You are on page 1of 7

Review

Emerging applications of
nanomedicine for the diagnosis and
treatment of cardiovascular diseases
Biana Godin1, Jason H. Sakamoto1, Rita E. Serda1, Alessandro Grattoni1,
Ali Bouamrani1 and Mauro Ferrari1,2,3
1
The University of Texas Health Science Center at Houston (UTHSC-H), Department of NanoMedicine and Biomedical Engineering,
1825 Pressler, Suite 537, Houston, TX 77030, USA
2
The University of Texas MD Anderson Cancer Center, Department of Experimental Therapeutics, Unit 422, 1515 Holcombe Blvd.,
Houston, TX 77030
3
Rice University, Department of Bioengineering, Houston, TX 77005, USA

Nanomedicine is an emerging field that utilizes nano- or its vital components; (ii) man-made nature and unique
technology concepts for advanced therapy and diagnos- characteristics of new material that arise due to its nano-
tics. This convergent discipline merges research areas scopic size [1,2]. Nanotechnology represents a convergent
such as chemistry, biology, physics, mathematics and discipline in which the margins separating various
engineering. It therefore bridges the gap between mol- research areas, such as chemistry, biology, physics, math-
ecular and cellular interactions, and has the potential to ematics and engineering, become blurred. Cardiovascular
revolutionize medicine. This review presents recent nanomedicine is likely to face and address current chal-
developments in nanomedicine research poised to have lenges in CVDs. It will also improve detection and therapy
an important impact on the treatment of cardiovascular by advancing the ex-vivo and in-vivo detection and imaging
disease. This will occur through improvement of the of biomarkers, as well as by directed/improved delivery of
diagnosis and therapy of cardiovascular disorders as drugs and tissue regeneration [3].
atherosclerosis, restenosis and myocardial infarction. We will summarize and discuss recent developments in
Specifically, we discuss the use of nanoparticles for nanotechnology for the detection and therapy of CVDs
molecular imaging and advanced therapeutics, specially focusing on nanoparticles, specially designed therapeutic
designed drug eluting stents and in vivo/ex vivo early and tissue regeneration devices, and in-vivo/ex-vivo early
detection techniques. detection techniques (Figure 1).

Introduction Nanoparticles for advanced diagnostics and treatment


Despite significant clinical advancements, cardiovascular of CVDs
diseases (CVDs), which include various disorders of blood Various nanoparticle-based drug-delivery systems have
vasculature and the heart, as well as stroke, remain the been and are being developed for applications in cancer,
leading cause of death in the USA. Based on statistics CVDs and other conditions. These have different features
provided by the National Institutes of Health (NIH) and and multiple-functionalities [2,4–6], exhibiting differences
the American Heart Association, 80 million people in the in: (i) sizes, ranging from few tens of nanometers (e.g.
USA suffer from CVDs and >35% of deaths in North dendrimers, gold and iron-oxide nanoparticles) to few hun-
America are attributed to CVDs (http://www.nhlbi.nih.gov; dreds of nanometers (e.g. polymeric and lipid-based
www.americanheart.org). The last breakthrough technol- particles) to micron-sized particles; (ii) shapes, from the
ogy to impact CVDs occurred over a decade ago with the classical spherical particles to discoidal, hemispherical,
introduction of the coronary stent by Palmaz and Schatz, cylindrical and conical; (iii) surface functionalizations, with
which was approved by the Food and Drug Administration a broad range of electrostatic charges and bio-molecule
(FDA) in 1994. Since then, medicine has relied upon new conjugations. Use of nanocarriers for these conditions allows
‘blockbuster’ therapeutics (e.g. statins, beta-blockers, for local or directed delivery, prolonged effect of the drug,
diuretics) and refinements of surgical procedures such as facilitated delivery into target cells, and reduction of the
percutaneous transluminal coronary angioplasty (PTCA), shear effects of blood flow. During the course of develop-
coronary artery bypass grafts (CABG) and stenting to treat ment, atherosclerotic plaques and the neointima display
CVDs. However, current methods for early detection and various stage-specific molecules that can be used as target-
advanced therapies of CVDs are limited, and their effi- ing moieties in CVDs (avb3-integrin, VCAM-1, YIGSR).
ciency in preventing CVDs questionable.
By definition, nanotechnology involves two inter-related Nanoparticles for advanced diagnostics of CVDs
constituents: (i) nanoscale dimensions of the whole system One of the major foci of the application of nanotechnology
for cardiovascular research has been the directed imaging
Corresponding author: Ferrari, M. (Mauro.Ferrari@uth.tmc.edu). and therapy of atherosclerosis, restenosis and over cardi-
0165-6147/$ – see front matter . Published by Elsevier Ltd. doi:10.1016/j.tips.2010.01.003 Available online 19 February 2010 199
Review Trends in Pharmacological Sciences Vol.31 No.5

Figure 1. Nanotechnology approaches for the advanced diagnosis and treatment of CVDs (schematic): Nanoparticles for (A) multimodal image contrast and (B) improved
treatment of CVDs can be targeted to immune cells or specific ligands on the inflamed endothelium of the atherosclerotic plaque; (C) in-vivo sensors implanted in the
pericardial region or on one of the main blood vessels and techniques for ex-vivo detection of biomarkers; (D) nanostructured drug/nanoparticle-eluting stents.

ovascular conditions. Nanoscale contrast agents have application and the weight of sensitivity, specificity and
emerged as multifaceted modalities that can be used to artifact minimization such as bright contrast originating
identify and characterize the early stages of disease before from perivascular fat on atherosclerotic plaque images [9].
the development of pathological manifestations. Contrast- Nanoparticles have size-dependent imaging properties.
generating nanomaterials for cardiovascular imaging in- For example, intrinsically fluorescent nanoparticles
clude fluorescent, radioactive, paramagnetic, superpara- (‘quantum dots’) emit light over a broad range from
magnetic, electron-dense and light-scattering particles near-ultraviolet to mid-infrared. Increases in particle size
(Table 1). are positively correlated to increases in emission wave-
Cardiovascular imaging by magnetic resonance imaging length [12]. Microparticle-based contrast agents for ima-
(MRI) requires powerful magnetic fields and radiofre- ging include porous silicon particles that encapsulate
quency waves to generate images of internal structures. abundant IO nanoparticles in a single unit for enhanced
Energy changes in tissue in response to the magnetic field contrast [13]. These multistage particles (as well as other
are detected and the presence of contrast agents amplifies particles that are candidates for phagocytosis by macro-
these changes. Three MRI techniques are T1, T2*, and off- phages) offer imaging of inflamed areas where macro-
resonance [7]. Off-resonance imaging relies on pulse phages accumulate (e.g. atherosclerotic plaques) [14].
sequences that excite and refocus off-resonance water, The feasibility of multi-modal imaging with nanoparticles
leading to positive contrast [8]. Paramagnetic contrast containing multiple contrast agents such as 18F-cross-
agents such as gadolinium chelates (e.g. gadolinium- linked iron oxide (18F-CLIO) nanoparticles [15,16] has
diethylenetriaminopentaacetic acid (DTPA)) enhance T1 been demonstrated. 18F-CLIO agents consist of a cross-
contrast, resulting in bright contrast in MR images [9]. linked dextran shell formed on a superparamagnetic IO
Manganese nanoparticles represent another, recently core and functionalized with the radionuclide 18F. These
introduced, example of T1-enhancing contrast agent particles can be detected with positron emission tomogra-
[10,11]. Superparamagnetic contrast agents such as iron phy (PET), fluorescence molecular tomography, and MRI.
oxide (IO) nanoparticles, predominately magnetite (Fe2O3/ Chen et al. [17] examined in-vivo MRI contrast of vulner-
Fe3O4), typically enhance T2 contrast and produce dark able plaque high-density lipoprotein (rHDL) nanoparticles
contrast [8]. The technique of choice is dependent upon the enriched with Gd-based amphiphiles and a targeting

Table 1. Examples of contrast-enhancing nanoparticles for imaging of CVDs


Category Agent (examples) Imaging techniques References
Fluorescent Quantum dots Fluorescence tomography [12]
18
Radioactive F CLIO, 111In nanoparticles PET, SPECT [15]
Paramagnetic Gd-DTPA MRI [17]
Superparamagnetic Iron oxide nanoparticles MRI [21]
Electron-dense Gold or I-based nanoparticles CT [18,19,78,79]
Light-scattering Gold nanoshells Optical coherent tomography [80]
Photoacoustic Colloidal nanobeacons Photoacoustic tomography [81]
Multimodal Copper-CLIO PET, MRI, NIRF [16]
Perfluorocarbon nanoparticles MRI, Molecular imaging [33,82]
CLIO: cross-linked iron oxide; PET: positron emission tomography; SPECT: single-photon emission computed tomography; Gd-DTPA: gadolinium chelated with diethylene-
triamine pentetic acid; MRI: magnetic resonance imaging; CT: computed tomography; NIRF: near-infrared fluorescence.

200
Review Trends in Pharmacological Sciences Vol.31 No.5

moiety to intra-plaque macrophages (apolipoprotein E- well as tissue factor, are targets for imaging arterial
derived lipopeptide, P2fA2). Data showed a significant thrombi by ultrasound [25] and MRI [26]. One example
enhancement in MRI signal of the atherosclerotic wall of angiogenesis targeting is the use of nanoparticles con-
24 h after the 50 mmol Gd/kg injection of rHDL-P2fA2 jugated to ligands that specifically interact with avb3-
relative to administration of rHDL (90% vs 53% enhance- integrin [27].
ment, respectively).
An interesting set of studies explored the use of nano- Therapeutic and theranostic nanoparticles
liposomes as carriers for contrast agents such as iodine for Various nanotechnology applications are being investi-
MRI and computed tomography (CT) [18,19]. These sys- gated for the treatment of atherosclerosis and restenosis,
tems were shown to efficiently prevent a rapid clearance of including nanocarriers for drug delivery (Figure 1B) and
the contrast agent from the body, significantly improving devices such as mechanical stents, possessing nanoscale
capabilities of the total blood pool and cardiac imaging in components (Figure 1D; see below). Among the drugs used
animal models. For example, with improved liposomal to prevent restenosis are cytotoxics that inhibit the growth
formulation of iodine, time–attenuation curves showed of smooth muscle cell (e.g. paclitaxel, cytarbine, etoposides,
an initial enhancement of about 900 H in the aorta doxorubicin), platelet-derived growth factor (PDGF) recep-
and plateau levels of 800 H were achieved after 2 h, tor antagonists (e.g. tyrphostins), inhibitors of the inflam-
indicating a high iodine concentration in the blood pool. matory response/immunomodulators (e.g. steroids,
These blood levels of liposomal iodine enabled excellent bisphosphonates, cyclosporine A), and antibiotics (e.g.
contrast discrimination between the myocardium and fumagillin). Other promising therapeutics affect specific
blood in the right and left ventricles, aorta, pulmonary gene targets responsible for thrombosis or intimal hyper-
trunk, and inferior vena cava with substantially lower plasia (e.g. prostacyclin synthase and thymidine kinase).
liver and spleen contrast, as would be expected from the In the case of genetic materials and other biomolecules,
delayed clearance of polyethylene glycol (PEGylated) lipo- their encapsulation in nanoparticles provides protection
somal iodine formulation via the reticulo-endothelial sys- from enzymatic degradation and allows for prolonged
tem. The long residence time at stable, high opacity makes release profiles. These therapeutic strategies have been
liposomal iodine a promising effective micro-CT agent for thoroughly reviewed. [28,29].
contrast enhancement within sub-millimeter vessels, and The main nanocarrier classes investigated as thera-
no significant renal clearance [18,19] making CT another peutic and ‘theranostic’ (i.e. particles integrating diagnos-
emerging field where nanoparticles have been shown to tic imaging and therapeutic components) agents for
increase imaging contrast [20]. restenosis are liposomes with different surface character-
FDA-approved nanoparticles for imaging are limited to istics, polymeric nanoparticles and micelles, perfluorocar-
three IO formulations (AMI-121 (FerumoxsilTM), OMP50, bon nano-emulsions and cross-linked iron oxide (CLIO)
and AMI-25 (FeridexTM)) targeted to the gastrointestinal particles conjugated to therapeutic molecules [30–34].
tract, liver and spleen. Injection of high doses of iron was Examples of these classes are given in Table 2.
shown to be non-toxic in the nanoparticle formulation due Theranostic agents have gained much recent interest as
to slow release of free iron and assimilation into iron- a valuable advance for drug delivery [35]. This strategy for
containing substances [21]. CVD applications is still in its infancy, but it has numerous
Targets for imaging of atherosclerotic plaques include potential advantages which are being extensively investi-
endothelia, macrophages [22], fibrin [23], collagen III [24], gated in cancer nanomedicine. Combining a diagnostic
and markers of angiogenesis (Figure 1A). Fibrin deposition imaging moiety with a targeted therapeutic nanoparticle
is one of the earliest signs of plaque rupture, and fibrin, as allows for precise, temporal and spatial monitoring of the

Table 2. Examples of nanocarriers for CVD therapy


Nanocarrier Example of agent Experimental model Outcomes References
Neutral liposomes Bisphosphonates Injured carotid artery in rats Macrophage depletion, [30]
(clodronate, alendronate) reduced inflammation
Cationic liposomes Chloramphenicol acetyl Balloon-injured artery in Increased expression [31,32]
transferase (CAT) encoding Yorkshire pigs, local delivery of CAT
gene [31]
Vascular endothelial Clinical trial, patients with Significant improvement
growth factor (VEGF) 60–99% stenosis in major in myocardial perfusion
encoding viral vector arteries, local delivery
through catheter
Hemaglutin virus of Japan Tissue factor pathway Iliac artery of hyperlipidemic Reduction of intimal [83]
(HVJ) liposomes inhibitor gene rabbits after angioplasty. hyperplasia
Local delivery through catheter.
Perfluorocarbon nanoparticles Surface bound Human plasma clots, In-vitro fibrinolysis, [82]
streptokinase, a3b hyperlipidemic animals theranostic in vivo
integrins, others
Polyelectrolyte nanoparticles Gene encoding for Rat carotid artery High transfection efficiency [84]
(RNA or polyvinyl sulfate with urokinase plasminogen
polyethylene imine/DNA complex) activator
Polymeric (PLA or PLGA) nanoparticles AG-1295 and AGL-2043 Balloon-injured Inhibition of restenosis [85,86]
carotid artery in rats

201
Review Trends in Pharmacological Sciences Vol.31 No.5

therapeutic agent as well as treatment outcomes. The detection approach) only moderately improves standard
imaging capabilities of theranostic nanoparticles can serve assessment. There is, therefore, an urgent need for tech-
to verify delivery of an active compound to its intended site nological advances in biomarker strategies to improve the
of action; monitor and quantify the efficacy of the thera- detection of markers and to discover new biomarkers.
peutics at the molecular or cellular level; design dosing In medical applications, the use of nanotechnology pre-
regiments; and identify the population of responders/non- senting unique physical and chemical properties has the
responders for a specific therapy. For example, a prolonged potential to dramatically advance diagnostic methods and
anti-angiogenesis therapy was reported using theranostic provide innovative devices for more efficient molecular
avb3-integrin-targeted paramagnetic nanoparticles in detection. Tunable nanoporous materials have been used
hyperlipidemic animals [33,36,37]. MRI data showing a to selectively harvest low-molecular-weight proteins, pro-
reduction of 50–75% in the neovascular signal for 3 weeks viding a unique opportunity to detect and identify new
corresponded well with histological evaluation, pointing circulating biomarkers after fractionation of body fluids
toward the potential of this strategy for efficient anti- [47,48]. Nanowires offer great potential for diagnosis by
angiogenic therapy and simultaneously evaluating plaque measuring pH variations or detecting trace amounts of
stability. Another example is a system based on fibrin- biological and chemical species [49]. Nanocantilevers can
coated perfluorocarbon nanoparticles which can be used for be used as multiple label-free assays for the detection of
acoustic or MRI imaging with targeted thrombolysis [38]. DNA species or circulating protein biomarkers [50].
There is also the opposite side of interaction of nano- A high-specificity immunoassay-based diagnostics
particles with the walls of blood vessel. The vascular device combining impedimetric analysis nanoelectrodes
endothelium could be a barrier and unwanted target for and microfluidics has been developed to measure the con-
nanoparticles to be delivered to other organs [39]. In this centration of D-dimer (a recognized biomarker of increased
respect, CVDs may affect the transport of nanoparticles blood clotting activity in deep-vein thrombosis (DVT)) in
across vessel walls, organelle-targeted delivery of nano- whole blood [51]. This device could improve the accuracy
particles, and other effects of nanoparticles on vessel cells. and reliability of early assessment of patients at risk of
These factors should be taken into account in future nano- DVT. Other ‘lab-on-a-chip’ approaches have been devel-
medical research. oped in combination with nanotechnology to improve the
sensitivity and accuracy of biomarker detection. Associ-
Devices for ex-vivo and in-vivo early detection of CVD ated with analysis by electrocardiography (ECG), a saliva-
indicators based biomarker test within a lab-on-a-chip platform
Along with the development and adoption of novel strat- exceeded the screening capacity of ECG alone, providing
egies for treatment and prevention of CVDs, efforts are rapid screening for acute myocardial infarction (AMI) [52].
being spent to apply nanotechnologies for ex-vivo and in- Another study demonstrated the use of a rapid fluoro-
vivo detection of CVD signals (Figure 1C). The ability to phore-mediated immuno-sensing system for simultaneous
monitor for precursor signals of CVDs could potentially quantification of four cardiac markers in AMI patients.
reduce the many fatalities associated with the diseases. This technology is based on a micro-electro-mechanical
For example, monitoring thrombotic or hemorrhagic system (MEMS) and nanoparticle reagents that increase
events could facilitate the diagnosis and treatment of detection sensitivity [53].
stroke and embolisms. Measurement of variations in the Isoforms of troponins are structural proteins that are
pressure and flow of blood, biomolecule concentration or unique to cardiac myocytes (cardiac troponin I and T).
ion concentration could provide insights in the understand- These proteins are tissue-specific, and the immuno-detec-
ing of cardiovascular events. tion of their cardiac forms has become a standard in the
diagnosis of myocardial infarction [54]. A clinical report
Nanotechnology for the ex-vivo harvesting and using a novel ultra-sensitive nanoparticle assay for cardiac
detection of biomarkers troponin I demonstrated the ability to detect pg/mL con-
Identification of biomarkers provides a powerful approach centrations of the protein in serum, revealing a significant
for screening, diagnosis, prognosis and therapeutic increase of the sensitivity of detection and providing a
monitoring [40]. Addressing the underlying causes of promising earlier detection of myocardial injury [55].
CVDs and improving the detection of early-stage disease Using engineered viral nanoparticles combining troponin
permitd early intervention with more efficient disease antibodies and nickel nanohairs, a different study reported
management and a significant reduction in premature a detection limit of troponin in human serum with 6–7
mortality. Development of high-performance ‘point of care’ orders of magnitude lower than conventional immunoas-
methods will improve the prognosis for CVD patients by says [56].
obtaining more sensitive, more specific, and faster assess-
ment of diagnostic markers. Several authors have reported In-vivo sensors for CVDs
an interest in using individual biomarkers of CVDs to Myocardial ischemia is the deadliest form of CVD. It affects
better identify high-risk populations. These markers in- millions of people and causes many fatalities. Several
clude C-reactive protein [41], B-type natriuretic peptide studies have focused on the development of nanosensors
[42], fibrinogen [43], D-dimer [44], and homocysteine [45]. for in-situ rapid detection of ions such as K+, H+, Na+ and
Wang et al. reported simultaneous measurement of several Ca2+ and demonstrated the role of the activity of K+ and H+
biomarkers to enhance risk stratification [46]. They con- ions as potential indicators of the onset of AMI [57]. The in-
cluded that the use of current biomarkers (even in a multi- vivo analysis was conducted using epicardial and arterial

202
Review Trends in Pharmacological Sciences Vol.31 No.5

implantation of sensors. Flexible nanoelectrode sensors for enhancement of stent technology. The integration of nano-
K+ were also developed to address the mechanism of technology into stent design has provided novel strategies
ischemic heart disease [58]. In addition, a multi-nanosen- for drug delivery from mesoporous substrates and
sor silicon needle was developed in vitro for the detection of enhanced biocompatibility from nano-textured surfaces.
myocardial ischemia during cardiac surgery by employing The classical challenges of deploying stents in an attempt
field effect transistors (FET) [59]. Nanosensors were also to revascularize arteries narrowed by disease are in-stent
developed for the in-vitro detection and analysis of real- restenosis as the result of intimal hyperplasia [66], and
time sodium concentration during action potentials [60] in late-stage thrombosis mediated by activated platelets [67].
HEK PN1 cells. The flux of Na+ ions across the cell mem- When clinical advancements were thought to be exhausted
brane plays a fundamental part in the generation of action through variations of stent geometry, the next degree of
potentials and regulation of membrane excitability in cells device sophistication established the stent as a drug-deliv-
such as cardiomyocytes. Diseases such as long QT syn- ery platform. Industry giants such as Boston Scientific,
drome and heart failure have been correlated to an altera- Johnson & Johnson, Medtronic, and Guidant began man-
tion of sodium-channel function. Functionalized nanowires ufacturing drug-eluting stents (DESs) that released drugs
were developed for real-time detection of Ca2+ ions import- such as paclitaxel and sirolimus to exploit their anti-pro-
ant in the context of CVDs [49]. Ca2+ ions are known for liferative effects. DESs have demonstrated a lower inci-
activating biological process such as muscle contraction, dence of restenosis 6 months post-procedure compared
protein secretion, cell death and cell development. with their bare-metal counterparts, but recent longer-term
Nanosensors for the detection of other molecules which studies have raised major concerns over the long-term
play an important part in the physiology of the cardiovas- benefit of DESs [68–71]. The anti-proliferative nature of
cular system are also in development (Table 3). Near- the eluted drugs inhibit the cell cycle, thereby prohibiting
infrared fluorescence sensors for nitric oxide (NO) were normal vessel remodeling that results in the integration of
developed with single-walled carbon nanotube technology the stent within the vessel wall. The exposed structure of
[61]. A porphyrinic nanosensor for in-situ measurement of the stent as a result of incomplete neointimal coverage
NO in endothelial cells or in the beating heart allowed facilitates thrombus formation, which leads to increased
understanding the effect of hypertension and ischemia– mortality due to late-stage thrombosis [69]. This outcome
reperfusion on NO release [62]. In2O3 nanowire-based FET is commonly observed after early cessation of dual anti-
sensors were employed as lab-on-a-chip devices for detec- platelet therapy (aspirin/clopidogrel) [70].
tion of oxidized low-density lipoprotein (oxLDL) cholesterol In light of the challenges of DESs, nanotechnology is
[63], which is considered to be a biomarker for acute heart being applied to stent design to improve outcomes. Nano-
attack in patients with coronary artery disease (CAD). porous platforms have demonstrated the ability to provide
Other studies have explored the employment of controlled drug-release profiles over a predetermined
nanotechnology for monitoring physical–mechanical period; such nanoporous technology has been applied to
parameters such as pressure and blood flow as potential cancer applications [4]. Investigators are exploring the
CVD indices. utility of nanoporous stent surfaces of: aluminum oxide
Many bio-MEMS pressure sensors have been developed to deliver tacrolimus [72], carbon–carbon nanoparticle
in-vitro for the in-situ measurement of blood pressure [64]. matrices for the elution of paclitaxel [73], gold [74] or
These include an implantable device for telemetric real- titanium oxide[75] for the delivery of various therapies.
time monitoring of blood pressure with the potential to To ameliorate the problem of impaired revascularization of
diagnose myocardial infarction [65]. Additionally, wireless vessels mediated by the physical presence of stents, surface
bio-MEMs sensors for continuous monitoring of blood flow nano-texturing is being investigated to enhance the inter-
in-situ reveal opportunities for surveillance strategies to action of endothelial cells with stent surfaces. Studies
detect stenosis and to prevent impending graft failure. suggest that nanoscale roughness/topography on nickel
titanium [67] and hydroxyapatite [76] substrates may
Nanotechnology for therapeutic and tissue- mimic the natural structure of vascular tissue and improve
regeneration devices cell adhesion; this will subsequently enhance endothelia-
The utility offered by nanotechnology for cardiovascular lization of stent struts and articulations for the reduction of
device applications is being primarily investigated as an thrombosis [77]. The next evolutionary stent advancement

Table 3. Nanotechnology based on in-vivo CVD sensors


Sensor targets Technology Applications Refs
K+, H+ ions Field effect transistor (FET) Myocardial ischemia [57–59]
Na+ ions Fluorescent nanosensors QT syndrome, heart failure
Ca2+ ions Boron-doped silicon nanowires (SiNWs) Multiple CVDs
Nitric oxide Single-walled carbon nanotube (SWNT) Hypertension Ischemia/reperfusion [60]
[49]
oxLDL Cholesterol Porphyrinic nanosensor Acute heart attack [61]
Blood pressure In2O3 nanowire-based FET Pressure monitoring, [62]
Piezoelectric-BioMEMS Myocardial infarction [63]
Chip-embedded flexible packaging (CEFP) Stenosis in heart bypass Grafts [64]
Blood flow Piezoelectric-BioMEMS [65]
oxLDL: oxidized low-density lipoprotein; BioMEMS: BIO-micro-electro-mechanical system.

203
Review Trends in Pharmacological Sciences Vol.31 No.5

may be realized through a combinatory integration of 8 Cunningham, C.H. et al. (2005) Positive contrast magnetic resonance
imaging of cells labeled with magnetic nanoparticles. Magn. Reson.
nano-porous stent surfaces for the controlled time release
Med. 53, 999–1005
of anti-proliferative agents with nanotextured features. 9 Sosnovik, D.E. et al. (2008) Magnetic nanoparticles for MR imaging:
This will promote vessel endothelialization to lower the agents, techniques and cardiovascular applications. Basic Res. Cardiol.
incidence of restenosis and occurrence of late-stage 103, 122–130
mortality attributed to thrombosis. 10 Pan, D. et al. (2009) Sensitive and efficient detection of thrombus with
fibrin-specific manganese nanocolloids. Chem. Commun. (Camb.) 22,
3234–3236
Conclusions and perspectives 11 Pan, D. et al. (2008) Ligand-directed nanobialys as theranostic agent
In ‘developed’ countries, CVDs represent an enormous for drug delivery and manganese-based magnetic resonance imaging of
burden on the healthcare system and economy because vascular targets. J. Am. Chem. Soc. 130, 9186–9187
they are the leading cause of morbidity and mortality. This 12 Michalet, X. et al. (2005) Quantum dots for live cells, in vivo imaging,
and diagnostics. Science 307, 538–544
becomes even more important considering the increasing 13 Serda, R.E. et al. (2009) Mitotic trafficking of silicon microparticles.
older and obese population. Rapid evolution in genetics, Nanoscale 1, 250–259
proteomics, molecular and cellular biology, material 14 Kooi, M.E. et al. (2003) Accumulation of ultrasmall superparamagnetic
science and bioengineering, make nanotechnology, which particles of iron oxide in human atherosclerotic plaques can be
bridges the gap between interactions on the molecular and detected by in vivo magnetic resonance imaging. Circulation 107,
2453–2458
microscopic levels, one of the major potential players in the 15 Devaraj, N.K. et al. (2009) 18F labeled nanoparticles for in vivo PET-
progress of the treatment and detection of CVDs. Though CT imaging. Bioconjug. Chem. 20, 397–401
still in the very early developmental stages, cardiovascular 16 Nahrendorf, M. et al. (2008) Nanoparticle PET-CT imaging of
nanomedicine is likely to meet the high demand for break- macrophages in inflammatory atherosclerosis. Circulation 117, 379–
387
through innovation in the diagnosis and treatment of
17 Chen, W. et al. (2008) Incorporation of an apoE-derived lipopeptide in
CVDs. Different from conventional molecular thera- high-density lipoprotein MRI contrast agents for enhanced imaging of
peutics, nanomedicine enables the design of multi-com- macrophages in atherosclerosis. Contrast Media Mol. Imaging 3, 233–
ponent, multi-tasking, multi-modular agents that can 242
simultaneously and precisely detect and treat disease. 18 Kao, C.Y. et al. (2003) Long-residence-time nano-scale liposomal
iohexol for X-ray-based blood pool imaging. Acad. Radiol. 10, 475–483
For example, we can envision smart nano-sensors inte-
19 Mukundan, S., Jr et al. (2006) A liposomal nanoscale contrast agent for
grated in existing implants such as defibrillators, stents or preclinical CT in mice. AJR Am. J. Roentgenol. 186, 300–307
pacemakers that may trigger warnings, or perhaps acute 20 Pan, D. et al. (2009) Detecting vascular biosignatures with a colloidal,
release of drug, if required. Another nanomedicine solution radio-opaque polymeric nanoparticle. J. Am. Chem. Soc. 131, 15522–
for CVDs could be projected for vulnerable plaques; ‘click 15527
21 Corot, C. et al. (2006) Recent advances in iron oxide nanocrystal
chemistry’ or highly controlled cross-linking strategies technology for medical imaging. Adv. Drug Deliv. Rev. 58, 1471–1504
targeting and ‘securing’ the plaque before subsequent 22 Amirbekian, V. et al. (2007) Detecting and assessing macrophages in
AMI without danger of occluding the vessel could be uti- vivo to evaluate atherosclerosis noninvasively using molecular MRI.
lized. Proc. Natl. Acad. Sci. U S A 104, 961–966
In summary, we gave a brief overview on the current 23 Botnar, R.M. et al. (2004) In vivo magnetic resonance imaging of
coronary thrombosis using a fibrin-binding molecular magnetic
developments in cardiovascular nanomedicine, but we resonance contrast agent. Circulation 110, 1463–1466
believe that these pale into comparison with future oppor- 24 Cyrus, T. et al. (2006) MR three-dimensional molecular imaging of
tunities for the application of nanotechnology for the diag- intramural biomarkers with targeted nanoparticles. J. Cardiovasc.
nosis and treatment of CVDs. Magn. Reson. 8, 535–541
25 Lanza, G.M. et al. (1998) In vitro characterization of a novel, tissue-
targeted ultrasonic contrast system with acoustic microscopy. J.
Acknowledgments Acoust. Soc. Am. 104, 3665–3672
Matt Landry is gratefully recognized for his artistry in the preparation of
26 Morawski, A.M. et al. (2004) Targeted nanoparticles for quantitative
Figure 1. The authors acknowledge financial support from the following
imaging of sparse molecular epitopes with MRI. Magn. Reson. Med. 51,
sources: DODW81XWH-09-1-0212, DODW81XWH-07-2-0101; NASA
480–486
NNJ06HE06A; NIH RO1CA128797, NIH – R33 CA122864, NIH
27 Winter, P.M. et al. (2003) Molecular imaging of angiogenesis in early-
U54CA143837 and State of Texas, Emerging Technology Fund and by
stage atherosclerosis with alpha(v)beta3-integrin-targeted
the grant-awarding organizations: Department of Defense (DoD),
nanoparticles. Circulation 108, 2270–2274
National Institutes Health (NIH) and National Aeronautics and Space
28 Brito, L. and Amiji, M. (2007) Nanoparticulate carriers for the
Administration (NASA).
treatment of coronary restenosis. Int. J. Nanomedicine 2, 143–161
29 Buxton, D.B. (2009) Nanomedicine for the management of lung and
References blood diseases. Nanomedicine 4, 331–339
1 Theis, T. et al. (2006) nan’o.tech.nol’o.gy n. Nat. Nanotechnol. 1, 8–10 30 Danenberg, H.D. et al. (2002) Macrophage depletion by clodronate-
2 Riehemann, K. et al. (2009) Nanomedicine—challenge and containing liposomes reduces neointimal formation after balloon injury
perspectives. Angew Chem. Int. Ed. Engl. 48, 872–897 in rats and rabbits. Circulation 106, 599–605
3 Kong, D.F. and Goldschmidt-Clermont, P.J. (2005) Tiny solutions for 31 Stephan, D. et al. (1997) Direct gene transfer in the rat kidney in vivo.
giant cardiac problems. Trends Cardiovasc. Med 15, 207–211 Arch. Mal. Coeur Vaiss 90, 1127–1130 [In French]
4 Ferrari, M. (2005) Cancer nanotechnology: opportunities and 32 Hedman, M. et al. (2003) Safety and feasibility of catheter-based
challenges. Nat. Rev. Cancer 5, 161–171 local intracoronary vascular endothelial growth factor gene transfer
5 Peer, D. et al. (2007) Nanocarriers as an emerging platform for cancer in the prevention of postangioplasty and in-stent restenosis and in
therapy. Nat. Nanotechnol. 2, 751–760 the treatment of chronic myocardial ischemia: phase II results of the
6 Ferrari, M. (2008) Nanogeometry: beyond drug delivery. Nat. Kuopio Angiogenesis Trial (KAT). Circulation 107, 2677–2683
Nanotechnol. 3, 131–132 33 Lanza, G.M. et al. (2006) Nanomedicine opportunities for
7 Smith, R.C. and McCarthy, S. (1992) Physics of magnetic resonance. J. cardiovascular disease with perfluorocarbon nanoparticles.
Reprod. Med. 37, 19–26 Nanomedicine 1, 321–329

204
Review Trends in Pharmacological Sciences Vol.31 No.5

34 Jaffer, F.A. et al. (2009) Optical and multimodality molecular imaging: 60 Dubach, J.M. et al. (2009) Visualizing sodium dynamics in isolated
insights into atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 29, cardiomyocytes using fluorescent nanosensors. Proc. Natl. Acad. Sci. U
1017–1024 S A 106, 16145–16150
35 Cyrus, T. et al. (2008) Intramural delivery of rapamycin with 61 Kim, J-H. et al. (2009) The rational design of nitric oxide selectivity in
alphavbeta3-targeted paramagnetic nanoparticles inhibits stenosis single-walled carbon nanotube near-infrared fluorescence sensors for
after balloon injury. Arterioscler. Thromb. Vasc. Biol. 28, 820–826 biological detection. Nat. Chem. 1, 473–481
36 Winter, P.M. et al. (2008) Antiangiogenic synergism of integrin- 62 Sato, M. et al. (2006) Cell-based indicator to visualize picomolar
targeted fumagillin nanoparticles and atorvastatin in dynamics of nitric oxide release from living cells. Anal. Chem. 78,
atherosclerosis. JACC Cardiovasc. Imaging 1, 624–634 8175–8182
37 Winter, P.M. et al. (2006) Endothelial alpha(v)beta3 integrin-targeted 63 Rouhanizadeh, M. et al. (2006) Differentiation of oxidized low
fumagillin nanoparticles inhibit angiogenesis in atherosclerosis. density lipoproteins by nanosensors. Sensors Actuators B: Chem.
Arterioscler. Thromb. Vasc. Biol. 26, 2103–2219 114, 788–798
38 Marsh, J.N. et al. (2007) Fibrin-targeted perfluorocarbon nanoparticles 64 Kovacs, G.T.A. (1998) Micromachined Transducers Handbook.,
for targeted thrombolysis. Nanomedicine 2, 533–543 McGraw Hill, (New York)
39 Lukyanenko, V. (2007) Delivery of nano-objects to functional sub- 65 Shin, K.H. et al. (2005) Flexible wireless pressure sensor module.
domains of healthy and failing cardiac myocytes. Nanomedicine 2, Sensors Actuators A. 123–124, 30–35
831–846 66 Hoffmann, R. et al. (1996) Patterns and mechanisms of in-stent
40 Vasan, R.S. (2006) Biomarkers of cardiovascular disease: molecular restenosis. A serial intravascular ultrasound study. Circulation 94,
basis and practical considerations. Circulation 113, 2335–2362 1247–1254
41 Danesh, J. et al. (2004) C-reactive protein and other circulating 67 Samaroo, H.D. et al. (2008) Enhanced endothelial cell density on NiTi
markers of inflammation in the prediction of coronary heart disease. surfaces with sub-micron to nanometer roughness. Int. J.
N. Engl. J. Med. 350, 1387–1397 Nanomedicine 3, 75–82
42 Wang, T.J. et al. (2004) Plasma natriuretic peptide levels and the risk of 68 Kastrati, A. et al. (2007) Analysis of 14 trials comparing sirolimus-
cardiovascular events and death. N. Engl. J. Med. 350, 655–663 eluting stents with bare-metal stents. N. Engl. J. Med. 356, 1030–1039
43 Danesh, J. et al. (2005) Plasma fibrinogen level and the risk of major 69 Lagerqvist, B. et al. (2007) Long-term outcomes with drug-eluting
cardiovascular diseases and nonvascular mortality: an individual stents versus bare-metal stents in Sweden. N. Engl. J. Med. 356,
participant meta-analysis. JAMA 294, 1799–1809 1009–1019
44 Cushman, M. et al. (1999) Fibrinolytic activation markers predict 70 Mauri, L. et al. (2007) Stent thrombosis in randomized clinical trials of
myocardial infarction in the elderly. The Cardiovascular Health drug-eluting stents. N. Engl. J. Med. 356, 1020–1029
Study. Arterioscler. Thromb. Vasc. Biol. 19, 493–498 71 Stone, G.W. et al. (2007) Safety and efficacy of sirolimus- and
45 Mangoni, A.A. and Jackson, S.H. (2002) Homocysteine and paclitaxel-eluting coronary stents. N. Engl. J. Med. 356, 998–1008
cardiovascular disease: current evidence and future prospects. Am. 72 Wieneke, H. et al. (2003) Synergistic effects of a novel nanoporous stent
J. Med. 112, 556–565 coating and tacrolimus on intima proliferation in rabbits. Catheter
46 Wang, T.J. et al. (2006) Multiple biomarkers for the prediction of first Cardiovasc. Interv. 60, 399–407
major cardiovascular events and death. N. Engl. J. Med. 355, 2631–2639 73 Bhargava, B. et al. (2006) A novel paclitaxel-eluting porous carbon-
47 Gaspari, M. et al. (2006) Nanoporous surfaces as harvesting agents for carbon nanoparticle coated, nonpolymeric cobalt-chromium stent:
mass spectrometric analysis of peptides in human plasma. J. Proteome evaluation in a porcine model. Catheter Cardiovasc. Interv. 67,
Res. 5, 1261–1266 698–702
48 Luchini, A. et al. (2008) Smart hydrogel particles: biomarker 74 Erlebacher, J. et al. (2001) Evolution of nanoporosity in dealloying.
harvesting: one-step affinity purification, size exclusion, and Nature 410, 450–453
protection against degradation. Nano. Lett. 8, 350–361 75 Ayon, A.A. et al. (2006) Drug loading of nanoporous TiO2 films. Biomed.
49 Cui, Y. et al. (2001) Nanowire nanosensors for highly sensitive and Mater. 1, L11–L15
selective detection of biological and chemical species. Science 293, 76 Liu, D.M. et al. (2002) Sol-gel hydroxyapatite coatings on stainless steel
1289–1292 substrates. Biomaterials 23, 691–698
50 McKendry, R. et al. (2002) Multiple label-free biodetection and 77 Caves, J.M. and Chaikof, E.L. (2006) The evolving impact of
quantitative DNA-binding assays on a nanomechanical cantilever microfabrication and nanotechnology on stent design. J. Vasc. Surg.
array. Proc. Natl. Acad. Sci. U S A 99, 9783–9788 44, 1363–1368
51 McMurray, A.A. et al. (2007) A novel point of care diagnostic device: 78 Kim, D. et al. (2007) Antibiofouling polymer-coated gold nanoparticles
impedimetric detection of a biomarker in whole blood. Conf. Proc. IEEE as a contrast agent for in vivo X-ray computed tomography imaging. J.
Eng. Med. Biol. Soc. 115–118 Am. Chem. Soc. 129, 7661–7665
52 Floriano, P.N. et al. (2009) Use of saliva-based nano-biochip tests for 79 Hyafil, F. et al. (2007) Noninvasive detection of macrophages using a
acute myocardial infarction at the point of care: a feasibility study. nanoparticulate contrast agent for computed tomography. Nat. Med.
Clin. Chem. 55, 1530–1538 13, 636–641
53 Wang, J. et al. (2009) Mini sensing chip for point-of-care acute 80 Pan, D. et al. (2009) Molecular photoacoustic tomography with colloidal
myocardial infarction diagnosis utilizing micro-electro-mechanical nanobeacons. Angew Chem. Int. Ed. Engl. 48, 4170–4173
system and nano-technology. Adv. Exp. Med. Biol. 645, 101–107 81 Loo, C. et al. (2004) Nanoshell-enabled photonics-based imaging and
54 Jaffe, A.S. et al. (2000) It’s time for a change to a troponin standard. therapy of cancer. Technol. Cancer Res. Treat. 3, 33–40
Circulation 102, 1216–1220 82 Kaneda, M.M. et al. (2009) Perfluorocarbon nanoemulsions for
55 Wilson, S.R. et al. (2009) Detection of myocardial injury in patients quantitative molecular imaging and targeted therapeutics. Ann.
with unstable angina using a novel nanoparticle cardiac troponin I Biomed. Eng. 37, 1922–1933
assay: observations from the PROTECT-TIMI 30 Trial. Am. Heart J. 83 Yin, X. et al. (2009) HVJ-AVE liposome-mediated Tissue Factor
158, 386–391 Pathway Inhibitor (TFPI) gene transfer with recombinant TFPI
56 Park, J.S. et al. (2009) A highly sensitive and selective diagnostic assay (rTFPI) irrigation attenuates restenosis in atherosclerotic arteries.
based on virus nanoparticles. Nat. Nanotechnol. 4, 259–264 Int. J. Cardiol. 135, 245–248
57 Vogt, S. et al. (2004) Efficacy of ion-selective probes in early epicardial 84 Zaitsev, S. et al. (2004) Polyelectrolyte nanoparticles mediate vascular
in vivo detection of myocardial ischemia. Physiol. Meas. 25, N21–26 gene delivery. Pharm. Res. 21, 1656–1661
58 Ji, T. et al. (2008) In vitro evaluation of flexible pH and potassium ion- 85 Fishbein, I. et al. (2000) Local delivery of platelet-derived growth factor
sensitive organic field effect transistor sensors. Appl. Phys. Lett. 92, receptor-specific tyrphostin inhibits neointimal formation in rats.
233–304 Arterioscler. Thromb. Vasc. Biol. 20, 667–676
59 Errachid, A. et al. (2001) New technology for multi-sensor silicon 86 Banai, S. et al. (2005) Locally delivered nanoencapsulated tyrphostin
needles for biomedical applications. Sensors Actuators B: Chem. 78, (AGL-2043) reduces neointima formation in balloon-injured rat carotid
279–284 and stented porcine coronary arteries. Biomaterials 26, 451–461

205

You might also like