug delivery (generally requiring endocytosis), the mechanism of drug transport in this case involves lipid exchange or
lipid mixing between the emulsion vesicle and the targeted
cell membrane,64,65 which depends on the extent and frequency of contact between 2 lipidic surfaces.30,64 The rate of
lipid exchange and drug delivery can be greatly increased by
the application of clinically safe levels of ultrasound energy
that increase the propensity for fusion or enhanced contact
between the nanoparticles and the targeted cell
membrane.64,65

Limitations
Although both molecular imaging and targeted therapeutics
are attractive subjects for clinical evaluation, the ultimate role
of these technical advances must be established in clinical
trials. To date, no solid proof of efficacy has been provided
with respect to altering courses of therapy or patient outcomes. The choice of imaging modalities also is broad and
remains to be worked out in practice, and it will depend on
cost, availability, and the specific application. As is the case
for any novel pharmacological agent undergoing clinical
trials, the use of nanoparticles also will require thorough
evaluation for pharmacokinetics, biodistribution, and toxicity.
However, in the era of molecular medicine where early
diagnosis and personalized therapeutics may reduce risk and
save lives, the promise of these novel technologies and
approaches represents a new avenue to disease control that
appears extraordinarily compelling.

Conclusion
The combination of targeted drug delivery and molecular
imaging with MRI has the potential to revolutionize the field
of cardiology, as well as many other fields. Drug delivery
agents that are also quantifiable at the targeted site based on
imaging readouts may ultimately permit serial characterization of the molecular epitope expression and confirmation of
therapeutic efficacy, thereby promoting truly personalized
medical regimens. Rapid developments in genomics, molecular biology, and nanotechnology have energized the multidisciplinary field of molecular imaging, and we anticipate

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

440

Arterioscler Thromb Vasc Biol.

March 2006

that clinical applications are at hand for these powerful


agents.

References
1. Britz-Cunningham SH, Adelstein SJ. Molecular targeting with radionuclides: state of the science. J Nucl Med. 2003;44:19451961.
2. Tsien RY. Imagining imagings future. Nat Rev Mol Cell Biol. 2003;
(suppl):SS16-SS21.
3. Lanza G, Wickline S. Targeted ultrasonic contrast agents for molecular
imaging and therapy. Prog Cardiovasc Dis. 2001;44:1331.
4. Lanza GM, Wickline SA. Targeted ultrasonic contrast agents for
molecular imaging and therapy. Curr Prob Cardiol. 2003;28:625 653.
5. Wickline SA, Lanza GM. Molecular imaging, targeted therapeutics, and
nanoscience. J Cell Biochem. 2002;39:90 97.
6. Wickline SA, Lanza GM. Nanotechnology for molecular imaging and
targeted therapy. Circulation. 2003;107:10921095.
7. Morawski AM, Winter PM, Yu X, Fuhrhop R, Scott M, Hockett F,
Robertson JD, Gaffney PJ, Lanza GM, Wickline SA. Quantitative
magnetic resonance immunohistochemistry with ligand-targeted 19F
nanoparticles. Magn Reson Med. 2004;52:12551262.
8. Buxton DB, Lee SC, Wickline SA, Ferrari M, for the Working Group
Members. Recommendations of the National Heart, Lung, and Blood
Institute Nanotechnology Working Group. Circulation. 2003;108:
27372742.
9. Demos SM, Alkan-Onyuksel H, Kane BJ, Ramani K, Nagaraj A, Greene
R, Klegerman M, McPherson DD. In vivo targeting of acoustically
reflective liposomes for intravascular and transvascular ultrasonic
enhancement. J Am Coll Cardiol. 1999;33:867 875.
10. Sipkins DA, Cheresh DA, Kazemi MR, Nevin LM, Bednarski MD, Li
KC. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted
magnetic resonance imaging. Nature Medicine. 1998;4:623 626.
11. Lanza GM, Winter P, Caruthers S, Schmeider A, Crowder K, Morawski
A, Zhang H, Scott MJ, Wickline SA. Novel paramagnetic contrast agents
for molecular imaging and targeted drug delivery. Curr Pharm Biotechnol. 2004;5:495507.
12. Flacke S, Fischer S, Scott MJ, Fuhrhop RJ, Allen JS, McLean M, Winter
P, Sicard GA, Gaffney PJ, Wickline SA, Lanza GM. Novel MRI contrast
agent for molecular imaging of fibrin: implications for detecting vulnerable plaques. Circulation. 2001;104:1280 1285.
13. Deleted in proof.
14. Frias JC, Williams KJ, Fisher EA, Fayad ZA. Recombinant HDL-like
nanoparticles: a specific contrast agent for MRI of atherosclerotic
plaques. J Am Chem Soc. 2004;126:16316 16317.
15. Li H, Gray BD, Corbin I, Lebherz C, Choi H, Lund-Katz S, Wilson JM,
Glickson JD, Zhou R. MR and fluorescent imaging of low-density
lipoprotein receptors. Acad Radiol. 2004;11:12511259.
16. Hawker CJ, Wooley KL. The convergence of synthetic organic and
polymer chemistries. Science. 2005;309:1200 1205.
17. Kobayashi H, Kawamoto S, Jo SK, Bryant HL Jr, Brechbiel MW, Star
RA. Macromolecular MRI contrast agents with small dendrimers: pharmacokinetic differences between sizes and cores. Bioconjug Chem. 2003;
14:388 394.
18. Sato N, Kobayashi H, Hiraga A, Saga T, Togashi K, Konishi J, Brechbiel
MW. Pharmacokinetics and enhancement patterns of macromolecular
MR contrast agents with various sizes of polyamidoamine dendrimer
cores. Magn Reson Med. 2001;46:1169 1173.
19. Schmitz SA, Coupland SE, Gust R, Winterhalter S, Wagner S, Kresse M,
Semmler W, Wolf KJ. Superparamagnetic iron oxide-enhanced MRI of
atherosclerotic plaques in Watanabe hereditable hyperlipidemic rabbits.
Invest Radiol. 2000;35:460 471.
20. Koch AM, Reynolds F, Merkle HP, Weissleder R, Josephson L. Transport
of surface-modified nanoparticles through cell monolayers. Chembiochem. 2005;6:337345.
21. Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, Rivera B, Price RE,
Hazle JD, Halas NJ, West JL. Nanoshell-mediated near-infrared thermal
therapy of tumors under magnetic resonance guidance. Proc Natl Acad
Sci U S A. 2003;100:13549 13554.
22. Loo C, Lin A, Hirsch L, Lee MH, Barton J, Halas N, West J, Drezek R.
Nanoshell-enabled photonics-based imaging and therapy of cancer.
Technol Cancer Res Treat. 2004;3:33 40.
23. Cherukuri P, Bachilo SM, Litovsky SH, Weisman RB. Near-infrared
fluorescence microscopy of single-walled carbon nanotubes in phagocytic
cells. J Am Chem Soc. 2004;126:15638 15639.

24. Barone PW, Baik S, Heller DA, Strano MS. Near-infrared optical sensors
based on single-walled carbon nanotubes. Nat Mater. 2005;4:86 92.
25. Akerman ME, Chan WC, Laakkonen P, Bhatia SN, Ruoslahti E. Nanocrystal targeting in vivo. Proc Natl Acad Sci U S A. 2002;99:
1261712621.
26. Chen L, Zurita AJ, Ardelt PU, Giordano RJ, Arap W, Pasqualini R.
Design and validation of a bifunctional ligand display system for receptor
targeting. Chem Biol. 2004;11:10811091.
27. Gao X, Nie S. Quantum dot-encoded beads. Methods Mol Biol. 2005;
303:6171.
28. Lanza GM, Wallace KD, Scott MJ, Cacheris WP, Sheehan CK,
Abendschein DR, Christy DH, Sharkey AM, Miller JG, Gaffney PJ,
Wickline SA. A novel site-targeted ultrasonic contrast agent with broad
biomedical application. Circulation. 1996;95:3334 3340.
29. Lanza GM, Abendschein DR, Hall CS, Scott MJ, Scherrer DE, Houseman
A, Miller JG, Wickline SA. In vivo molecular imaging of stretch-induced
tissue factor in carotid arteries with ligand-targeted nanoparticles. J Am
Soc Echocardiogr. 2000;13:608 614.
30. Lanza GM, Yu X, Winter PM, Abendschein DR, Karukstis KK, Scott MJ,
Fuhrhop RJ, Scherer DE, Wickline SA. Targeted antiproliferative drug
delivery to vascular smooth muscle cells with an MRI nanoparticle
contrast agent: implications for rational therapy of restenosis. Circulation.
2002;106:28422847.
31. Oltrona L, Speidel CM, Recchia D, Wickline SA, Eisenberg PR,
Abendschein DR. Inhibition of tissue factor-mediated coagulation
markedly attenuates stenosis after balloon-induced arterial injury in
minipigs. Circulation. 1997;96:646 652.
32. Morawski AM, Winter PM, Crowder KC, Caruthers SD, Fuhrhop RW,
Scott MJ, Robertson JD, Abendschein DR, Lanza GM, Wickline SA.
Targeted nanoparticles for quantitative imaging of sparse molecular
epitopes with MRI. Magnc Reson Med. 2004;51:480 486.
33. Hamilton A, Huang SL, Warnick D, Stein A, Rabbat M, Madhav T, Kane
B, Nagaraj A, Klegerman M, MacDonald R, McPherson D. Left ventricular thrombus enhancement after intravenous injection of echogenic
immunoliposomes: studies in a new experimental model. Circulation.
2002;105:27722778.
34. Hamilton AJ, Huang SL, Warnick D, Rabbat M, Kane B, Nagaraj A,
Klegerman M, McPherson DD. Intravascular ultrasound molecular
imaging of atheroma components in vivo. J Am Coll Cardiol. 2004;43:
453 460.
35. Kelly KA, Allport JR, Tsourkas A, Shinde-Patil VR, Josephson L,
Weissleder R. Detection of vascular adhesion molecule-1 expression
using a novel multimodal nanoparticle. Circ Res. 2005;96:327336.
36. Lanza GM, Trousil RL, Wallace KD, Rose JH, Hall CS, Scott MJ, Miller
JG, Eisenberg PR, Gaffney PJ, Wickline SA. In vitro characterization of
a novel, tissue-targeted ultrasonic contrast system with acoustic
microscopy. J Acoust Soc Am. 1998;104:36653672.
37. Botnar RM, Buecker A, Wiethoff AJ, Parsons EC Jr, Katoh M,
Katsimaglis G, Weisskoff RM, Lauffer RB, Graham PB, Gunther RW,
Manning WJ, Spuentrup E. In vivo magnetic resonance imaging of
coronary thrombosis using a fibrin-binding molecular magnetic resonance
contrast agent. Circulation. 2004;110:14631466.
38. Spuentrup E, Fausten B, Kinzel S, Wiethoff AJ, Botnar RM, Graham PB,
Haller S, Katoh M, Parsons EC Jr, Manning WJ, Busch T, Gunther RW,
Buecker A. Molecular magnetic resonance imaging of atrial clots in a
swine model. Circulation. 2005;112:396 399.
39. Kerr JS, Mousa SA, Slee AM. Alpha(v)beta(3) integrin in angiogenesis
and restenosis. Drug News Perspect. 2001;14:143150.
40. Bishop GG, McPherson JA, Sanders JM, Hesselbacher SE, Feldman MJ,
McNamara CA, Gimple LW, Powers ER, Mousa SA, Sarembock IJ.
Selective alpha(v)beta(3)-receptor blockade reduces macrophage infiltration and restenosis after balloon angioplasty in the atherosclerotic
rabbit. Circulation. 2001;103:1906 1911.
41. Corjay MH, Diamond SM, Schlingmann KL, Gibbs SK, Stoltenborg JK,
Racanelli AL. Alphavbeta3, alphavbeta5, and osteopontin are coordinately upregulated at early time points in a rabbit model of neointima
formation. J Cell Biochem. 1999;75:492504.
42. Brooks PC, Stromblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA.
Antiintegrin alpha v beta 3 blocks human breast cancer growth and
angiogenesis in human skin. J Clin Invest. 1995;96:18151822.
43. Anderson SA, Rader RK, Westlin WF, Null C, Jackson D, Lanza CM,
Wickline SA, Kotyk JJ. Magnetic resonance contrast enhancement of
neovasculature with alpha(v)beta(3)-targeted nanoparticles. Magn Reson
Med. 2000;44:433 439.

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

Wickline et al
44. Winter PM, Caruthers SD, Kassner A, Harris TD, Chinen LK, Allen JS,
Lacy EK, Zhang H, Robertson JD, Wickline SA, Lanza GM. Molecular
imaging of angiogenesis in nascent Vx-2 rabbit tumors using a novel
alpha(nu)beta3-targeted nanoparticle and 1.5 tesla magnetic resonance
imaging. Cancer Res. 2003;63:5838 5843.
45. Winter PM, Morawski AM, Caruthers SD, Fuhrhop RW, Zhang HY,
Williams TA, Allen JS, Lacy EK, Robertson JD, Lanza GM, Wickline
SA. Molecular imaging of angiogenesis in early-stage atherosclerosis
with alpha(v)beta(3)-Integrin-targeted nanoparticles. Circulation. 2003;
108:2270 2274.
46. Moulton KS, Heller E, Konerding MA, Flynn E, Palinski W, Folkman J.
Angiogenesis inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque growth in apolipoprotien E-defficient mice. Circulation. 1999;99:1726 1732.
47. Tenaglia AN, Peters KG, Sketch MH Jr, Annex BH. Neovascularization
in atherectomy specimens from patients with unstable angina: implications for pathogenesis of unstable angina. Am Heart J. 1998;135:
10 14.
48. Wilson SH, Hermann J, Lermann LO, Holmes DR Jr, Napoli C, Ritman
EL, Lerman A. Simvastatin preserves the structure of coronary adventitial
vasa vasorum in experimental hypercholesterolemia independent of lipid
lowering. Circulation. 2002;105:415 418.
49. Zhang Y, Cliff WJ, Schoefl GI, Higgins G. Immunohistochemical study
of intimal microvessels in coronary atherosclerosis. Am J Pathol. 1993;
143:164 172.
50. Ruehm SG, Corot C, Vogt P, Kolb S, Debatin JF. Magnetic resonance
imaging of atherosclerotic plaque with ultrasmall superparamagnetic particles of iron oxide in hyperlipidemic rabbits. Circulation. 2001;103:
415 422.
51. Corot C, Petry KG, Trivedi R, Saleh A, Jonkmanns C, Le Bas J-F, Blezer
E, Rausch M, Brochet B, Foster-Gareau P, Baleriaux D, Gaillard S,
Dousset V. Macrophage imaging in central nervous system and in carotid
atheroscerotic plaque using ultrasmall superparamagnetic iron oxide in
magnetic resonance imaging. Invest Radiol. 2004;39:619 625.
52. Kooi ME, Cappendijk VC, Cleutjens KB, Kessels AG, Kitslaar PJ,
Borgers M, Frederik PM, Daemen MJ, van Engelshoven JM. Accumulation of ultrasmall superparamagnetic particles of iron oxide in human
atherosclerotic plaques can be detected by in vivo magnetic resonance
imaging. Circulation. 2003;107:24532458.
53. Trivedi RA, U-King-Im JM, Graves MJ, Cross JJ, Horsley J, Goddard
MJ, Skepper JN, Quartey G, Warburton E, Joubert I, Wang L, Kirkpatrick
PJ, Brown J, Gillard J. In vivo detection of macrophages in human carotid
atheroma: temporal dependence of ultrasmall superparamagnetic particles
of iron oxide-enhanced MRI. Stroke. 2004;35:16311635.
54. Sirol M, Itskovich VV, Mani V, Aguinaldo JG, Fallon JT, Misselwitz B,
Weinmann HJ, Fuster V, Toussaint JF, Fayad ZA. Lipid-rich athero-

55.

56.

57.

58.
59.

60.

61.
62.

63.

64.

65.

Applications of Nanotechnology

441

sclerotic plaques detected by gadofluorine-enhanced in vivo magnetic


resonance imaging. Circulation. 2004;109:2890 2896.
Arbab AS, Bashaw LA, Miller BR, Jordan EK, Bulte JW, Frank JA.
Intracytoplasmic tagging of cells with ferumoxides and transfection agent
for cellular magnetic resonance imaging after cell transplantation:
methods and techniques. Transplantation. 2003;76:11231130.
Frank JA, Miller BR, Arbab AS, Zywicke HA, Jordan EK, Lewis BK,
Bryant LH Jr, Bulte JW. Clinically applicable labeling of mammalian and
stem cells by combining superparamagnetic iron oxides and transfection
agents. Radiology. 2003;228:480 487.
Funovics MA, Kapeller B, Hoeller C, Su HS, Kunstfeld R, Puig S,
Macfelda K. MR imaging of the her2/neu and 9.2.27 tumor antigens using
immunospecific contrast agents. Magn Reson Imaging. 2004;22:
843 850.
Bulte JW, Kraitchman DL. Monitoring cell therapy using iron oxide MR
contrast agents. Curr Pharm Biotechnol. 2004;5:567584.
Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ,
Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of
mesenchymal stem cells in myocardial infarction. Circulation. 2003;107:
2290 2293.
de Vries IJM, Lesterhuis WJ, Barentsz JO, Verdijk P, van Krieken JH,
Boerman OC, Oyen WJG, Bonenkamp JJ, Boezeman JB, Adema GJ,
Bulte JWM, Scheenen TWJ, Punt CJA, Heerschap A, Figdor CG.
Magnetic resonance tracking of dendritic cells in melanoma patients for
monitoring of cellular therapy. Nat Biotech. 2005;23:14071413.
Ahrens ET, Flores R, Xu H, Morel PA. In vivo imaging platform for
tracking immunotherapeutic cells. Nat Biotechnol. 2005;23:983987.
Winter PM, Morawski AM, Caruthers SD, Harris TD, Fuhrhop RW,
Zhang HY, Allen JS, Lacy EK, Williams TA, Wickline SA, Lanza GM.
Antiangiogenic therapy of early atherosclerosis with paramagnetic
alpha(v)beta(3)-integrin-targeted fumagillin nanoparticles. J Am Coll
Cardiol. 2004;43:322A323A.
Kolodgie FD, John M, Khurana C, Farb A, Wilson PS, Acampado E,
Desai N, Soon-Shiong P, Virmani R. Sustained reduction of in-stent
neointimal growth with the use of a novel systemic nanoparticle paclitaxel. Circulation. 2002;106:11951198.
Crowder KC, Hughes MS, Marsh JN, Scott MJ, Fuhrhop RW, Lanza GM,
Wickline SA. Augmented and selective delivery of liquid perfluorocarbon
nanoparticles to melanoma cells with noncavitational ultrasound. Presented at the 2003 IEEE Symposium on Ultrasonics, Honolulu, Hawaii, 2003.
Crowder KC, Hughes MS, Marsh JN, Barbieri AM, Fuhrhop R, Lanza
GM, Wickline SA. Sonic activation of molecularly-targeted nanoparticles
accelerates transmembrane lipid delivery to cancer cells through contactmediated mechanisms: implications for enhanced local drug delivery.
Ultrasound Med Biol. 2005;31:16931700.

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

Applications of Nanotechnology to Atherosclerosis, Thrombosis, and Vascular Biology


Samuel A. Wickline, Anne M. Neubauer, Patrick Winter, Shelton Caruthers and Gregory Lanza
Arterioscler Thromb Vasc Biol. 2006;26:435-441; originally published online December 22,
2005;
doi: 10.1161/01.ATV.0000201069.47550.8b
Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272
Greenville Avenue, Dallas, TX 75231
Copyright 2005 American Heart Association, Inc. All rights reserved.
Print ISSN: 1079-5642. Online ISSN: 1524-4636

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://atvb.ahajournals.org/content/26/3/435

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the
Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for
which permission is being requested is located, click Request Permissions in the middle column of the Web
page under Services. Further information about this process is available in the Permissions and Rights
Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online
at:
http://atvb.ahajournals.org//subscriptions/

Downloaded from http://atvb.ahajournals.org/ by guest on January 8, 2015

You might also like