ArticlePDF AvailableLiterature Review

Recent Advancement in the Treatment of Cardiovascular Diseases: Conventional Therapy to Nanotechnology

Authors:
  • Centre for Food Biology & Environment Studies
  • University of Iowa, Iowa City, USA

Abstract and Figures

Cardiovascular disease (CVD), accounting around 30% of deaths worldwide, collectively comprised of disorders affecting the heart and blood vessels as well as their associated adverse conditions. Despite outstanding progress in the area of the treatments of CVDs, significant challenges remain in designing of efficient delivery systems for myocardial therapy. Moreover, current therapy for CVDs is limited due to various clinical complications such as systemic toxicity, stent thrombosis, etc. Molecular and nanotechnology approaches provide the tools to explore such frontiers of biomedical science at the cellular level and thus offer unique features for potential application in the field of cardiac therapy. In this review, recent advances in CVD related risk factors, chronic inflammation, and their therapeutic modalities such as stem cell therapy, gene delivery, tissue factor (TF) inhibitors, miRNAs, leukotriene modifiers, thrombolytic agents etc., in modern molecular aspects are discussed. Moreover, nanoparticle based drug delivery, nanocarriers as molecular imaging, and the various challenges of myocardial tissue engineering aspects have been summarized. All these aspects may provide additional therapeutic substitutes in clinical trials for the registration of new drugs.
Content may be subject to copyright.
Send Ord ers for Reprints to reprints@benthamscience.ae
Current Pharmaceutical Design, 2015, 21, 4479-4497 4479
Recent Advancement in the Treatment of Cardiovascular Diseases: Conventional
Therapy to Nanotechnology
Sudhanshu S. Beheraa, Krishna Pramanika and Manasa K. Nayaka,b*
aDepartment of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008,
India; bDepartment of Internal Medicine, University of Iowa, Iowa city, 52242, USA
Abstract: Cardiovascular disease (CVD), accounting around 30% of deaths worldwide, collectively comprised of
disorders affecting the heart and blood vessels as well as their associated adverse conditions. Despite outstanding
progress in the area of the treatments of CVDs, significant challenges remain in designing of efficient delivery sys-
tems for myocardial therapy. Moreover, current therapy for CVDs is limited due to various clinical complications
such as systemic toxicity, stent thrombosis, etc. Molecular and nanotechnology approaches provide the tools to ex-
plore such frontiers of biomedical science at the cellular level and thus offer unique features for potential applica-
tion in the field of cardiac therapy. In this review, recent advances in CVD related risk factors, chronic inflamma-
tion, and their therapeutic modalities such as stem cell therapy, gene delivery, tissue factor (TF) inhibitors, miR-
NAs, leukotriene modifiers, thrombolytic agents etc., in modern molecular aspects are discussed. Moreover,
nanoparticle based drug delivery, nanocarriers as molecular imaging, and the various challenges of myocardial tissue engineering aspects
have been summarized. All these aspects may provide additional therapeutic substitutes in clinical trials for the registration of new drugs.
Keywords: Cardiovascular disease (CVD), inflammation, potential therapeutics, nanotechnology.
1. INTRODUCTION
Cardiovascular diseases (CVDs) are a group of disorders of the
heart, the associated blood vessels (BVs) (such as arteries, veins,
and capillaries) or both [1]. These comprise of coronary heart dis-
eases, cerebrovasular diseases, peripheral arterial diseases, rh euma-
toid heart diseases, congenital heart diseases, deep vein thrombosis
(DVT) and pulmonary embolism (PE) [2]. Moreover, heart attacks
and strokes are usually acute events, caused by a blockage that
prevents blood from flowing to the heart and brain, respectively
[3,4]. The major cause for this is a build-up of fatty deposits on the
inner walls of the BVs that supply blood to the heart and brain.
Strokes can also be caused by bleeding from BVs in the brain or
from blood clots [5]. The causes of CVD are diverse but atheroscle-
rosis and hypertension are the most common. In addition, with ag-
ing comes a number of morphological and physiological changes
that alter cardiovascular function and lead to increased risk of CVD,
even in healthy asymptomatic individuals [6,7]. The common types
of CVDs, symptoms and their risk factors are mentioned in Table.
1. CVD is the leading cause of deaths worldwide, though, since the
1970s, cardiovascular mortality rates have declined in many high-
income countries [8]. Meanwhile, cardiovascular deaths and disease
have elevated at a faster rate in low- and middle-income countries
[9] and it has been estimated that over 80% of the world’s deaths
from CVDs occur in low- and middle-income countries [10-13].
Many people in these countries die younger from CVDs and other
non-communicable diseases, often in their most productive years.
On the basis of the global status of non-communicable diseases,
WHO reported that CVDs are the number one cause of deaths glob-
ally; as estimated, 17.3 million people died from CVDs in 2008,
indicative of 30% of all global deaths [14]. Of these deaths, the
Global atlas on CVD prevention and control, WHO has estimated
that 7.3 million were due to coronary heart disease and 6.2 million
were due to stroke [14,15]. Therefore, non-commu-nicable disease,
including CVD and diabetes are estimated to reduce GDP by up to
6.77% in low- and middle-income countries experiencing rapid
*Address correspondence to this author at the Department of Internal Medi-
cine, University of Iowa, Iowa city, 52242, USA;
E-mail: manaskumarnayakbiotech@gmail.com
economic growth, as many people die prematurely [16]. Mathers et
al. reported that CVDs (mainly from heart attacks and strokes) are
projected to remain the single leading cause of death and will in-
crease to reach 23.3 million by 2030 [8]. In addition, Lim et al.
analysed a systematic and comparative risk assessment of burden of
disease and injury attributable to risk factors and observed that each
year 9.4 million deaths or 16.5% of all deaths are caused due to
high blood pressures, among which 51% of deaths include stokes
and 45% of deaths attributed to CVDs [10]. Most CVDs can be
prevented by addressing risk factors such as tobacco use, unhealthy
diet, obesity, physical inactivity, high blood pressure, diabetes, and
raised lipid contents [7,17,18].
The recent advent of nanotechnology has had a tremendous
impact on many areas in science and engineering, especially in
advancing medical science and health care [19,20]. In nanotechnol-
ogy, engineered materials or devices are used for the smallest func-
tional organisation on the nanometer scale (1-100 nm) in atleast one
dimension [21]. The nanomaterials and nanodevices can interact
with biological entities at molecular levels with a high degree of
specificity and reactivity [22]. Thus, they can stimulate, respond to
and interact with target cells and tissues in controlled ways to in-
duce desired physiological responses while minimizing undesirable
effects. With all this potential, nanotechnology could have a revolu-
tionary impact on diagnosis and therapy of cardiovascular diseases
(CVDs) [19,20,22]. In this review, we provide an overview of the
most exciting advances in nanomaterials and nanodevices that have
been implemented as a therapeutic agent in the treatment and diag-
nosis of CVD.
2. RISK FACTORS FOR CARDIOVASCULAR DISEASES
(CVDs)
Behavioral risk factors such as unhealthy diet, physical inactiv-
ity, tobacco use and harmful use of alcohol are responsible for
about 80% of coronary heart disease (CHD) and cerbrovascular
disease (CD) [23]. These risk factors are responsible for increased
blood pressure, raised blood glucose and blood lipid levels and
caused overweight and obesity in individuals. The cessation of
tobacco use, reduction of salts in the diet, consuming fruits, vegeta-
bles, regular physical activity and avoiding harmful use of alcohols
Manasa K. Nayak
1873-4286/15 $58.00+.00 © 2015 Bentham Science Publishers
4480 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
have been shown to reduce the risk of CVDs. Moreover, the cardio-
vascular risk can also be reduced by preventing or treating hyper-
tension, diabetes, and raised blood lipids [24]. In addition, there are
a number of underplaying instigations such as globalizations, ur-
banization, population ageing as well as poverty, stress and heredi-
tary factors causes of the CVDs [25].
Anderson et al. [26] predicted the equations for several cardio-
vascular disease endpoints, which are based on measurement of
several known risk factors. The equations to predict the risk of
CVDs were developed considering myocardial infarction, coronary
heart diseases (CHDs), death from CHDs, CVDs and death from
CVDs and stroke. The equations demonstrated the potential impor-
tance of controlling multiple risk factors such as blood pressure,
total cholesterol, high-density lipoprotein cholesterol (H-DLC),
smoking, glucose intolerance, and left ventricular hypertrophy as
opposed to focussing or controlling single risk factor. Ahmed et al.
[25] reported that Asian Indians are known to be at a high risk for
cardiovascular disease (CVD), type-2 diabetes and metabolic syn-
drome (MetS) which is attributed to the ‘Asian phenotype’. Several
studies indicated that the MetS is closely related to insulin resis-
tance (IR) and also linked with iron overload. Increased serum fer-
ritin (SF) is often associated with measures of IR, such as elevated
blood glucose and insulin levels [27, 28]. With this connection,
Ewang-Emukowhate et al. [29] reported that genetic and environ-
mental factors together, play a role in the lipid patterns. Dyslipide-
mia in the MetS associated with insulin resistance is characterized
by an atherogenic lipid profile comprising increased low density
cholesterol (LDL-C), triglycerides and inclined numbers of small
dense low density lipoprotein particles. The pattern of dyslipidemia
varies across different ethic groups with increases in triglycerides
and a reduction in LDL-C being the commonest pattern in non-
Caucasians (South Asia and West African population). Gabrielsen
et al. investigated the mechanism underlying the relationships
among SF, adiponectin, and MetS in mice and human. Studies in
cell culture, demonstrated that iron plays a direct and causal role in
determining adiponectin levels and diabetes risk [27]. Epidemiol-
ogical studies provided the increased iron stores are a risk factor for
developing cardiovascular and metabolic abnormalities [28]. Al-
though mechanistic insights have been limited, in diabetes and
MetS, iron may contribute to risk following deposition in the liver,
skeletal muscle and pancreas, where it can enhance oxidative dam-
age, contribute to insulin deficiency and resistance [30]. However,
Toss et al. reported that several positive acute phase reactants, such
as C-reactive protein (CRP) and ferritin are accompanied by the
elevation of CVDs [31]. With this connection, He et al. [32] re-
ported that the relative risk (RR) existing in recurrent cardiovascu-
lar events or death associated with C-reactive proteins (CRP) within
72h from acute coronary syndromes (ACS) onset in a cohort and
secondary analysis of randomized controlled trials. Moreover,
Table 1. Types of cardiovascular diseases (CVDs), symptoms and risk factors.
Types of CVD Description Symptoms Risk factors
I. Coronary heart diseases
Ischemic heart disease (IHD); most
common type
(i) Heart attack;
(ii) Angina at chronic condition
High BP, high BC, tobacco use, unhealthy
diet, physical inactivity, diabetes,
advancing age, inherited disposition
II. Stroke Common form of CVD and three
categories:
(i) Ischemic stroke; (ii) hemorrhagic
stroke; (iii) Transient ischemic attack
Brain damage, leading to a
sudden impairments; weakness
often on one side of the body
High BC, tobacco use, unhealthy diet,
physical inactivity, diabetes, and
advancing age
III. Rheumatic heart dis-
ease and Rheumatic fever
Inflammation of the heart valves and
heart muscle caused rheumatic fever
(streptococcal bacteria); begins as a sore
throat or tonsillitis in children
(i) Shortness of breath, fatigue,
irregular heart beats, chest pain
and fainting.
(ii) Fever, pain and swelling of
the joints, nausea, stomach
cramps and vomiting
-
IV. Congenital heart
disease
Malformations of heart or central blood
vessel at birth or during gestation (e.g.,
hole in heart, abnormal valves, and
abnormal heart chambers)
Breathlessness or a failure to
attain normal growth and
development
Maternal alcohol and medicines use; ma-
ternal infection (e.g., rubella); poor mater-
nal nutrition; close blood relationship
between parents consanguinity
V. Peripheral vascular
disease
Peripheral arterial disease;
Two important forms;
(i) Atherosclerosis (ii) Abdominal aortic
aneurysm
- Long-standing high BP; Marfan
syndrome; tangential heart disorders,
syphilis, and other infectious and
inflammatory disorders
VI. Deep venous thrombo-
sis (DVT) and pulmonary
embolism
The blood clots in the leg veins, which
can dislodge and move to the heart and
lungs
- Surgery, obesity, cancer, recent childbirth,
use of contraceptive and hormone
replacement therapy, long periods of im-
mortality and previous episode of DVT
VII. Other cardiovascular
diseases
Tumors of the heart; vascular tumors of
the brain; disorders of heart muscle
(cardiomyopathy); heart valve diseases
- -
[BP: Blood pressure; BC: Blood cholesterol]
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4481
Lipoprotein (a) (Lp (a)), considered an emerging risk factor by
National Cholesterol Education Programme’s Adult Treatment
panel III (NCEP ATP III), has been implicated in the development
of the premature atherosclerotic disease seen in South Asians [33].
Serum uric acid (UA) is implicated and often been considered as
one of the potential risk factors underlying the development of both
MetS and CVDs [34, 35]. Chatterjee et al. reported that the levels
of novel risk factors namely high sensitivity C-reactive protein
(hsCRP), ferritin, Lp (a) and UA in metabolic syndrome (MetS)
subjects with and without coronary artery disease (CAD) and found
out their association with components of MetS as well as other
traditional risk factors, which could be helpful in preventing future
morbidity and mortality and thereby reducing the burden of CVDs
[36].
3. CARDIOVASCULAR DISEASES (CVDs) AND INFLAM-
MATION
The role of inflammation has become well established over the
past decade or more in theories describing the CVD process [37].
Inflammation provides a common link between many risk factors
for atherosclerosis and altered arterial biology [38]. It has been
reported that modification of these risk factors can exert a clinical
benefit by reducing inflammation and its effects [39]. Emerging
evidence from clinical trials supports the use of inflammatory status
as a guide to therapy that can reduce cardiovascular diseases. Abso-
lutely, the concept of inflammation has materialized from the do-
main of theory and laboratory investigations to assume a promising
role as a useful tool in the clinic to aid the prevention and manage-
ment of cardiovascular disease [40]. The CVD and related disorders
are characterized by an inflammatory component at some stage of
the pathology. The risk factors for CVDs give rise to a variety of
stimuli that draw out the secretion of leukocyte soluble adhesion
molecules such as E-selectin, which promote the attachment of
monocytes to endothelial cells (endothelial cells also activate and
express a variety of adhesion molecules) and facilitate monocytes to
migrate into the sub-endothelial space. Moreover, in the sub-
endothelial space, the transformation of monocytes into macro-
phages and the uptake of cholesterol lipoproteins are thought to
emerge fatty streaks. Furthermore, injurious stimuli may continue
the accumulation and attraction of macrophages, mast cells, and
activated T cells within the growing atherosclerotic lesion. Fig. (1)
shows mechanism of cardiovascular diseases (CVDs) and related
inflammation. The non-fatal incidents of stroke and myocardial
infarction that result from a thrombus blocking the cerebral or
coronary vessels, respectively, result in ischemic injury that is fol-
lowed by reperfusion of the infarcted area. During the reperfusion,
leukocytes are activated and release a variety of oxidative stress-
response molecules and pro-inflammatory lipid and peptide media-
tors [41].
Oxidized low-density lipoproteins (LDL-oxidized) may be one
of several factors that donate to loss of smooth muscle cells through
apoptosis in the atherosclerotic plaque cap. Discharge of metallo-
proteinase and other connective tissue enzymes by activating
macrophages may break downcollagen, weakening the cap and
making it prone to rupture [42]. This crack-up of the atherosclerotic
plaque induces thrombosis. Therefore, every stage of atherosclero-
sis is believed to involve cytokines, other bioactive molecules, and
cells that are characteristics of inflammation [43, 44].
Since inflammation is believed to have a role in the pathogene-
sis of cardiovascular events, the markers of inflammation such as
lipids and peptide mediators, cytokines, macrophages, mast cells,
and activated T cells and other bioactive components are essentials
for prediction of CVDs. [45].
The updated information about inflammations of CVD such as
causative agents, sequential events, and possible mechanisms will
benefit in diagnostics and clinical applications of CVDs.
4. CONVENTIONAL THERAPEUTICS STRATEGIES FOR
CVDs
Despite of all advances in pharmacological and clinical treat-
ment, CVDs and their related causes are leading cause of morbidity
and mortality worldwide.
Fig. (1). Cardiovascular diseases and related inflammation. The risk factors give rise to a variety of stimuli, which draw out the secretion of leukocyte
soluble adhesion molecules such as E-selectin. The adhesion molecules further promote the attachment of monocytes to endothelial cells and facilitate mono-
cytes to migrate into the sub-endothelial space. In the sub-endothelial space, the transformation of monocytes into macrophages and the uptake of cholesterol
lipoproteins are thought to emerge fatty streaks. Furthermore, injurious stimuli may continue the accumulation and attraction of macrophages, mast cells, and
activated T cells within the growing atherosclerotic lesion.
Endothelial Cells
E-Selectin
Rolling
Adhesion
Transmigration
Endothelial Space
Macrophage
Lipoprotein Uptake
Mast Cells
T-C e ll
T-Cell activated
Chemokines
Activation
4482 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
4.1. Molecular Therapeutic Approaches
A range of molecular therapeutic advance strategies, including
cell transplantation and stem cell therapy, gene delivery or therapy,
tissue factor inhibitors, micro RNSs or other small molecules have
been researched to treat CVDs [46].
4.1.1. Cell Transplantation and Stem Cell Therapy
Stem cell-based therapies have rapidly emerged as a potential
novel therapeutic approach in CVDs [47]. Bone marrow-derived
progenitor cells and other progenitor cells can differentiate into
vascular cell types, restoring blood flow [48, 49]. They may con-
tribute to the regeneration of infarcted myocardium and enhance
neovascularization of ischemic myocardium [48]. Several research-
ers have been reported that autologous bone marrow cells (BMCs)
transplanted into ventricular scar tissue may differentiate into car-
diomyocytes and restore myocardial function [50-54]. In contempt
of all advancements in the therap eutic use of stem/progenitor cells
for ischaemic heart diseases. The effect of the implanted cells on
heart function is probably occurring at a very low rate, due to un-
certainties in proliferation and differentiation potential of cardiac
cells [55]. Moreover, there is strong evidence that endothelial cells
are renewed by bone marrow- derived progenitor cells [56], but the
idea that cardiomyocytes are renewed by such cells is vigorously
debated [49].
4.1.2. Gene Delivery or Therapy
The advancement of the molecular mechanisms in CVDs has
increased dramatically in recent years [57]. To understand the mo-
lecular pathology of CVDs, the evolution of vector technology has
significant potential to target the underlying molecular processes
affecting failing cardiomyocytes [58]. Gene therapy offers an at-
tractive alternative to current pharmacological therapies and may be
beneficial in treatments for CVDs [59]. In addition, advances in
recombinant DNA technology, including gene transfer, have stimu-
lated hope that this technology can be used to improve the practice
of cardiovascular medicine [58].
The development of molecular genetics involvements to treat
CVDs depends on technical advances in the development of meth-
ods of gene delivery. The objective of various developed methods
of gene transfer is to achieve a long-term, highly efficient, and tar-
geted expression in relevant cells of the cardiovascular system [60].
The modes of gene transfer include; viral vectors (adenoviruses and
retroviruses), and nonviral vectors (cationic liposomes, polymers,
and injection of plasmid DNA) and delivery method (direct,
transvascular, and ex vivo) have been used. Moreover, gene therapy
with isoforms of growth factors such as vascular endothelial growth
factors (VEGFs), fibroblast growth factors (FGFs) and hepatocyte
growth factors (HGFs) induces angiogenesis, decreases apoptosis
and leads to protection in the ischemic heart [61]. Gene therapy
coding for antioxidants, heat shock proteins (HSPs), mitogen acti-
vated protein kinase (MAPK) and numerous other anti apoptotic
proteins have demonstrated significant cardio-protection in animal
models [62].
However, it has been reported that a cardiovascular gene ther-
apy for stimulating angiogenesis in patients, the peripheral vascular
disease has been initiated. Moreover, the application of gene trans-
fer to CVDs has proceeded at a slower rate [58, 59]. In addition,
lack of persistence of gene expression may result from cytolytic
responses directed against infected cells.
4.1.3. Tissue Factors (TF) Inhibitors
Tissue factor, formerly known as thromboplastin, is the key
initiator of the coagulation cascade, has been implicated in the
pathogenesis of several cardiovascular disorders [63]. Elevated
levels of TF are observed in patients with CVDs and related dis-
eases. TF may certainly be involved in the pathogenesis of athero-
sclerosis by promoting thrombus formation and act through diverse
signal transduction mechanisms, including MAP kinases, PI3-
kinase, and protein kinase C [63, 64]. In addition, various cytoki-
nes, growth factors, and biogenic amines have been recognized to
induce TF expression in endothelial cells (ECs), vascular smooth
muscle cells (VSMCs), and monocytes [64].
To maintain steady-state and healthy conditions, several inhibi-
tory pathways may be practiced to counteract the activity of the TF
mediated coagulation system [64]. Molecular approaches such as
ribozymes or antisense oligonucleotides specifically inhibit TF
production. Monoclonal or polyclonal anti-TF antibodies directly
target and inactivate the TF protein. Recombinant anticoagulant
protein interferes with the TF complex, leading to the formation of
a quaternary inhibitory complex and thus, inactive the TF forma-
tion. Although, several promising therapeutic strategies have been
developed for targeting the action of TF; at present, the relative
contribution of TF to thrombus formation and/or propagation and
associated with atherogenesis is still debated [64].
4.1.4. MicroRNAs (miRNAs)
MicroRNA (abbreviated miRNA) is a small non-protein coding
regulatory RNA molecules (containing about 20-23 nucleotides)
that exist in virtually all organisms and which regulates in RNA
silencing and post translational regulation of gene expression [65].
Moreover, they are highly evolutionary conserved, suggesting a
superior role in essential biological [66]. Details about the biogene-
sis and regulation of cardiovascular miRNAs have been reviewed
[67]. In brief, initial primary miRNAs (pri-miRNA) are processed
to form precursor miRNAs (pre-miRNA) and further processed by
various factors (such as, RNase type-III, Ribonuclease Dicer) into
small 20-23 nt long miRNA duplexes. Finally, miRNAs are in-
volved in silencing the gene expression at the post translational
level with the result of mRNA degradation or by translational inhi-
bition finally leading to target protein repression. These miRNA
expression patterns change in various CVDs, such as myocardial
infarction, cardiac hypertrophy and heart failure [68, 69]. This has
raised the possibility that miRNAs may be probed in the circulation
and can serve as novel diagnostic markers [70]. It has been quickly
recognized that miRNAs can be efficiently inhibited for prolonged
periods by antisense technologies. These technilogies have fueled a
growing interest in the inhibition of specific miRNAs (referred to as
antimiRs) and triggered enthusiasm for miRNAs as novel therapeu-
tic targets [71].
Disease-inducing cardiac microRNAs can be persistently si-
lenced in vivo through the systemic delivery of antimiRs, allowing
for the direct therapeutic modulation of disease mechanisms [71].
However, nature of the stability of miRNAs that circulate in the
bloodstream raised potential challenges of microRNA-based ap-
proaches drug-based therapies [72-76].
4.2. Other Small Molecules
4.2.1. Microparticles (MPs)
Microparticles (MPs) are membrane vesicles released from
many different cell types [77]. There are different pathways in-
volved in the MPs generation (i.e., cell activation and apoptosis).
MPs vary in protein composition, size and phospholipids contents,
and have a robust pro-inflammatory effect and promote coagulation
cascade pathways [78]. In addition, circulating levels of MPs is
augmented in pathogenesis of cardiovascular diseases [79]. There-
fore, MPs inhibited based therapies may be followed in the under-
standing of the pathogenesis, prevention and treatment of these
CVDs. Treatment with antioxidants, (e.g., Vitamin C or Carvedilol)
decreases circulating endothelial MPs in patients with heart failure
[80].
Thus, several researchers claimed for the treatment and also
formulated therapeutics of CVD which affect MPs numbers. How-
ever, these effects on MPs contributing to the therapeutic action
remains to be established [80,81].
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4483
Table 2. A global vision of potential therapeutics for CVDs.
Potential Therapeutics Study design Results References
Conventional therapeutic strategies for CVDs - - -
High-density lipoprotein cholesterol (HDL-C) - - -
HDL-C and CVDs: four prospective American
studies
Men and women (aged between 30 and 69
years)
HDL-C levels unrelated to non-
CVDs mortality
[81]
Raising HDL with CETP inhibitor HDL-C levels and 3H-tracer macrophages Significantly reduced TG, blood
glucose and plasma insulin levels
[84]
Leukotriene (LT) modifiers - - -
5-LOX inhibitor inhibits neuronal apoptosis
and focal cerebral ischemia
Adult male Sprague–Dawley rats Neuroprotection against ischemic
stroke
[94]
Zileuton on inflammatory injury to myocar-
dium
3mg/kg zileuton was given to rat model Significant decrease in myocardial
apoptosis
[95]
Micro RNA (miRNA) - - -
miRNAs in atherosclerosis in vivo -
Elevated miR-155 levels in athero-
sclerotic lesions
[68]
MicroRNA-126-5p - Rescued EC proliferation and limits
atherosclerosis
[69]
miR-181 family - Vascular inflammation [70]
Angiotensin-converting enzyme -2 (ACE-2) - - -
ACE2/Ang-(1-7)/Mas pathway Used three-month and eight-month transgenic
mice
Prevents brain from ischemic injury [107]
Nanotechnology for therapeutic strategies for
CVDs
- - -
Streptokinase Target-sensitive liposomes and evaluation
with in vivo and in vitro studies
Treatment of thromboembolism [171]
Functionalized nanoporticles (PLGA) nanoparticles Cardio-protection and clinical ther-
apy for ICD
[162]
Functionalized nanofibrous scaffold - - -
Elctrospun matrices for cardiomyogenic stem
cell differentiation
For differentiation of ESCs towards a cardio-
myogenic lineage
Promoted differentiation and
maturation of cardiomyocytes
[186]
Nanofibrous scaffold Nanofibrous scaffold from Hemoglo-
bin/gelatin/fibrinogen (Hb/gel/fib)
Regeneration of the ischemic myo-
cardium
[183]
Tissue engineered bioprosthetic heart valve Heart valve leaflets/scaffold Feasible to construsct allogenic
heart valve tissue
[190]
Tissue engineered Biomaterials for treatment
of CVDs
- - -
PLGA-(70/30) microtubular orientation-
structured blood vessel
Cell affinity of the scaffolds was evaluated in
vitro
Cells was found and showed high
viability and rapid proliferation
[180]
PPLA scaffolds to better engineered blood
vessels of small-diameter
Better engineered blood vessels of small-
diameter
Facilitate blood-vessel regeneration [182]
Co-electrospinning of PLGA, gelatine, and
elastin (PGE) for vascular tissue engineering
Tune-fine fibrous samples were prepared Potential in vascular tissue engi-
neering
[183]
Hydroxyl-functionalized PCL and loaded with
VEGF
Scaffolds were prepared by coaxial
electrospinning
Bioactive scaffolds for TE applica-
tions
[187]
4484 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
(Table 2) Contd….
Potential Therapeutics Study design Results References
PCL tubular freeze-dried scaffolds for small
diameter blood vessel replacements
A PCL tubular freeze-dried scaffold was
fabricated with freeze-dried composite struc-
ture
For a small diameter vascular pros-
thesis
[192]
PEG - QK (Gln-Lys) based composite RGD sequences and the novel Both in vitro
and in vivo in a mouse cornea micropocket
angiogenesis assay was performed
Provided an angiogenic signalling [194]
Biomimetic PEG based hydrogels A collagen type I-derived peptide (GIA) con-
taining PEGDA (PEGDA-GIA) was synthe-
sized
Formed a capillary-like networks [195]
hCASMCs seeded on polyurethane scaffold Cell viability as well as growth has been as-
sessed
Elastomeric tissue engineering
scaffolds
[201]
Gradient nanofibrous for vascular tissue
engineering
Constructed a biomimetic were characterized
by seeding HUVECs
Heparinozed CS/PCL nanofibrous
creates vessel grafts with similar
natural blood vessels
[199]
Effects of endothelial cell adhesion to SPUUs
containing amino acids
MTT tests as well as cell adhesion, spreading
and viability was determined
Potential for CVDs and angiogene-
sis
[200]
Porous SF scaffolds for potential use as
vascular grafts
In growth and neovascularisation were also
examined in vivo by subcutaneous implanta-
tion in rats
Potential vascular grafts
[202]
[CETP: Cholesteryl ester transfer protein; TG: triglycerides; 5-LOX: 5-Lipoxygenase; ICD: Ischemic cardiovascular diseases; PLGA: Poly (lactide-co-glycolide); PCL: poly(-
caprolactone); TE: Tissue engineering; hCASMCs: Human coronary artery smooth muscle cells; HUVECs: Human umbilical vein endothelial cells; SPUUs: segmented poly (urea)
urethanes; SF: silk fibroin]
4.2.2. High-Density Lipoprotein Cholesterol (HDL-C)
Higher plasma levels of high-density lipoprotein cholesterol
(HDLC) and decreased incidence of cardiovascular disease end-
points have been observed in epidemiological studies conducted by
several groups of researchers [82-84]. Therefore, more successful
therapeutic interventions for CVD, have been reduced plasma con-
centrations of low-density lipoprotein cholesterol (LDL-C) and the
inverse relationship of HDL-C raising therapy as a novel target [85-
92]. The underlining mechanism through which raising HDL-C
levels may exert its atherosclerotic effects is due to inhibition of
cholesteryl ester transfer protein (CETP) [85]. Rader et al. [92]
reported that HDL is a key component of predicting cardiovascular
risk. However, in spite of its properties, rational with atheroprotec-
tion, the relation between HDL and atherosclerosis is uncertain.
Human genetics and failed clinical trials have created doubt about
the HDL hypothesis. Nevertheless, drugs that raise HDL-C concen-
trations, cholesteryl ester transfer protein (CETP) inhibitors, are in
late stage clinical development, and other approaches that enhance
HDL function, such as reverse cholesterol transport, are in early
stage clinical development. Remaley et al. briefly discussed the
currently established the two main classes of drugs that raise HDL-
C is nicotinic acid and fibrates [93]. Nicotinic acid is more effective
in raising HDL-C, wh ereas fibrates are more effective in decreasing
elevated triglyceride (TG) levels, which are often inversely associ-
ated with HDL levels.
However, both drugs have some of their limitations such as
uricosuria, increased glucose tolerance, flushing for nicotinic acid,
and problematic pharmacokinetic interactions for fibrates, there-
fore, limits their use.
4.2.3. Leukotriene (LT) Modifiers
Leukotrienes (LT) are short-lived lipid mediators that have
potent pro-inflammatory biological activities. They may form at the
nuclear membrane of inflammatory cells, such as macrophages,
neutrophils, eosinophils and mast cells in response to diverse im-
mune and inflammatory stimuli [95]. Several evidences from ex-
perimental and genetic studies suggested that there is an existence
of potential link between the LT signalling cascade, and the patho-
genesis/progression of atherosclerosis [94-105]. LTs are then se-
creted into the extracellular space through specific transporters [95,
96]. Owing to their anti-inflammatory properties, LTs have been
used for the primary therapeutics in asthma management for several
years. Although blocking the inflammatory component of human
disease is a long-lasting and established concept, the use of LTs
modifiers in treating the inflammatory component of CVDs (athe-
rosclerosis, myocardial infarction, stroke and aortic aneurysm) has,
surprisingly, only been seriously reflected in the past few years [95-
97]. The LTs modifiers such as 5-lipoxygenase (5-LO) inhibitor
(ziluton), the leukotriene receptor antagonists (pranlukast, zafirlu-
kast and montelukast) and two members of the 5-LO- activating
protein (FLAP) antagonist family (BayX-1005/DG-031 and MK-
0591) that underwent extensive preclinical development [96]. Fur-
ther experimental and clinical studies are needed to determine the
potential therapeutic strategies targeting the LT pathway in CVD to
increase a potential use of these drugs in the context of subclinical
atherosclerosis [105].
4.2.4. Angiotensin-Converting Enzyme 2 (ACE2)
It has been well established that the rennin-angiotensin system
(RAS) is an important regulator of cardiovascular function and has
an important role in the pathophysiological development of various
CVDs [106,107]. Shi et al. [108] reported that angiotensin-
converting enzyme (ACE) is a major target in the treatment of
CVDs. Moreover, angiotensin-converting enzyme-2 (ACE-2), the
homolog of ACE, which promotes the degradation of angiotensin II
(Ang II) to Ang (1-7) has been recognized recently as a potential
therapeutic target in the management of CVDs. More recently,
Reddy Gaddam et al. [109] reported that ACE involved in the syn-
thesis of bioactive components of RAS and the main active peptides
of the RAS include angiotensin II (Ang II), Ang III, Ang IV, and
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4485
angiotensin-(1-7) [Ang-(1-7)] among which Ang II and Ang-(1-7)
are much more important in health and disease. More specifically,
the hydrolysation of angiotensin (Ang) I to produce Ang-(1-9),
which is subsequently converted into Ang-(1-7) by a neutral en-
dopeptidase and ACE. The Ang-(1-7), so formed is considered to
be a beneficial peptide of the RAS cascade in the cardiovascular
system and control cardiovascular physiology. Zheng et al. [110]
investigated the effect of angiotensin (Ang) converting enzyme-2
(ACE-2)/Ang-(1-7)/Mas receptor pathway (component of RAS) on
the beneficial effect in ischemic stroke. The results found that ACE-
2/Ang-(1-7)/Mas axis prevents the brain from ischemic injury via
the Nox/ROS signalling pathway. However, it has been difficult to
understand the complex relationship of two-arm RAS hypothesis
(of ACE-Ang II-receptor and ACE2-Ang(1-7)-Mas axis) [111].
4.2.5. Thrombolytic Agent
Vascular thrombosis (VT) or thrombolytics play an important
role in several diseases and is a major clinical problem, particularly
in developed Western countries. Indeed, VT (thrombolytics) ac-
counts for about half of all deaths in these countries as a result of
consequential myocardial infarction, ischemic stroke, pulmonary
embolism (PE) and likewise other vascular obstructions (peripheral
arterial diseases). The process of fibrinolysis acts by activation of
plasminogen to plasmin; plasmin splits fibrinogen and fibrin and
lyses the clot, which then allows reperfusion of the ischemic brain.
The best way to improve patient survival and decrease morbid-
ity is prompt detection and treatment of thrombosis with throm-
bolytic agent(s). A variety of thrombolytic agents such as strepto-
kinase (SK), urokinase and tissue plasminogen activators (t-PA) are
pharmacologically active and available. These thrombolytic agents
comprise proteolytic components of the blood clotting cascade, and
as they circulated throughout the cardiovascular system they do not
selectively target specific organs or tissues. The systemic side ef-
fects of these agents such as fibrinogenolysis and bleeding [112].
These are currently in clinical use for dissolving arterial thrombi,
particularly in cardiac blood vessels. These thrombolytic agents
work by activating the protein, plasminogen into plasmin [113].
Streptokinase (SK) is one of the most commonly used throm-
bolytic agents for the treatment of thromboembolism. However,
short half-life of the SK due to rapid inactivation of plasminogen
activators such as plasminogen activator inhibitor-1 (PAI-1) and
circulating antibodies requires administration of higher dose which
results in various side effects, including systemic haemorrhage due
to activation of systemic plasmin. Various techniques have been
employed to enhance stability of plasminogen activators against
these components and hence to increase the half-life, which ulti-
mately increase the accumulation at the site of thrombus. PEGy-
altion and novel carrier constructs, including liposome and polym-
eric particles are most frequently used [114]. Murray et al. [115]
reported that thrombolytic agents such as streptokinase (SK), re-
combinant tissue-type plasminogen activator (rt-PA) among prom-
ising agents under development and in clinical trials. SK is nonfi-
brin-specific, has a longer half-life than tissue-type palsminogen
activator (t-PA). SK avoids re-occlusion and is degraded enzymati-
cally in the circulation. However, rt-PA is more fibrin-specfic and
has clot-dissolving activity and is metabolized during the first pas-
sage in the liver. So far the fibrin-specific rt-PA is the only agent to
be approved for use in stroke. This may be due to its short half-life
and its absence of any specific amount of circulating fibrinogen
degradation products, thereby leaving platelet functionally intact.
More specifically, Campbell et al. [116] reported that recomibinant
thrombolytic peptides are mainly represented by recombinant forms
of tissue-type palsminogen activator (t-PA) (a proteolytic enzyme)
that catalyzes the conversion of plasminogen into active plasmin,
which then functions to dissolve clots. The three clinically suitable
recombinant thrombolytic peptides are alteplase (t-PA), reteplase
(r-PA), and tenecteplase (TNK). r-PA and TNK have been structur-
ally modified from native t-PA to increase their half-life and fibrin
specificity. Moreover, thrombolytic therapy is the treatment of
choice for ischemic stroke in patients who present 3 hours follow-
ing the onset of symptoms. Furthermore, thrombolytic therapy is
used to restore function to stenotized central venous access devices
as well as occluded hemodialysis access grafts. Zhang et al. [117]
reported that the addition of a neuroprotective agent can increase
the effectiveness of thrombolytic therapy, increase the therapeutic
time window, and reduce cerebral ischemia-reperfusion injury. It is
hoped that thrombolytic agents with neuroprotective effects can be
developed for clinical use. Vaidya et al. [118] developed target-
sensitive liposomes which were prepared by using moderate affinity
peptide c (RGDfK) acylated with palmitic acid. In vitro results
revealed that target-sensitive liposomes beneficial in the targeted
delivery of thrombolytic agents to the site of blood clot where acti-
vated platelets are embedded. However, for in vivo application,
targeting ligand with higher affinity to the target site would be more
effective than moderate affinity ligand. Therefore, c (RGD) peptide
[CNPRGDY (OEt) RC] has been investigated and found to have a
higher affinity for GP IIb/IIIa receptors on the platelet surface as
compared to other receptors. Thus, developed liposomes were
found to release streptokinase in vitro following binding with acti-
vated platelets. In addition, the prepared liposomes were character-
ized for in vivo targeting by intravital microscopy and therapeutic
efficacy was evaluated using human clot inoculated rat model and
revealed higher accumulation in the thrombus area. In vivo throm-
bolytic study was performed in the human clot inoculated rat
model. Moreover, the overall results of the study showed that tar-
get-sensitive liposomes dissolved 28.27±1.56% thrombus as com-
pared to 17.18±1.23% non-liposomal streptokinase. Du et al. [119]
synthesized a hybrid hydrogels based on a genetically modified tax
interactive protein-1 (TIP1) by introducing two or four cysteine
residues in the primary structure of TIP1. The cysteine residues
were cross-linked with four armed- poly (ethylene glycol) (PEG)
capped with maleimide residues (4-armed-PEG-Mal) to form hy-
drogels. Further, AD293 cells were allowed to divide and displayed
a polyhedron or spindle-shape during 3-day culture period. The
cells were recovered from the TIP1 2C RGD gel grew well in the
conventional 2D cell culture, suggesting its great potential for
large-scale culture.
5. NANOTECHNOLOGY ASPECTS OF THERAPEUTIC
STRATEGIES FOR CVDS
Nanotechnology represents a convergent discipline of various
research areas, such as biology, chemistry, physics, mathematics
and engineering is becoming complicated [120]. Recent develop-
ments in nanotechnology have created considerable potential to-
ward diagnosis and therapeutic treatments for CVDs. Fig. (2) high-
lights the nanotechnology approaches for the benefits of CVDs that
will be discussed in this review. Though, the use of nanotechnology
in the treatment and diagnosis of CVD remains largely unexplored.
Nevertheless, cardiovascular nanomedicine is likely to face and
address current challenges in CVDs. It will also improve detection
and therapy by advancing techniques, to improve delivery of drugs
and for tissue regeneration [121]. Therefore, the main objective of
this review article is addressing the problems associated with con-
ventional therapeutics such as nonspecific effects and poor stability
and to focus on nano-carriers advancement for the benefits of
CVDs.
Nanotechnology mainly provides following benefits for CVDs
5.1. In Vivo Imaging Technique:
Offers the visualization of cellular functions using “smart” im-
aging agents or targeted imaging nanoprobes (especially magnetic
nanoparticles, and quantum dots) rather than biomarkers in tradi-
tional imaging techniques.
4486 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
5.2. NanoBiosensors and In Vitro Diagnostics:
Improved analytical devices/ Biomedical or Biological Micro-
Electro-Mechanical Systems (BioMEMS) for accurate detection of
small quantities of disease biomarkers is critical for the clinical
diagnosis of disease.
5.3. Controlled D rug Delivery or Targeted Therapeutics:
Nanoscale delivery of drugs enhances therapeutic efficacy and
develop an effective therapeutic modality.
5.4. Tissue Engineering:
Nanotechnology can provide an engineered biocompatible ma-
terials resembling extracellular matrix (ECM), support for building
new and implanting tissue [122,123].
5.5. In Vivo Imaging Technique
The lesions in atherosclerosis represent the most important
cause of acute cardiovascular incidents and best described as vascu-
lar inflammation [124]. The hydrolytic enzymes secreted by differ-
ent cell types (monocyte-derived macrophages and T lymphocytes)
during cardiovascular inflammation may play a central role in dif-
ferent stages of atherogenesis. Conventional imaging techniques
such as development of enzyme (Cathepsin B as a model prote-
olytic enzyme) sensing near-infrared imaging probes and become
possible due to a developed imaging technique known as fluores-
cence-mediated tomography (FMT). Moreover, FMT technique
shares sensing of picomole to femtomole quantities of fluoro-
chromes in deep tissues at macroscopic scale [125]. Despite the
encouraging results of this technique, several points will require
further investigation. The most importan t is the animals routinely
develop accelerated and lively inflammation-rich atherosclerosis
leading to aortic aneurysms [125, 126]. Therefore, many molecular
imaging studies of atherosclerosis target inflammation, involve a
critical process underlying the progression and rupture of athero-
sclerotic lesions [127]. In addition, the diagnosis, monitoring, and
prognostication of CVDs are based on techniques that measure
changes in metabolism, blood flow, and biological function [125-
127].
Nanoscale contrast agents have emerged as multifaceted ap-
proaches that can be used to label and characterize the early stages
of disease before the development of pathological symptoms. Con-
trast generating nanomaterials for cardiovascular imaging include
radioactive, fluorescent, paramagnetic, super-paramagnetic, elec-
tron-dense and light-scattering particles [128].
5.5.1. Nanoparticles (NPs) -Enhanced Imaging
Nanoparticles (NPs) are emerging as potentially powerful
probes for in-vivo imaging in medical and biological diagnostics
[129]. They can fulfill a number of critical requirements required
for imaging techniques. The NPs are not influenced by various local
in-vivo environments such as ionic strength, pH, solvent polarity or
temperature. Moreover, they can easily dispersible, resist aggrega-
tion and form stable imaging agent. Ideally, these NPs are suitable
for long-term quantitative imaging at low doses and can be safely
cleared from the body after imaging is complete [129].
5.5.1.1. Magnetic Nanoparticles (MNPs)
The magnetic nanoparticles (MNPs) are classified (based on
their size) as magnetic iron oxide nanoparticles (MIONs, m), su-
perparamagnetic iron oxide nanoparticles (SPIONs, hundreds of
nm), and ultra-small paramagnetic iron oxide nanoparticles (USPI-
ONs, <50 nm) [130]. These MNP-based probes have been devel-
oped for Magnetic Resonance Imaging (MRI) to achieve high tissue
contrast and to improve the imaging sensitivity [131]. Moreover,
they have been directed imaging and therapy of atherosclerosis,
restenosis and related cardiovascular conditions. The superpar-
amagnetic contrast agents (SPIONs) predominately magnetite
(Fe2O3/Fe3O4), typically enhance and produce dark contrast [130].
The SPIONs encapsulated porous silica in a single unit enhanced
contrast at atherosclerotic plaques [132].
To fulfill the high resolution and high sensitivity requirements
for in-vivo imaging applications, biomimetic nanomaterial conju-
gated MNPs contrast agents with improved magnetic and physico-
chemical properties have been developed [133]. PLGA is usually
used in the construction of biomimetic materials and has extensive
application in medical imaging technology. Few reports have em-
ployed a new type of molecular probe in the treatment as well as in
the diagnosis of the thrombi. Zhou et al. constructed Fe3O4-based
poly (lactic-co-glycolic acid) (PLGA) nanoparticles to use in the
detection of thrombi and in targeted thrombolysis using MRI moni-
toring [134]. These biomimetic PLGA conjugated MNPs can serve
as a potential dual function tool in the early detection of thrombi
and in the dynamic monitoring of the thrombolytic efficiency using
MRI at the molecular level [134].
Macrophages, key effector inflammatory cells in atherosclero-
sis, abound in coronary plaques that have caused sudden cardiac
Fig. (2). The possible nanotechnology approaches in the therapeutics of CVDs.
Nanotechnology
as Therapeutic
strategies of
CVD
1.In vivo Imaging
technique
3.Controlled Drug
delivery or Targeted
therapeutics
2.NanoBiosensor
s and in vitro
Diagnostics
4.Tissue
Engineered
Biomaterials
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4487
death. Imaging of macrophages is an appealing approach to detect
inflammation in plaques prone to clinical complication. Dextran-
coated magnetic nanoparticles (MNPs) which are composed of
SPIONs (core) that induces strong MRI contrast and can easily
detect inflammatory cells (macrophages) at and around plaques
[135]. In addition, fibrin an attractive target for thrombosis (both
arterial and venous) imaging. The thrombin-mediated cleavage of
fibrinogen yields fibrin monomers, which then polymerize and
undergo cross-linking to form a stable clot. A recent progress in
fibrin imaging is the advancement of fibrin-targeted nanoparticles
(e.g., SPIONs) for computed tomography (CT) molecular imaging
of thrombus. Further, in vivo analysis with this agent could develop
the ability of CT to diagnose pulmonary embolism, stroke, or acute
coronary syndromes [136,137].
5.5.1.2. Quantum Dots (QDs)
One of the fastest moving and most exciting interfaces of
nanotechnology is the use of quantum dots (QDs) in biology [138].
Quantum dots (QDs) are tiny light-emitting particles on the
nanometer scale, and are emerging as a new class of fluorescent
labels in biology and medicine. In comparison with organic dyes
and fluorescent proteins, they have unique optical and electronic
properties [139]. They have proven themselves as powerful fluores-
cent probes, especially for their long-term, multiplexed and high
fluorescence quantum yields, high photostability, are a popular
choice for fluorescence imaging applications [140]. The fluorescent
quantum dots (QDs) emit light over a broad range from near-
ultraviolet to mid-infrared and have been used in contrasting agents
in CVDs [85]. Another important consideration for in vivo applica-
tions of QDs reducing the blood concentration and easily cleared
from the bloodstream [141]. Moreover, fluorescent QDs will pro-
vide important information in the rational design of biocompatible
drug carriers and will serve as a superior alternative to magnetic
and radioactive imaging contrast agents in preclinical drug screen-
ing, validation and delivery research [140]. So far, CdSe/ZnS and
CdTe/ZnS QDs are among the most extensively studied QDs for in
vivo imaging, due to their develop synthetic procedure and easily
available [142]. Concurrently, there are also several studies using
CdTe/CdS, PbS, and CdHgTe QDs, which are appealing for in vivo
applications because of their Near-infrared (NIR) emission [143-
145].
The ability to make QDs water soluble and target them to spe-
cific biomolecules potent surface functionalization is essential. In
addition, particularly for those QDs synthesized via the organomet-
allic route, surface modification of QDs can reduce the levels of
toxicity (e.g., Cd) [142]. The surface modification of bioconjugated
QDs has led to promising applications in cellular labelling, deep-
tissue imaging, and assay labelling and as efficient fluorescence
resonance energy transfer donors [138]. For example, carboxylate-
conjugates QDs (a mutant of the bioluminescent protein luciferase)
have a greatly enhanced sensitivity in small animal imaging. The
functionalized and bioconjugated QD probes can emit long-
wavelength bioluminescent light in cells and in animals, even in
deep tissues, and are suitable for multiplexed in vivo imaging [146].
Liu et al. fabricated surface functionalized QDs through covalent
modification via simple carbodiimide coupling chemistry. The sur-
face functionalization of QDs can be achieved by incorporating
ligand dihydrolipoic acid (DHLA), a short poly (ethylene glycol)
(PEG) spacer, and with an amine or carboxylate terminus. The
functionalized QDs (DHLA-PEG-ligand) produced aqueous de-
rivatizable QDs with small hydrodynamic size, low nonspecific
binding, high quantum yield, and good solution stability across a
wide pH range for cell-labeling applications [147]. In addition to
PEG, other polymer coating or QD-entrapped materials such as
poly (lactic-co-glycolic acid) PLGA and liposomes have also been
investigated because of their increased stability and reduced pho-
tooxidation and photobleaching in an in vivo experiments [142,
148]. Recently, Cao, et al. [149] developed a long term and sustain-
able QDs-functionalized with amphiphilic polymer with hydropho-
bic inner core, named as N-succinyl-N’-octyl nanomicelles (SOC),
used to incorporate oil-soluble PbS for tracking in an in vivo local
conditions. [149,150].
5.3. NanoBiosensors and In Vitro Diagnostics
Biosensor is an analytical device that accounts a biological
perception system to target molecules. This device can be conjoined
to a physiochemical transducer which transforms recognition into a
measurable amplified output signal in an appropriate format [151].
The establishment and advancement of micro- and nanoscale tech-
nologies for biology has a great potential to lead to the development
of next generation biosensors with improved sensitivity with re-
duced costs [151]. In addition, usually nanomaterials are exploited
to enhance the sensitivity of sensors, as they are easy to functionali-
ties with the appropriate sensing probes, and also act as signal
transducers. By restraining the material into the nanoscale dimen-
sion the extraordinary sensitivity can be achieved [152].
To date, all most all biochemical methods for acute myocardial
infarction (AMI) diagnosis are based on Enzyme-Linked Immu-
noSorbent Assay (ELISA). ELISA is an antibody (Ab) -based tech-
nique using a surface immobilized Ab1 and an enzyme-linked Ab2
to sandwich the target, converting each target into an Ab1/target/
Ab2 sandwich for sensitive enzymatic detection [153,154]. Al-
though ELISA been renowned as one of the fastest growing tech-
nologies used in clinical diagnostics for detecting specific proteins
associated with disease; however, for clinical diagnoses of AMI,
cardiac markers need additional chemicals. Moreover, compact test-
strips are able to give qualitative, but not quantitative information
on cardiac biomarkers. In addition, compared with commercially
available ELISA systems, nanobiosensor (electrochemical immu-
nosensor) is cheaper [155].
Suprun, et al. developed an electrochemical nanosensor with
immobilized anti-myoglobin (Mb) [146]. The fabricated nanobio-
sensor was quantified Mb (biomarker) for acute myocardial infarc-
tion (AMI) diagnosis. The detection system based on electron trans-
fer between Fe (III) -heme and electrode surface modified with gold
nanoparticles/didodecyldimethylammonium bromide (DDAB/Au)
and antibodies. The method proposed does not require signal en-
hancement or amplification; nor does it require labeled secondary
antibodies. Immunosensor has a detection limit of 10 ng/ml (0.56
nM) and a broad range of working concentrations (10-1780 ng/ml;
0.56-100 nM). The whole procedure takes 30 min and can be used
for express diagnosis of acute myocardial infarction [155].
5.3.1. Biomedical or Biological Micro-Electro-Mechanical
Systems (BioMEMS)
In recent years, the biological and biomedical applications of
micro- and nanotechnology (commonly referred to as Biomedical
or Biological Micro-Electro-Mechanical Systems [BioMEMS])
have become increasingly prevalent and have found widespread use
in a wide variety of applications such as diagnostics, therapeutics,
and tissue engineering [156,157]. In general, BioMEMS can be
defined as ‘‘devices or systems, constructed using techniques in-
spired from micro/nano-scale fabrication, that are used for process-
ing, delivery, manipulation, analysis, or construction of biological
and chemical entities’’[157,158]. These devices and systems en-
compass all interfaces of the life sciences and biomedical disci-
plines with micro- and nanoscale systems. Areas of research and
applications in BioMEMS range from diagnostics, such as DNA
and protein micro-arrays, to novel materials for Bio-MEMS, micro-
fluidics, tissue engineering, surface modification, implantable
BioMEMS, systems for drug delivery, etc. [156-160]. The recent
progress in BioMEMS and nanosensors crates very effective tools
for the early detection and diagnosis of CVDs. For example, the
nanowire integrated potassium and dopamine sensors are optimized
for the monitoring and diagnosis of acute myocardial infarction
[161].
4488 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
5.4. Controlled Drug Delivery or Targeted Therapeutic
Research in the areas of drug delivery has endorsed enormous
progress in recent years due to their unlimited potential to improve
human health. The limitations of current drug delivery systems
include suboptimal bioavailability, limited effective targeting and
potential cytotoxicity. Meanwhile, the development of nanotech-
nology provides opportunities to synthesize with controlled compo-
sition, shape, size and morphology, characterize, manipulate the
matter systematically at the nanometer scale [107]. In particular,
they can enhance the therapeutic activity by prolonging drug half-
life, improving the solubility of hydrophobic drugs and releasing
drugs at a sustained rate. Moreover, their surface properties can be
manipulated to enhance solubility, immunocompatibility and cellu-
lar uptake. In addition, nanoscale particles can passively accumu-
late in specific tissues (e.g., tumors) through the enhanced perme-
ability and retention (EPR) effect [162].
The promising and versatile nano-scale drug-delivery systems
include nanoparticles, nanospheres, nanocapsules, nanotubes, nano-
gels and colloidal carriers such as liposomes or dendrimers. They
can be used to deliver small-molecule drugs, growth factors, en-
zymes and/or biomacromolecules, such as peptides, proteins, plas-
mid DNA and synthetic oligodeoxynucleotides which provide a
sustained therapeutic stimulus at the injured tissue [107, 163].
5.4.1. Nanopaticles (NPs) Based Controlled Drug Delivery
The NPs have been exploited for both diagnostic and therapeu-
tic purposes. They have been used as a controlled drug delivery
vehicle for a variety of diseases; however, in the cardiac field, most
NP work has focused on detection of myocardial infarction [164].
The ideal size of NPs used as drug delivery systems ranges from 10
to 100 nm [164]. Further, these NPs based control drug release, can
targeting and increase the effectiveness or bioavailability of many
diagnostic or therapeutic agents. Among the materials most com-
monly used for cardiovascular drug-delivery systems are the NPs
made of synthetic or natural polymers, such as liposomes, dextrans,
poly (lactic-co-glycolic acid) (PLGA), polyaccrylates, as well as
metal or metal oxide nanoparticles (e.g., gold, silver, SPION), and
quantum dots. Several of the commonly tested drug-carrier systems
are briefly outlined below.
Among the NPss based drug-delivery systems, liposomes have
relatively low toxicity and a good therapeutic index. Liposomes are
composed of a lipid bilayer consisting of amphipathic phospholip-
ids (primarily phosphatidylcholine) that enclose an interior aqueous
space [165]. The head groups of phospholipids are usually func-
tionalized with polymerizable moieties to improve their stability.
Liposome mediated site-directed drug delivery of plasminogen
activators have been explored by various research groups for CVDs
treatments [166,167]. For example, the activation of platelets is
mediated by the binding of fibrinogen through RGD (Arg-Gly-Asp)
motif and therefore, it has been reported that RGD-peptides conju-
gated liposomes have an affinity towards activated platelets and
may be useful for the targeted delivery of thrombolytic agents
[168,169]. However, the liposomes, originally used as transfection
reagents for siRNA or gene delivery, can be functionalized with
ligands (e.g antibodies or polymers) offers low immunogenicity,
which is expected to enable safe and repeated administration [170].
Poly (lactic-co-glycolic acid) (PLGA), is a common and widely
used polymer due to its biocompatibility and biodegradability.
PLGA have approved by the U.S. Food and Drug Administration
(FDA) and the European Medicine Agency (EMA) for parenteral
administration of a drug carrier system [171]. The PLGA can easily
metabolize and hence eliminated from the body due to smooth deg-
radation of PLGA into their constituent products (lactic acid and
glycolic acid). Thus, expected systemic toxicity associated with
PLGA administration is low and safe [172]. However, Chang et al.
[173] have recently developed the insulin like growth factor 1 (IGF-
1) complexes with PLGA nanoparticles, and injected into the
ischemic myocardium, has shown a significant decrease in myocar-
dial apoptosis by inducing (protein kinase B) Akt phosphorylation.
Moreover, it is suggested that, as compared with larger particles,
the smallest (60 nm) sized PLGA-IGF-1 NPs carried more IGF-1
and induced more Akt phoshorylation in cultured cardiomyocytes.
Kona et al. [174] developed targeted NPs as a drug carrier system
that mimics platelets binding to the injured vessel wall under
physiological fluid flow (shear stress) conditions. Glycoprotein Ib-
alpha (GPIb-) of platelets was chosen as the targeting ligand and
conjugated to dexamethasone (drug) -loaded biodegradable poly-
(D, L lactic- co-glycolic acid) (PLGA) nanoparticles. The NPs were
formulated using a standard emulsion method. The results demon-
strate that conjugated NPs (GPIb-PLGA) have increased particle
adhesion onto target surfaces (P-selectin) of damaged endothelial
cells (ECs). In addition, under shear stress condition, cellular up-
take of these NPs by activated endothelial cells found increased.
Therefore, drug-loaded, conjugated NPs (GPIb-PLGA) could be
used as a targeted and controlled drug delivery system to the site of
vascular injury for treatment of CVDs.
They reported that vascular carrier may accumulate in the
thrombus and decrease the systemic degradation of the drug as well.
For optimal drug delivery, drug should be localized at the site of
thrombus and should avoid its non-specific uptake of unwanted
tissue. Thus, a well designed drug delivery system; surface modi-
fied with targeting moiety for targeted delivery of thrombolytic
agents to the site of thrombus may provide an effective alternative
[175]. Platelets play an important role in various thromboembolic
diseases. Under normal physiological conditions, platelets circulate
independently inside the blood vessels without interacting with
other factors or the vascular endothelium. However, at the action of
endothelial damage, blood platelets found contact with the subendo-
thelial extracellular matrix, perform a series of reactions and leads
to the formation of a platelets-rich hemostatic plug [176].
5.5. Tissue Engineered Biomaterials for Treatment of CVDs
The present treatments for the loss or failure of cardiovascular
functions, including organ transplantation, mechanical devices,
surgical reconstruction, or the administration of metabolic products
are although have been routinely used; these treatments are under
constraints and complications [177]. Tissue engineering is an alter-
native method for the treatment of CVDs (Fig. 3). Tissue engineer-
ing seeks to restore the function of diseased or damaged tissues
through the use of cells and biomaterial scaffolds [178]. In recent
years, progress has been made in the engineering the various com-
ponents of cardiovascular system, such as heart valves, blood ves-
sels, and cardiac muscle. Moreover, many pivotal studies are con-
ducted for the widespread applications of tissue engineered therapy
for CVDs. In engineered skeletal and myocardial tissue constructs,
designed scaffolds should match to the native tissue mechanical
properties as well as to enhance cell alignment [179]. The next
generation of functional tissue replacements such as skeletal and
myocardial tissue constructs for the treatment CVDs will require
advanced material strategies to achieve many of the important re-
quirements for long-term success. Therefore, viewing the rapid
evolution in the field of tissue engineering, it is essential to consider
the use of advanced materials in light of the emerging role of genet-
ics, growth factors, bioreactors, and other technologies [180].
Cardiovascular tissue engineering has the potential to improve
the ability to treat CVDs and the single greatest cause of mortality
in the United States. Recently, a significant step could be taken
towards the improvement in the type of heart valves used to replace
diseased and damaged heart valves. Significant progress of the
tissue engineering has been obtained in recent years. However, the
major limitations in tissue engineering are the cultivation and for-
mation of complex tissue constructs to keep viable in vitro as well
as in vivo during implantation [181]. Moreover, replacement struc-
tures manufactured from autologous cells and bioabsorbable poly-
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4489
mer scaffolds using engineering techniques to contain living cells,
these structures have growth potential, imparts less infection and
calcification, and decrease incidence of thrmboembolic complica-
tions [182]. One of the first materials used for tissue engineering of
the allogenic heart valve was biodegradable and biocompatible
polymers of polylactic acid, polyglycolic acid (PGA) and their co-
polymer polylactic-co-glycolic acid (PLGA) or of poly (-
caprolactone) (PCL) [180-182].
5.5.1. Copolymer Polylactic-Co-Glycolic Acid (PLGA)
Hu et al. [181] fabricated a kind of poly (lactide-co-glycolide-
70/30) (PLGA-70/30) microtubular orientation-structured blood
vessel mimicking natural structure by an improved thermal-induced
phase separation (TIPS) technique. By adjusting various parameters
of TIPS techniques such as temperature, concentration of polymer
solution,and inner-and outer-diameter of the polyethylene (PE)
mould (for manufacturing the orientation-structured blood vessel
scaffolds), various microtubular orientations-structured blood ves-
sels scaffolds with different wall thickness was obtained. The cell
affinity of the scaffolds was evaluated in vitro by using A10 cells as
model cells. The results showed that the A10 cells grew (seeded
and migrated) in the vessel scaffolds which were modified by am-
monia plasma treatment and then anchored with collagen. The cells
were found along the direction of the microtubules and showed
high viability and rapid proliferation. With this connection, Ma et
al. fabricated a type of novel scaffolds to better engineered blood
vessels of small-diameter from biodegradable poly (L-lactic acid)
(PPLA) by means of thermally induced phase separation (TIPS)
techniques [183]. By applying the differences in thermal conductiv-
ities of the mould materials and using benzene as the solvent scaf-
folds with oriented gradient microtubular structures in the axial or
radial direction can be prepared. The structural properties of such
scaffolds can be conventionally adjusted by varying the solvent
ratio, phase-separation temperature, and polymer concentration to
mimic the nanofibrous features of an extracellular matrix. These
scaffolds were fabricated for the tissue engineering of small-
diameter blood vessels by using their advantageous structural prop-
erties to facilitate blood-vessel regeneration. Han et al. fabricated a
novel biomimetic scaffolds for application in vascular tissue engi-
neering from co-electrospinning tertiary blends of poly (lactide-co-
glycolide) (PLGA), gelatine, and elastin (PGE). By varying the
ratios of PLGA and gelatin, fine-tune fibers, swelling (on hydra-
tion) and enhanced mechanical properties of the scaffold was ob-
tained [184]. Moreover, the effect of the varying ratios of PLGA
and gelatin on vascular cell morphology and growth on and pene-
tration into the scaffolds was investigated. Further, the electrospun
PGE scaffolds supported the attachment and metabolization of hu-
man endothelial cells (ECs) and bovine aortic smooth muscle cells
(SMCs) and their organotypic distribution on and within the scaf-
folds. Taken together, the results indicated that the PGE scaffolds
were potential in vascular tissue engineering. Nguyen et al. devel-
oped multilayered scaffolds composed of polycaprolactone (PCL) -
gelatin/poly (lactic-co-glycolic acid) (PLGA)-gelatin/PLGA-
chitosan with different diameters using the double-ejection elcetro-
spinning system [185]. The effects of the cross-linking process on
the mechanical behaviors, microstructure, and biocompatibility of
the fibers were investigated. The cross-linked elctrospun scaffolds
satisfied the three requirements for artificial blood vessels (ABVs),
such as resistance to intermediate pressure, flex ibility, and the abil-
ity to promote cell growth and proliferation. Moreover, the biocom-
patibility of the cross-linked ABV scaffolds were examined using
the MTT assay and by evaluating cell attachment and cell prolifera-
tion. In addition, the multilayered ABVs imparted high tensile
strength (2.3MPa of stress and 100% of strain) and liquid strength
(340 mmHg), which are essential for implantation. Taken together,
the use of an elctrospinning array to form multicomponent nanofi-
brous membranes will lead to the creation of novel scaffolds and
holds great promise for eventual use in ABVs.
5.5.2. Poly (-Caprolactone) (PCL)
Cardiovascular diseases (CVDs) are the leading cause of death
in the United States with many patients requiring coronary artery
bypass grafting. The recent years autografts such as using intimal
mammary artery or saphenous vein, could be replaced by creating a
synthetic grafts for eliminating inconvenient procedures. In addi-
tion, large diameter grafts have long been established with materials
such as teflon and dacron, however, these materials have not proved
successful in small diameter (< 6 mm) grafts [186].
Fig. (3). A Basic of tissue engineering (TE) construct for CVDs. For the construction of 3D cardiovascular tissue engineered grafts, nanofiberous scaffolds
were seeded with vascular and cardiac muscle cells. Further, the seeded scaffolds are functioning in addition of growth factors or signalling molecules.
Tissue Engineering
Construct for CVDs
Nanofibers for TE Scaffold
Signalling Molecules or Growth Factors
Stem Cell Differentiation of Stem Cell
Vascular Cells
Cardiac Muscle Cells
4490 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
Gupta et al. have hypothesized that polymeric biomaterials
scaffolds with distinct chemical and mechanical properties could be
employed to enhance the differentiation of embryonic stem cells
(ESCs) to cardiomyocytes as a potential patch for cardiac repair
[187]. A combinatorial polymer was prepared by copolymerizing
three distinct subunits at varying molar ratios to tune the physical
properties of the resulting polymer such as, hydrophilic polyethyl-
ene glycol (PEG), hydrophobic poly (-caprolactone) (PCL), and
negatively-charged, carboxylated PCL (CPCL). Interestingly, Mur-
ine ESCs on the most compliant substrates, 4%PEG-86%PCL-
10%CPCL (electrospun polymeric scaffolds), exhibited the highest
myosin heavy chain (-MHC) expression as well as intracellular
Ca2+ signalling dynamics (depolarization and repolarization path-
way) and optimally facilitated the differentiation of ESCs into func-
tional cardiomyocytes. These results are promising for the treatment
of cardiac ischemia, where the ensuing myocardial hypoxia and
fibrosis may be treated through the delivery of a tissue-engineered
myocardial patch. Seyednejad et al. [188] fabricated nanofibrous
scaffolds based on blends of a hydroxyl functionalized polyster
(poly (hydroxymethylglycolide-co--caprolactone), pHMGCL) and
poly (-caprolactone) (PCL) by means of a coaxial electrospinning
technique. The scaffolds were loaded with bovine serum albumin
(BSA) as a protein stabilizer and vascular endothelial growth factor
(VEGF) as a potent angiogenic factor for tissue engineering appli-
cations. Moreover, the effect of releasing protein (VEGF) on the
attachment and proliferation of endothelial cells was investigated.
The fabricated pHMGCL scaffolds showed an enhanced protein
release (VEGF) as well as increased hydrolysis rate as compared to
PCL scaffolds. The incorporated proteins preserved in pHMGCL
scaffolds showed its biological activity and higher numbers of ad-
hering cells. Thus, these bioactive scaffolds based on blends of
pHMGCL/PCL loaded with VEGF can be considered as potential
candidates for tissue engineering applications. Lee CHY. synthe-
sized a novel tri-layered tissue engineered blood vessels contain
microchannels from biodegradable polymer (polycaprolactone) by
electrospinning and laser ablation techniques [189]. The tri-layered
blood vessels to mimic native arteries that have an endothelium to
protect thrombosis in the inner layer, aligned, smooth muscle cells
in the middle to control constriction and vasodilation, and a me-
chanically robust outer layer. The mechanical properties such as
tensile, fatigue, brust pressue, and suture retention strength were
investigated. In addition, the biological interactions between the co-
culture of endothelial and smooth muscle cells with elctrospun PCL
scaffolds were evaluated. The results concluded that the elctrospun
tri-layers provided an adequate mechanical strength and the micro-
channels are able to contain the smooth muscle cells, and that cells
are able to adhere to PCL fibers. Han et al. [190] developed elc-
trospun-multilayered vascular scaffold in 1.5 mm diameter with
sufficient mechanical properties of poly (ethylene glycol) -b-poly
(lactide-co- -caprolactone) (PELCL), poly (ethylene glycol)
(PLGA), poly (-caprolactone) (PCL) and gelatine. The combina-
tion of PELCL, PLGA and PCL in three layers possessed 5.2 ± 0.7
MPa tensile strength, 146.7 ± 0.6% elongation and 6534 ± 926 mm
Hg burst pressure, higher than single component and comparable to
natural vessels. Spatio-temporal releases of growth factors, such as
vascular endothelial growth factor (VEGF) and platelet-derived
growth factor-bb (PDGF) and mechanical stability were controlled
by inner (PELCL), middle (PLGA) and outer (PCL) layers in that
order. However, gelatin significantly increased vascular endothelial
cell (VECs) adhesion on lumen and promoted VSMCs infiltration
from the outer layer in the middle layer. Cell activities indicated
dual release of growth factors (VEGF and PDGF). The improved
vascular grafts with dual-loading growth factors could maintain
potency in rabbit left common carotid artery for 8 weeks, compared
to other sample loading one kind of growth factor or without load-
ing any growth factor. Therefore, it is concluded that the specially
prepared vascular graft could benefit the blood vessel reconstruc-
tion.
Wang et al. fabricated a PCL tubular freeze-dried scaffold for
small diameter blood vessel replacements. The PCL scaffold was
prepared by dissolving PCL in acetic acid (15wt %) solution [191].
Further, the PCL scaffold was reinforced by embedding a tubular
fabric that was knitted from polyethylene terepthalate (PET) yarns
within the freeze-dried composite structure. The mechanical proper-
ties of reinforced scaffolds such as tensile strength, initial modulus,
radial compliance, compression recovery, and suture retention
force, were significantly improved compared to those of the unrein-
forced sample. Moreover, the reinforced composite structure is a
candidate for use as a tissue engineered scaffold for a future small
diameter vascular prosthesis. Furthermore, Wang et al. fabricated
vascular grafts from mesoporous elctrospun polycaprolactone
(PCL) scaffolds with thicker fibers (5-6 m) and large pores (~30
m) for macrophage polarization and arterial regeneration [192].
Further, demonstrated that thicker-fiber elctospun PCL grafts with
large pore sizes significantly enhanced cell infiltration, migration,
and vascularisation. In addition, the thicker fiber pores also stimu-
late the macrophage polarization into M2 phenotype (in vitro),
which subsequently mediated the regeneration process of vascular
grafts (neo-arteries in vivo) in comparison with the regular thinner-
fiber PCL grafts. In vitro culture showed that macrophages cultured
on thicker-fiber scaffolds tended to polarize into the immunomodu-
latory and tissue remodelling (M2) phenotype, whereas those cul-
tured on thinner-fiber scaffolds expressed pro-inflammatory (M1)
phenotype. In vivo implantation by replacing rat abdominal aorta
was performed and followed up for 7, 14, 28, and 100d. The results
demonstrated that the macroporous grafts significantly enhanced
cell infiltration and extracellular matrix (ECM) secretion. All grafts
showed satisfactory potency for up to 100 days. At day 100, the
endothelium coverage was complete, and the regenerated smooth
muscle layer was correctly organized with abundant ECM similar to
those in the native arteries. Moreover, the regerenated arteries dem-
onstrated contractile response to adrenaline and acetylcholine-
induced relaxation. In addition, cellularization process revealed that
the thicker-fiber scaffolds induced a large number of M2 macro-
phages to infiltration and vascularization into the graft wall. The
findings demonstrated that the combined biomaterial-immuno-
modulatory approach of thicker-fiber elctrospun PCL vascular
grafts should help with better design next generation of vascular
grafts and suggesting a promising candidate for future in vivo
evaluation. Yao et al. [193] developed a hybrid small-diameter
vascular graft (inner diameter of 1.5 mm) from synthetic polymer
poly (-caprolactone) (PCL) and natural polymer chitosan (CS) by
the co-elctrospinning technique. The prepared PCL/CS vascular
grafts had a tri-layer structure with a mass ratio varying from 5/4,
5/2 to 5/1 (w/w) for inner, middle and outer layers, respectively.
The vascular graft was further functionalized by immobilization of
heparin. The release heparin was monitored, and the effect of hepa-
rin release on the hemocompatibility, vascular cell proliferation and
inhibition of thrombus formation was investigated in vitro and in
vivo. The in vitro cell proliferation assay showed that heparin can
promote the growth of human umbilical vein endothelial cells
(HUVECs), while moderately inhibiting the proliferation of vascu-
lar smooth muscle cells (VMCs), a principal factor for neointimal
hyperplasia. Implantation in rat abdominal aorta for 1 month dem-
onstrated the heparin functionalized PCL/CS grafts sustained re-
lease of heparin with optimal anti-thrombogenic effect by reducing
thrombus formation. Furthermore, functionalized graft provided
better endothelialization and potency in comparison with the con-
trol group. In conclusion, heparin functionalized PCL/CS grafts
provided a facile and useful technique for the development of hea-
parinized medical devices, including vascular grafts. Wang et al.
[194] prepared a hybrid scaffold that consists of synthetic PCL
fibers and native gelatin (Gel) fibers by co-elctrospinning tech-
nique. The PCL provides optimal mechanical strength, while gela-
tin was heparinised in order to deliver VEGF in a controlled manner
which is monitored by in vtro releasing test. The mechanical prop-
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4491
erties and surface hydrophilicity was investigated. The effect of
releasing VEGF from the heparinised PCL/Gel scaffolds on im-
proving vascularisation has been investigated by both in vitro cell
experiment and in vivo subcutaneous implantation. The mechanical
properties and surface hydrophilicity was found satisfied as require
to the tissue engineering scaffolds. In vitro cell assay indicated that
VEGF release significantly enhanced the proliferation of endothe-
lial cells. More importantly, in vivo subcutaneous implantation
showed that the neovascularisation significantly improved in
PCl/Gel scaffolds compared with PCL counterpart due to sustained
release of VEGF. Thus, taken together the above properties, the
developed heparinised PCL/Gel scaffolds are a promising candidate
for the regeneration of complex tissues with sufficient vascularioza-
tion.
5.5.3. Poly (Ethylene Glycol) (PEG)
Leslie-Barbick et al. prepared a microvasularization of tissue
engineered constructs by using a VEGF (vascular endothelial
gro wth factor) -mimicking peptide, QK (Gln-Lys), covalently
bound to a poly (ethylene glycol) (PEG) hydrogel matrix [195]. The
PEGyalation of peptides (QK) increased its bioactivity and solubil-
ity, as evidenced by endothelial cell proliferation assay. In addition,
PEGyaltion of peptides showed the equal or superior ability to
promote angiogenesis in vitro within 3-D collagenase hydrogels
and on the surface of hydrogels, compared to PEG-VEGF hydro-
gels or only RGD (Arg-Gly-Asp) peptide sequences. Thus, PEG-
QK provided an alternative to the use of growth factor proteins in
angiogenic signalling. Zhu et al. [196] reported on the synthesis of
extra cellular matrix (ECM) -mimetic poly (ethylene glycol) (PEG)
hydrogels for inducing endothelial cell (EC) adhesion and capillary-
like network formation. A collagen type I-derived peptide (GIA)
containing PEGDA (PEGDA-GIA) was synthesized with the colla-
genase-sensitive GIA sequence attached in the middle of the
PEGDA chain, which was then copolymerized with RGD capped-
PEG monoacrylate (RGD-PEGMA) to form biomimetic hydrogels.
In addition, biomimetic RGD-PEGDA/GIA-PEGDA hydrogels,
showed an induced capillary like organization, on human umbilical
vein ECs (HUVECs) seeded hydrogel surface, compared to
RGD/PEGDA and GIA/PEGDA hydrogels. These results indicated
that the both cell adhesion and biodegradability of scaffolds paly an
important role in formation of capillary-like networks.
5.5.4. Polyurethane
There are few synthetic elastomeric biomaterials that simulta-
neously provide the required biological conditions and translate
biomechanical stimuli to vascular smooth muscle cells (VSMCs)
[197]. Polyurethanes are the class of synthetic polymers or elastom-
ers that have been performed various vascular tissue engineering
applications, such as ability to control high degree of different me-
chanical properties, tailored biochemical signalling, and specific
cell and material interactions, which ultimately influences extracel-
lular matrix (ECM) synthesis and tissue regeneration [198].
Sharifpoor et al. [199] developed a customized bioreactor used to
investigate the effects of uniaxial cyclic mechanical strain (CMS)
on human coronary artery smooth muscle cells (hCASMCs). The
cells (hCASMCs) were cultured in a porous degradable po-
lar/hydrophobic/ionic (D-PHI) polyurethane scaffold and its cell
viability as well as growth has been assessed. In addition, distribu-
tion, proliferation and protein expression of cells (hCASMCs) in
the scaffolds were then analyzed and compared to those grown
under control conditions. The resulted CMS-scaffolds showed a
greater DNA mass, more cell area coverage, and a better distribu-
tion of cells deeper within the scaffold construct. Moreover, CMS-
scaffolds provided enhanced tensile mechanical properties and
showed improved expression of contractile proteins (actin, myosin
and calponin), suggesting that hCASMCs retained the contractile
behavior on these CMS-scaffolds biomaterials. Du et al. [200] de-
veloped a 3D gradient heparinised nanofibrous scaffolds from ver-
tical graded elctrospun chitosan/poly -caprolactone (CS/PCL)
aiding endothelial cells lined on the lumen of blood vessel to pre-
vent thrombosis. Moreover, scaffolds were designed such a way
that, modify the surface properties of the scaffolds using heparin,
since it has been shown that heparinization and immobilization can
control the vascular endothelial growth factor (VEGF) delivery by
potentially increasing endothelial cells bioaffinity, proliferation and
anticoagulation properties. Furthermore, the quantity of heparinised
chitosan nanofibers increased gradually from the tunica adventitia
to the lumen surfaces in the gradient CS/PCL wall of tissue engi-
neered vessel. More heparin reacted to chitosan nanofibers in gradi-
ent CS/PCL than in uniform CS/PCL nonofibrous scaffolds. The
adhesion and proliferation of human umbilical vein endothelial
cells (HUVECs) were increased on the gradient CS/PCL scaffolds.
In addition, HUVECs grew and formed an entire monolayer on the
top side of the gradient CS/PCL scaffold. Taken together, the use of
vertical gradient heparinozed CS/PCL nanofibrous scaffolds could
provide an approach to create small-diameter blood vessel grafts
with similar natural blood vessels.
Although polyurethanes very often suffer from poor in endo-
thelization, are freqently used in the cardiovascular field due to
their tunable physiochemical properties and acceptable hemocoma-
patibility. For which, polyurethanes are synthesized as degradable
segmented products with help of amino acids as chain extenders.
Chan-Chan et al. [201] synthesized a cost-effective and straight-
forward method to enhance endothelial cell adhesion to the degrad-
able segmented poly (urea) urethanes (SPUUs) containing amino
acids (either L-arginine, glycine or L-aspartic acid) as chain ex-
tenders. These amino acids are selected on the basis of their differ-
ent pH behavior and their ability to form charged species since it
has been shown that positively charged surfaces can promote cell
adhesion while a slightly hydrophilic surface can enhance prolifera-
tion of cells. The alkaline L-arginine, neutral glycine and acidic L-
aspartic acid were incorporated separately into the polyurethane
backbone to provide both bulk and surface properties of the poly-
mer. All polymers showed a little mass loss (slow degradation) in
PBS (pH 7.4) but a higher mass loss (accelerated degradation)
showed under alkaline, acidic and oxidative media. The polyure-
thanes with L-aspartic acids exhibited poor mechanical properties.
Moreover, MTT tests on polyurethanes with L-arginine showed no
adverse effect on the metabolism of human umbilical vein endothe-
lial cells (HUVECs). However, polyurethanes with L-arginine
showed an enhanced higher level of HUVECs adhesion, spreading
and viability after 7 days compared to commonly used Tecoflex
polyurethane as a control. Taken together, along with a slow degra-
dation rate of polyurethanes containing L-arginine would be a po-
tential candidate for cardiovascular applications and angiogenesis.
Jing et al. [202] fabricated a thermoplastic polyurethane (TPU)
/grapheme oxide (GO) composite membrane and tubular scaffolds
(inner diameter of 3.18 mm) via elctrospinning at different GO
contents (0.5%, 1% and 2%) as potential candidates for small di-
ameter vascular grafts. In addition, the structure, properties and cell
response on the TPU/GO elctrospun scaffolds were investigated.
For investigation of cell response, mouse fibroblasts (3T3) and
human umbilical vein endothelial cells (HUVECs) were tested on
the TPU/GO elctrospun fibers. Further, suture retention assets, cy-
clic tensile tests, and burst pressure tests were conducted on
TPU/GO scaffolds to verify their ability to be used in vascular scaf-
fold applications. In terms of mechanical and surface properties, the
tensile strength, Young’s modulus, and hydrophobicity of scaffolds
(on plasma treatment) were found to increase with an increase of
GO contents. It was found that cell viability for both types of cells,
fibroblast (3T3) proliferation, and HUVECs attachment were the
highest at a 0.5 wt. % GO loading whereas oxygen plasma treat-
ment also enhanced HUVECs viability and attachment signifi-
cantly. Further, the suture retention strength and burst pressure of
tubular TPU/GO scaffolds containing 0.5 wt. % GO was found to
resemble with the native blood vessel and also endothelial cells
were able to attach to the inner surface of the tubular scaffolds. The
4492 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
platelet adhesion tests indicated that vascular scaffolds containin g
0.5 wt. % GO had low platelet adhesion and activation, and is un-
likely to cause thrombosis on the scaffolds. Therefore, the electros-
pun TPU/GO tubular scaffolds have the potential to be used in vas-
cular tissue engineering.
5.5.5. Silk
Zhu et al. [203] designed and fabricated porous 3-D silk fibroin
(SF) scaffolds and tubular scaffolds by modifying the freeze-drying
method which could be used as potential vascular grafts. The de-
veloped SF scaffolds provided the remarkable mechanical property,
excellent biocompatibility and biodegradability, and low immuno-
genicity. Furthermore, the anticoagulant activity of the heparin-
loaded SF scaffolds was investigated. In vitro assays were per-
formed to assess the effects of heparin-loaded SF scaffolds on hu-
man smooth muscle cell (hSMC) proliferation and infiltration. In
addition, the impact of the SF scaffolds on the tissue ingrowth and
neovascularisation were also examined in vivo by subcutaneous
implantation in rats. The incorporation of heparin during fabrication
possessed significant anticoagulant property and released in a sus-
tain manner for approximately 7 days. The heaparin-loaded scaf-
folds, showed inhibition of hSMCs proliferation within scaffold (in
vitro) and while significantly promoted hemocompatibility and
neovascularisation (in vivo) in subcutaneous implantation in rats.
Thus, heparin-loaded SF scaffolds considered as potential vascular
grafts.
6. DISCUSSION
Nanotechnology and nanomedicine have recently emerged as
alternative and promising therapeutic method for the treatment of
CVDs and related diseases. The potential technology utilizes
nanoscale dimension materials and bridges the gap between mo-
lecular and cellular interactions, and has the potential to revolution-
ize medicine [83]. In addition, as compared to traditional medicine,
nanomedicine offers early diagnosis, to treat as effectively as possi-
ble with minimal side effects and an improved clinical result [204].
Various nanotechnology applications are being studied for the
treatment of atherosclerosis and restenosis, and related CVDs in-
cluding nanocarriers for drug delivery. The major nanocarrier
classes explored as therapeutic and theranostic (i.e., particles inte-
grating diagnostic imaging and therapeutic components). Nanopati-
cles (NPs) such as SPIONs have been used as a prolonged anti-
angiogenesis therapy and MRI imaging with targeted thrombolysis
[205]. There is also the reversed side of interaction of NPs with the
walls of blood vessel. The vascular endothelium could be an obsta-
cle and undesirable target for NPs to be delivered to other organs
[92]. In this regard, CVDs may affect the transport of NPs across
vessel walls, organelle-targeted delivery of NPs, and other effects
of NPs on vessel cells. These factors should be taken into account
in future nanomedical research.
Gene delivery and therapy is the most notable therapy, hold
great promise as treatment and prevention methods for CVDs [206].
However, the systemic administration of these therapeutics is af-
fected by several barriers such as uptake by the reticuloendothelial
system, enzymatic degradation, kidney filtration, and limited intra-
cellular entry. Therefore, nanocarriers have been widely used to
overcome and best alternative to make therapeutics of CVDs be-
comes more efficient [206]. In addition, site-targeted drug delivery
holds promise in the treatment of vascular injury-associated throm-
botic and occlusive events caused by cardiovascular diseases (e.g.
atherosclerosis) or interventional procedure (e.g. angioplasty and
stenting). However, conventional techniques are expensive and
require experienced personnel for their use [95]. Thus, a novel car-
rier based targeted delivery of thrombolytic agents is a desirable
promising strategy for the thrombolytic agents is a desirable prom-
ising strategy for the treatment of thromboembolic diseases. How-
ever, the systemic side effects of these thrombolytic agents
(systemic fibrinogenolysis and bleeding) raise and impose unavoid-
able clinical difficulties. The side effects of therapeutic agents
comprise proteolytic components of the blood clotting cascade, and
thus their thrombolytic abilities are also responsible for their capac-
ity to disrupt haemostasis systemically. To reduce the incidence of
the systemic side effects thrombolytic agents could specifically
delivered targeted to the thrombus sites in vivo, or particular dos-
ages which are required and used for therapeutically effective ef-
fects of drugs at the thrombus site [118].
On the other side, quantum dots (QDs) contain toxic compo-
nents (such as Cd). The coating/ functionalization with QDs have
been used with nontoxic materials such as organic mole-
cules/polymers (e.g. PEG) or inorganic layers (e.g. ZnS and silica)
to reduce the levels of toxicity. Therefore, several groups of re-
searchers have been studied the water soluble QDs encapsulated by
amphiphilic polymers (with PEG), claimed very low toxicity to
cells [207-209]. In recent years, tissue-engineering (TE) scaffolds
have been fabricated by emphasized the control over cell behaviors.
The tissue formation can be functionalized by tissue engineered
scaffolds that closely mimics the natural extracellular matrix
(ECM). By understanding the behaviors of natural ECM, research-
ers have been developing/ constructed nanofibrous scaffolds/nano-
composites through electrospinning or self-assembly techniques
[210].
Beside all these facts, nanotechnology based formulated drugs
has made a significant impact on the treatment of CVD. The field of
cardiovascular therapeutic and theranostic holds tremendous prom-
ise and excitement for basic and clinical scientists to work together
to explore the possibilities of nanoscale technologies for the treat-
ment of CVDs.
CONCLUSION
In developed and developing countries, CVDs symbolize an
enormous burden on the healthcare system and economy because
they are the leading cause of morbidity and mortality [138]. The
most challenging goal in the field of cardiovascular tissue engineer-
ing is the creation/ regeneration of an engineered tissue for re-
placement or transplant. Recent advances in cell scaffolds provide a
platform for delivery of bioactive molecules as well as a physical
environment to allow for cell attachment thus preventing cells from
being carried away. A variety of materials have been used as cell
scaffolds including synthetic and natural polymers and hydrogels,
native extracellular matrix (ECM) proteins, and processed native
ECM following myocardial infarction towards cardiovascular tissue
regeneration [93]. Moreover, in recent years, more advances in
methods of stem cell isolation, culture in bioreactors, and the syn-
thesis of bioactive materials promise to create engineered tissue ex
vivo. Furthermore, new modalities are perceived that explore ways
to induce cardiovascular tissue regeneration after injury.
The rapid evolution of nanotechnology, which connects the gap
between interactions on the nanoscale and macroscopic levels, one
of the major promising players in the development of the detection
and treatment of CVDs. Though still in the very early developmen-
tal stages, cardiovascular nanomedicine is likely to meet the high
demand for breakthrough innovation in the diagnosis and treatment
of CVDs. Nanomedicine enables the design of multi-functional
agents, which can simultaneously and precisely detect and treat
CVDs and related diseases. Thus, there remains a significant oppor-
tunity to develop new methods and techniques for the generation of
cardiovascular tissue engineering grafts, to promote therapeutic
strategies for CVDs treatments and to create fully functioning car-
diovascular tissues.
CONFLICT OF INTEREST
The authors confirm that this article content has no conflict of
interest.
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4493
ACKNOWLEDGEMENTS
The authors acknowledge the financial support from Ministry of
Human Resource Development, Government of India, and TEQIP-
II programme funded by the World Bank.
REFERENCES
[1] Poirier P, Giles TD, Bray GA, et al. Obesity and cardiovascular
disease: pathophysiology, evaluation, and effect of weight loss an
update of the 1997 American Heart Association Scientific state-
ment on obesity and heart disease from the obesity committee of
the council on nutrition, physical activity, and metabolism. Circula-
tion 2006; 113: 898-918.
[2] Goldhaber SZ, Bounameaux H. Pulmonary embolism and deep
vein thrombosis.Lancet 2012; 379: 1835-46.
[3] Yang Q, Cogswell ME, Flanders WD, et al. Trends in cardiovascu-
lar health metrics and associations with all-cause and CVD mortal-
ity among US adults.JAMA 2012; 307: 1273-83.
[4] Bauer UE, Briss PA, Goodman RA, Bowman BA. Prevention of
chronic disease in the 21st century: elimination of the leading pre-
ventable causes of premature death and disability in the U SA.Lan-
cet 2014; 384: 45-52.
[5] Ross R, Glomset JA. Atherosclerosis and the arterial smooth mus-
cle cell.Science 1973; 180: 1332-9.
[6] Anderson KM, Odell PM, Wilson PW, Kannel WB. Cardiovascular
disease risk profiles.Am Heart J 1991; 121: 293-8.
[7] Sidney S, Rosamond WD, Howard VJ, Luepker RV. The “heart
disease and stroke statistics—2013 update” and the need for a na-
tional cardiovascular surveillance system.Circulation 2013; 127:
21-3.
[8] Mathers CD, Loncar D. Projections of global mortality and burden
of disease from 2002 to 2030.PLoS Med 2006; 3: e442.
[9] Cai H, Harrison D G. Endothelial dysfunction in cardiovascular
diseases: the role of oxidant stress. Circ Res 2000; 87: 840-4.
[10] Lim SS, Vos T, Flaxman AD, et al. A comparative risk assessment
of burden of disease and injury attributable to 67 risk factors and
risk factor clusters in 21 regions, 1990-2010: a systematic analysis
for the Global Burden of Disease Study 2010. Lancet 2012; 380:
2224-60.
[11] Yusuf S, Reddy S, Ounpuu S, Anand S. Global burden of cardio-
vascular diseases part I: general considerations, the epidemiologic
transition, risk factors, and impact of urbanization.Circulation
2001; 104: 2746-53.
[12] Teo K, Chow CK, Vaz M, Rangarajan S, Yusuf S. The Prospective
Urban Rural Epidemiology (PURE) study: examining the impact of
societal influences on chronic noncommunicable diseases in low-,
middle-, and high-income countries.Am Heart J 2009; 158: 1-7.
[13] Gaziano TA, Bitton A, Anand S, Abrahams-Gessel S, Murphy A.
Growing epidemic of coronary heart disease in low-and middle-
income countries.Curr Problems Cardiol 2010; 35: 72-115.
[14] Mendis S, Puska P, Norrving B (eds). Global Atlas on Cardiovas-
cular Disease Prevention and Control. Word Health Organization,
Geneva, 2011. Available from: (apps.who.int)
[15] Thom T, Haase N, Rosamond W, et al. Heart disease and stroke
statistics—2006 update a report from the American Heart Associa-
tion Statistics Committee and Stroke Statistics Subcommit-
tee.Circulation 2006; 113: 85-151.
[16] Zhao Y, Malik S, Wong ND. Evidence for coronary artery calcifi-
cation screening in the early detection of coronary artery disease
and implications of screening in developing countries.Global Heart
2014; 9: 399-407.
[17] Gupta R, Joshi P, MohanV, Reddy KS, Yusuf S. Epidemiology and
causation of coronary heart disease and stroke in India. Heart 2008;
94: 16-26.
[18] Patel V, Chatterji S, Chisholm D, et al. Chronic diseases and inju-
ries in India.Lancet 2011; 377: 413-28.
[19] Linazasoro G. Nanotechnologies for Neurodegenerative Diseases
Study Group of the Basque Country (NANEDIS): Potential appli-
cations of nanotechnologies to Parkinson's disease ther-
apy. Parkinson Related Disord 2008; 14: 383-92.
[20] Fang J, Chen YC. Nanomaterials for photohyperthermia: a review.
Curr Pharm Des 2013; 19: 6622-6634.
[21] Behera SS, Jha S, Arakha M, Panigrahi TK. Synthesis of Silver
Nanoparticles from microbial source-a green synthesis approach,
and evaluation of its antimicrobial activity against Escherichia coli.
Int J Eng Res Appl (IJERA)2013; 3: 058-62.
[22] Silva GA. Neuroscience nanotechnology: progress, opportunities
and challenges. Nat Rev Neurosci 2006; 7: 65-74.
[23] Mohan V, Sandeep S, Deepa R, Shah B, Varghese C. Epidemiol-
ogy of type 2 diabetes, Indian scenario. Indian J Med Res 2007;
125: 217-30.
[24] Chobanian AV, Bakris GL, Black HR, et al. Seventh report of the
joint national committee on prevention, detection, evaluation, and
treatment of high blood pressure.Hypertension 2003; 42: 1206-52.
[25] Ahmed MS, Jadhav AB, Hassan A and Meng QH. Acute phase
reactants as novel predictors of cardiovascular disease. ISRN In-
flam 2012; 2012: 1-18.
[26] Aronson D, Bartha P, Zinder O, et al. Obesity is the major determi-
nant of elevated C-reactive protein in subjects with the metabolic
syndrome, Int J Obes Relat Metab Disord 2004; 28: 674-9.
[27] Gabrielsen JS, Gao Y, Simcox JA, et al. Adipocyte iron regulates
adiponectin and insulin sensitivity.J Clin Invest 2012; 122: 3529-
40.
[28] Basuli D, Stevens RG, Torti FM, Torti SV. Epidemiological asso-
ciations between iron and cardiovascular disease and diabe-
tes.Front Pharmacol 2014; 5.
[29] Ewang-Emukowhate M, Perera DS, Wierzbicki DMA. Dyslipi-
daemia related to insulin resistance and cardiovascular disease in
South Asian and West African populations.Curr Pharm Des 2014;
20(40): 6270-5.
[30] Eguchi A, Naito Y, Iwasaku T, et al. Association of dietary iron
restriction with left ventricular remodeling after myocardial infarc-
tion in mice.Heart Vessels 2015, 1-8.
[31] Toss H, Lindahl B, Siegbahn A, et al. Prognostic influence of in-
creased fibrinogen and C-reactive protein levels in unstable coro-
nary artery disease. Circulation 1997; 96(12): 4204-10.
[32] He LP, Tang XY, Ling WH, Chen WQ, Chen YM. Early C-
reactive protein in the prediction of long-term outcomes after acute
coronary syndromes: a meta-analysis of longitudinal studies.Heart
2010; 96(5): 339-46.
[33] Eapen D, Kalra GL, Merchant N, Arora A, Khan BV. Metabolic
syndrome and cardiovascular disease in South Asians. Vasc Health
Risk Manag 2009; 5: 731- 43.
[34] Sanghamitra P, Mohapatra PC.The role of uric acid in cardiovascu-
lar Disease and its clinical implications. Orissa J Med Biochem
2004: 1; 39-43
[35] Fang J, Alderman MH. Serum uric acid and cardiovascular mortal-
ity in the NHANES I epidemiologic follow-up study, 1971-1992.
National Health and Nutrition Examination Survey. JAMA 2000;
283: 2404-10.
[36] Chatterjee B, Shah T, Trivedi A, Mahant H, Katwa V, Gosai, K.
(2014). Novel cardiovascular risk factors in metabolic syndrome
with and without coronary artery disease. J Res Med Dental Sci
2014; 2: 29-36.
[37] Pearson TA, Mensah GA, Alexander RW, et al. Markers of in-
flammation and cardiovascular disease application to clinical and
public health practice: a statement for healthcare professionals from
the centers for disease control and prevention and the American
Heart Association.Circulation 2003; 107: 499-511.
[38] Libby P, Okamoto Y, Rocha VZ, Folco E. Inflammation in athero-
sclerosis.Circ J 2010; 74: 213-20.
[39] Wang Z, Nakayama T. (2010). Inflammation, a link between obe-
sity and cardiovascular disease.Mediat Inflam 2010; 2010: 535918.
[40] Kones R. Primary prevention of coronary heart disease: integration
of new data, evolving views, revised goals, and role of rosuvastatin
in management: A comprehensive survey.Drug Des Dev Ther
2011; 5: 325.
[41] Sierevogel MJ, Pasterkamp G, De Kleijn DPV, Strauss B. Matrix
metalloproteinases: a therapeutic target in cardiovascular disease.
Curr Pharm Des 2003; 9(13): 1033-1040.
[42] Athyros GV, Tziomalos K, Katsiki N, Mikhailidis DP. Editorial
(Thematic Issue: Novel Data on the Pathogenesis of Atherosclero-
sis, Treatment Targets, and New Therapeutic Interventions in
Lipid-Related Cardiovascular Risk Factors). Curr Pharm Des
20(40), 6215-9.
[43] Rocha M, Apostolova N, Hernandez-Mijares A, Herance R, M
Victor V. Oxidative stress and endothelial dysfunction in cardio-
vascular disease: mitochondria-targeted therapeutics. Curr Med
Chem 2010; 17(32): 3827-41.
4494 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
[44] Deftereos S, Bouras G. Editorial (Thematic Issue: Targeting In-
flammation in Cardiovascular Disease).Med Chem 2014; 10(7):
641-2.
[45] Ridker PM, Hennekens C H, Buring JE, Rifai N. C-reactive protein
and other markers of inflammation in the prediction of cardiovas-
cular disease in women. New Eng J Med 2000; 342(12): 836-43.
[46] Aranega A. Therapeutic approaches in regenerative medicine of
cardiovascular diseases: from bench to bedside. Adv Regen Med
2011; 61.
[47] Tongers J, Losordo DW, Landmesser U. Stem and progenitor cell-
based therapy in ischaemic heart disease: promise, uncertainties,
and challenges. Eur Heart J 2011; 32(10): 1197-206.
[48] Assmus B, Schachinger V, Teupe C, et al. Transplantation of pro-
genitor cells and regeneration enhancement in acute myocardial in-
farction. Circulation 2002; 106(24): 3009-17.
[49] Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature
2008; 451(7181): 937-42.
[50] Tomita S, Li RK, Weisel RD, et al. Autologous transplantation of
bone marrow cells improves damaged heart function. Circulation
1999; 100(suppl 2): II-247.
[51] Yoder MC, Mead LE, Prater D, et al. Redefining endothelial pro-
genitor cells via clonal analysis and hematopoietic stem/progenitor
cell principals. Blood 2007; 109(5): 1801-9.
[52] Loffredo FS, Steinhauser M L, Gannon J, Lee RT. Bone marrow-
derived cell therapy stimulates endogenous cardiomyocyte progeni-
tors and promotes cardiac repair. Cell Stem Cell 2011; 8(4): 389-
98.
[53] Mahmoodzadeh S, Leber J, Zhang X, et al. Cardiomyocyte-specific
estrogen receptor alpha increases angiogenesis, lymphangiogenesis
and reduces fibrosis in the female mouse heart post-myocardial in-
farction. J Cell Sci Therapy 2014; 5(1): 153.
[54] Dilley RJ, Morrison WA. Vascularisation to improve translational
potential of tissue engineering systems for cardiac repair. Int J Bio-
chem Cell Biol 2014; 56: 38-46.
[55] Casieri V, Agostini S, Lionetti V. Epigenetic modulation of myo-
cardial angiogenic balance: an emerging therapeutic perspective for
adult failing heart. Curr Angiog 2014; 3(1): 3-10.
[56] Laflamme MA, Murry CE. Regenerating the heart. Nat Biotechnol
2005; 23(7): 845-56.
[57] Fang H, Lai NC, Gao MH, et al. Comparison of adeno-associated
virus serotypes and delivery methods for cardiac gene trans-
fer. Human Gene Therapy, Part B: Methods 2012; 23(4): 234-41.
[58] Katz MG, Fargnoli AS, Tomasulo CE, Pritchette LA, Bridges CR.
Modelspecific selection of molecular targets for heart failure gene
therapy. J Gene Med 2011; 13(10): 573-586.
[59] Katz MG, Fargnoli AS, Williams RD, Bridges CR. Gene therapy
delivery systems for enhancing viral and nonviral vectors for car-
diac diseases: current concepts and future applications. Hum Gene
Ther 2013; 24(11): 914-27.
[60] Tang T, Hammond HK. Gene transfer for congestive heart failure:
update 2013. Transl Res 2013; 161(4): 313-20.
[61] Lu ZH, Kaliberov S, Zhang J, et al. The myeloid-binding peptide
adenoviral vector enables multi-organ vascular endothelial gene
targeting. Lab Invest 2014; 94(8): 881-92.
[62] Guellich A, Mehel H, Fischmeister R. Cyclic AMP synthesis and
hydrolysis in the normal and failing heart. Pflügers Arc Eur J
Physiol 2014; 466(6): 1163-75.
[63] Steffel J, Lüscher TF, Tanner FC. Tissue factor in cardiovascular
diseases molecular mechanisms and clinical implications. Circula-
tion 2006; 113(5): 722-31.
[64] Breitenstein A, Tanner FC, Luscher TF. Tissue factor and cardio-
vascular disease: quo vadis?. Circulation J 2010; 74(1): 3-12.
[65] Ambros V. The functions of animal microRNAs.Nature 2004; 431:
350
[66] Bartel, DP. MicroRNAs: genomics, biogenesis, mechanism, and
function.Cell116: 281-97.
[67] Bonauer A, Carmona G, Iwasaki M, et al. MicroRNA-92a controls
angiogenesis and functional recovery of ischemic tissues in mice.
Science 2009; 324: 1710-3.
[68] Fiedler J, Jazbutyte V, Kirchmaier BC, et al. MicroRNA-24 regu-
lates vascularity after myocardial infarction. Circulation 2011; 124:
720-30.
[69] Da Costa Martins PA, Salic K, Gladka MM, et al. MicroRNA-199b
targets the nuclear kinase Dyrk1a in an auto-amplication loop
promoting calcineurin/NFAT signalling. Nat Cell Biol 2010; 12:
1220-7.
[70] Creemers EE, Tijsen AJ, Pinto YM. Circulating microRNAs novel
biomarkers and extracellular communicators in cardiovascular
disease?. Circ Res 2012; 110(3): 483-95.
[71] Van Rooij E, Marshall WS, Olson EN. Toward MicroRNA-based
therapeutics for heart disease the sense in antisense. Circ Res 2008;
103(9): 919-28.
[72] Reid G, Kirschner MB, Van Zandwijk N. Circulating microRNAs:
Association with disease and potential use as biomarkers. Crit Rev
Oncol Hematol 2011; 80(2): 193-208.
[73] Ajit SK. Circulating microRNAs as biomarkers, therapeutic targets,
and signaling molecules. Sensors 2012; 12(3): 3359-69.
[74] Raitoharju E, Oksala N, Lehtimaki T. MicroRNAs in the athero-
sclerotic plaque. Clin Chem 2013; 59(12): 1708-21.
[75] Fan X, Wang E, Wang X, Cong X, Chen X. MicroRNA-21 is a
unique signature associated with coronary plaque instability in hu-
mans by regulating matrix metalloproteinase-9 via reversion-
inducing cysteine-rich protein with Kazal motifs. Exp Mol Pathol
2014; 96(2): 242-9.
[76] Lu X, Kakkar V. The roles of microRNAs in atherosclerosis. Curr
Med Chem 2014; 21(13): 1531-43.
[77] VanWijk MJ, VanBavel E, Sturk A, Nieuwland R. Microparticles
in cardiovascular diseases. Cardiovasc Res 2003; 59(2): 277-87.
[78] Biasucci L M, Porto I, Di Vito L, et al. Differences in microparticle
release in patients with acute coronary syndrome and stable an-
gina. Circ J 2012; 76(9): 2174-82.
[79] Boulanger CM, Amabile N, Tedgui A. Circulating microparticles a
potential prognostic marker for atherosclerotic vascular dis-
ease. Hypertension 2006; 48(2): 180-6.
[80] Rossig L, Hoffmann J, Hugel B, et al. Vitamin C inhibits endothe-
lial cell apoptosis in congestive heart failure. Circulation 2001;
104(18): 2182-7.
[81] Hugel B, Martínez MC, Kunzelmann C, Freyssinet JM. (2005).
Membrane microparticles: two sides of the coin. Physiology 2005;
20(1): 22-7.
[82] Gordon DJ, Probstfield JL, Garrison RJ, et al. High-density lipo-
protein cholesterol and cardiovascular disease. Four prospective
American studies. Circulation 1989; 79(1): 8-15.
[83] Boden WE. High-density lipoprotein cholesterol as an independent
risk factor in cardiovascular disease: assessing the data from
Framingham to the Veterans Affairs High-Density Lipoprotein In-
tervention Trial. Am J Cardiol 2000; 86(12): 19-22.
[84] Sharrett A R, Ballantyne CM, Coady SA, et al. Coronary heart
disease prediction from lipoprotein cholesterol levels, triglycerides,
lipoprotein (a), apolipoproteins AI and B, and HDL density sub-
fractions the atherosclerosis risk in communities (ARIC)
study. Circulation 2001; 104(10): 1108-13.
[85] Bochem AE, Kuivenhoven J A, Stroes ESG. The promise of cho-
lesteryl ester transfer protein (CETP) inhibition in the treatment of
cardiovascular disease.Curr Pharm Des 2013; 19(17): 3143-9.
[86] Elseweidy MM, Abdallah FR, Younis NN, Aldohmy S, Kassem
HM. 10-Dehydrogingerdione raises HDL-cholesterol through a
CETP inhibition and wards off oxidation and inflammation in dys-
lipidemic rabbits. Atherosclerosis 2013; 231: 334-40.
[87] Gordon DJ, Probstfield JL, Garrison R J, et al. High-density lipo-
protein cholesterol and cardiovascular disease. Four prospective
American studies.Circulation1989; 79: 8-15.
[88] Poss J, Custodis F, Werner C, Weingartner O, Bohm, M, Laufs U.
Cardiovascular disease and dyslipidemia: beyond LDL.Curr Pharm
Des 2011; 17: 861-70.
[89] Gouni-Berthold I. Editorial [Hot Topic: Cardiovascular Disease:
Focus on Dyslipidemia (Executive Guest Editor: I. Gouni-
Berthold)].Curr Pharm Des 2011; 17: 850-1.
[90] Van Capelleveen JC, Brewer HB, Kastelein JJ, Hovingh GK.
Novel therapies focused on the high-density lipoprotein parti-
cle.Circ Res 2014; 114: 193-204.
[91] Briand F, Prunet-Marcassus B, Thieblemont Q, et al. Raising HDL
with CETP inhibitor torcetrapib improves glucose homeostasis in
dyslipidemic and insulin resistant hamsters. Atherosclerosis 2014;
233: 359-62.
[92] Rader DJ, Hovingh GK. HDL and cardiovascular disease.The
Lancet 2014; 384: 618-25.
[93] Remaley AT, Norata GD, Catapano AL. Novel concepts in HDL
pharmacology.Cardiovasc Res 2014; 103: 423-8.
[94] Riccioni G, Back M, Capra V. Leukotrienes and atherosclerosis.
Curr Drug Targets 2010; 11: 882-7.
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4495
[95] Funk CD. Leukotriene modifiers as potential therapeutics for car-
diovascular disease.Nat Rev Drug Discov 2005; 4: 664-72.
[96] Mastalerz L, Sanak M, Szczeklik A. Pharmacological inhibitors of
cysteinyl leukotrienes biosynthesis: therapeutic implications.Curr
Med Chem-Anti-Inflam Anti-Allergy Agents 2004; 3: 157-65.
[97] Sanak M, Dropinski J, Sokolowska B, Faber J, Rzeszutko M,
Szczeklik A. Pharmacological inhibition of leukotriene biosynthe-
sis: effects on the heart conductance.J Physiol Pharmacol 2010; 61:
53.
[98] Szczeklik W, Gorka J, Kozka M, et al. Leukotrienes biosynthesis in
vascular surgery patients during perioperative period.J Physiol
Pharmacol 2014; 65: 705-8.
[99] Stary HC, Chandler AB, Glagov S, et al. A definition of initial,
fatty streak, and intermediate lesions of atherosclerosis: a report
from the Committee on Vascular Lesions of the Council on Arte-
riosclerosis, American Heart Association. Circulation 1994; 89:
2462-78.
[100] Haeggström JZ, Funk CD. Lipoxygenase and leukotriene path-
ways: biochemistry, biology, and roles in disease.Chem Rev 2011;
111: 5866-98.
[101] Arvan DA, Spitalnik PF. Robbins Pathological Basis of Disease.
In: Ramzi S. Cotran, Vinay Kumar, and Tucker Collins, Eds. Clini-
cal Chemistry. Philadelphia, PA: WB Saunders Co.1999; pp.1583-
4.
[102] Garcia C, Boyce BF, Gilles J, et al.Leukotriene B4 stimulates
osteoclastic bone resorption both in vitro and in vivo. J Bone Miner
Res 1996; 11: 1619-27.
[103] Shi SS, Yang WZ, Tu XK, Wang CH., Chen CM, Chen Y. 5-
lipoxygenase inhibitor zileuton inhibits neuronal apoptosis follow-
ing focal cerebral ischemia. Inflammation 2013; 36: 1209-17.
[104] Qin HD, Qin CC, Zhang HT, Yu X J, Chang J. Inhibitory effect of
zileuton on inflammatory injury to myocardium following ische-
mia/reperfusion in rats.Med J Chinese People's Liberation Army
2014; 39: 192-6.
[105] Tu XK, Yang WZ, Chen JP, et al. Curcumin inhibits TLR2/4-NF-
B signaling pathway and attenuates brain damage in permanent
focal cerebral ischemia in rats. Inflammation 2014; 1-8.
[106] Paul M, Poyan Mehr A, Kreutz R. Physiology of local renin-
angiotensin systems. Physiol Rev 2006; 86: 747-803.
[107] Santos RA, Jose Campagnole-Santos M, Pinheiro SV, Ferreira A J.
The renin-angiotensin system: emerging concepts.Curr Hyper Rev
2006; 2: 219-26.
[108] Shi L, Mao C, Xu Z, Zhang L. Angiotensin-converting enzymes
and drug discovery in cardiovascular diseases.Drug Discov Today
2010; 15: 332-41.
[109] Reddy Gaddam R, Chambers S, Bhatia M. ACE and ACE2 in
inflammation: a tale of two enzymes. Inflam Allergy-Drug Targets
2014; 13: 224-34.
[110] Zheng JL, Li G Z, Chen SZ, et al. Angiotensin converting enzyme
2/Ang(1-7)/Mas axis protects brain from ischemic injury with a
tendency of agedependence.CNS Neurosci Ther 2014; 20: 452-9.
[111] Santos RA, Ferreira AJ, Simoes e Silva AC. Recent advances in the
angiotensinconverting enzyme 2-angiotensin (1-7)-Mas axis. Exp
Physiol 2008; 93(5): 519-27.
[112] Absar S, Nahar K, Kwon YM, Ahsan F. Thrombus-targeted nano-
carrier attenuates bleeding complications associated with conven-
tional thrombolytic therapy.Pharm Res2013; 30: 1663-76.
[113] Kunamneni A, Taleb T, Abdelghani A, Ellaiah P. Streptokinase-the
drug of choice for thrombolytic therapy. J Thromb Thrombolysis
2007; 23: 9-23.
[114] Nguyen PD, O’rear EA, Johnson AE, et al. Thrombolysis using
liposomal-encapsulated streptokinase: an in vitro study. Proc Soc
Exp Biol Med 1989; 192: 261-9.
[115] Murray V, Norrving B, Sandercock PAG, Terént A, Wardlaw JM,
Wester P. The molecular basis of thrombolysis and its clinical ap-
plication in stroke.J Internal Med 2010; 267: 191-208.
[116] Campbell J, Hilleman D. Recombinant peptides in thrombolysis.
In: Seminars in thrombosis and hemostasis.Thieme Medical Pub-
lishers, 2010; 36: pp. 529-36.
[117] Zhang J, Ma G, Lv Z, et al. Targeted thrombolysis strategies for
neuroprotective effect.Neural Regen Res 2014; 9: 1316.
[118] Vaidya B, Nayak MK, Dash D, Agrawal GP, Vyas SP. Develop-
ment and characterization of site specific target sensitive liposomes
for the delivery of thrombolytic agents.Int J Pharm 2011; 403:
254-61.
[119] Du X, Wang J, Diao W, Wang L, Long J, Zhou H. A genetically
modified protein-based hydrogel for 3D culture of AD293
Cells.PloS One 2014; 9: 107949.
[120] Behera SS, Patra JK, Pramanik K, Panda N, Thatoi H. Characteri-
zation and Evaluation of Antibacterial Activities of Chemically
Synthesized Iron Oxide Nanoparticles.World J Nano Sci Eng
2012; 2: 196-200.
[121] Godin B, Sakamoto JH, Serda RE, Grattoni A, Bouamrani A, Fer-
rari M. Emerging applications of nanomedicine for the diagnosis
and treatment of cardiovascular diseases. Trends Pharmacol Sci
2010; 31: 199-205.
[122] Arayne MS, Sultana N,Qureshi F. Review: nanoparticles in deliv-
ery of cardiovascular drugs. Pakistan J Pharm Sci 2007; 20(4): 340-
8.
[123] Shi J, Votruba AR, Farokhzad OC, Langer R. Nanotechnology in
drug delivery and tissue engineering: from discovery to applica-
tions. Nano Lett 2010; 10(9): 3223-30.
[124] Chen J, Tung C H, Mahmood U, et al. In vivo imaging of prote-
olytic activity in atherosclerosis. Circulation 2002; 105(23): 2766-
71.
[125] Ntziachristos V, Yodh AG, Schnall M, Chance B. Concurrent MRI
and diffuse optical tomography of breast after indocyanine green
enhancement. Proc Natl Acad Sci 2000; 97(6): 2767-72.
[126] Alessi P, Ebbinghaus C, Neri D. Molecular targeting of angiogene-
sis. Biochim Biophys Acta (BBA)-Rev Cancer 2004; 1654(1): 39-
49.
[127] Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis.
Circulation 2002; 105(9): 1135-43.
[128] Cunningham C H, Arai T, Yang PC, et al. Positive contrast mag-
netic resonance imaging of cells labeled with magnetic nanoparti-
cles.Magn Reson Med 2005; 53: 999-1005.
[129] Hahn MA, Singh AK, Sharma P, Brown SC, Moudgil BM.
Nanoparticles as contrast agents for in-vivo bioimaging: current
status and future perspectives. Anal Bioanal Chem 2011; 399(1): 3-
27.
[130] Semelka RC, Helmberger TK. Contrast Agents for MR Imaging of
the Liver 1. Radiology 2001; 218(1): 27-38.
[131] Na HB, Song IC, Hyeon T. Inorganic nanoparticles for MRI con-
trast agents. Adv Mater 2009; 21(21): 2133-48.
[132] Serda RE, Ferrati S, Godin B, Tasciotti E, Liu X, Ferrari M.
(2009). Mitotic trafficking of silicon microparticles.Nanoscale
2009; 1: 250-9.
[133] Schlorf T, Meincke M, Kossel E, et al. Biological properties of iron
oxide nanoparticles for cellular and molecular magnetic resonance
imaging. Int J Mol Sci 2010; 12(1): 12-23.
[134] Zhou J, Guo D, Zhang Y, Wu W, Ran H, Wang Z. Construction
and evaluation of Fe3O4-based PLGA nanoparticles carrying rtPA
used in the detection of thrombosis and in targeted thromboly-
sis. ACS Appl Mater Interf 2104; 6(8): 5566-76.
[135] Jaffer FA, Libby P, Weissleder R. Molecular imaging of cardiovas-
cular disease. Circulation 2007; 116(9): 1052-61.
[136] Winter PM, Shukla HP, Caruthers SD, et al. Molecular imaging of
human thrombus with computed tomography. Acad Radiol 2005;
12(5): S9-S13.
[137] Winter PM. Molecular Imaging at Nanoscale with Magnetic Reso-
nance Imaging. Nanomed A Soft Matter Persp 2014; 75.
[138] Medintz IL, Uyeda H T, Goldman ER, Mattoussi H. Quantum dot
bioconjugates for imaging, labelling and sensing. Nat Mater 2005;
4(6): 435-46.
[139] Smith A M, Duan H, Mohs A. M, Nie S. Bioconjugated quantum
dots for in vivo molecular and cellular imaging. Adv Drug Deliv
Rev 2008; 60(11): 1226-40.
[140] Qi L, Gao X. Quantum dot amphipol nanocomplex for intracellu-
lar delivery and real-time imaging of siRNA. ACS Nano 2008;
2(7): 1403-10.
[141] Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted sys-
tems for cancer therapy. Adv Drug Deliv Rev 2012; 64, 206-12.
[142] Zhu Y, Hong H, Xu Z P, Li Z, Cai W. Quantum dot-based nano-
probes for in vivo targeted imaging. Curr Mol Med 2013; 13(10):
1549-67.
[143] He Y, Zhong Y, Su Y, et al. Waterdispersed
nearinfraredemitting quantum dots of ultrasmall sizes for in vitro
and in vivo imaging. Angew Chem Int Ed 2011; 50(25): 5695-98.
[144] Probst CE, Zrazhevskiy P, Bagalkot V, Gao X. Quantum dots as a
platform for nanoparticle drug delivery vehicle design. Adv Drug
Deliv Rev 2013; 65(5): 703-18.
4496 Current Pharmaceutical Design, 2015, Vol. 21, No. 30 Behera et al.
[145] Xu H, Li Q, Wang L, et al. Nanoscale optical probes for cellular
imaging. Chem Soc Rev 2014; 43(8): 2650-61.
[146] So MK, Xu C, Loening AM, Gambhir SS, Rao J. Self-illuminating
quantum dot conjugates for in vivo imaging. Nat Biotechnol 2006;
24(3): 339-43.
[147] Liu W, Howarth M, Greytak A B, et al. Compact biocompatible
quantum dots functionalized for cellular imaging. J Am Chem Soc
2008; 130(4): 1274-84.
[148] Wen CJ, Zhang LW, Al-Suwayeh SA, Yen TC, Fang JY. Ther-
anostic liposomes loaded with quantum dots and apomorphine for
brain targeting and bioimaging. Int J Nanomed 2012; 7: 1599.
[149] Cao J, Zhu H, Deng D, et al. In vivo NIR imaging with PbS quan-
tum dots entrapped in biodegradable micelles. J Biomed Mater Res
Part A 2012; 100(4): 958-68.
[150] Pichaandi J, Van Veggel FC. Near-infrared emitting quantum dots:
Recent progress on their synthesis and characterization. Coordin
Chem Rev 2014; 263: 138-50.
[151] Rai M, Gade A, Gaikwad S, Marcato PD, Durán N. Biomedical
applications of nanobiosensors: the state-of-the-art. J Brazilian
Chem Soc 2012; 23(1): 14-24.
[152] Travas-Sejdic J, Aydemir N, Kannan B, Williams DE, Malmstrom
J. Intrinsically conducting polymer nanowires for biosensing. J Ma-
ter Chem B 2014; 2(29): 4593-609.
[153] Nam J M, Thaxton CS, Mirkin CA. Nanoparticle-based bio-bar
codes for the ultrasensitive detection of proteins. Science 2003;
301(5641): 1884-6.
[154] Zhang Y, Zhou D.Magnetic particle-based ultrasensitive biosensors
for diagnostics. 2012; 12 (6): 565-71.
[155] Suprun E, Bulko T, Lisitsa A, et al. Electrochemical nanobiosensor
for express diagnosis of acute myocardial infarction in undiluted
plasma. Biosens Bioelectr 2010; 25(7): 1694-8.
[156] Bashir R. BioMEMS: state-of-the-art in detection, opportunities
and prospects.Adv Drug Deliv Rev 2004; 56(11): 1565-86.
[157] Wilson AD, Baietto M. Advances in electronic-nose technologies
developed for biomedical applications. Sensors 2011; 11(1): 1105-
76.
[158] Greulich T, Hattesohl A, Grabisch A, et al. Detection of obstructive
sleep apnoea by an electronic nose.Eur Respirat J 2013; 42(1):
145-55.
[159] Ardito R, Corigliano A, Frangi A. Multiscale finite-element models
for predicting spontaneous adhesion in MEMS. Mécanique Indust
2010; 11(3-4): 177-82.
[160] Ardito R, Frangi A, Corigliano A, De Masi B, Cazzaniga G. The
effect of nano-scale interaction forces on the premature pull-in of
real-life Micro-Electro-Mechanical Systems.Microelectr Reliab
2012; 52(1): 271-81.
[161] VaradanVK. The role of Nanotechnology and Nano and Micro-
Electronics in monitoring and control of Cardiovascular Diseases
and Neurological Disorders. In The 14th International Symposium
on: Smart Structures and Materials & Nondestructive Evaluation
and Health Monitoring 2007; 652813-652813.
[162] Goldberg M, Langer R, Jia X. Nanostructured materials for appli-
cations in drug delivery and tissue engineering. J Biomater Sci Po-
lym Ed 2007; 18(3): 241-68.
[163] Suarez S, Almutairi A, Christman KL. Micro-and nanoparticles for
treating cardiovascular disease. Biomater Sci 2015; 3: 564-80.
[164] Sy JC, Davis ME. Delivering regenerative cues to the heart: cardiac
drug delivery by microspheres and peptide nanofibers.J Cardiovasc
Transl Res 2010; 3: 461-8.
[165] Paul A, Shum-Tim D, Prakash S. Angiogenic nanodelivery systems
for myocardial therapy. Cell Cardiomyopl 2013; 137-49.
[166] Erdogan S, Özer AY, Volkan B, Caner B, Bilgili H. Thrombus
localization by using streptokinase containing vesicular systems.
Drug Deliv 2006; 13: 303-9.
[167] Leach JK, O’rear EA, Patterson E, Miao Y, Johnson AE. Acceler-
ated thrombolysis in a rabbit model of carotid artery thrombosis
with liposomeencapsulated and microencapsulated streptokinase.
Thromb Haemost 2003; 90: 64-70.
[168] Gupta AS, Huang G, Lestini BJ, Sagnella S, Kottke-Marchant K,
Marchant RE. RGD-modified liposomes targeted to activated plate-
lets as a potential vascular drug delivery system. Thromb Haemost
2005; 93: 106-14.
[169] Huang G, Zhou Z, Srinivasan R, et al. Affinity manipulation of
surface-conjugated RGD peptide to modulate binding of liposomes
to activated platelets. Biomaterials 2008; 29: 1676-85.
[170] Huwyler J, Drewe J, Krahenbuhl S. Tumor targeting using liposo-
mal antineoplastic drugs. Int J Nanomed 2008; 3(1): 21.
[171] Zhou J, Patel TR, Sirianni RW, et al. Highly penetrative, drug-
loaded nanocarriers improve treatment of glioblastoma. Proc Natl
Acad Sci 2013; 110(29): 11751-6.
[172] Danhier F, Ansorena E, Silva J M, Coco R, Le Breton A, Préat V.
PLGA-based nanoparticles: an overview of biomedical applica-
tions. J Control Release 2012; 161(2): 505-22.
[173] Chang MY, Yang YJ, Chang CH, et al. Functionalized nanoparti-
cles provide early cardioprotection after acute myocardial infarc-
tion. J Control Release 2013; 170: 287-94.
[174] Kona S, Dong JF, Liu Y, Tan J, Nguyen KT. Biodegradable
nanoparticles mimicking platelet binding as a targeted and con-
trolled drug delivery system. Int J Pharm 2012; 423(2): 516-24.
[175] Vyas SP, Vaidya B. Targeted delivery of thrombolytic agents: role
of integrin receptors. Exp Opin Drug Deliv 2009; 6: 409-508.
[176] Langer HF, Gawaz M. Platelet-vessel wall interactions in athero-
sclerotic disease. Thromb Haemost 2008; 99: 480-6.
[177] Nugent HM, Edelman ER. Tissue engineering therapy for cardio-
vascular disease.Circ Res 2003; 92: 1068-78.
[178] Sankaran KK, Subramanian A, Krishnan UM, Sethuraman S.
Nanoarchitecture of scaffolds and endothelial cells in engineering
small diameter vascular grafts.Biotechnol J 2015; 10: 96-108.
[179] Eschenhagen T, Zimmermann WH. (2005). Engineering myocar-
dial tissue. Circ Res 2005; 97: 1220-31.
[180] Zund G, Breuer CK, Shinoka T, et al. The in vitro construction of a
tissue engineered bioprosthetic heart valve. Eur J Cardiothor Surg
1997; 11: 493-7.
[181] Hu X, Shen H, Yang F, Bei J, Wang S. Preparation and cell affinity
of microtubular orientation-structured PLGA (70/30) blood vessel
scaffold.Biomaterials 2008; 29: 3128-36.
[182] Ikada Y. Challenges in tissue engineering.J Royal Soc Interface
2006; 3: 589-601.
[183] Ma H, Hu J, Ma PX. Polymer scaffolds for smalldiameter vascular
tissue engineering.Adv Funct Mater 2010; 20: 2833-41.
[184] Han J, Lazarovici P, Pomerantz C, Chen X, Wei Y, Lelkes PI. Co-
electrospun blends of PLGA, gelatin, and elastin as potential
nonthrombogenic scaffolds for vascular tissue engineer-
ing.Biomacromolecules 2010; 12: 399-408.
[185] Nguyen TH, Lee BT. The effect of cross-linking on the microstruc-
ture, mechanical properties and biocompatibility of electrospun
polycaprolactone-gelatin/PLGA-gelatin/PLGA-chitosan hybrid
composite. Sci Technol Advd Mater 2012; 13: 035002.
[186] Heidenreich PA, Trogdon J G, Khavjou OA, et al. Forecasting the
future of cardiovascular disease in the United States a policy state-
ment from the American heart association.Circulation 2011; 123:
933-44.
[187] Gupta MK, Walthall JM, Venkataraman R, et al. Combinatorial
polymer electrospun matrices promote physiologically-relevant
cardiomyogenic stem cell differentiation.PloS One 2011; 6:
e28935.
[188] Seyednejad H, Ji W, Yang F, et al. Coaxially electrospun scaffolds
based on hydroxyl-functionalized poly (-caprolactone) and loaded
with VEGF for tissue engineering applications.Biomacromolecules
2012; 13: 3650-60.
[189] Lee CHY. Electrospun Polycaprolactone Scaffolds for Small-
Diameter Tissue Engineered Blood VesselsDoctoral dissertation,
The Ohio State University, 2013.
[190] Han F, Jia X, Dai D, et al. Performance of a multilayered small-
diameter vascular scaffold dual-loaded with VEGF and
PDGF.Biomaterials 2013; 34: 7302-13.
[191] Wang F, Mohammed A, Li C, Ge P, Wang L, King MW. Degrad-
able/non-degradable polymer composites for in-situ tissue engi-
neering small diameter vascular prosthesis application.Bio-med
Mater Eng 2014; 24: 2127-33.
[192] Wang Z, Cui Y, Wang J, et al. The effect of thick fibers and large
pores of electrospun poly (-caprolactone) vascular grafts on
macrophage polarization and arterial regeneration. Biomaterials
2014; 35: 5700-10.
[193] Yao Y, Wang J, Cui Y, et al. Effect of sustained heparin release
from PCL/chitosan hybrid small-diameter vascular grafts on anti-
thrombogenic property and endothelialization.Acta Biomater 2014;
10: 2739-49.
[194] Wang K, Chen X, Pan Y, et al. Enhanced vascularization in hybrid
PCL/gelatin fibrous scaffolds with sustained release of VEGF.
BioMed Res Int 2014; ID 865076.
Recent Advancement in the Treatment of Cardiovascular Diseases Current Pharmaceutical Design, 2015, Vol. 21, No. 30 4497
[195] Leslie-Barbick JE, Saik JE, Gould DJ, Dickinson ME, West JL.
The promotion of microvasculature formation in poly (ethylene
glycol) diacrylate hydrogels by an immobilized VEGF-mimetic
peptide.Biomaterials 2011; 32: 5782-9.
[196] Zhu J, He P, Lin L, Jones DR, Marchant RE. Biomimetic poly
(ethylene glycol)-based hydrogels as scaffolds for inducing endo-
thelial adhesion and capillary-like network formation. Biomacro-
molecules 2012; 13: 706-13.
[197] Stankus JJ, Guan J, Fujimoto K, Wagner WR. Microintegrating
smooth muscle cells into a biodegradable, elastomeric fiber matrix.
Biomaterials 2006; 27: 735-44.
[198] Lee M, Wu BM, Dunn JC. Effect of scaffold architecture and pore
size on smooth muscle cell growth. J Biomed Mater Res A 2008;
87: 1010-6.
[199] Sharifpoor S, Simmons CA, Labow RS, Paul Santerre J. Functional
characterization of human coronary artery smooth muscle cells un-
der cyclic mechanical strain in a degradable polyurethane scaffold.
Biomaterials 2011; 32: 4816-29.
[200] Du F, Wang H, Zhao W, et al. Gradient nanofibrous chitosan/poly
-caprolactone scaffolds as extracellular microenvironments for
vascular tissue engineering.Biomaterials 2012; 33: 762-70.
[201] Chan-Chan L H, Tkaczyk C, Vargas-Coronado RF, Cervantes-Uc
JM, Tabrizian M, Cauich-Rodriguez JV. Characterization and bio-
compatibility studies of new degradable poly (urea) urethanes pre-
pared with arginine, glycine or aspartic acid as chain extenders.J
Mater Sci Mater Med 2013; 24: 1733-44.
[202] Jing X, Mi HY, Salick MR, Cordie TM, Peng XF, Turng LS. Elec-
trospinning thermoplastic polyurethane/graphene oxide scaffolds
for small diameter vascular graft applications.Mater Sci Eng C
2014; 49: 40-50.
[203] Zhu M, Wang K, Mei J, et al. Fabrication of highly interconnected
porous silk fibroin scaffolds for potential use as vascular
grafts.Acta Biomater 2014; 10: 2014-23.
[204] Singh B, Garg T, Goyal AK, Rath G. Recent advancements in the
cardiovascular drug carriers. Artif Cell Nanomed Biotechnol 2014;
(0): 1-10.
[205] Godin B, Sakamoto JH, Serda RE, Grattoni A, Bouamrani A, Fer-
rari M. Emerging applications of nanomedicine for the diagnosis
and treatment of cardiovascular diseases. Trends Pharmacol Sci
2010; 31(5): 199-205.
[206] Ryden L, Scherer M, Syvanne M. European Guidelines on cardio-
vascular disease prevention in clinical practice. Eur Heart J 2012;
33: 1635-701.
[207] ZhuY, Hong H, Xu ZP, Li Z,Cai W. Quantum dot-based nano-
probes for in vivo targeted imaging. Curr Mol Med 2013; 13(10):
1549-67.
[208] Jamieson T, Bakhshi R, Petrova D, Pocock R, Imani M, Seifalian
AM. Biological applications of quantum dots. Biomaterials 2007;
28(31): 4717-32.
[209] William WY, Chang E, Drezek R, Colvin V L. Water-soluble
quantum dots for biomedical applications. Biochem Biophys Res
Commun 2006; 348(3): 781-6.
[210] Ravichandran R, Seitz V, Reddy Venugopal J, et al. Mimicking
native extracellular matrix with phytic acid-crosslinked protein
nanofibers for cardiac tissue engineering. Macromol Biosci 2013;
13: 366-75.
Received: April 14, 2015 Accepted: July 31, 2015
... Finally, nanoparticle-based imaging is leveraged to gauge the effectiveness of monotherapy, as highlighted in reference (Tang et al., 2012). In cardiology, nanomedicine finds substantial utility in four critical domains: molecular imaging, tissue engineering, targeted drug delivery, and diagnostics, serving as a proactive approach to prevent and manage CVDs (S Behera et al., 2015). Leveraging the inherent capabilities of nanocarriers offers many advantages, including precise drug delivery, heightened bioavailability of therapeutic agents, and controlled release to specific targets, as expounded in Fig. 7. ...
... China and India have made significant progress already in this race. Only six of the 57 Islamic nations, namely Iran, Turkey, Egypt, Saudi Arabia, Pakistan, and Malaysia, appear to be exhibiting encouraging tendencies in the advancement of nanotechnology (S Behera et al., 2015). Pakistan, Turkey, and Egypt are the only three of these six countries with no national nanotechnology programmed. ...
Article
The increasing burden of cardiovascular disease (CVD) remains responsible for morbidity and mortality worldwide; their effective diagnostic or treatment methods are of great interest to researchers. The use of NPs and nanocarriers in cardiology has drawn much interest. The present comprehensive review provides deep insights into the use of current and innovative approaches in CVD diagnostics to offer practical ways to utilize nanotechnological interventions and the critical elements in the CVD diagnosis, associated risk factors, and management strategies of patients with chronic CVDs. We proposed a decision tree-based solution by discussing the emerging applications of NPs for the higher number of rules to increase efficiency in treating CVDs. This review-based study explores the screening methods, tests, and toxicity to provide a unique way of creating a multi-parametric feature that includes cutting-edge techniques for identifying cardiovascular problems and their treatments. We discussed the benefits and drawbacks of various NPs in the context of cost, space, time and complexity that have been previously suggested in the literature for the diagnosis of CVDs risk factors. Also, we highlighted the advances in using NPs for targeted and improved drug delivery and discussed the evolution toward the nano-cardiovascular potential for medical science. Finally, we also examined the mixed-based diagnostic approaches crucial for treating cardiovascular disorders, broad applications and the potential future applications of nanotechnology in medical sciences.
... Its physiology includes four chambers, which consist of two atria and two ventricles, as shown in Fig. 1. The cardiovascular system distributes blood, nutrition, oxygen, hormones, and other essentials to cells, tissues, and organs [9]. ...
Article
Full-text available
Globally, high death rates due to heart failure are an essential topic in medical research. Cardiovascular disease is the leading cause of cardiac dysfunction and collapse, with high mortality and morbidity rates. Early diagnosis and prognosis of CVD will reduce the risk of cardiovascular conditions. It is essential to develop various tools that provide accurate, real-time insight into the heart's physiology, functionality, and cardiac events. Due to the dispersed nature of the information and the reported results, a comprehensive literature review is required because there is a shortage of data about the hemodynamics analysis of blood flow in the ventricular region. Therefore, reviewing the status of hemodynamics analysis of ventricle blood f low is the prime importance of this review article. This article reviews the numerous investigations conducted over the past 15 years to simulate ventricular blood flow using experimental and computational techniques on patient-specific models or idealized models with or without specific medical conditions. This article discusses the fundamentals of hemodynamic analysis, such as the geometry types of a particular cardiac phase, medical conditions, and medical imaging methods. Recent developments in hemodynamic analysis, such as AI, HPC, and digital twins, were also mentioned in this comprehensive review study. This review article concluded that improvements in medical image processing and data acquisition techniques are needed to obtain accurate information regarding the functioning of the heart. Our review of previous studies shows that blood flow simulation is developing into a proper medical tool for instant heart function diagnosis.
... Improved healthcare system and the advancement of medical practice are additional possible contributing factors to the observed decreased burden of lead-associated CVD and CKD. The treatment of IHD, stroke, and hypertensive heart diseases, as major subclasses of CVDs, depend upon timely intervention, adequately advanced medical equipment, and trained medical personnel 26 . A similar framework is also applicable to the treatment and care of CKD, as the disease requires meticulous acute and chronic care 27,28 . ...
Article
Full-text available
This study aimed to investigate the estimated burden attributed to lead exposure (LE), at the national and subnational levels from 1990 to 2019 in Iran. The burden attributed to LE was determined through the estimation of deaths, disability-adjusted life years (DALYs), years of life lost (YLLs) and years lived with disability (YLDs) using the comparative risk assessment method of Global Burden of Disease (GBD) study presenting as age-standardized per 100,000 person year (PY) with 95% uncertainty intervals (95% UI). Furthermore, the burden of each disease were recorded independently. Eventually, the age-standardized YLLs, DALYs, deaths and YLDs rates attributed to LE demonstrated a decrease of 50.7%, 48.9%, 38.0%, and 36.4%, respectively, from 1990 to 2019. The most important causes of LE burden are divided into two acute and chronic categories: acute, mainly causes mental disorders (DALYs rate of 36.0 in 2019), and chronic, results in cardiovascular diseases (CVDs) (DALYs rate of 391.8) and chronic kidney diseases (CKDs) (DALYs rate of 26.6), with CVDs bearing the most significant burden. At the sub-national level, a decrease in burden was evident in most provinces; moreover, low and low-middle SDI provinces born the highest burden. The burden increased mainly by ageing and was higher in males than females. It was concluded that although the overall decrease in the burden; still it is high, especially in low and low-middle SDI provinces, in advanced ages and in males. Among IDID, CKDs and CVDs that are the most important causes of LE-attributed burden in Iran; CVDs bear the highest burden.
... Because of the high morbidity, mortality, and rehospitalization rates, researchers have been compelled to seek the most effective methods of diagnosing, risk-stratifying, and managing patients with suspected cardiovascular illnesses (Garcia and Spyropoulos, 2008). CVDs are a set of conditions that affect the heart, the blood vessels (capillaries, arteries, and veins), or both (Behera et al., 2015;Buller et al., 2005;Hyers 1999). These include congenital heart disorders, rheumatic heart disorders, coronary heart disorders, cerebrovascular disorders, peripheral arterial disorders, and venous thromboembolism (VTE), of which we emphasize the VTE in this review (Wang et al., 2020;Wells et al., 2014). ...
... It is instantaneous to find an optimal solution for this potentially fatal issue [2]. Cardiovascular diseases (CVD) are classified into four types [3]. First, the most common type is CAD, which occurs due to plaque buildup in coronary arteries, leading to reduced blood flow to the heart muscle, causing heart attacks, chest pain, and shortness of breath [4]. ...
Article
Full-text available
Coronary Artery Disease (CAD) is an increasingly prevalent disorder that significantly affects both longevity and quality of life, particularly among people aged 30 to 60. Lifestyle, genetics, nutrition, and stress are contributing factors to the increasing mortality rates. Therefore, there is a need for low-cost automated technology to diagnose CAD early and help medical practitioners treat chronic illnesses effectively. However, machine learning methods are typically designed to perform well with large datasets and may not be well-suited for smaller clinical datasets that contain categorical features. To address this issue, alternative approaches such as Data Preprocessing (DP), feature selection techniques, and Hyperparameter tuning (HP) are necessary to achieve optimal performance and accuracy on such datasets. Data preprocessing is also crucial to obtain accurate results by eliminating noise, handling missing values, and dealing with outliers. To address the challenges associated with feature selection, manual selection of hyperparameter tuning, and optimization, we have developed a novel model called BSOXGB (BorutaShap feature selection based Optuna hyperparameter tuning of eXtremely Gradient Boost). The proposed model achieves a remarkable accuracy of 97.70%, outperforming other classifiers like Random Forest (RF), AdaBoost (AB), CatBoost (CB), and ExtraTrees (ET) on the publicly available Z-Alizadeh Sani dataset. BSOXGB, with only 9 relevant features out of 56, has the highest classification accuracy, demonstrating its potential as a practical solution for automatically detecting and diagnosing CAD in the real world.
... The potential use of nanotechnology in treating CVDs(146).International Journal of Pharmaceutical Research and ApplicationsVolume 8, Issue 4 July-Aug 2023, pp: 2222-2251 www.ijprajournal.com ISSN: 2249-7781 DOI: 10.35629/7781-080422222251 | Impact Factor value 7.429 | ISO 9001: 2008 Certified Journal Page 2231 (i) ...
Article
Full-text available
The phrase "cardiovascular disease" (CVD) refers to all conditions that affect the heart and vasculature, the organs responsible for the displacement and transportation of the blood, respectively. This complex condition includes a variety of inherited and acquired illnesses. Cardiovascular disease includes rheumatic heart disease (damage to the myocardium and cardiac valves caused by streptococci bacteria), congenital heart disease, peripheral artery disease, cerebral disease, and peripheral artery disease with its subtypes (coronary, cerebral, and deep vein thrombosis), which are more common than hemorrhagic stroke.The leading cause of death is cardiovascular disease in both highly industrialized and undeveloped countries. 30% of all-cause mortality in 2008, or 17.5 million people, were attributable to CVDs. It is anticipated to reach 23.3 million by 2030.The needs of the patient will be met during therapy. Infectious diseases and cardiovascular medicine are currently beginning to adopt these designs as well. Until recently, oncology was the only field using clinical trial designs that assessed a biomarker for prognostic or predictive utility.For treating patients with CVDs, using nanoparticles in medicine provides significant advantages over both traditional and cutting-edge medical techniques.
Article
Thrombus has long been the major contributor of death and disability because it can cause adverse effects to varying degrees on the body, resulting in vascular blockage, embolism, heart valve deformation, widespread bleeding, etc. However, clinically, conventional thrombolytic drug treatments have hemorrhagic complication risks and easy to miss the best time of treatment window. Thus, it is an urgent need to investigate newly alternative treatment strategies that can reduce adverse effects and improve treatment effectiveness. Drugs based on nanomaterials act as a new biomedical strategy and promising tools, and have already been investigated for both diagnostic and therapeutic purposes in thrombus therapy. Recent studies have some encouraging progress. In the present review, we primarily concern with the latest developments in the areas of nanomedicines targeting thrombosis therapy. We present the thrombus’ formation, characteristics, and biomarkers for diagnosis, overview recent emerging nanomedicine strategies for thrombus therapy, and focus on the future design directions, challenges, and prospects in the nanomedicine application in thrombus therapy.
Article
Full-text available
THE PURPOSE of the "Sixth Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure" (JNC VI) is to provide guidance for primary care clinicians. The committee recognizes that the responsible clinician's judgment of the individual patient's needs remains paramount. Therefore, this national guideline should serve as a tool to be adapted and implemented in local and individual situations. Using evidence-based medicine and consensus, the report updates contemporary approaches to hypertension control. Among the issues covered are the important need for prevention of high blood pressure by improving lifestyles, the cost of health care, the use of self-measurement of blood pressure, the role of managed care in the treatment of high blood pressure, the introduction of new combination antihypertensive medications and angiotensin II receptor blockers, and strategies for improving adherence to treatment. The JNC VI report places more emphasis than earlier reports on absolute
Article
Objective To investigate the protective effect of zileuton, an inhibitor of 5-lipoxygenase (5-LO), on ischemia/ reperfusion (I/R) injury of rat myocardium, and its probable mechanisms. Methods Thirty female SD rats were randomly divided into 3 groups (10 each): sham group (S group), ischemic/reperfusion group (I/R group), and zileuton group (Z group). The I/ R model was reproduced by left anterior descending (LAD) artery occlusion for 45min followed by 120-min reperfusion. To rats of Z group 3mg/kg zileuton was given 15min before ischemia. Animals were sacrificed after reperfusion. The degree of myocardial damage was determined by means of pathological examination. Myocardial apoptosis was identified by TUNEL assay. The levels of leukotriene B4 (LTB4), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in plasma were determined by ELISA; the distribution of 5-LO in neutrophils was determined by immunofluorescence and Western blotting. Results Compared with I/ R group, zileuton attenuated the I/R-induced myocardial damage significantly. TUNEL assay demonstrated a significant decrease in myocardial apoptosis by zileuton (P<0.05). Inflammatory cytokines, including LTB4, IL-1β and TNF-α in the plasma were significantly decreased in Z group than in I/R group (P<0.05). Additionally, immunofluorescence and Western blotting showed that zileuton significantly inhibited the activation and redistribution of 5-LO (P<0.05). However, no significant difference in total 5-LO protein expression was found among the three groups (P>0.05). Conclusion Zileuton may protect myocardium from I/R injury in rats through suppressing myocardial apoptosis and inflammation by inhibiting the 5-LO activation and redistribution.
Article
Atherosclerosis, formerly considered a bland lipid storage disease, actually involves an ongoing inflammatory response. Recent advances in basic science have established a fundamental role for inflammation in mediating all stages of this disease from initiation through progression and, ultimately, the thrombotic complications of atherosclerosis. These new findings provide important links between risk factors and the mechanisms of atherogenesis. Clinical studies have shown that this emerging biology of inflammation in atherosclerosis applies directly to human patients. Elevation in markers of inflammation predicts outcomes of patients with acute coronary syndromes, independently of myocardial damage. In addition, low-grade chronic inflammation, as indicated by levels of the inflammatory marker C-reactive protein, prospectively defines risk of atherosclerotic complications, thus adding to prognostic information provided by traditional risk factors. Moreover, certain treatments that reduce coronary risk also limit inflammation. In the case of lipid lowering with statins, this anti-inflammatory effect does not appear to correlate with reduction in low-density lipoprotein levels. These new insights into inflammation in atherosclerosis not only increase our understanding of this disease, but also have practical clinical applications in risk stratification and targeting of therapy for this scourge of growing worldwide importance.