You are on page 1of 26

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/265968880

Heart-Targeted Nanoscale Drug Delivery Systems

Article  in  Journal of Biomedical Nanotechnology · September 2015


DOI: 10.1166/jbn.2014.1894

CITATIONS READS

18 2,749

7 authors, including:

Hai-Sheng Peng Qun Wang


Harbin Medical University Iowa State University
42 PUBLICATIONS   786 CITATIONS    76 PUBLICATIONS   2,927 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Exploring the mechanism and biomedical application of electricity generating hydrogel View project

Applications of Porous Silicon Materials in Drug Delivery View project

All content following this page was uploaded by Hai-Sheng Peng on 16 October 2014.

The user has requested enhancement of the downloaded file.


Review
Journal of
Biomedical Nanotechnology
Copyright © 2014 American Scientific Publishers
All rights reserved Vol. 10, 2038–2062, 2014
Printed in the United States of America www.aspbs.com/jbn

Heart-Targeted Nanoscale Drug Delivery Systems


Meifang Liu1 , Minghui Li1 , Guangtian Wang1 , Xiaoying Liu1 , Daming Liu1 ,
Haisheng Peng1 2 ∗ , and Qun Wang2 3 ∗
1
Department of Pharmaceutics, Daqing Campus of Harbin Medical University, Daqing, 163319, China
2
Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
3
Department of Civil, Construction and Environmental Engineering, Iowa State University, Ames, IA 50011, USA

The efficacious delivery of drugs to the heart is an important treatment strategy for various heart diseases. Nanocarriers
have shown increasing promise in targeted drug delivery systems. The success of nanocarriers for delivering drugs to
therapeutic sites in the heart mainly depends on specific target sites, appropriate drug delivery carriers and effective
targeting ligands. Successful targeted drug delivery suggests the specific deposition of a drug in the heart with mini-
mal effects on other organs after administration. This review discusses the pathological manifestations, pathogenesis,
therapeutic limitations and new therapeutic advances in various heart diseases. In particular, we summarize the recent
advances in heart-targeted nanoscale drug delivery systems, including dendrimers, liposomes, polymer-drug conjugates,
microparticles, nanostents, nanoparticles, micelles and microbubbles. Current clinical trials, the commercial market and
future perspective are further discussed in the conclusions.
KEYWORDS: Heart-Targeted, Nanoscale, Materials, Drug Delivery Systems, Clinical Trials.

CONTENTS INTRODUCTION
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2038 Cardiovascular disease, particularly ischemic impairment,
Cardiovascular Physiological Properties . . . . . . . . . . . . . . . . . 2040 is an important cause of morbidity and mortality world-
Cardiac Physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2040
wide and will become most common cause of death after
Cardiac Cells for Drug Targeting . . . . . . . . . . . . . . . . . . . 2042
Cardiovascular Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . 2043 10 years.1 The heart is an organ of concern at the molec-
Ischemia and Infarction . . . . . . . . . . . . . . . . . . . . . . . . . 2043 ular level and is easily affected by disease in both child
Heart Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2045 and adult populations.2–4 Human cardiac disease involves
Myocardial Hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . 2045 abnormalities in morphogenesis, muscle repair and func-
Restenosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2045 tion and cardiac rhythm.5 A promising solution to heart
Heart-Targeted Drug Delivery Systems . . . . . . . . . . . . . . . . . . 2046
disease involves directly delivering cardioprotective drugs
Dendrimers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2046
Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2047 into the infarcted myocardium and cardiovascular system.
Polymeric Drug Conjugates . . . . . . . . . . . . . . . . . . . . . . . 2048 The need for improved therapies has led to the emergence
Microparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2049 of targeted drug delivery to the heart.
Nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2049 In 1906, Erhlich et al. originally proposed the concept
Micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2052 of targeted delivery in the form of a magic bullet that
Stents with Nano-Coating . . . . . . . . . . . . . . . . . . . . . . . . 2053
would attack affected cells but without any influence on
Microbubbles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2054
Clinical Trials and Commercial Market . . . . . . . . . . . . . . . . . 2054 normal tissues.6 The strategies of targeted drug delivery
Conclusions and Future Direction . . . . . . . . . . . . . . . . . . . . . 2054 refer to the direction of active agents and their carriers into
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2054 the affected tissue with minimal effects on surrounding
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2055 healthy tissues after systemic administration.7–11 In addi-
tion, the old therapeutics always require higher doses

to acquire enough effect-site concentration to achieve a
Authors to whom correspondence should be addressed.
Emails: fisher1688@163.com, qunwang@iastate.edu
valid therapeutic effect due to the limited solubility of
Received: 13 January 2014 the drugs.12 In addition, biomolecules, such as oligonu-
Accepted: 26 January 2014 cleotides, proteins and peptides and small chemicals with

2038 J. Biomed. Nanotechnol. 2014, Vol. 10, No. 9 1550-7033/2014/10/2038/025 doi:10.1166/jbn.2014.1894


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

a short half-life in the circulation require the adminis- All of these drawbacks drive researchers forward to
tration of repeated doses to maintain active levels in the further develop and optimize new drug carriers. Targeted
vessels. Under some circumstances, drug levels in the drug delivery enhances the specific deposition of drugs
body exceed the minimum toxic concentration and gen- in the abnormal foci after administration and simultane-
erate adverse effects for the patient. Overall, the conven- ously decreases the adverse effects in healthy organs.16–22
tional application of drugs is limited by several hurdles, Drug carriers are vehicles for the protection of drugs dur-
such as weak effectiveness, poor biodistribution and low ing transportation after administration and for the mainte-
selectivity.13–15 nance of controlled-release in the body.23 24 The promising

Meifang Liu is a graduate student in pharmaceutics at Harbin Medical University


(Daqing). She received her bachelor’s degree from WeiFang Medical University in 2011.
She has 7 publications in international journals. Currently she is working on exploring
the use of antibody-modified liposomes loaded with AMO-1 to deliver oligonucleotides
to ischemic myocardium for arrhythmia therapy; the corresponding research paper has
been accepted by the Journal of Biomaterials.

Minghui Li works as a lecturer and a vice director in the Department of Pharmaceutics


at Harbin Medical University (Daqing). She got her Master degree in Pharmaceutical
Engineering from Dalian University of Technology in 2010. She got Bachelor degree in
Harbin Medical University in 2007. Li Minghui’s research interests include controlled
release formulations and targeted drug delivery.

Guangtian Wang is a graduate student in Harbin Medical University (Daqing). He


received a B.S. degree in Harbin University of Science and Technology. Currently, he
is doing research in the laboratory of Dr. Haisheng Peng. His areas of interests include
biomaterials, nanotechnology and drug delivery.

Xiaoying Liu is a graduate student in Harbin Medical University (Daqing). She got
the bachelor degree in Harbin Medical University (Daqing) in 2013. Currently, she is
doing research in the laboratory of Dr. Haisheng Peng. Her areas of interests include
biomaterials, nanotechnology and ischemic myocardial delivery.

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2039


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Daming Liu is a graduate student in the 2nd Hospital Affiliated of Harbin Medical
University. He got his bachelor degree in Harbin Medical University in 2006. He had
been working as a neurosurgeon in Harbin Red Cross Hospital for 6 years. He will con-
tinue his study for M.D. majored in Neurology and Pharmacy. Daming Liu’s research
focuses on targeted drug delivery in the treatment of ischemia and tumor.

Haisheng Peng works as an Associate Professor in the Department of Pharmaceutics


at Harbin Medical University (Daqing) and is a Postdoctoral Fellow in the Department
of Chemical and Biological Engineering at Iowa State University. He finished his first
postdoctoral training in cardiovascular pharmacology at Harbin Medical University in
2011. He earned his Ph.D. in neural biology from Harbin Medical University in 2008.
Dr. Peng’s areas of interest include the design of drug delivery systems, the investigation
of distribution behavior and mechanisms, stem cell engineering and biomaterials.

Qun Wang works as an Assistant Professor with a joint appointment in the Department
of Chemical and Biological Engineering and the Department of Civil, Construction and
Environmental Engineering at Iowa State University. He is also an Associate Scientist
at the Ames National Laboratory of Department of Energy. He earned his Ph.D. in
chemical and petroleum engineering from the University of Kansas in 2010. He also
received another Ph.D. in environmental science and engineering from Wuhan Univer-
sity in 2007. Dr. Wang’s areas of interest include biomaterials, intestinal engineering,
nanotechnology and drug delivery. At Iowa State University, Dr. Wang BINDs his
research in these areas to provide innovative solutions and products for human health.

drug carriers include osmotic pumps, liposomes, hydro- appropriate drug delivery carriers and to select effective
gels and polymeric microparticles.25–28 Successful targeted targeting ligands that will ensure a specific interaction of
drug delivery can decrease toxicity and improve the solu- carriers with the complementary molecules on the targeted
bility and stability of the loaded drug.29 30 Classical target- cell membrane. The tissue-recognition ligands in the heart
ing nanoscale carriers usually consist of several functional have been widely studied and include PECAM-1, myosin,
elements, including drugs, targeting ligands and the bio- P-selection, cTnI and cTnC. This review summarizes the
compatible coating of nanoparticles.31–34 These elements properties, symptoms and therapies of various heart dis-
could avoid the quick clearance of nanocarriers by the eases and discusses the emerging developments of heart-
reticuloendothelial system. A conceptual illustration of the targeted nanoscale drug delivery systems.
design of drug-loaded nanoparticle targeting to the heart
is shown in Figure 1.
Recent therapeutic approaches to prevent heart fail- CARDIOVASCULAR PHYSIOLOGICAL
ure after myocardial infarction (MI) mainly rely on the PROPERTIES
systemic injection of proangiogenic peptides and stem Cardiac Physiology
cell therapy.35–37 Unfortunately, some data have confirmed Rhythmicity
the absence of effectiveness of these active biomacro- The rhythm of the heart refers to the heart beating.
molecules due to their short half-life and low stability. It should be very homogeneous in a healthy heart.
Therefore, a heart-targeted nanoparticle drug delivery sys- The cardiac pacemaker is the sinoatrial nodal cell,
tem is emerging and looks promising. The major task for which generates normal, homogeneous heart contractions
fabricating heart-targeted nanoparticles is to choose the (Fig. 2). The electrical signals derived from the sinoatrial

2040 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

Figure 1. Heart-targeted nanoscale drug delivery systems (DDS).

node are distributed over the heart surface by a special- catecholaminergic polymorphous ventricular tachycardia,
ized conduction system; these signals can be recorded long-QT or short-QT syndrome and Brugada syndrome.
as an electrocardiogram (ECG). Moreover, the sinoatrial However, there are ECG abnormalities with no explana-
cells are highly resistant to cardiac failure and ischemia.38 tion, such as prolonged QT interval, ventricular extrasys-
However, when the heart pacemaker, resulting heart rate tole on stress ECG, ST segment elevation in the precordial
and rhythm and impulse conduction exhibit pathological leads and negative or abnormal T waves.40 The clinical
changes, arrhythmias occur in the form of bradycardia, symptoms of various arrhythmias have been described by
tachycardia and other rhythm abnormalities. Standard lead previous investigators.41 42 The diagnosis and therapy of
II electrocardiograms are used to record the rhythm of the arrhythmias are carried out in many ways.43 44 In 2008,
heart in vivo for diagnosing arrhythmias. In 1988, Curtis Churchill et al. proposed that PKC isozymes may be the
and Walker defined the arrhythmia score to evaluate this therapeutic targets of chronic cardiac diseases.45 Moreover,
disease.39 it has been reported that an irregular ventricular rhythm
Many ion channel diseases are diagnosed through causes an alteration in excitation-contraction coupling,
ECG recordings. The ECG abnormalities mainly include which contributes to the progression of heart failure.46

Figure 2. Heart structures and associated cells for drug targeting. SAN: sinoatrial node, AVN: atrioventricular node, AMC: aor-
tomitral continuity, L&R BD: left and right bundle branches, PF: Purkinje fibers, EC: endothelial cells, SC: smooth cells, VM:
ventricular myocytes and NHA: normal human artery.

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2041


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Blood Pressure and have shown potential application value for restoring
Blood pressure refers to the lateral pressure from blood blood flow.64–67
flow per unit area in blood vessels. It can be divided into
diastolic blood pressure (DBP) and systolic blood pressure Cardiac Cells for Drug Targeting
(SBP). Clinical diagnostics have shown that abnormalities Cardiac Muscle Cells
in SBP and DBP are associated with some heart diseases, Cardiomyocytes are highly specialized cells that are
such as stroke, myocardial infarction and heart failure. responsible for the bioelectric variations that generate
Hypertension is a major risk factor that increases the inci- autorhythmicity, excitability and conductibility. Autorhyth-
dence of cardiovascular disease (CVD), which impairs micity is the property that cardiomyocytes acquire
quality of life and usually leads to higher mortality.47–49 spontaneous diastolic depolarization followed by an elec-
In addition, hypertension in the elderly contributes to trical impulse in the absence of external electrical
numerous structural and functional changes to the vascu- stimuli.68 Under normal conditions, the sinoatrial node has
lature during arterial aging. These changes usually lead to the highest autonomy and determines the heart rate in var-
isolated systolic hypertension, diastolic heart failure and ious mammalian species, followed by the atrioventricular
small vessel disease in the brain and other organs.50 Other junction area and then the Purkinje fibers. Spontaneously
risk factors are involved in hypertension, such as dimin- originated action potentials (APs) are propagated via car-
ished kidney function, increasing age, diabetes mellitus, diomyocytes when the cardiac cells are exposed to a stim-
higher body mass index and genetic factors.51–54 ulus with a generative AP, which refers to the excitability
In the 1960s, safe and effective antihypertensive drugs of cardiac cells.69
were first developed and led to dramatic improvements in Gap junctions are specialized membrane structures.
the prognosis of patients with malignant hypertension.55 Intercellular ion channels in gap junctions form a path-
Over the next few decades, the use of an expanding arma- way of cell-to-cell communication. Gap junctions play an
mentarium of blood pressure-lowering drugs has effec- important role in the propagation of cardiac impulses.70
tively eradicated the risk of malignant hypertension.56 In addition, the suppression or enhancement of pacemaker
However, the issues related to drug interactions and the activities and automaticity may cause clinical arrhythmias.
side effects of antihypertensive medications, such as ortho- The individual ion channels, gap junctions and exchanger
static hypotension in the setting of autonomic dysreg- activities determine cardiac excitability and electrical
ulation, are of increasing concern. Recent studies have activity. Previous reports have demonstrated that primary
indicated that renal denervation was a new interventional heart disease, which has an increased arrhythmia risk, and
approach that significantly reduced blood pressure with- primary arrhythmia syndromes are caused by dysfunction
out causing major complications in patients with resistant of the ion channels in the cardiac muscle.71–74 Many data
hypertension.57 58 In addition, there is class I, level B evi- have supported that cardiovascular abnormalities, such as
dence that 150 minutes of weekly physical activity may coronary artery disease, stroke and heart failure, are asso-
be used to complement antihypertensive medication.59 ciated with changes in cardiac electrical excitability due to
transcriptional and post-translational modifications of ion
Blood Flow and Output channels and gap junction proteins.75 76 Typical nanoparti-
Blood flow is well known as a basis of the diagnosis of cle systems target thrombi in lining of artery are demon-
cardiac disease states and can be influenced by numer- strated in Figure 3.
ous factors.60 The evaluation of cardiac blood flow is use- In addition, some previous studies have also indicated
ful in the hemodynamic management of neonates. It is that the interactions and fine balance between various
an effective indicator of health or disease and a pathway transmembrane ion currents regulate cardiac repolarization
to assess the reaction to various stimuli and pharmaceuti- and, consequently, the duration of the ventricular action
cal interventions.61 62 Although the measurement of flow
is more difficult than the measurement of pressure, most
organs require flow rather than pressure. Blood flow can
be estimated by functional echocardiography by measur-
ing upper body systemic blood flow.63 Estimating car-
diac blood flow offers a clearer understanding of the
pathophysiology underling the various clinical conditions
and guidance in the management of these conditions. For
example, thrombus resulting from pathological conditions
in blood vessel contributes to ischemia, myocardial infarc-
tion, atherosis, restenosis, etc. To treat these diseases,
the biodegradable polymer-coated stent and ultrasound- Figure 3. Nanoparticles target thrombi in the lining of an
mediated microbubble destruction have been developed artery.

2042 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

potential.77 78 First, K+ channels play an important role in smooth muscle cells (VSMC) and releases mediators that
determining the morphology and repolarization of the car- regulating vascular tone and growth, blood fluidity, platelet
diac action potential. At least eleven K+ currents have been function and coagulation.91 Under pathological conditions,
characterized pharmacologically by molecular cloning.79 endothelial damage or dysfunction generally leads to a loss
For example, the cardiac delayed rectifier K+ current (Ik ) of anti-aggregative, anti-thrombotic, anti-inflammatory and
mainly consists of two components, the rapidly activat- anti-VSMC activation/growth properties.92 Designed inter-
ing (Ikr ), which is blocked by the class III antiarrhyth- ference in these processes may yield optimistic therapeu-
mic agent E4031, and the slowly activating (Iks ), which is tic benefit in the treatment of diseases that are associated
inhibited by chromanol 293B.80 81 In addition, the inward with endothelial damage and dysfunction. Therefore,
rectifier K+ current (Ik1 ) influences terminal repolarization many diseases, including angiogenesis, atherosclerosis,
and stabilization of the resting membrane potential in car- tumor growth, myocardial infarction and limb and car-
diac cells.82 Changes in Ik1 currents density are relevant to diac ischemia, can be treated by the regulation of
arrhythmogenesis.83 endothelium.93–95 Vascular endothelial cells (VECS) are
Second, high-voltage, L-type Ca2+ channels and low- particular important targets for circulating drug delivery
voltage, T -type Ca2+ channels have been identified in the systems because of their large population and contiguity
sarcolemma of cardiac cells.84 85 The influx of Ca2+ ions with the blood stream.96
through Ca2+ channels contributes to cardiac excitability Various therapeutic approaches have been developed
and excitation-contraction coupling.12 Braz et al. proposed to fight against vascular diseases.97 Recently, some
that protein kinase C may affect cardiac excitation- researchers have proposed the targeted delivery of
contraction coupling through a negative feedback of sar- endothelial cells by overexpressing interleukin-8 receptors
coplasmic reticulum Ca2+ load in addition to its effects on A and B (IL8-RA and -RB). This approach prevented
cardiomyocyte excitability, further affecting cardiac per- inflammatory responses and promoted the structural recov-
formance during cardiac dysfunction and heart failure.86 ery of arteries following endothelial injury by reducing
It is implied that transient low-voltage, activated currents IL-8 in injured or infected tissues.96 However, only a small
(ICaT ) could trigger Ca2+ ion release from the sarcoplas- fraction of injected therapeutic agents bind to endothelial
mic reticulum and induce contraction of Purkinje cells.62 87 cells (EC) due to a lack of affinity for the endothelium
Previous studies have reported that the levels of re- or removal by hemopsonin. Another challenge for deliv-
expression of T -type Ca2+ channels are increased in cer- ering drugs to the endothelium is that many therapeutic
tain pathologic states, such as ventricular hypertrophy.63 88 agents require precise delivery to specific subcellular com-
Third, voltage-gated Na+ channels are the primary partments. The goal of endothelial targeting is to specially,
channels that control the rising phase of the action safely and effectively transport a drug to specific parts of
potential in excitable cells. Some researchers have dis- ECs to achieve local effects, thereby, improving pharma-
covered that the down-regulation of Ca2+ and Na+ chan- cological interventions, including those for metabolic and
nel currents through the protein kinase isozyme (II, ) oncological diseases.98 99 The design determinant of tar-
may lead to cardiac hypertrophy, heart failure and a geted drug delivery systems (DDS) is shown in Figure 4.100
disruption in cardiac excitability.79 Voltage-gated sodium A large number of molecules can serve as specific
channels (VGSCs) are a family of proteins with 9 iso- ligands for EC targeting, such as vascular endothelial
forms that regulate the inward Na+ current upon mem- growth factor (VEGF), cell adhesion molecule (CAM)
brane depolarization. Various inherited arrhythmias may and immunoglobulin G (IgG)-type antibodies.101 102 Drugs
be caused by VGSC dysfunction, such as long QT syn- loaded into liposomes or polymer vehicles or conjugated
drome and Brugada syndrome.89 Fibroblast growth factor with affinity moieties for targeting to ECs may also
homologous factors (FGFHFs), which are comprised of improve the treatment of vascular diseases. Recent research
four genes (FGF11–14), are intracellular proteins that has attempted to explore efficient targeted drug delivery
modulate VGSCs and are potential regulators responsible vehicles to the endothelium, such as liposomes, polymeric
for cardiac conduction abnormalities.90 Additionally, gap nanocarriers and ultrasound-mediated microbubbles.103–105
junctions also play an essential role in the electrical cell-to-
cell coupling and impulse propagation between cells. The
Na+ /Ca2+ exchanger is responsible for regulating intracel- CARDIOVASCULAR DISEASES
lular Ca2+ homeostasis to maintain the normal electrical Ischemia and Infarction
and mechanical activities of the heart. Cardiovascular diseases are usually characterized by lower
blood perfusion. Coronary arteriosclerosis due to coro-
Endothelial Cells nary artery stenosis or occlusion is the leading cause
As a dynamic component of the cardiovascular system, of coronary heart disease. Coronary arteriosclerosis may
the endothelium lines the luminal surface of blood ves- result in myocardial infarction with ischemic death of
sels and plays an important role in cardiac health and dis- cardiomyocytes.106 107 To salvage the myocardium from
ease. It is a barrier between the blood vessels and vascular acute or chronic ischemia, reperfusion is the essential

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2043


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Figure 4. A conceptual illustration of the design for drug-loaded nanoparticle targeting to the heart in vivo and in vitro, and the
clinical application of nanoparticles in humans. Reprinted with permission from [100], K. T. Fitzgerald, et al., Standardization of
models and methods used to assess nanoparticles in cardiovascular applications. Small 7, 705 (2011). © 2011, John Wiley and
Sons.

strategy and is achieved through thrombolysis, percuta- (I/R) injury is another important factor responsible for
neous coronary angioplasty and/or coronary bypass.75 108 infarct size and cardiac dysfunction. The negative effects
Previous studies have shown that a pathological change of I/R mainly included intracellular calcium overload and
in the ischemic myocardium was expressed by cardiac oxidative stress, which eventually lead to cell death.112
troponin I (cTnI). As a current gold-standard marker, In addition, myocardial I/R also leads to arrhythmias and
cTnI is uniquely expressed only when cardiac damage has cardiac contractile dysfunction.113 The new mechanisms
occurred.109 Therefore, the pathological overexpression of and microRNA pathways of myocardial I/R injury are
cTnI can, as a target for the myocardium, be designed shown in Figure 5.114 It has been reported that the pri-
as a drug delivery system for targeting ischemic arrhyth- mary determinant of the outcome of an I/R insult is
mias. However, injury to the endothelium and cardiomy- the pressure, and associated mechanical stress/load, on
ocytes occurs after reperfusion.110 111 Ischemia-reperfusion the myocardium.115 The evaluation of I/R injury mainly

Figure 5. Signal pathways of microRNAs after myocardial ischemia-reperfusion (I/R). miR-92a is expressed in vascular endothe-
lia and inhibits the expression of proangiogenic proteins to block ischemic angiogenesis. In contrast, the levels of miR-499 and
miR-24 were decreased after I/R injury. MiR499 represses cells apoptosis via calcineurin, while miR-24 decreases apoptosis via
the down-regulation of BIM levels. Therefore, the down-regulation of miR-92a expression or the up-regulation of miR-499 or miR-
24 expression may be of benefit to myocardial ischemia and reperfusion. Calcineurin (CN); endothelial cell (EC). Reprinted with
permission from [114], H. K. Eltzschig and T. Eckle, Ischemia and reperfusion [mdash] from mechanism to translation. Nat. Med.
17, 1391 (2011). © 2011, Nature Publishing Group.

2044 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

depends on hemodynamic parameters, including heart rate,


aortic flow, coronary flow, perfusion pressure and cardiac
output.97 Recent studies have shown that cardioprotective
drugs, such as ginsenoside-Re, pioglitazone and follistatin-
like 1, significantly inhibited the cardiac injury caused
by I/R.116–118

Heart Failure
Heart failure (HF) is a complex pathophysiological syn-
drome. It arises from the debilitated function of the Figure 6. Hypertrophied heart.
heart to fill and/or eject blood sufficiently. The clini-
cal manifestations of HF are associated with myocardial to as myocardial hypertrophy. The cellular responses of
insult, including coronary artery disease, genetic factors cardiomyocytes to various signaling pathways are used to
or hypertension, and their attendant sequelae. It is esti- maintain cardiac homeostasis and to prevent pathologi-
mated that HF is the primary cause of over 55,000 deaths cal cardiac hypertrophy. In general, cardiomyocytes ini-
each year, and the incidence of symptomatic HF rises tiate a hypertrophic response to adapt to stress and to
with the increasing age.119 120 Chronic HF (CHF) generally improve cardiac function when the heart is suffering from
occurs due to continued left ventricular (LV) remodeling various stimuli, including mechanical, hemodynamic, hor-
and the progressive loss of function, leading to abnormali- monal and pathologic variations. The adaptive response is
ties in diastolic and/or systolic function.121 The myocardial triggered by a complex cascade of signaling pathways.126
injury evokes an increased orchestrated cascade of remod- Compared to the physiologically normal myocardium, the
eling stimuli within the heart and leads to alterations in hypertrophic myocardium needs more oxygen to main-
neurohormones, vascellum, the kidney and skeletal mus- tain blood circulation. Under this condition, the coronary
cle. Lymperopoulos et al. have articulated the idea that G arteries cannot provide enough blood supply, which even-
protein-coupled receptors (GPCRs) are the major neuro- tually leads to heart failure.
hormonal receptors that control cardiac function and phys- Myocardial hypertrophy is usually accompanied by the
iology. For example, the -adrenergic receptors (1 - and increased synthesis of proteins, assembly of sarcomeres,
2 -ARs) in the membranes of cardiomyocytes dominate perivascular and interstitial fibrosis and increased expres-
cardiac contractile function. Angiotensin II (AngII) type 1 sion of embryonic genes. As a result, hypertrophy even-
receptors (AT1 Rs), which are mainly present in the mem- tually leads to heart failure.127 Cardiac hypertrophy is an
branes of endothelial cell and cardiac fibroblasts, regulate important risk factor for the development of heart fail-
the cardiac structure and morphology. Heart rate is regu- ure with increased mortality.128 129 Recent studies have
lated by the balance between muscarinic cholinergic recep- revealed that the expression signatures of miRNAs have
tors and -adrenergic receptors.122 123 been linked to pathological cardiac hypertrophy.130 The
Given the weak potential of the available therapeutic data indicate that miRNAs are a new regulator in cardiac
approaches, it is essential to design new drug delivery sys- development and disease.
tems for the treatment of HF.124 Some studies have revealed
that CHF increases action potential duration (APD), which Restenosis
leads to early after depolarizations (EAD) and lethal ven- Restenosis is a serious complication of the vascular inter-
tricular tachyarrhythmias.2 123 The molecular mechanism ventional procedures that aim to restore blood flow across
may lie in that tumor necrosis factor- (TNF-) prolongs obstructed arteries (Fig. 7).131 Although angioplasty and
APD. This mechanism is a potential reason for electro- stent implants remove the occlusion and expand the inner
physiological abnormalities and sudden death in HF.125 diameter of the artery with improved hemodynamic flow
Recently, many researchers have focused on strategies rate, restenosis remains a limitation to the overall effi-
based on regulatory RNA (related to heart diseases) or gene cacy of vascular reconstruction and has a 30%–40%
transfer to the heart. For instance, in 2007 Yang et al. dis- incidence.132 Restenosis is characterized as an inflam-
covered that microRNA-1 (miR-1) had important physio- matory process that occurs after injury caused by stent
logical effects in relieving arrhythmias.83 replacement and balloon dilation during angioplasty.133 134
In general, restenosis is associated with neointimal for-
Myocardial Hypertrophy mation from smooth muscle cell (SMC) proliferation
In adult mammalian heart, the cardiomyocytes possess and migration. In addition, some new pathophysiologi-
poor proliferative capacity. The heart increases in size cal mechanisms of restenosis have been well elucidated.
in response to increased workload or stress, such as For example, some new signaling molecules (microRNAs)
aortic stenosis, hypertension, vascular heart disease and have been deemed to control the formation of restenosis.
myocardial infarction. A hypertrophied heart is showed in In the 1970s, the prevention of restenosis through
Figure 6. The increase in cardiomyocyte size is referred systemic drug therapy was first investigated. However,

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2045


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Figure 8. A self-assembly schematic of fluorine-


Figure 7. Therapeutic effects of miR-143/145 on in-stent
modified poly(amidoamine) dendrimers. Amine groups of
restenosis: vascular smooth muscle cell (VSMC) proliferative
(a) poly(amidoamine) dendrimers were conjugated with
capacity is reduced by miR-143/145, which may relieve in-
(b) HFAA to form (c) hepta-fluoroacylated poly(amidoamine)
stent restenosis. Reprinted with permission from [131], J. F.
terminal branches. The branch terminus and blue sphere the
O’Sullivan, et al., Microribonucleic acids for prevention of
represent the terminal primary amine and the hepta-fluoroacyl
plaque rupture and in-stent restenosis “a finger in the dam.”
substituent, respectively. The partially fluorinated dendrimers
J. Am. Coll. Cardiol. 57, 383 (2011). © 2011, Elsevier Limited.
(d) self-assembled in water and produced (e) dendrimers
under the appropriate conditions. The representative diameter
systemic administration has been plague by poorly tol- of the dendrimers in (e) shows the highly compressed cross
network of the fluorine-substituted dendrimers. Reprinted with
erated dosages, narrow therapeutic ranges and dimin-
permission from [146], J. M. Criscione, et al., Self-assembly of
ished efficacy.135 136 Given these disadvantages of systemic pH-responsive fluorinated dendrimer-based particulates for
drug delivery, local drug delivery systems were proposed drug delivery and noninvasive imaging. Biomaterials 30, 3946
based on different approaches. For instance, higher doses (2009). © 2009, Elsevier Limited.
of a therapeutic agent were administered directly to a
treated artery or vein without causing adverse systemic design a new, targeted drug carrier to increase the local
effects.137 138 Other promising approaches have been inves- drug accumulation and to avoid the potential adverse
tigated to prevent restenosis, such as drug-eluting balloons, effects on neighboring tissues. Furthermore, heart-target
periadventitial drug delivery and targeted-nanoparticle sys- vehicles play a crucial role in protecting therapeutics from
temic therapies.139 immediate degradation in the blood. In this section, we
summarize the emerging research progress and results
HEART-TARGETED DRUG on heart-targeted nanoscale drug delivery systems. The
DELIVERY SYSTEMS chemical structure of the loaded molecules, types of tar-
Normally, cardiovascular diseases result from a decrease get foci and pathways of nanoparticle administration into
in blood perfusion. In the heart, the activation of neo- the organ determined the options for the polymers for
angiogenesis around the ischemic area promotes the deliv- nanoparticles.144
ery of oxygen and nutrients, leading to reduced cell
death and scar formation.77 Various therapeutic strategies Dendrimers
have been developed to protect the cardiac vasculature. Dendrimers are repeatedly branched molecules that are
In 1991, Nabel et al. demonstrated that the endothelium characterized by their aesthetic three-dimensional structure.
was a vital target for vascular therapy. The endothelium The aesthetic structure is based on the evaluation of sym-
is associated with normal and abnormal conditions, such metry and polydispersity. Branching units extend radially
as tumor growth, angiogenesis, myocardial infarction, car- from a central core to form dendrimers. They were synthe-
diac and limb ischemia, atherosclerosis and restenosis.65 sized in a stepwise and repetitive manner (Fig. 8).145 146
The clinical outcomes of patients with heart disease were The terminal groups are diversified and multi-
significantly improved after targeted therapeutics to the functionalized. The external active groups can be utilized
infarcted myocardium.140 Recently, a number of pharma- for the modification of dendrimers. The internal organiza-
cological interventions for targeting the endothelium have tion is the branching points, which are of crucial impor-
increased exponentially.141–143 Because vascular diseases tance. The preparations and characterization of dendrimers
have been widely studied, various drugs have been inves- determine their shapes, sizes and structures.147 The iden-
tigated for potential clinical therapies, such as microRNA, tification and characterization are performed via scan-
vascular endothelium growth factor (VEGF), serum and ning tunneling microscopy, atomic force microscopy and
glucocorticoid inducible kinase 1 (SGIK1), and various photoluminescence.148 In 1973, the first dendrimer with
cytokines. Despite years of study, many therapies have some functional groups was prepared.149 Since then, var-
been evaluated to be poorly tolerated, have many side ious modifications of the dendritic branches have been
effects and have diminished efficacy. It is essential to performed in different fields.150 Dendrimers can entrap

2046 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

small chemicals and nanoscale particles in their inte- heart-targeted liposomal carriers is promising to lower the
rior to form host-guest complexes through electrostatic incidence of heart diseases.
or hydrophobic interactions.151 152 Furthermore, the sur- In 2006, Verma et al. reported that anti-myosin mon-
face of dendrimers can bind drugs through electrostatic oclonal antibody-doped liposomes loaded with ATP were
absorption due to similar polarity, such as occurs with car- locally injected into the isolated rat heart before global
boxyl and amine groups. The desired functional group can ischemia-reperfusion. The results confirmed the improved
also be conjugated to the external terminal of dendrimers post-ischemic contractile recovery.163 Recently, both Scott
via non-covalent or covalent modification. With excellent et al. and Wang et al. confirmed that anti-P-selectin-
biocompatibility, dendrimers are often used as drug deliv- conjugated immunoliposomes containing VEGF could
ery systems to deliver therapeutics to targeted tissues. significantly improve vascularization and cardiac func-
Recently, studies have shown that Starbust dendrimers tion based on the overexpression of P-selection in the
are non-immunogenic and can regulate the transfer and infarcted myocardium.164 165 Currently, my team is work-
expression of gene in vitro and in vivo.153 Bella Chanyshev ing on designing anti-cTnI antibody-modified liposomes
and her colleagues demonstrated the conjugation of chem- containing antisense oligodeoxynucleotide of microRNA-1
ically functionalized nucleosides onto poly amidoamine (AMO-1) to target the ischemic myocardium based on
dendrimeric polymers to enhance cardioprotective potency the overexpression of cTnI in the infarcted myocardium.
via the activation of A3 adenosine receptor (A3AR) In addition, diagnostic probes targeting v 3 integrin have
on the cardiomyocyte surface.154 Other researchers have been used in various animal studies on atherosclerosis and
conjugated appropriate cardiac-targeted ligands to the ischemia. Imaging of v 3 expression in ischemic myocar-
surface of dendrimers to achieve cardiac-targeted thera- dial tissue has been performed in dogs and rats. An intra-
peutic effects. Wang et al. combined the technique of venous injection of RPT748 was administered, combining
DNA/dendrimer complexes and electroporation to enhance a 111 In-complex with a non-peptide v 3 ligand.166 167 The
gene transfection in murine cardiac grafts.155 Johnson uptake of 111 In-complex colocalized with neoangiogenesis
et al. fabricated S-nitroso-N -acetylpenicillamine-modified in the ischemic foci, as demonstrated using in vivo and ex
polyamidoamine dendrimers (G4-SNAP) to reduce I/R vivo imaging techniques. Kohane’s group modified lipo-
injury in an isolated, perfused rat heart. They found that somes with a ligand targeted to angiotensin II type1 (AT1).
glutathione was able to enhance NO release from the Their results demonstrated that the carriers were able to
dendrimers to protect the affected tissue against radial deliver active drug to the affected heart tissue after sys-
oxidation.156 tematic administration in vivo (Fig. 9).168
Takahama et al. investigated the influence of various
Liposomes liposomes on the distribution and activity of the ischemic/
Liposomes are structures made up of phospholipid bilay- reperfused myocardium. Stealth liposomes loaded with
ers and contain closed vesicles or concentric spheres. adenosine were prepared by the thin film hydration
Liposomes can be classified as unilamellar or multilamel- method. They assessed the targeting efficiency of lipo-
lar vesicles. In liposomes, the hydrophilic environment somes and myocardial infarction (MI) size after a 30-min
enclosed by the lipid bilayers can be used to entrap water- ligation followed by a 3-h reperfusion. Pharmacokinetic
soluble drugs. Meanwhile, the hydrophobic environment data confirmed that the liposomes were able to effectively
between the bilayers can be used to entrap lipid-soluble accumulate in the foci and prolong the blood residence
drugs.157 158 Liposome drug delivery systems have many time of adenosine. The results from biological studies
advantages in the formulation of potent drug to improve showed that PEGylated liposomal adenosine was able to
therapeutic efficacy.159–161 These properties include bio- enhance the cardioprotective effects of adenosine against
compatibility, flexibility, controlled hydration, various I/R injury and to reduce its unfavorable hemodynamic
administration routes and stabilization of the entrapped effects (Fig. 10).169 170
drug from hostile environments. However, liposomal drug Torchilin et al. constructed a rabbit model of acute
delivery suffers from rapid clearance by the reticuloen- myocardial infarction and fabricated 111 In-labeled stealth
dothelial system if administered by parenteral injection into liposomes modified with antimyosin antibody. They
the bloodstream.162 The conjugation of polyethyleneglycol observed the accumulation of PEG-coated immunolipo-
(PEG) and distearoyl phosphatidylethanolamine (DSPE) to somes in the infarcted rabbit myocardium. The data con-
the liposomes can reduce their recognition by the reticu- firmed that the proportion of PEG in the membrane of
loendothelial system. Recently, liposomes were modified the immunoliposomes was a vital factor. PEG influenced
by targeted ligands to decrease toxicity and improve depo- the half time and targeting efficiency of carriers in the
sition in the desired tissues. These modifications led to new body.171 These authors further investigated the effect of
advancements, including the specific delivery of liposomes various parameters, such as liposomes size, the presence
loaded with drugs to a desired cell and in the target tis- or absence of PEGylation and infarct-specific antimyosin
sue through high-affinity ligands, such as peptides, pro- antibody, on carrier behavior in terms of biodistribu-
teins and antibodies.158 In particular, the development of tion and infarct accumulation. The influence of various

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2047


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Figure 9. Images of nanoparticles targeting the infarcted myocardium. (A) The accumulation of nanoparticles in ischemic hearts,
suffered from 1, 4 and 7 days of ligation, injected with AT1 or scrambled (S) nanoscale particles. (B) Distribution of AT1 nanopar-
ticles in the LV of the infarcted heart 1 day after injection. Pink, nanoparticles; green, autofluorescence. Bar: 200 m. (C) Fluores-
cence intensity in isolated hearts 1 day after administration. Reprinted with permission from [168], T. Dvir, et al., Nanoparticles
targeting the infarcted heart. Nano Lett. 11, 4411 (2011). © 2011, American Chemical Society.

parameters from strong to weak was PEG, antibody and delivery systems exhibited some novel features compared
size.172 with the parent drugs, decreasing adverse reaction and
increased clinical efficacy, ease of drug administra-
Polymeric Drug Conjugates tion and patient compliance.177 The improved clinical
Polymeric drug conjugates are characterized by the conju- efficacy mainly depends on the enhanced permeabil-
gation between drug molecules and water-soluble polymer ity and retention effect (EPR) of stealth polymers.178
carriers. The employed polymers include methacrylamide, Polymer-drug conjugates produce a 100-fold higher
amino acid, PEG and some linear polysaccharides.173–175 concentration of the drug in the target tissue than
In the mid-1970s, Ringsdrof first proposed the concept free drug.179 Thereby, the myocardium-recognition lig-
of the covalent conjugation of drugs to a hydrophilic ands, such as antibodies, proteins and peptides, that
polymer, which was envisioned to achieve active target- are conjugated on the water-soluble polymers can
ing and the modulation of pharmacokinetics.176 Through enhance the targeted delivery of drugs into the infarcted
years of studies on polymer-drug conjugates, these new myocardium.

Figure 10. The in vivo images of mouse hearts after acute myocardial infarction (MI) before and after injection of Gd-DTPA,
liposomes and micelles as captured by MRI. Typical MR images confirm the time-dependent deposition of the drugs in the
infarcted myocardium. The vessel portion of contrast enhancement is marked with arrows. Lateral (L), anterior (A), septal (S),
inferior (I) and septal (S) walls. Reprinted with permission from [170], L. E. Paulis, et al., Distribution of lipid-based nanoparticles
to infarcted myocardium with potential application for MRI-monitored drug delivery. J. Controlled Release (2012). © 2012, Elsevier
Limited.

2048 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

Microparticles heart after surface functionalization.140 Galagudza et al.


Microparticles can be described as spherical structures evaluated the targeting efficiency of silica nanocarriers
containing a matrix that is loaded with drugs through to cardiac tissue. They confirmed that silica nanocarriers
encapsulation or entrapment methods. Accordingly, the exhibited no acute toxicity after administration. The parti-
microparticles can be classified as either microsphere, cles accumulated in the foci within the affected heart tissue
in which the drugs are dispersed into the matrix, or as with ischemia/reperfusion based on the passive targeting
microcapsules, in which the drugs are confined in the inner mechanisms.197 In addition, they proposed that annexin V
core. In this approach, the physicochemical properties of attached to the surface of the silica nanocarriers was able
the loaded drugs are determined through their encapsula- to achieve local deposition of nanocarriers in the affected
tion inside polymers or dispersion into a polymer matrix. myocardium.177
These features allow them to easily penetrate through Targeting nanocarriers to ischemic myocardium can sig-
different defensive barriers in the body.180 In addition, nificantly improve the clinical outcome of individuals with
microparticles are more stable in vivo than are liposomes. heart disease. However, the toxicities may be a drawback
Microparticles can be fabricated by reproducible formu- of the silica nanoparticles in clinical applications. To over-
lation processes that entrap water-soluble and fat-soluble come this limitation, the Shlyakhto group investigated the
drugs and broaden their overall clinical application.181 surface modification and characterization of silica nanocar-
Recently, the intracardiac injection of polymeric riers, evaluated the acute hemodynamic effects of nanopar-
microparticles of anti-inflammatory drugs blocked the ticles, and examined the biodistribution of the carriers
activation of macrophages and reduced the apoptosis or in vivo. The results indicated that silica nanoparticles were
necrosis of cardiomyocytes. The polymeric microparti- not toxic during acute intravenous administration.140
cles of anti-inflammatory drugs showed excellent results Duan et al. investigated the cardiovascular toxicity of
in the clinical therapy of myocardial infarction and other silica nanocarriers to endothelial cells and zebra fish. Their
inflammatory heart abnormalities.182 183 In another study, results confirmed that the silica nanoparticles reduced the
to improve the half-life and stability of cardioprotective
level of p-VDGFR2 and p-ERK1/2. Meanwhile, the car-
drugs, such as VEGF,165 Formiga et al. proposed the syn-
riers also repressed the levels of NKX2.5 and MEF2C
thesis of PLGA microparticles loaded with VEGF;165 they
(Fig. 11). All the data suggest that silica particles may
then validated the use of these microparticles on vasculo-
pose a risk to the cardiovascular system.198
genesis and cardiac tissue remodeling.184
Magnetic Nanoparticles
Nanoparticles
Magnetic nanoparticles (MNP) are widely studied carri-
Silica Nanoparticles
ers that exhibit a variety of attributes. First, they can be
Silica nanoparticles are promising drug carriers due to their
easily handled with the aid of an external magnetic field.
excellent properties, such as chemical and mechanical sta-
Second, it is possible for MNPs to be used in passive and
bility, hydrophilicity and biocompatibility. The particles’
active drug delivery strategies after modification. Third,
surface reactivity and zeta potential can also be tailored
MNPs can be visualized with magnetic resonance imag-
through surface modification.185–187 Silica is bio-resorbed
ing (MRI). Fourth, the enhancement of drug accumulation
via the hydrolysis of siloxane bonds into Si(OH)4 , which
disperses into the blood and lymph system and is eventu- in the target tissue leads to effective treatment at the
ally excreted through the kidneys.188 Given that negatively
charged particles are repelled by the similarly charged cell
membrane, silica nanoparticles are useful to avoid fast
elimination from the body due to their nearly −40 mV zeta
potential at neutral pH.189 190 In addition, silica nanoparti-
cles allow for the functional groups (e.g., amines, thiol) to
be modified on the surface; these groups can then react with
the corresponding functional groups of enzymes, proteins,
DNA and pharmaceutical substances.191 192
It has been reported that mesoporous silica nanoparti-
cles had no influence on cell viability or on the integrity
of the plasma membrane when they were used to deliver Figure 11. Cardiovascular influences of silica particles on
cysteine, hydrophobic dyes and genetic sequences into early, developing zebra fish embryos. The expression of
cells.193–195 A recent study suggested that porous silica VEGFR2, p-VEGFR2, ERK1/2, p-ERK1/2, NKX2.5 and MEF2C
were evaluated by Western blot analysis (-actin as an inter-
microparticles could be rapidly metabolized by the body
nal control). Reprinted with permission from [198], J. Duan,
through the erosion of the plasma-soluble silicic acid et al., Cardiovascular toxicity evaluation of silica nanoparticles
on the surface of the nanoparticles.196 Recently, silica in endothelial cells and zebra fish model. Biomaterials (2013).
nanoparticles have been designed to deliver drugs into © 2013, Elsevier Limited.

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2049


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

Figure 12. The in vivo imaging of MNB/PEI/DNA complexes 2 h after injection via the tail vein. The fluorescent signals were
overlaid in the image. The fluorescence signal was stronger in the left chest of the Mag+group (A) than the Mag−group (B).
Reprinted with permission from [209], W. Li, et al., Enhanced thoracic gene delivery by magnetic nanobead-mediated vector.
J. Gene. Med. 10, 897 (2008). © 2008, John Wiley and Sons.

therapeutically optimal doses.199 However, the applications they are biocompatible and non-toxic at physiological
of MNPs are limited by some problems, such as inappro- concentrations.
priate features and inadequate magnet systems. Therefore, Active targeting of drug-loaded MNPs to diseased heart
it is necessary to consider many factors when designing tissues relies on the attachment between ligands conju-
heart-targeted magnetic drug delivery systems. These fac- gated to the surface of MNPs and the diseased sites with
tors include the magnetic properties and the size of the the adjustment of an external magnetic field.207 Recently,
particles, the strength of the magnetic field, the drug load- a new method of magnetic force-improved gene delivery
ing capacity, the accessibility to the target tissue and the exhibited more feasibility for gene transfer in the cardio-
rate of blood flow.200
vascular system (Fig. 12).208–210 In addition, in 2012 Zhang
Ferric oxide nanoparticles are the only type of MNPs
et al. developed stable magnetic nanoparticle-adenoviral
approved for clinical use by the FDA. Their favorable
vector complexes. They confirmed the possibility of deliv-
features include their facile single step synthesis, chemi-
cal stability in physiological conditions and flexibility of ering therapeutics into the infracted heart in the exter-
chemical modification by coating of the iron oxide cores nal magnetic field for the treatment of acute myocardial
with various shells.201 The common shells for MNPs are infarction.211
gold, polymer, silane or dendrimer.202–205 In addition, it
has been demonstrated that iron oxides (magnetite and Cerium Oxide Nanoparticles
maghemite) exist naturally in the human heart, spleen Cerium oxide nanoparticles (CeO2 NPs) consist of a
and liver.206 Their existence in the body indicates that cerium core surrounded by an oxygen lattice. They have

2050 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

Figure 13. Influence of CeO2 nanocarriers on the inflammatory myocardium. (A) Histopathological staining of left ventricular
sections from control and vehicle- and CeO2 -treated MCP mice. First panel: H&E staining; second panel: Masson’s trichrome
staining, blue indicated collagen deposition. (B) Monocyte/macrophage infiltration in the myocardium (monocytes/macrophages,
green). (C), (D) Statistical analysis of MAB1852-positive cells and TUNEL-positive cells in the myocardium, respectively. ∗ P < 0001
compared with wild-type controls; # P < 005 compared with vehicle-treated MCP mice (n = 5). Reprinted with permission from
[219], J. Niu, et al., Cardioprotective effects of cerium oxide nanoparticles in a transgenic murine model of cardiomyopathy.
Cardiovasc. Res. 73, 549 (2007). © 2007, Oxford University Press.

shown various applications in industrial and commercial due to their antioxidant properties.215 In healthy cells, the
products, such as automotive catalytic converters and oxy- cellular levels of reactive oxygen species (ROS) are finely
gen sensors.212 213 CeO2 NPs have both Ce3+ and Ce4+ controlled. The accumulation of ROS is generally asso-
oxidation states, which are able to perform an autoregen- ciated with undesired effects, such as neurodegenerative
erative redox cycle. Oxygen defects on their surface offer diseases, diabetes, atherosclerosis and aging.216 Moreover,
many active sites for free radical scavenging.214 The CeO2 the elevation of ROS may lead to the increased production
NPs are gaining much attention in the biomedical field of inflammatory cytokines, which are key players in the

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2051


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

development of cardiomyopathy through activated nuclear


factor-B (NF-B).217 218 Recently, studies have demon-
strated that the administration of CeO2 NPs could protect
the heart from oxidative and inflammatory injury induced
by monocyte chemotactic protein-1 (MCP-1) (Fig. 13).219
In addition, Flemming et al. have demonstrated that CeO2
NPs could reduce the atherosclerotic burden associated
with exposure to standard diesel fuel.220

Pt-Modified TiO2 Nanoparticles


Titanium dioxide (TiO2 ) has been extensively investigated
for its catalytic and electrochemical properties and has
been applied as a photocatalyst and gas sensor.221 TiO2 has
also been used in solar energy cells and has been exam-
ined for use as an antitumor agent. TiO2 and modified
TiO2 nanomaterials have shown potential as drug carri-
ers and molecular imaging vehicles in the cardiovascular
system.222 223 However, some studies have shown that TiO2
Figure 14. The influence of four distinct PECAM-1 extracel-
nanoparticles exhibit a certain cytotoxicity due to differ- lular epitopes on the trafficking of anti-PECAM/nanocarriers
ent physical and chemical properties.224 It was reported (NCs) in endothelial cells (ECs). ECs were incubated for 1 h at
that Pt-doped TiO2 nanoparticles exhibited an enhance- 37  C in media containing FITC-labeled anti-PECAM/NCs fol-
ment in photocatalytic efficiency. higher photocatalytic and lowed by incubation with Texas red goat anti-mouse IgG to
efficiency and promise for cardiac imaging and targeted counterstain non-internalized NCs on the cell surface. Merged
images illuminate the localization of single-marked anti-
medical treatments.225 226
PECAM/NCs (green, arrows) compared with double-marked
anti-PECAM/NCs nanoparticles (yellow, arrowheads) in the
Polymeric Nanoparticles cells. The dashed line was used to mark the cell bor-
The size of nanoparticles is between 10–100 nm.227–230 ders. Bar: 10 m. Reprinted with permission from [238],
Major attention has been focused on biodegradable poly- C. Garnacho, et al., Differential intra-endothelial delivery of
polymer nanocarriers targeted to distinct PECAM-1 epitopes.
meric nanoparticles due to their easy penetration across J. Controlled Release 130, 226 (2008). © 2008, Elsevier Limited.
barriers, easy absorption into cells, controlled release and
increased stability of drugs, flexibility to target particular
Micelles
organs/tissues and biocompatibility.231–236
Self-assembly is a method used to fabricate nanoscale Micelles are small, spherical or globular structures. They
particles using the diblock or triblock copolymer with var- are usually prepared by self-assembly with amphiphilic
ious hydrophobicities. In solution, these copolymers auto- block copolymers. In an aqueous environment, these poly-
matically develop micelles.237 Recently, Garnacho et al. mers automatically create a thermodynamically stable col-
fabricated platelet-endothelial cell adhesion molecule 1 loidal solution in the shape of a “core–shell” structure
(PECAM-1) antibody-modified nanocarriers with the when the threshold concentration of polymers is more
ability to target ECs (Fig. 14).238 Somasuntharam than the critical micelle concentration (CMC).241 The core
et al. designed polyketal nanoparticles loaded with is composed of hydrophobic materials, including poly-L-
Nox2-NADPH oxidase siRNA and evaluated cardiac func- lysine, poly(-caprolactone), polylactide and polyglycol-
tion in infarcted mice after administration. They found ide. It serves as a container for water-insoluble drugs.
that the acid-degradable polyketal particles were able to The hydrophilic shell mainly consists of polyethylene gly-
deliver Nox2-siRNA to the affected heart sites. Nox2- col, which can be modified to load water-soluble drugs.
siRNA nanoparticles could be effectively internalized by Polymeric drug carriers can carry low molecular weight,
macrophages, producing the significant inhibition of Nox2 hydrophobic drugs and biomacromolecules, such as nucleic
expression and activity in vitro. The particles prevented the acids and proteins.242 Based on previous studies, poly-
upregulation of Nox2 and significantly improved the heart meric micelle drug carriers were able to improve the sol-
function in vivo.239 Lipinski et al. fabricated gadolinium ubility of hydrophobic drugs, control the release of drugs,
(Gd)-containing lipid-based nanoparticles (NPs) modified increase the stability of drugs and ensure biocompatibil-
with CD36 antibody to target macrophages in the affected ity. They can be designed to target particular organs or
heart. Compared with Gd-containing unmodified NPs and tissues. The polymeric micelle drug delivery system has
Fc-NPs, targeted NPs were able to accumulate in the resi- been used to deliver anti-tumor drugs, such as tamoxifen,
dent macrophages in the plaque based on confocal fluores- paclitaxel, doxorubicin and cisplatin, to tumor cells.243–245
cent microscopy; they also had a stronger contrast-to-noise However, sometimes it is difficult to control the release
ratio.240 kinetics under specific conditions, and the micelles may

2052 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

Figure 15. Ex vivo micrographs of the infarct area in acute MI (one day old) after systematic administration with Gd-DTPA,
micelles and liposomes (red) by confocal laser scanning microscopy. Cyan = CD18 or CD31; green = CD68 or laminin. Bar =
100 m. The co-localization of liposomes (red) with blood vessels (cyan) was performed at higher magnification (the insets in D
and E). Reprinted with permission from [170], L. E. Paulis, et al., Distribution of lipid-based nanoparticles to infarcted myocardium
with potential application for MRI-monitored drug delivery. J. Controlled Release (2012). © 2012, Elsevier Limited.

be degraded in the presence of hostile enzymes. In addi- of anti-remodeling and cardioprotective therapeutics
tion, a polymeric micelle-based drug delivery system has (Fig. 15).170
not yet been applied to heart-targeted therapy in the clinic.
Paulis et al. compared the distribution between micelles and Stents with Nano-Coating
liposomes in the infarcted heart in mice. They used in vivo Metal stents have limited application due to the inflamma-
MRI of paramagnetic lipids in both carriers. The micelles
tory response, thrombosis and restenosis. It was reported
(∼15 nm) and liposomes (∼100 nm) were used to evalu-
that drug-eluting stents (DES) could reduce the inci-
ate differences in the distribution patterns. The data con-
firmed that liposomes showed slower and more restricted dence of restenosis to less than 10% in clinical trials.64 66
extravasation from the vessels, which indicates that lipo- Although DES reduced the percentage of thrombosis and
somes are a promising delivery system for pro-angiogenic restenosis, drawbacks similar to those seen with metal
drug. However, micelles were able to penetrate the ves- stents eventually appear after the drugs on the stent are
sels and went into the infarcted area, which suggests completely released. In addition, in 2004 Virmani et al.
that micelles are more attractive for the targeted delivery proposed that the polymer coating on the stent surface is

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2053


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

involved in an inflammatory response at the site of injury thrombosis, the remission of arterial ischemia and the
and leads to potential restenosis.246 relief of acute coronary syndromes and acute ischemic
To overcome these shortcomings, biodegradable poly- stroke.95 264–266
mer stents were designed to inhibit restenosis before
implantation.247 They are degraded in situ after the ves-
sel is stabilized. Moreover, the vessel recovered its nor- CLINICAL TRIALS AND
mal physiological vasomotor tone with the absence of COMMERCIAL MARKET
a permanent rigid object fixed in the arterial wall.248 Several drugs with different mechanisms have been used
Lincoff et al. demonstrated that stents composed of to deliver drugs to the heart with appropriate nanoscale
poly-L-lactide (PLLA) produced minimal inflammation carriers. The good news is that some heart-targeted
and durable results in a porcine model.249 A limitation nanoscale drug carriers have successfully passed clin-
of polymer-based stents is their low mechanical rigidity. ical trials.140 Furthermore, some of them are already
Bioresorbable stents are mainly constructed from poly- commercially available on the market. For instance, the
mer and metallic alloys. It was reported that a magnesium intramuscular transplantation of the ultrasound-mediated
destruction of microbubbles combined with bone marrow-
stent was absorbed within 3 weeks after implantation.250
derived mononuclear cells (BM-MNCs) has been clinically
In addition, porous aluminum oxide-coated stents encap-
applied for inducing angiogenesis in the ischemic tissue.267
sulated with drugs have exhibited inhibition to neointimal
In addition, stents are widely accepted as an effective treat-
growth.251 Although biodegradable polymeric stents are
ment for occluded arteries.268 269 The drug-eluting stent
often used in the interventional cardiology field, they still
(DES) has been reported to effectively reduce the per-
need improvement to meet clinical requirements.
centage of restenosis to less than 10% in initial clinical
trials.181 182 Currently, there are two DES on the market:
Microbubbles the Cypher® stent and the Taxus® stent.
Recently, various studies have demonstrated that the
ultrasound-targeted microbubble destruction (UTMD)
technique shows excellent features for drug and gene CONCLUSIONS AND FUTURE DIRECTION
delivery.252 253 Microbubbles used in UTMD consist of We have reviewed the physiological properties of the
albumin, saccharide, biocompatible polymers, lipids and cardiovascular system, various heart diseases and new
other materials. Ultrasound contrast agent is one of advances in heart-targeted nanoscale drug delivery sys-
the materials used because of its capacity to reflect tems. A successful design for heart-targeted nanoscale
ultrasound.254–256 Active agents can be encapsulated into drug delivery systems is based on three parameters. First,
the microbubbles using different approaches, such as coat- it is necessary to understand the fundamentals of various
ing or binding to the microbubble surface. The sonopora- heart diseases, including their etiology, pathological man-
tion produced by UTMD can transfer genes into cells.257 ifestation, clinical features and desired therapies. Second,
Microbubbles will oscillate and rupture when they are suitable heart-targeted drug carriers need to be selected
and the active targeting properties of these carriers then
exposed to ultrasound. Then the entrapped gene therapy
needs to be regulated to improve the stability of loaded
vector will be released from the microbubbles, producing
drugs. Finally, researchers should choose the appropriate
high local concentrations at the site of interest. Meanwhile,
targeting ligands to ensure the specific interaction of the
the destruction of microbubbles may transiently induce
nanocarriers with the complementary molecules on the
holes in the membrane, which increase the permeability
surface of the targeted cells. Successful targeted delivery
of vessels and biomembranes, thus facilitating the entry of
of nanocarriers to the heart is helpful to reduce toxicity
drugs or genes into cells.258 259
and to increase the availability of the drugs.171 Although
It was reported that specific ligands for endothelial cell the heart-targeted delivery of nanocarriers is promising
adhesion could be modified on the surface of microbub- for transporting drugs to affected cardiac tissue and for
bles for delivery.260 261 Recently, a large number of studies controlling drug release in the body, these systems suf-
have supported the idea that UTMD is a feasible method in fer from limitations, such as toxicity, immunogenicity
the clinical therapy of cardiovascular diseases by improv- and unwanted pharmacokinetic behavior.270–272 The desired
ing gene transfection.262 For example, Chen et al. have techniques of nanocarrier fabrication is aimed at ensuring
successfully transfected the human TB4 gene into nor- better uptake in the target cells, more desirable accumu-
mal rat hearts using UTMD.263 In another study, Chen lation of drugs in affected tissue than in unaffected sites,
et al. confirmed that the combination of UTMD with PEI and more effective avoidance of drug toxicity. Eventually,
could improve the transfection efficiency of naked genes an ideal tool for the targeted delivery of drugs into the
into the myocardium without causing any apparent adverse heart is emerging.
reaction.263 Moreover, ultrasound-mediated microbubble
drug therapy for thrombolysis has already reached the Acknowledgments: This research was supported by
clinical arena, such as in the treatment of deep venous grants from the Heilongjiang Provincial Natural Science

2054 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

Foundation (D201031), the Heilongjiang Provincial Health 17. Z. Gu, A. A. Aimetti, Q. Wang, T. T. Dang, Y. Zhang, O. Veiseh,
Bureau Foundation (2009-265 and 2011-236) and the H. Cheng, R. S. Langer, and D. G. Anderson, Injectable nano-
network for glucose-mediated insulin delivery. ACS Nano 7, 4194
Innovative Fund of Harbin Medical University Graduate
(2013).
Student (YJSCX2012-202HLJ). 18. B. Büyüktimkin, Q. Wang, P. Kiptoo, J. M. Stewart, C. Berkland,
and T. J. Siahaan, Vaccine-like controlled-release delivery of
an immunomodulating peptide to treat experimental autoimmune
REFERENCES encephalomyelitis. Mol. Pharm. 9, 979 (2012).
1. B. Ostadal, The past, the present and the future of experimental 19. Q. Wang, J. Wang, Q. Lu, M. S. Detamore, and C. Berkland,
research on myocardial research on myocardial ischemia and pro- Injectable PLGA based colloidal gels for zero-order dexamethasone
tection. Pharmacol. Rep. 61, 3 (2009). release in cranial defects. Biomaterials 31, 4980 (2010).
2. M. Buckingham, S. Meilhac, and S. Zaffran, Building the mam- 20. J. Ye, Q. Wang, X. Zhou, and N. Zhang, Injectable actarit-loaded
malian heart from two sources of myocardial cells. Nat. Rev. Genet. solid lipid nanoparticles as passive targeting therapeutic agents for
6, 826 (2005). rheumatoid arthritis. Int. J. Pharm. 352, 273 (2008).
3. E. N. Olson, Gene regulatory networks in the evolution and devel- 21. R. Bi, W. Shao, Q. Wang, and N. Zhang, Spray-freeze-dried dry
opment of the heart. Science 313, 1922 (2006). powder inhalation of insulin-loaded liposomes for enhanced pul-
4. T. Thom, N. Haase, W. Rosamond, V. J. Howard, J. Rumsfeld, monary delivery. J. Drug Target 16, 639 (2008).
T. Manolio, Z. J. Zheng, K. Flegal, C. O’Donnell, S. Kittner, 22. F. Jia, X. Liu, L. Li, S. Mallapragada, B. Narasimhan, and Q. Wang,
D. Lloyd-Jones, D. C. Goff, Jr., Y. Hong, R. Adams, G. Friday, Multifunctional nanoparticles for targeted delivery of immune acti-
K. Furie, P. Gorelick, B. Kissela, J. Marler, J. Meigs, V. Roger, vating and cancer therapeutic agents. J. Controlled Release 172,
S. Sidney, P. Sorlie, J. Steinberger, S. Wasserthiel-Smoller, 1020 (2013).
M. Wilson, and P. Wolf, C. American Heart Association Statistics 23. Q. Wang, N. Zhang, X. Hu, J. Yang, and Y. Du, Algi-
and S. Stroke Statistics, Heart disease and stroke statistics–2006 nate/polyethylene glycol blend fibers and their properties for drug
update: A report from the American Heart Association Statistics controlled release. J. Biomed. Mater. Res. A 82, 122 (2007).
Committee and Stroke Statistics Subcommittee. Circulation 113, 24. Q. Wang, N. Zhang, X. Hu, J. Yang, and Y. Du, Chi-
e85 (2006). tosan/polyethylene glycol blend fibers and their properties for drug
5. Y. Zhao, J. F. Ransom, A. Li, V. Vedantham, M. von Drehle, controlled release. J. Biomed. Mater. Res. A 85, 881 (2008).
A. N. Muth, T. Tsuchihashi, M. T. McManus, R. J. Schwartz, and 25. W. T. Al-Jamal and K. Kostarelos, Liposomes: From a clinically
D. Srivastava, Dysregulation of cardiogenesis, cardiac conduction, established drug delivery system to a nanoparticle platform for ther-
and cell cycle in mice lacking miRNA-1-2. Cell 129, 303 (2007). anostic nanomedicine. Acc. Chem. Res. 44, 1094 (2011).
6. P. Ehrlich and C. Bolduan, Collected Studies on Immunity, Wiley, 26. A. S. Hoffman, Hydrogels for biomedical applications. Adv. Drug
New York (1906). Deliv. Rev. 54, 3 (2002).
7. T. Lammers, F. Kiessling, W. E. Hennink, and G. Storm, Nanoth- 27. W. Jiang, R. K. Gupta, M. C. Deshpande, and S. P. Schwendeman,
eranostics and image-guided drug delivery: Current concepts and Biodegradable poly(lactic-co-glycolic acid) microparticles for
future directions. Mol. Pharm. 7, 1899 (2010). injectable delivery of vaccine antigens. Adv. Drug Deliv. Rev. 57,
8. Z. Liu, Y. Wang, J. Zhang, M. Li, Y. Liu, and N. Zhang, Pluronic 391 (2005).
P123-docetaxel conjugate micelles: Synthesis, characterization, and 28. J. Urquhart, Internal medicine in the 21st century: Controlled drug
antitumor activity. J. Biomed. Nanotechnol. 9, 2007 (2013). delivery: Therapeutic and pharmacological aspects. J. Intern. Med.
9. D. Lin, G. Li, L. Qin, Z. Wen, J. Wang, and X. Sun, Preparation, 248, 357 (2000).
characterization and uptake of PEG-coated, muco-inert nanoparti- 29. E. B. Lages, M. B. de Freitas, I. M. B. Goncalves, R. J. Alves,
cles in HGC-27 cells, a mucin-producing, gastric-cancer cell line. C. D. Vianna-Soares, L. A. M. Ferreira, M. C. de Oliveira, and
J. Biomed. Nanotechnol. 9, 2017 (2013). R. B. de Oliveira, Evaluation of antitumor activity and development
10. X. Jia, J. Yin, D. He, X. He, K. Wang, M. Chen, and Y. Li, Poly- of solid lipid nanoparticles of metronidazole analogue. J. Biomed.
acrylic acid modified upconversion nanoparticles for simultaneous Nanotechnol. 9, 1939 (2013).
pH-triggered drug delivery and release imaging. J. Biomed. Nano- 30. V. Saxena and M. D. Hussain, Polymeric mixed micelles for deliv-
technol. 9, 2063 (2013). ery of curcumin to multidrug resistant ovarian cancer. J. Biomed.
11. N. S. Elbialy, B. M. Abdol-Azim, M. W. Shafaa, L. H. El Shazly, Nanotechnol. 9, 1146 (2013).
A. H. El Shazly, and W. A. Khalil, Enhancement of the ocular 31. A. L. Miranda-Vilela, R. C. A. Peixoto, J. P. F. Longo, F. A.
therapeutic effect of prednisolone acetate by liposomal entrapment. Portilho, K. L. C. Miranda, P. P. C. Sartoratto, S. N. Bao, R. B.
J. Biomed. Nanotechnol. 9, 2105 (2013). de Azevedo, and Z. G. M. Lacava, Dextran-functionalized mag-
12. R. Ottenbrite, Encyclopedia of Polymer Science and Engineering, netic fluid mediating magnetohyperthermia combined with preven-
edited by J. I. Kroschwitz, Wiley, New York (1990), pp. 164–187. tive antioxidant pequi-oil supplementation: Potential use against
13. D. Nevozhay, U. Kanska, R. Budzynska, and J. Boratynski, Current cancer. J. Biomed. Nanotechnol. 9, 1261 (2013).
status of research on conjugates and related drug delivery systems 32. Q. Long, Y. Xie, Y. Huang, Q. Wu, H. Zhang, S. Xiong, Y. Liu,
in the treatment of cancer and other diseases. Postepy. Hig. Med. L. Chen, Y. Wei, and X. Zhao, Induction of apoptosis and inhibition
Dosw. (Online) 61, 350 (2007). of angiogenesis by PEGylated liposomal quercetin in both cisplatin-
14. J. Cao, S. Zhai, C. Li, B. He, Y. Lai, Y. Chen, X. Luo, and Z. Gu, sensitive and cisplatin-resistant ovarian cancers. J. Biomed. Nan-
Novel pH-sensitive micelles generated by star-shape copolymers otechnol. 9, 965 (2013).
containing zwitterionic sulfobetaine for efficient cellular internal- 33. K.-J. Lee, J. H. An, J.-R. Chun, K.-H. Chung, W.-Y. Park, J.-S.
ization. J. Biomed. Nanotechnol. 9, 1847 (2013). Shin, D.-H. Kim, and Y. Y. Bahk, In vitro analysis of the anti-
15. P. Ojer, L. Neutsch, F. Gabor, J. M. Irache, and A. L. de cancer activity of mitoxantrone loaded on magnetic nanoparticles.
Cerain, Cytotoxicity and cell interaction studies of bioadhe- J. Biomed. Nanotechnol. 9, 1071 (2013).
sive poly(anhydride) nanoparticles for oral antigen/drug delivery. 34. G. Zhang, X. Zeng, and P. Li, Nanomaterials in cancer-therapy drug
J. Biomed. Nanotechnol. 9, 1891 (2013). delivery system. J. Biomed. Nanotechnol. 9, 741 (2013).
16. X. Ye and D. Yang, Recent advances in biological strategies for 35. N. Maulik and M. Thirunavukkarasu, Growth factor/s and cell ther-
targeted drug delivery. Cardiovasc. Hematol. Disord. Drug Targets apy in myocardial regeneration. J. Mol. Cell. Cardiol. 44, 219
9, 206 (2009). (2008).

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2055


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

36. R. Passier, L. W. van Laake, and C. L. Mummery, Stem-cell-based 57. Simplicity HTN-2 Investigators, M. D. Esler, H. Krum, P. A.
therapy and lessons from the heart. Nature 453, 322 (2008). Sobotka, M. P. Schlaich, R. E. Schmieder, and M. Bohm, Renal
37. V. F. Segers and R. T. Lee, Stem-cell therapy for cardiac disease. sympathetic denervation in patients with treatment-resistant hyper-
Nature 451, 937 (2008). tension (The Symplicity HTN-2 Trial): A randomised controlled
38. H. Satoh, Sino-atrial nodal cells of mammalian hearts: Ionic cur- trial. Lancet 376, 1903 (2010).
rents and gene expression of pacemaker ionic channels. J. Smooth 58. H. Krum, M. Schlaich, R. Whitbourn, P. A. Sobotka, J. Sadowski,
Muscle Res. 39, 175 (2003). K. Bartus, B. Kapelak, A. Walton, H. Sievert, and S. Thambar,
39. M. J. Curtis and M. J. Walker, Quantification of arrhythmias using Catheter-based renal sympathetic denervation for resistant hyper-
scoring systems: An examination of seven scores in an in vivo tension: A multicentre safety and proof-of-principle cohort study.
model of regional myocardial ischaemia. Cardiovasc. Res. 22, 656 Lancet 373, 1275 (2009).
(1988). 59. L. Mosca, E. J. Benjamin, K. Berra, J. L. Bezanson, R. J.
40. B.-M. Beckmann, A. Pfeufer, and S. Kääb, Inherited cardiac Dolor, D. M. Lloyd-Jones, L. K. Newby, I. L. Piña, V. L. Roger,
arrhythmias: Diagnosis, treatment, and prevention. Dtsch. Arztebl. L. J. Shaw, D. Zhao, T. M. Beckie, C. Bushnell, J. D’Armiento,
Int. 108, 623 (2011). P. M. Kris-Etherton, J. Fang, T. G. Ganiats, A. S. Gomes, C. R.
41. C. Antzelevitch, P. Brugada, M. Borggrefe, J. Brugada, R. Brugada, Gracia, C. K. Haan, E. A. Jackson, D. R. Judelson, E. Kelepouris,
D. Corrado, I. Gussak, H. LeMarec, K. Nademanee, and A. R. P. C. J. Lavie, A. Moore, N. A. Nussmeier, E. Ofili, S. Oparil,
Riera, Brugada syndrome: Report of the second consensus con- P. Ouyang, V. W. Pinn, K. Sherif, S. C. Smith Jr, G. Sopko,
ference endorsed by the Heart Rhythm Society and the European N. Chandra-Strobos, E. M. Urbina, V. Vaccarino, and N. K. Wenger,
Heart Rhythm Association. Circulation 111, 659 (2005). Effectiveness-based guidelines for the prevention of cardiovascu-
42. A. J. Moss, J. L. Robinson, L. Gessman, R. Gillespie, W. Zareba, lar disease in women-2011 update: A guideline from the American
P. J. Schwartz, G. M. Vincent, J. Benhorin, E. L. Heilbron, and J. A. Heart Association. Circulation 123, E624 (2011).
Towbin, Comparison of clinical and genetic variables of cardiac 60. J. S. Petrofsky, Resting blood flow in the skin: Does it exist, and
events associated with loud noise versus swimming among subjects what is the influence of temperature, aging, and diabetes? J. Dia-
with the long QT syndrome. Am. J. Cardiol. 84, 876 (1999). betes Sci. Technol. 6, 674 (2012).
43. W. Shimizu, The long QT syndrome: Therapeutic implications of 61. N. Banait, P. Suryawanshi, N. Malshe, R. Nagpal, and S. Lalwani,
a genetic diagnosis. Cardiovasc. Res. 67, 347 (2005). Cardiac blood flow measurements in stable full term small for ges-
44. D. J. Tester and M. J. Ackerman, Genetic testing for potentially tational age neonates. J. Clin. Diagn. Res. 7, 1651 (2013).
lethal, highly treatable inherited cardiomyopathies/channelopathies
62. Z. Zhou and C. T. January, Both T - and L-type Ca2+ channels
in clinical practice. Circulation 123, 1021 (2011).
can contribute to excitation-contraction coupling in cardiac Purkinje
45. E. Churchill, G. Budas, A. Vallentin, T. Koyanagi, and D. Mochly-
cells. Biophys. J. 74, 1830 (1998).
Rosen, PKC isozymes in chronic cardiac disease: Possible thera-
63. H. B. Nuss and S. R. Houser, T -type Ca2+ current is expressed in
peutic targets? Annu. Rev. Pharmacol. Toxicol. 48, 569 (2008).
hypertrophied adult feline left ventricular myocytes. Circ. Res. 73,
46. L.-H. Ling, O. Khammy, M. Byrne, F. Amirahmadi, A. Foster,
777 (1993).
G. Li, L. Zhang, C. dos Remedios, C. Chen, and D. M. Kaye, Irreg-
64. M. Degertekin, E. Regar, K. Tanabe, P. C. Smits, W. J. van der
ular rhythm adversely influences calcium handling in ventricular
Giessen, S. G. Carlier, P. de Feyter, J. Vos, D. P. Foley, and J. M.
myocardium implications for the interaction between heart failure
Ligthart, Sirolimus-eluting stent for treatment of complex in-stent
and atrial fibrillation. Circ. Heart Fail 5, 786 (2012).
restenosisthe first clinical experience. J. Am. Coll. Cardiol. 41, 184
47. P. B. Gorelick, R. L. Sacco, D. B. Smith, M. Alberts, L. Mustone-
(2003).
Alexander, D. Rader, J. L. Ross, E. Raps, M. N. Ozer, and L. M.
65. C. Di Mario, H. Griffiths, O. Goktekin, N. Peeters, J. Verbist,
Brass, Prevention of a first stroke. JAMA 281, 1112 (1999).
48. D. K. Hayes, C. H. Denny, N. L. Keenan, J. B. Croft, and K. J. M. Bosiers, K. Deloose, B. Heublein, R. Rohde, and V. Kasese,
Greenlund, Health-related quality of life and hypertension status, Drug-eluting bioabsorbable magnesium stent. J. Intervent. Cardiol.
awareness, treatment, and control: National health and nutrition 17, 391 (2004).
examination survey, 2001–2004. J. Hypertens. 26, 641 (2008). 66. G. W. Stone, S. G. Ellis, D. A. Cox, J. Hermiller,
49. C. M. Lawes, S. V. Hoorn, and A. Rodgers, Global burden of blood- C. O’Shaughnessy, J. T. Mann, M. Turco, R. Caputo, P. Bergin, and
pressure-related disease, 2001. Lancet 371, 1513 (2008). J. Greenberg, A polymer-based, paclitaxel-eluting stent in patients
50. M. G. Denker and D. L. Cohen, What is an appropriate blood with coronary artery disease. N. Engl. J. Med. 350, 221 (2004).
pressure goal for the elderly: Review of recent studies and practical 67. H. Tamai, K. Igaki, E. Kyo, K. Kosuga, A. Kawashima, S. Matsui,
recommendations. Clin. Interv. Aging 8, 1505 (2013). H. Komori, T. Tsuji, S. Motohara, and H. Uehata, Initial and
51. B. M. Egan, Y. Zhao, R. N. Axon, W. A. Brzezinski, and K. C. 6-month results of biodegradable poly-l-lactic acid coronary stents
Ferdinand, Uncontrolled and apparent treatment resistant hyperten- in humans. Circulation 102, 399 (2000).
sion in the United States, 1988 to 2008. Circulation 124, 1046 68. N. Peters, C. Cabo, and A. Wit, Cardiac Electrophysiology, from
(2011). Cell to Bedside, 3rd edn., edited by D. P. Zipes and J. Jalife,
52. C.-J. Huang, H. E. Webb, M. C. Zourdos, and E. O. Acevedo, Car- Saunders, Philadelphia (2000), pp. 345–355.
diovascular reactivity, stress, and physical activity. Front. Physiol. 69. A. O. Grant and S. Durrani, Textbook of Cardiovascular Medicine,
4, 314 (2013). 3rd edn., edited by E. Topol, Lippincott, Philadelphia (2006),
53. S. D. Persell, Prevalence of resistant hypertension in the United pp. 950–963.
States, 2003–2008. Hypertension 57, 1076 (2011). 70. G. Tomaselly and D. Roden, Electrophysiological Disorders of the
54. N. Redmond, H. J. Baer, and L. S. Hicks, Health behaviors and Heart, edited by S. Saksena and A. J. Camm, Elsevier, New York
racial disparity in blood pressure control in the national health and (2005), pp. 27–50.
nutrition examination survey. Hypertension 57, 383 (2011). 71. C. Basso, M. Burke, P. Fornes, P. J. Gallagher, R. H. de Gouveia,
55. E. R. Mohler Jr and E. D. Freis, Five-year survival of patients with M. Sheppard, G. Thiene, and A. van der Wal, Guidelines for
malignant hypertension treated with antihypertensive agents. Am. autopsy investigation of sudden cardiac death. Virchows Arch. 452,
Heart J. 60, 329 (1960). 11 (2008).
56. V. Perkovic, R. Huxley, Y. Wu, D. Prabhakaran, and S. MacMahon, 72. E. R. Behr, C. Dalageorgou, M. Christiansen, P. Syrris, S. Hughes,
The burden of blood pressure-related disease a neglected priority M. T. T. Esteban, E. Rowland, S. Jeffery, and W. J. McKenna,
for global health. Hypertension 50, 991 (2007). Sudden arrhythmic death syndrome: Familial evaluation identifies

2056 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

inheritable heart disease in the majority of families. Eur. Heart J. 93. C. R. Dass and T. Su, Delivery of lipoplexes for genotherapy of
29, 1670 (2008). solid tumours: Role of vascular endothelial cells. J. Pharm. Phar-
73. S. Kauferstein, N. Kiehne, T. Neumann, H.-F. Pitschner, and macol. 52, 1301 (2000).
H. Bratzke, Cardiac gene defects can cause sudden cardiac death 94. E. G. Nabel, Biology of the impaired endothelium. Am. J. Cardiol.
in young people. Dtsch. Arztebl. Int. 106, 41 (2009). 68, 6 (1991).
74. H. L. Tan, N. Hofman, I. M. van Langen, A. C. van der Wal, and 95. S. A. Parikh and E. R. Edelman, Endothelial cell delivery for car-
A. A. Wilde, Sudden unexplained death heritability and diagnostic diovascular therapy. Adv. Drug Deliv. Rev. 42, 139 (2000).
yield of cardiological and genetic examination in surviving rela- 96. D. Xing, P. Li, K. Gong, Z. Yang, H. Yu, F. G. Hage, S. Oparil, and
tives. Circulation 112, 207 (2005). Y.-F. Chen, Endothelial cells overexpressing interleukin-8 receptors
75. T. Aiba and G. F. Tomaselli, Electrical remodeling in the failing reduce inflammatory and neointimal responses to arterial injury.
heart. Curr. Opin. Cardiol. 25, 29 (2010). Circulation 125, 1533 (2012).
76. I. Imanaga, Pathological remodeling of cardiac gap junction con- 97. Y. Xu, H. Lee, Y. Hu, J. Huang, S. Kim, and M. Yun, Detec-
nexin 43-with special reference to arrhythmogenesis. Pathophysiol- tion and identification of breast cancer volatile organic compounds
ogy 17, 73 (2010). biomarkers using highly-sensitive single nanowire array on a chip.
77. P. Carmeliet and R. K. Jain, Molecular mechanisms and clinical J. Biomed. Nanotechnol. 9, 1164 (2013).
applications of angiogenesis. Nature 473, 298 (2011). 98. V. R. Muzykantov, Biomedical aspects of targeted delivery of drugs
78. A. Varro, D. Lathrop, S. Hester, P. Nanasi, and J. Papp, Ionic cur- to pulmonary endothelium. Expert Opin. Drug Deliv. 2, 909 (2005).
rents and action potentials in rabbit, rat, and guinea pig ventricular 99. B.-S. Ding, T. Dziubla, V. V. Shuvaev, S. Muro, and V. R.
myocytes. Basic Res. Cardiol. 88, 93 (1993). Muzykantov, Advanced drug delivery systems that target the vas-
79. J. C. B. Ferreira, D. Mochly-Rosen, and M. Boutjdir, Regulation cular endothelium. Mol. Interventions 6, 98 (2006).
of cardiac excitability protein kinase C isozymes. Front. Biosci. 100. K. T. Fitzgerald, C. A. Holladay, C. McCarthy, K. A. Power,
(Schol. Ed.) 4, 532 (2012). A. Pandit, and W. M. Gallagher, Standardization of models and
80. A. Busch, H. Suessbrich, S. Waldegger, E. Sailer, R. Greger, H.-J. methods used to assess nanoparticles in cardiovascular applications.
Lang, F. Lang, K. Gibson, and J. Maylie, Inhibition of IKs in guinea Small 7, 705 (2011).
pig cardiac myocytes and guinea pig IsK channels by the chromanol 101. D. J. Lefer, D. M. Flynn, D. C. Anderson, and A. J. Buda, Com-
293B. Pflügers Arch. 432, 1094 (1996). bined inhibition of P-selectin and ICAM-1 reduces myocardial
81. M. C. Sanguinetti and N. Jurkiewicz, Two components of cardiac injury following ischemia and reperfusion. Am. J. Physiol. Heart
delayed rectifier K+ current. Differential sensitivity to block by Circ. Physiol. 271, H2421 (1996).
class III antiarrhythmic agents. J. Gen. Physiol. 96, 195 (1990). 102. R. L. Roberts, R. E. Fine, and A. Sandra, Receptor-mediated endo-
82. N. Nagy, K. Acsai, A. Kormos, Z. Sebõk, A. S. Farkas, N. Jost, cytosis of transferrin at the blood-brain barrier. J. Cell Sci. 104,
P. P. Nánási, J. G. Papp, A. Varró, and A. Tóth, [Ca2+ ] i-induced 521 (1993).
augmentation of the inward rectifier potassium current (IK1) in 103. A. Scherpereel, R. Wiewrodt, M. Christofidou-Solomidou,
canine and human ventricular myocardium. Pflügers Arch. 465, R. Gervais, J.-C. Murciano, S. M. Albelda, and V. R. Muzykantov,
1621 (2013). Cell-selective intracellular delivery of a foreign enzyme to endothe-
83. B. Yang, H. Lin, J. Xiao, Y. Lu, X. Luo, B. Li, Y. Zhang, C. Xu, lium in vivo using vascular immunotargeting. FASEB J. 15, 416
Y. Bai, and H. Wang, The muscle-specific microRNA miR-1 reg- (2001).
ulates cardiac arrhythmogenic potential by targeting GJA1 and 104. R. Stahn, C. Grittner, R. Zeisig, U. Karsten, S. Felix, and
KCNJ2. Nat. Med. 13, 486 (2007). K. Wenzel, Sialyl Lewisx-liposomes as vehicles for site-directed,
84. A. S. Amin, H. L. Tan, and A. A. Wilde, Cardiac ion channels in E-selectin-mediated drug transfer into activated endothelial cells.
health and disease. Heart Rhythm 7, 117 (2010). Cell. Mol. Life Sci. 58, 141 (2001).
85. F. Hofmann, L. Lacinova, and N. Klugbauer, Reviews of Physiol- 105. K. Zen, M. Okigaki, Y. Hosokawa, Y. Adachi, Y. Nozawa,
ogy, Biochemistry and Pharmacology, edited by M. P. Blaustein, M. Takamiya, T. Tatsumi, N. Urao, K. Tateishi, and T. Takahashi,
R. Greger, and A. Miyajima, Springer, Dordrecht (1999), Vol. 139, Myocardium-targeted delivery of endothelial progenitor cells by
pp. 33–87. ultrasound-mediated microbubble destruction improves cardiac
86. J. C. Braz, K. Gregory, A. Pathak, W. Zhao, B. Sahin, R. Klevitsky, function via an angiogenic response. J. Mol. Cell. Cardiol. 40, 799
T. F. Kimball, J. N. Lorenz, A. C. Nairn, and S. B. Liggett, PKC-á (2006).
regulates cardiac contractility and propensity toward heart failure. 106. P. Ferdinandy, R. Schulz, and G. F. Baxter, Interaction of cardio-
Nat. Med. 10, 248 (2004). vascular risk factors with myocardial ischemia/reperfusion injury,
87. K. R. Sipido, E. Carmeliet, and F. Van de Werf, T -type Ca2+ current preconditioning, and postconditioning. Pharmacol. Rev. 59, 418
as a trigger for Ca2+ release from the sarcoplasmic reticulum in (2007).
guinea-pig ventricular myocytes. J. Physiol. 508, 439 (1998). 107. F. Leuschner and M. Nahrendorf, Molecular Imaging of coronary
88. B. Huang, D. Qin, L. Deng, M. Boutjdir, and N. El-Sherif, Reex- atherosclerosis and myocardial infarction considerations for the
pression of T -type Ca2+ channel gene and current in post-infarction bench and perspectives for the clinic. Circ. Res. 108, 593 (2011).
remodeled rat left ventricle. Cardiovasc. Res. 46, 442 (2000). 108. M. Magro, S. Garg, and P. W. Serruys, Revascularization treatment
89. C. Wang, J. A. Hennessey, R. D. Kirkton, C. Wang, V. Graham, of stable coronary artery disease. Expert Opin. Pharmacother. 12,
R. S. Puranam, P. B. Rosenberg, N. Bursac, and G. S. Pitt, Fibrob- 195 (2011).
last growth factor homologous factor 13 regulates Na+ channels and 109. A. Wu, The role of cardiac troponin in the recent redefinition of
conduction velocity in murine hearts. Circ. Res. 109, 775 (2011). acute myocardial infarction. Clin. Lab. Sci. 17, 50 (2004).
90. M. Goldfarb, Fibroblast growth factor homologous factors: Evolu- 110. G. J. Gross and J. A. Auchampach, Reperfusion injury: Does it
tion, structure, and function. Cytokine Growth Factor Rev. 16, 215 exist? J. Mol. Cell. Cardiol. 42, 12 (2007).
(2005). 111. H. M. Honda, P. Korge, and J. N. Weiss, Mitochondria and
91. M. Butt, G. Dwivedi, A. Blann, O. Khair, and G. Y. H. Lip, ischemia/reperfusion injury. Ann. N. Y. Acad. Sci. 1047, 248 (2005).
Endothelial dysfunction: Methods of assessment implications for 112. A. Rodríguez-Sinovas, Y. Abdallah, H. M. Piper, and D. Garcia-
cardiovascular diseases. Curr. Pharm. Des. 16, 3442 (2010). Dorado, Reperfusion injury as a therapeutic challenge in patients
92. M. Pate, V. Damarla, D. S. Chi, S. Negi, and G. Krishnaswamy, with acute myocardial infarction. Heart Fail. Rev. 12, 207 (2007).
Endothelial cell biology: Role in the inflammatory response. Adv. 113. S. Verma, P. W. Fedak, R. D. Weisel, J. Butany, V. Rao,
Clin. Chem. 52, 109 (2010). A. Maitland, R.-K. Li, B. Dhillon, and T. M. Yau, Fundamentals

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2057


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

of reperfusion injury for the clinical cardiologist. Circulation 105, 136. S. Sharma, C. Christopoulos, N. Kukreja, and D. Gorog, Local drug
2332 (2002). delivery for percutaneous coronary intervention. Pharmacol. Ther.
114. H. K. Eltzschig and T. Eckle, Ischemia and reperfusion [mdash] 129, 260 (2011).
from mechanism to translation. Nat. Med. 17, 1391 (2011). 137. D. B. Fram, J. F. Mitchel, M. A. Azrin, M. S. Chow, D. D. Waters,
115. B. Baban, J. Y. Liu, and M. S. Mozaffari, Pressure overload reg- and R. G. McKay, Local delivery of heparin to balloon angioplasty
ulates expression of cytokines, ãH2AX, and growth arrest- and sites with a new angiotherapy catheter: Pharmacokinetics and effect
DNA-damage inducible protein 153 via glycogen synthase kinase- on platelet deposition in the porcine model. Cathet. Cardiovasc.
3â in ischemic-reperfused hearts. Hypertension 61, 95 (2013). Diagn. 41, 275 (1997).
116. K. H. Lim, D.-J. Lim, and J.-H. Kim, Ginsenoside-Re amelio- 138. D. W. Muller, E. J. Topol, G. D. Abrams, K. P. Gallagher, and
rates ischemia and reperfusion injury in the heart: A hemodynamics S. G. Ellis, Intramural methotrexate therapy for the prevention of
approach. J. Ginseng Res. 37, 283 (2013). neointimal thickening after ballon angioplasty. J. Am. Coll. Cardiol.
117. Y. Ogura, N. Ouchi, K. Ohashi, R. Shibata, Y. Kataoka, 20, 460 (1992).
T. Kambara, T. Kito, S. Maruyama, D. Yuasa, and K. Matsuo, Ther- 139. S. M. Seedial, S. Ghosh, R. S. Saunders, P. A. Suwanabol, X. Shi,
apeutic impact of follistatin-like 1 on myocardial ischemic injury B. Liu, and K. C. Kent, Local drug delivery to prevent restenosis.
in preclinical models clinical perspective. Circulation 126, 1728 J. Vasc. Surg. 57, 1403 (2013).
(2012). 140. M. M. Galagudza, D. V. Korolev, D. L. Sonin, V. N. Postnov, G. V.
118. H. Wang, Q. Zhu, P. Ye, Z. Li, Y. Li, Z. Cao, and L. Shen, Piogli- Papayan, I. S. Uskov, A. V. Belozertseva, and E. V. Shlyakhto,
tazone attenuates myocardial ischemia-reperfusion injury via up- Targeted drug delivery into reversibly injured myocardium with sil-
regulation of ERK and COX-2. Biosci. Trends. 6, 325 (2012). ica nanoparticles: Surface functionalization, natural biodistribution,
119. D. M. Kaye and H. Krum, Drug discovery for heart failure: A new and acute toxicity. Int. J. Nanomedicine 5, 231 (2010).
era or the end of the pipeline? Nat. Rev. Drug Discov. 6, 127 (2007). 141. I. A. Shehata, J. R. Ballard, A. J. Casper, D. Liu, T. Mitchell,
120. D. Levy, S. Kenchaiah, M. G. Larson, E. J. Benjamin, M. J. Kupka, and E. S. Ebbini, Feasibility of targeting atherosclerotic plaques by
K. K. Ho, J. M. Murabito, and R. S. Vasan, Long-term trends in high-intensity–focused ultrasound: An in vivo study. J. Vasc. Interv.
the incidence of and survival with heart failure. N. Engl. J. Med. Radiol. 24, 1880 (2013).
347, 1397 (2002). 142. M. Margaritis, K. Channon, and C. Antoniades, Statins as regula-
121. A. Siryk-Bathgate, S. Dabul, and A. Lymperopoulos, Current and tors of redox state in the vascular endothelium: Beyond lipid low-
future G protein-coupled receptor signaling targets for heart failure ering. Antioxid. Redox Signal (2014), doi:10.1089/ars.2013.5430.
therapy. Drug Des. Devel. Ther. 7, 1209 (2013).
143. N. R. Kuznetsova, E. V. Stepanova, N. M. Peretolchina, D. A.
122. A. Lymperopoulos and A. Bathgate, Arrestins in the cardiovascular
Khochenkov, I. A. Boldyrev, N. V. Bovin, and E. L. Vodovozova,
system. Prog. Mol. Biol. Transl. Sci. 118, 297 (2012).
Targeting liposomes loaded with melphalan prodrug to tumour vas-
123. A. Lymperopoulos, G. Rengo, and W. J. Koch, Adrenergic nervous
culature via the Sialyl Lewis X selectin ligand. J. Drug Target
system in heart failure pathophysiology and therapy. Circ. Res. 113,
(2013), doi:10.3109/1061186X.2013.862805.
739 (2013).
144. S. Ganta, H. Devalapally, A. Shahiwala, and M. Amiji, A review of
124. E. Marbán, Cardiac channelopathies. Nature 415, 213 (2002).
stimuli-responsive nanocarriers for drug and gene delivery. J. Con-
125. J. Wang, H. Wang, Y. Zhang, H. Gao, S. Nattel, and Z. Wang,
trolled Release 126, 187 (2008).
Impairment of HERG K+ channel function by tumor necrosis
145. A.-M. Caminade, R. Laurent, M. Zablocka, and J.-P. Majoral,
factor-á: Role of reactive oxygen species as a mediator. J. Biol.
Organophosphorus chemistry for the synthesis of dendrimers.
Chem. 279, 13289 (2004).
Molecules 17, 13605 (2012).
126. S. E. Iismaa and R. M. Graham, Dissecting cardiac hypertrophy
146. J. M. Criscione, B. L. Le, E. Stern, M. Brennan, C. Rahner,
and signaling pathways evidence for an interaction between multi-
functional G proteins and prostanoids. Circ. Res. 92, 1059 (2003). X. Papademetris, and T. M. Fahmy, Self-assembly of pH-responsive
127. N. Frey and E. Olson, Cardiac hypertrophy: The good, the bad, and fluorinated dendrimer-based particulates for drug delivery and non-
the ugly. Annu. Rev. Physiol. 65, 45 (2003). invasive imaging. Biomaterials 30, 3946 (2009).
128. M. J. Koren, R. B. Devereux, P. N. Casale, D. D. Savage, and J. H. 147. D. A. Tomalia, A. M. Naylor, and W. A. Goddard, Starburst den-
Laragh, Relation of left ventricular mass and geometry to morbidity drimers: Molecular-level control of size, shape, surface chemistry,
and mortality in uncomplicated essential hypertension. Ann. Intern. topology, and flexibility from atoms to macroscopic matter. Angew.
Med. 114, 345 (1991). Chem. Int. Ed. Engl. 29, 138 (1990).
129. T. A. McKinsey and D. A. Kass, Small-molecule therapies for car- 148. G. R. Newkome, H. J. Kim, K. H. Choi, and C. N. Moorefield, Syn-
diac hypertrophy: Moving beneath the cell surface. Nat. Rev. Drug thesis of neutral metallodendrimers possessing adamantane termini:
Discov. 6, 617 (2007). Supramolecular assembly with â-cyclodextrin. Macromolecules 37,
130. T. Thum, D. Catalucci, and J. Bauersachs, MicroRNAs: Novel reg- 6268 (2004).
ulators in cardiac development and disease. Cardiovasc. Res. 79, 149. E. Buhleier, W. Wehner, and F. Vögtle, Cascade and nonskid-
562 (2008). chain-like synthesis of molecular cavity topologies. Synthesis 2,
131. J. F. O’Sullivan, K. Martin, and N. M. Caplice, Microribonucleic 155 (1978).
acids for prevention of plaque rupture and in-stent restenosis “a 150. F. Vogtle, G. Richardt, and N. Werner, Dendrimer Chemistry: Con-
finger in the dam.” J. Am. Coll. Cardiol. 57, 383 (2011). cepts, Syntheses, Properties, Applications, Wiley-VCH, Weinheim
132. L.-J. Chen, S. H. Lim, Y.-T. Yeh, S.-C. Lien, and J.-J. Chiu, Roles (2009).
of microRNAs in atherosclerosis and restenosis. J. Biomed. Sci. 19, 151. E. Meijer, J. Jansen, and E. De Brabander-van den Berg, Encapsu-
79 (2012). lation of guest molecules into a dendritic box. Science 266, 1226
133. J. W. Jukema, J. J. Verschuren, T. A. Ahmed, and P. H. Quax, (1994).
Restenosis after PCI. Part 1: Pathophysiology and risk factors. Nat. 152. R. W. Scott, O. M. Wilson, and R. M. Crooks, Synthesis, character-
Rev. Cardiol. 9, 53 (2011). ization, and applications of dendrimer-encapsulated nanoparticles.
134. T. P. Smith, Atherosclerosis and restenosis: An inflammatory issue. J. Phys. Chem. B 109, 692 (2005).
Radiology 225, 10 (2002). 153. J. F. Kukowska-Latallo, A. U. Bielinska, J. Johnson, R. Spindler,
135. P. M. Hamon, E. Lécluse, J.-P. Monassier, G. Grollier, and J.-C. D. A. Tomalia, and J. R. Baker, Efficient transfer of genetic material
Potier, Pharmacological approaches to the prevention of restenosis into mammalian cells using Starburst polyamidoamine dendrimers.
after coronary angioplasty. Drugs Aging 13, 291 (1998). Proc. Natl. Acad. Sci. USA 93, 4897 (1996).

2058 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

154. B. Chanyshev, A. Shainberg, A. Isak, A. Litinsky, Y. Chepurko, 171. V. Torchilin, A. Klibanov, L. Huang, S. O’Donnell, N. Nossiff,
D. K. Tosh, K. Phan, Z.-G. Gao, E. Hochhauser, and K. A. and B. Khaw, Targeted accumulation of polyethylene glycol-coated
Jacobson, Anti-ischemic effects of multivalent dendrimeric A3 immunoliposomes in infarcted rabbit myocardium. FASEB J. 6,
adenosine receptor agonists in cultured cardiomyocytes and in the 2716 (1992).
isolated rat heart. Pharmacol. Res. 65, 338 (2012). 172. V. P. Torchilin, J. Narula, E. Halpern, and B. A. Khaw,
155. Y. Wang, Y. Bai, C. Price, P. Boros, L. Qin, A. U. Bielinska, J. F. Poly(ethylene glycol)-coated anti-cardiac myosin immunolipo-
Kukowska-Latallo, J. R. Baker, and J. S. Bromberg, Combination somes: Factors influencing targeted accumulation in the infarcted
of electroporation and DNA/dendrimer complexes enhances gene myocardium. Biochim. Biophys. Acta 1279, 75 (1996).
transfer into murine cardiac transplants. Am. J. Transplantation 1, 173. J. Sanchis, F. Canal, R. Lucas, and M. J. Vicent, Polymer-drug con-
334 (2001). jugates for novel molecular targets. Nanomedicine 5, 915 (2010).
156. T. A. Johnson, N. A. Stasko, J. L. Matthews, W. E. Cascio, E. L. 174. S. J. Williams, Q. Wang, R. R. MacGregor, T. J. Siahaan, L. Stehno-
Holmuhamedov, C. B. Johnson, and M. H. Schoenfisch, Reduced Bittel, and C. Berkland, Adhesion of pancreatic beta cells to
ischemia/reperfusion injury via glutathione-initiated nitric oxide- biopolymer films. Biopolymers 91, 676 (2009).
releasing dendrimers. Nitric Oxide 22, 30 (2010). 175. X. Hu, Y. Tang, Q. Wang, Y. Li, J. Yang, Y. Du, and J. F. Kennedy,
157. M.-R. Micheli, R. Bova, A. Magini, M. Polidoro, and C. Emiliani, Rheological behaviour of chitin in NaOH/urea aqueous solution.
Lipid-based nanocarriers for CNS-targeted drug delivery. Recent Carbohydr. Polym. 83, 1128 (2011).
Pat. CNS Drug Discov. 7, 71 (2012). 176. H. Ringsdorf. Structure and properties of pharmacologically active
158. A. Samad, Y. Sultana, and M. Aqil, Liposomal drug delivery sys- polymers. J. Polym. Sci. 51, 135 (1975).
tems: An update review. Curr. Drug Deliv. 4, 297 (2007). 177. C. Li and S. Wallace, Polymer-drug conjugates: recent development
159. M. Shao, S.-L. Sun, M.-H. Li, B.-X. Li, H. Yu, Z.-Y. Shen, Y.-C. in clinical oncology. Adv. Drug Deliv. Rev. 60, 886 (2008).
Ren, Z.-F. Hao, N.-D. Chang, and H.-S. Peng, The liposomal 178. H. Maeda, J. Wu, T. Sawa, Y. Matsumura, and K. Hori, Tumor
daunorubicin plus tamoxifen: Improving the stability, uptake, and vascular permeability and the EPR effect in macromolecular thera-
biodistribution of carriers. J. Liposome Res. 22, 168 (2012). peutics: A review. J. Controlled Release 65, 271 (2000).
160. H. Peng, D. Du, and J. Zhang, Liposomes modified with p- 179. M. Galagudza, Novel Strategies in Ischemic Heart Disease, edited
aminophenyl-á-D-mannopyranoside: A promising delivery system by U. Lakshmanadoss, InTech, Rijeka (2012), Chap. 14.
in targeting the brain. Ther. Deliv. 4, 1475 (2013). 180. R. Misra, M. Upadhyay, and S. Mohanty, Nanoparticles as carriers
161. Z.-F. Hao, Y.-X. Cui, M.-H. Li, D. Du, M.-F. Liu, H.-Q. Tao, S. Li, for chemotherapeutic drugs: A review. J. Nanopharm. Drug Deliv.
F.-Y. Cao, Y.-L. Chen, and X.-H. Lei, Liposomes modified with 1, 103 (2013).
P-aminophenyl-á-D-mannopyranoside: A carrier for targeting cere- 181. C. Vilos and L. Velasquez, Therapeutic strategies based on poly-
bral functional regions in mice. Eur. J. Pharm. Biopharm. 84, 505 meric microparticles. Biomed. Res. Int. 2012, 672760 (2012).
(2013). 182. J. C. Sy, G. Seshadri, S. C. Yang, M. Brown, T. Oh, S. Dikalov,
162. L. Peiser, S. Mukhopadhyay, and S. Gordon, Scavenger receptors N. Murthy, and M. E. Davis, Sustained release of a p38 inhibitor
in innate immunity. Curr. Opin. Immunol. 14, 123 (2002). from non-inflammatory microspheres inhibits cardiac dysfunction.
163. D. D. Verma, T. S. Levchenko, E. A. Bernstein, D. Mongayt, and Nat. Mater. 7, 863 (2008).
V. P. Torchilin, ATP-loaded immunoliposomes specific for cardiac 183. A. Lamprecht, H. R. Torres, U. Schäfer, and C.-M. Lehr,
myosin provide improved protection of the mechanical functions of Biodegradable microparticles as a two-drug controlled release for-
myocardium from global ischemia in an isolated rat heart model. mulation: A potential treatment of inflammatory bowel disease.
J. Drug Target. 14, 273 (2006). J. Controlled Release 69, 445 (2000).
164. R. C. Scott, J. M. Rosano, Z. Ivanov, B. Wang, P. L.-G. Chong, 184. F. R. Formiga, B. Pelacho, E. Garbayo, G. Abizanda, J. J. Gavira,
A. C. Issekutz, D. L. Crabbe, and M. F. Kiani, Targeting VEGF- T. Simon-Yarza, M. Mazo, E. Tamayo, C. Jauquicoa, and C. Ortiz-
encapsulated immunoliposomes to MI heart improves vascularity de-Solorzano, Sustained release of VEGF through PLGA micropar-
and cardiac function. FASEB J. 23, 3361 (2009). ticles improves vasculogenesis and tissue remodeling in an acute
165. B. Wang, J. Rosano, D. L. Crabbe, and M. F. Kiani, Oxygen Trans- myocardial ischemia–reperfusion model. J. Controlled Release 147,
port to Tissue XXXIV, edited by W. J. Welch, Springer, New York 30 (2010).
(2013), pp. 307–314. 185. M. Arruebo, M. Galán, N. Navascués, C. Téllez, C. Marquina,
166. L. Kalinowski, L. W. Dobrucki, D. F. Meoli, D. P. Dione, M. M. M. R. Ibarra, and J. Santamaría, Development of magnetic nanos-
Sadeghi, J. A. Madri, and A. J. Sinusas, Targeted imaging of tructured silica-based materials as potential vectors for drug-
hypoxia-induced integrin activation in myocardium early after delivery applications. Chem. Mater. 18, 1911 (2006).
infarction. J. Appl. Physiol. 104, 1504 (2008). 186. C. Barbe, J. Bartlett, L. Kong, K. Finnie, H. Q. Lin, M. Larkin,
167. D. F. Meoli, M. M. Sadeghi, S. Krassilnikova, B. N. Bourke, F. J. S. Calleja, A. Bush, and G. Calleja, Silica particles: A novel drug-
Giordano, D. P. Dione, H. Su, D. S. Edwards, S. Liu, and T. D. delivery system. Adv. Mater. 16, 1959 (2004).
Harris, Noninvasive imaging of myocardial angiogenesis follow- 187. B. G. Trewyn, J. A. Nieweg, Y. Zhao, and V. S.-Y. Lin, Biocompati-
ing experimental myocardial infarction. J. Clin. Invest. 113, 1684 ble mesoporous silica nanoparticles with different morphologies for
(2004). animal cell membrane penetration. Chem. Eng. J. 137, 23 (2008).
168. T. Dvir, M. Bauer, A. Schroeder, J. H. Tsui, D. G. Anderson, 188. P. Kortesuo, M. Ahola, S. Karlsson, I. Kangasniemi, A. Yli-Urpo
R. Langer, R. Liao, and D. S. Kohane, Nanoparticles targeting the and J. Kiesvaara, Silica xerogel as an implantable carrier for con-
infarcted heart. Nano Lett. 11, 4411 (2011). trolled drug delivery—evaluation of drug distribution and tissue
169. H. Takahama, T. Minamino, H. Asanuma, M. Fujita, T. Asai, effects after implantation. Biomaterials 21, 193 (2000).
M. Wakeno, H. Sasaki, H. Kikuchi, K. Hashimoto, and N. Oku, 189. T. Neuberger, B. Schöpf, H. Hofmann, M. Hofmann, and B. von
Prolonged targeting of ischemic/reperfused myocardium by lipo- Rechenberg, Superparamagnetic nanoparticles for biomedical appli-
somal adenosine augments cardioprotection in rats. J. Am. Coll. cations: Possibilities and limitations of a new drug delivery system.
Cardiol. 53, 709 (2009). J. Magn. Magn. Mater. 293, 483 (2005).
170. L. E. Paulis, T. Geelen, M. T. Kuhlmann, B. F. Coolen, M. Schäfers, 190. Y. Sun, L. Duan, Z. Guo, Y. DuanMu, M. Ma, L. Xu, Y. Zhang, and
K. Nicolay, and G. J. Strijkers, Distribution of lipid-based nanopar- N. Gu, An improved way to prepare superparamagnetic magnetite-
ticles to infarcted myocardium with potential application for MRI- silica core–shell nanoparticles for possible biological application.
monitored drug delivery. J. Controlled Release 162, 276 (2012). J. Magn. Magn. Mater. 285, 65 (2005).

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2059


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

191. D. J. Bharali, I. Klejbor, E. K. Stachowiak, P. Dutta, I. Roy, 210. W. Li, C. Nesselmann, Z. Zhou, L.-L. Ong, F. Öri, G. Tang,
N. Kaur, E. J. Bergey, P. N. Prasad, and M. K. Stachowiak, Organ- A. Kaminski, K. Lützow, A. Lendlein, and A. Liebold, Gene deliv-
ically modified silica nanoparticles: A nonviral vector for in vivo ery to the heart by magnetic nanobeads. J. Magn. Magn. Mater.
gene delivery and expression in the brain. Proc. Natl. Acad. Sci. 311, 336 (2007).
USA 102, 11539 (2005). 211. Y. Zhang, W. Li, L. Ou, W. Wang, E. Delyagina, C. Lux,
192. H. Mansur, R. Oréfice, W. Vasconcelos, Z. Lobato, and H. Sorg, K. Riehemann, G. Steinhoff, and N. Ma, Targeted delivery
L. Machado, Biomaterial with chemically engineered surface for of human VEGF gene via complexes of magnetic nanoparticle-
protein immobilization. J. Mater. Sci. Mater. Med. 16, 333 (2005). adenoviral vectors enhanced cardiac regeneration. PloS One 7,
193. R. Mortera, J. Vivero-Escoto, I. I. Slowing, E. Garrone, B. Onida, e39490 (2012).
and V. S.-Y. Lin, Cell-induced intracellular controlled release 212. P. Jasinski, T. Suzuki, and H. U. Anderson, Nanocrystalline
of membrane impermeable cysteine from a mesoporous silica undoped ceria oxygen sensor. Sensors Actuators B Chem. 95, 73
nanoparticle-based drug delivery system. Chem. Commun. 45, 3219 (2003).
(2009). 213. T. Masui, T. Ozaki, K.-I. Machida, and G.-Y. Adachi, Prepara-
194. J. M. Rosenholm, E. Peuhu, J. E. Eriksson, C. Sahlgren, and tion of ceria–zirconia sub-catalysts for automotive exhaust cleaning.
M. Lindén, Targeted intracellular delivery of hydrophobic agents J. Alloys Compd. 303, 49 (2000).
using mesoporous hybrid silica nanoparticles as carrier systems. 214. J. C. Yu, L. Zhang, and J. Lin, Direct sonochemical preparation of
Nano Lett. 9, 3308 (2009). high-surface-area nanoporous ceria and ceria–zirconia solid solu-
195. T. Xia, M. Kovochich, M. Liong, H. Meng, S. Kabehie, S. George, tions. J. Colloid Interface Sci. 260, 240 (2003).
J. I. Zink, and A. E. Nel, Polyethyleneimine coating enhances the 215. A. Karakoti, N. Monteiro-Riviere, R. Aggarwal, J. Davis,
cellular uptake of mesoporous silica nanoparticles and allows safe R. Narayan, W. Self, J. McGinnis, and S. Seal, Nanoceria as antiox-
delivery of siRNA and DNA constructs. ACS Nano 3, 3273 (2009). idant: Synthesis and biomedical applications. JOM 60, 33 (2008).
196. K. S. Finnie, D. J. Waller, F. L. Perret, A. M. Krause-Heuer, H. Q. 216. P. D. Ray, B.-W. Huang, and Y. Tsuji, Reactive oxygen species
Lin, J. V. Hanna, and C. J. Barbé, Biodegradability of sol–gel sil- (ROS) homeostasis and redox regulation in cellular signaling. Cell
ica microparticles for drug delivery. J. Solgel Sci. Technol. 49, 12 Signal. 24, 981 (2012).
(2009). 217. K. S. Ahn and B. B. Aggarwal, Transcription factor NF-r: A sen-
197. M. Galagudza, D. Korolev, D. Sonin, I. Alexandrov, S. Minasian, sor for smoke and stress signals. Ann. N. Y. Acad. Sci. 1056, 218
V. Postnov, E. Kirpicheva, G. Papayan, and I. Uskov, Passive and (2005).
active target delivery of drugs to ischemic myocardium. Bull. Exp.
218. G. Hall, J. D. Hasday, and T. B. Rogers, Regulating the regulator:
Biol. Med. 152, 105 (2011).
NF-êB signaling in heart. J. Mol. Cell. Cardiol. 41, 580 (2006).
198. J. Duan, Y. Yu, Y. Li, Y. Yu, and Z. Sun, Cardiovascular toxicity
219. J. Niu, A. Azfer, L. M. Rogers, X. Wang, and P. E. Kolattukudy,
evaluation of silica nanoparticles in endothelial cells and zebrafish
Cardioprotective effects of cerium oxide nanoparticles in a trans-
model. Biomaterials 34, 5853 (2013).
genic murine model of cardiomyopathy. Cardiovasc. Res. 73, 549
199. M. Arruebo, R. Fernández-Pacheco, M. R. Ibarra, and
(2007).
J. Santamaría, Magnetic nanoparticles for drug delivery. Nano
220. F. R. Cassee, A. Campbell, A. J. F. Boere, S. G. McLean, R. Duffin,
Today 2, 22 (2007).
P. Krystek, I. Gosens, and M. R. Miller, The biological effects of
200. Q. Cao, X. Han, and L. Li, Enhancement of the efficiency of mag-
subacute inhalation of diesel exhaust following addition of cerium
netic targeting for drug delivery: Development and evaluation of
oxide nanoparticles in atherosclerosis-prone mice. Environ. Res.
magnet system. J. Magn. Magn. Mater. 323, 1919 (2011).
115, 1 (2012).
201. R. Asmatulu, M. A. Zalich, R. O. Claus, and J. S. Riffle, Synthesis,
221. S. Shah, W. Li, C.-P. Huang, O. Jung, and C. Ni, Study of Nd3+ ,
characterization and targeting of biodegradable magnetic nanocom-
posite particles by external magnetic fields. J. Magn. Magn. Mater. Pd2+ , Pt4+ , and Fe3+ dopant effect on photoreactivity of TiO2
292, 108 (2005). nanoparticles. Proc. Natl. Acad. Sci. USA 99, 6482 (2002).
202. J. H. Chang, K. H. Kang, J. Choi, and Y. K. Jeong, High effi- 222. T. Paunesku, T. Rajh, G. Wiederrecht, J. Maser, N. S. S. Vogt,
ciency protein separation with organosilane assembled silica coated Cacute, Proti, M. Cacute, B. Lai, J. Oryhon, and G. W. Marion
magnetic nanoparticles. Superlattices Microstruct. 44, 442 (2008). Thurnauer, Biology of TiO2 –oligonucleotide nanocomposites. Nat.
203. J. Chomoucka, J. Drbohlavova, D. Huska, V. Adam, R. Kizek, and Mater. 2, 343 (2003).
J. Hubalek, Magnetic nanoparticles and targeted drug delivering. 223. G. Wu, T. Nishikawa, B. Ohtani, and A. Chen, Synthesis and char-
Pharmacol. Res. 62, 144 (2010). acterization of carbon-doped TiO2 nanostructures with enhanced
204. B.-F. Pan, F. Gao, and H.-C. Gu, Dendrimer modified magnetite visible light response. Chem. Mater. 19, 4530 (2007).
nanoparticles for protein immobilization. J. Colloid Interface Sci. 224. D. B. Warheit, T. R. Webb, K. L. Reed, S. Frerichs, and C. M.
284, 1 (2005). Sayes, Pulmonary toxicity study in rats with three forms of
205. U. Tamer, Y. Gündoǧdu, Ý. H. Boyacý, and K. Pekmez, Synthe- ultrafine-TiO2 particles: Differential responses related to surface
sis of magnetic core–shell Fe3 O4 -Au nanoparticle for biomolecule properties. Toxicology 230, 90 (2007).
immobilization and detection. J. Nanopart. Res. 12, 1187 (2010). 225. M. Hidalgo, M. Maicu, J. Navío, and G. Colón, Photocatalytic
206. P. Grassi-Schultheiss, F. Heller, and J. Dobson, Analysis of mag- properties of surface modified platinised TiO2 : Effects of particle
netic material in the human heart, spleen and liver. Biometals 10, size and structural composition. Catalysis Today 129, 43 (2007).
351 (1997). 226. G. N. Kryukova, G. A. Zenkovets, A. A. Shutilov, M. Wilde,
207. A. Figuerola, R. Di Corato, L. Manna, and T. Pellegrino, From iron K. Günther, D. Fassler, and K. Richter, Structural peculiarities of
oxide nanoparticles towards advanced iron-based inorganic materi- TiO2 and Pt/TiO2 catalysts for the photocatalytic oxidation of aque-
als designed for biomedical applications. Pharmacol. Res. 62, 126 ous solution of acid orange 7 dye upon ultraviolet light. Appl. Catal.
(2010). B 71, 169 (2007).
208. E. Delyagina, W. Li, N. Ma, and G. Steinhoff, Magnetic targeting 227. A. Gupta, S. Asthana, R. Konwar, and M. Chourasia, An insight
strategies in gene delivery. Nanomedicine 6, 1593 (2011). into potential of nanoparticles-assisted chemotherapy of cancer
209. W. Li, N. Ma, L. L. Ong, A. Kaminski, C. Skrabal, M. Ugurlucan, using gemcitabine and its fatty acid prodrug: A comparative study.
P. Lorenz, H. H. Gatzen, K. Lützow, and A. Lendlein, Enhanced J. Biomed. Nanotechnol. 9, 915 (2013).
thoracic gene delivery by magnetic nanobead-mediated vector. 228. C. A. Dehelean, S. Feflea, D. Gheorgheosu, S. Ganta, A. M.
J. Gene. Med. 10, 897 (2008). Cimpean, D. Muntean, and M. M. Amiji, Anti-angiogenic and

2060 J. Biomed. Nanotechnol. 10, 2038–2062, 2014


Liu et al. Heart-Targeted Nanoscale Drug Delivery Systems

anti-cancer evaluation of betulin nanoemulsion in chicken chorioal- 245. N. Nasongkla, E. Bey, J. Ren, H. Ai, C. Khemtong, J. S. Guthi,
lantoic membrane and skin carcinoma in balb/c mice. J. Biomed. S.-F. Chin, A. D. Sherry, D. A. Boothman, and J. Gao, Multifunc-
Nanotechnol. 9, 577 (2013). tional polymeric micelles as cancer-targeted, MRI-ultrasensitive
229. Z. He, Y. Yu, Y. Zhang, Y. Yan, Y. Zheng, J. He, Y. Xie, G. He, drug delivery systems. Nano Lett. 6, 2427 (2006).
Y. Wei, and X. Song, Gene delivery with active targeting to ovarian 246. R. Virmani, A. Farb, G. Guagliumi, and F. D. Kolodgie, Drug-
cancer cells mediated by folate receptor. J. Biomed. Nanotechnol. eluting stents: Caution and concerns for long-term outcome. Coron.
9, 833 (2013). Artery Dis. 15, 313 (2004).
230. A. Anitha, S. Uthaman, S. V. Nair, R. Jayakumar, and 247. X. Zheng, Y. Wang, Z. Lan, Y. Lyu, G. Feng, Y. Zhang, S. Tagusari,
V.-K. Lakshmanan, Enhanced delivery system of flutamide E. Kislauskis, M. P. Robich, and S. McCarthy, Improved biocom-
loaded chitosan-dextran sulphate nanoparticles for prostate cancer. patibility of poly(lactic-co-glycolic acid) and poly-L-lactic acid
J. Biomed. Nanotechnol. 9, 335 (2013). blended with nanoparticulate amorphous calcium phosphate in vas-
231. H.-S. Peng, X.-J. Liu, G.-X. Lv, B. Sun, Q.-F. Kong, D.-X. Zhai, cular stent applications. J. Biomed. Nanotechnol. 10, 900 (2014).
Q. Wang, W. Zhao, G.-Y. Wang, and D.-D. Wang, Voriconazole 248. P. W. Serruys, Y. Onuma, J. A. Ormiston, B. de Bruyne, E. Regar,
into PLGA nanoparticles: Improving agglomeration and antifungal D. Dudek, L. Thuesen, P. C. Smits, B. Chevalier, and D. McClean,
efficacy. Int. J. Pharm. 352, 29 (2008). Evaluation of the second generation of a bioresorbable everolimus
232. H.-Q. Tao, Q. Meng, M.-H. Li, H. Yu, M.-F. Liu, D. Du, S.-L. Sun, drug-eluting vascular scaffold for treatment of de novo coronary
H.-C. Yang, Y.-M. Wang, and W. Ye, HP-â-CD-PLGA nanopar- artery stenosis clinical perspective: Six-month clinical and imaging
ticles improve the penetration and bioavailability of puerarin and outcomes. Circulation 122, 2301 (2010).
enhance the therapeutic effects on brain ischemia–reperfusion 249. A. M. Lincoff, J. G. Furst, S. G. Ellis, R. J. Tuch, and E. J. Topol,
injury in rats. Naunyn Schmiedebergs Arch. Pharmacol. 386, 61 Sustained local delivery of dexamethasone by a novel intravascular
(2013). eluting stent to prevent restenosis in the porcine coronary injury
233. Q. Wang, Z. Gu, S. Jamal, M. S. Detamore, and C. Berkland, model. J. Am. Coll. Cardiol. 29, 808 (1997).
Hybrid hydroxyapatite nanoparticle colloidal gels are injectable 250. D. Böse, H. Eggebrecht, and R. Erbel, Absorbable metal stent in
fillers for bone tissue engineering. Tissue Eng. A 19, 2586 (2013). human coronary arteries: Imaging with intravascular ultrasound.
234. R. Wahab, S. Dwivedi, A. Umar, S. Singh, I. Hwang, H.-S. Shin, Heart 92, 892 (2006).
J. Musarrat, A. A. Al-Khedhairy, and Y.-S. Kim, ZnO nanoparticles 251. H. Wieneke, O. Dirsch, T. Sawitowski, Y. L. Gu, H. Brauer,
induce oxidative stress in Cloudman S91 melanoma cancer cells. U. Dahmen, A. Fischer, S. Wnendt, and R. Erbel, Synergistic
J. Biomed. Nanotechnol. 9, 441 (2013). effects of a novel nanoporous stent coating and tacrolimus on
235. M. Sharma, R. Malik, A. Verma, P. Dwivedi, G. S. Banoth, intima proliferation in rabbits. Cathet. Cardiovasc. Interv. 60, 399
N. Pandey, J. Sarkar, P. R. Mishra, and A. K. Dwivedi, Folic acid (2003).
conjugated guar gum nanoparticles for targeting methotrexate to 252. Z. Chen, M. Xie, X. Wang, Q. Lv, and S. Ding, Efficient gene deliv-
colon cancer. J. Biomed. Nanotechnol. 9, 96 (2013). ery to myocardium with ultrasound targeted microbubble destruc-
236. A. M. Prantner, C. V. Nguyen, and N. Scholler, Facile immuno- tion and polyethylenimine. J. Huazhong Univ. Sci. Technolog Med.
targeting of nanoparticles against tumor antigens using site-specific Sci. 28, 613 (2008).
biotinylated antibody fragments. J. Biomed. Nanotechnol. 9, 1686 253. Z.-Y. Chen, K. Liang, R.-X. Qiu, and L.-P. Luo, Ultrasound- and
(2013). liposome microbubble-mediated targeted gene transfer to cardiomy-
237. V. Torchilin, Micellar nanocarriers: Pharmaceutical perspectives. ocytes in vivo accompanied by polyethylenimine. J. Ultrasound
Pharm. Res. 24, 1 (2007). Med. 30, 1247 (2011).
238. C. Garnacho, S. M. Albelda, V. R. Muzykantov, and S. Muro, 254. E. Stride, Physical principles of microbubbles for ultrasound imag-
Differential intra-endothelial delivery of polymer nanocarriers tar- ing and therapy. Cerebrovasc. Dis. 27, 1 (2009).
geted to distinct PECAM-1 epitopes. J. Controlled Release 130, 255. H. Fujii, Z. Sun, S.-H. Li, J. Wu, S. Fazel, R. D. Weisel,
226 (2008). H. Rakowski, J. Lindner, and R.-K. Li, Ultrasound-targeted gene
239. I. Somasuntharam, A. V. Boopathy, R. S. Khan, M. D. Martinez, delivery induces angiogenesis after a myocardial infarction in mice.
M. E. Brown, N. Murthy, and M. E. Davis, Delivery of Nox2- JACC Cardiovasc. Imaging 2, 869 (2009).
NADPH oxidase siRNA with polyketal nanoparticles for improving 256. I. Lentacker, N. Wang, R. E. Vandenbroucke, J. Demeester, S. C. De
cardiac function following myocardial infarction. Biomaterials 34, Smedt, and N. N. Sanders, Ultrasound exposure of lipoplex loaded
7790 (2013). microbubbles facilitates direct cytoplasmic entry of the lipoplexes.
240. M. J. Lipinski, J. C. Frias, V. Amirbekian, K. C. Briley-Saebo, Mol. Pharm. 6, 457 (2009).
V. Mani, D. Samber, A. Abbate, J. G. S. Aguinaldo, D. Massey, and 257. Z.-Y. Chen, Y. Lin, F. Yang, L. Jiang, and S. P. Ge, Gene therapy for
V. Fuster, Macrophage-specific lipid-based nanoparticles improve cardiovascular disease mediated by ultrasound and microbubbles.
cardiac magnetic resonance detection and characterization of Cardiovasc. Ultrasound 11, 11 (2013).
human atherosclerosis. JACC Cardiovasc. Imaging 2, 637 (2009). 258. W. Li, F. Kong, X. Li, X. Dai, X. Liu, Q. Zheng, R. Wu,
241. Y. Nagasaki, T. Kutsuna, M. Iijima, M. Kato, K. Kataoka, X. Zhou, F. Lü, and B. Chang, Gene therapy following subreti-
S. Kitano, and Y. Kadoma, Formyl-ended heterobifunctional nal AAV5 vector delivery is not affected by a previous intravitreal
poly(ethylene oxide): Synthesis of poly(ethylene oxide) with a AAV5 vector administration in the partner eye. Mol. Vis. 15, 267
formyl group at one end and a hydroxyl group at the other end. (2009).
Bioconjug. Chem. 6, 231 (1995). 259. K. Stieger, J. Schroeder, N. Provost, A. Mendes-Madeira,
242. C. Perez, A. Sanchez, D. Putnam, D. Ting, R. Langer, and B. Belbellaa, G. Le Meur, M. Weber, J.-Y. Deschamps, B. Lorenz,
M. Alonso, Poly(lactic acid)-poly(ethylene glycol) nanoparticles as and P. Moullier, Detection of intact rAAV particles up to 6 years
new carriers for the delivery of plasmid DNA. J. Controlled Release after successful gene transfer in the retina of dogs and primates.
75, 211 (2001). Mol. Ther. 17, 516 (2008).
243. S. Bontha, A. V. Kabanov, and T. K. Bronich, Polymer micelles 260. J. R. Lindner, J. Song, J. Christiansen, A. L. Klibanov, F. Xu, and
with cross-linked ionic cores for delivery of anticancer drugs. K. Ley, Ultrasound assessment of inflammation and renal tissue
J. Controlled Release 114, 163 (2006). injury with microbubbles targeted to P-selectin. Circulation 104,
244. Z. Gao, A. N. Lukyanov, A. Singhal, and V. P. Torchilin, 2107 (2001).
Diacyllipid-polymer micelles as nanocarriers for poorly soluble 261. G. E. Weller, E. Lu, M. M. Csikari, A. L. Klibanov, D. Fischer,
anticancer drugs. Nano Lett. 2, 979 (2002). W. R. Wagner, and F. S. Villanueva, Ultrasound imaging of acute

J. Biomed. Nanotechnol. 10, 2038–2062, 2014 2061


Heart-Targeted Nanoscale Drug Delivery Systems Liu et al.

cardiac transplant rejection with microbubbles targeted to intercel- and H. Akashi, Therapeutic angiogenesis for patients with limb
lular adhesion molecule-1. Circulation 108, 218 (2003). ischaemia by autologous transplantation of bone-marrow cells:
262. C.-H. Su, Y.-J. Wu, H.-H. Wang, and H.-I. Yeh, Nonviral gene A pilot study and a randomised controlled trial. Lancet 360, 427
therapy targeting cardiovascular system. Am. J. Physiol. Heart Circ. (2002).
Physiol. 303, H629 (2012). 268. E. J. Topol, Coronary-artery stents–gauging, gorging, and gouging.
263. S. Chen, M. Shimoda, J. Chen, and P. Grayburn, Stimulation N. Engl. J. Med. 339, 1702 (1998).
of adult resident cardiac progenitor cells by durable myocardial 269. R. Tung, S. Kaul, G. A. Diamond, and P. K. Shah, Narrative
expression of thymosin beta 4 with ultrasound-targeted microbub- review: Drug-eluting stents for the management of restenosis:
ble delivery. Gene Ther. 20, 225 (2013). A critical appraisal of the evidence. Ann. Intern. Med. 144, 913
264. A. V. Alexandrov, C. A. Molina, J. C. Grotta, Z. Garami, S. R. (2006).
Ford, J. Alvarez-Sabin, J. Montaner, M. Saqqur, A. M. Demchuk, 270. B. J. Aungst, N. J. Rogers, and E. Shefter, Comparison of nasal,
and L. A. Moyé, Ultrasound-enhanced systemic thrombolysis for rectal, buccal, sublingual and intramuscular insulin efficacy and the
acute ischemic stroke. N. Engl. J. Med. 351, 2170 (2004). effects of a bile salt absorption promoter. J. Pharmacol. Exp. Ther.
265. U. Rosenschein, A. Roth, T. Rassin, S. Basan, S. Laniado, and H. I. 244, 23 (1988).
Miller, Analysis of coronary ultrasound thrombolysis endpoints in 271. Z. Shao and A. K. Mitra, Nasal membrane and intracellular protein
acute myocardial infarction (ACUTE trial) results of the feasibility and enzyme release by bile salts and bile salt-fatty acid mixed
phase. Circulation 95, 1411 (1997). micelles: Correlation with facilitated drug transport. Pharm. Res.
266. C. Wissgott, A. Richter, P. Kamusella, and H. J. Steinkamp, Treat- 9, 1184 (1992).
ment of critical limb ischemia using ultrasound-enhanced thrombol- 272. P. Tengamnuay and A. K. Mitra, Bile salt–fatty acid mixed micelles
ysis (PARES Trial): Final results. J. Endovasc. Ther. 14, 438 (2007). as nasal absorption promoters of peptides. I. Effects of Ionic
267. E. Tateishi-Yuyama, H. Matsubara, T. Murohara, U. Ikeda, strength, adjuvant composition, and lipid structure on the nasal
S. Shintani, H. Masaki, K. Amano, Y. Kishimoto, K. Yoshimoto, absorption of [D-Arg2] kyotorphin. Pharm. Res. 7, 127 (1990).

2062 J. Biomed. Nanotechnol. 10, 2038–2062, 2014

View publication stats

You might also like