You are on page 1of 11

Journal of Controlled Release 337 (2021) 494–504

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Review article

Advance cardiac nanomedicine by targeting the pathophysiological


characteristics of heart failure
Congcong Lin a, b, Huile Gao c, *, Liang Ouyang a, *
a
State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041,
China
b
Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China
c
Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, 610041, China

A R T I C L E I N F O A B S T R A C T

Keywords: Heart failure (HF) has continued to be a leading cause of morbidity and mortality worldwide. Nanomedicine,
Cardiac nanomedicine which can deliver therapeutic drugs/biomolecules specifically to damaged myocardium and overcome the
Targeted drug delivery limitations of conventional therapies, shows great potential in the treatment of HF. Although a number of
Biological barriers
preclinical studies of cardiac nanoformulations have been published, targeted nanomedicine for HF is yet to be
Pathophysiological changes
Heart failure
applied in clinical practice. Therefore, it is meaningful to sum up past experiences and deepen the understanding
of nanomedicine and HF. In this review, we first emphasized the key biological barriers to cardiac nanomedicine
that hinder its targeting effect. Since the rational design of nanoparticles should take into account the specific
characteristics of HF, we then summarized the key pathophysiological changes of HF to provide a clear un­
derstanding on HF, as well as the latest examples of nanotechnology-based delivery strategies for different
pathophysiological characteristics. Finally, the major challenges are discussed in detail, aiming to provide
guidance for future development of cardiac nanomedicine.

1. Introduction these beneficial effects in the application of nanoparticles, the clinical


translation of nanomedicine for the treatment of cardiac diseases is still
HF is a heterogeneous and complex syndrome that results from on the way. As such, summarizing the previous experiences and deep­
impairment of cardiac structure and function [1]. Although important ening the knowledge of nanomedicine and HF are necessary, which may
advances have been made in HF treatment, it remains a dominant cause contribute to finding innovative solutions to meet the current challenges
of death worldwide. Conventional methods including dilation of blood in the treatment of HF.
vessels, limiting inflammation and the use of antithrombotic measures HF is not so much a disease as a pathophysiological state caused by
are far from optimal, resulting in a 5-year mortality rate of up to 50%, myocardial injury. Following myocardial injury, a network involving
even 2–3 times that of breast cancer, colorectal cancer and other ma­ molecular, cellular and structural mechanisms is activated to maintain
lignant tumors [2,3]. It is worth noting that by 2030, the prevalence of the physiological function [9]. These coordinated, complicated pro­
HF is expected to increase by 46%, by then 1 in 33 Americans will suffer cesses accompanied with a series of pathophysiological changes
from HF [4,5]. Therefore, alternative therapies such as nanomedicine including activated oxidative stress, aberrant extracellular matrix
have been sought after in recent years. (ECM), overloaded intracellular calcium and activated immune system.
As the forefront of life sciences, nanomedicine represents a prom­ In view of this, it has great potential for the treatment of HF in response
ising approach for health care [6]. In line with this, over 100 nano­ to the key physiological changes caused by HF. Such potential can be
medicine applications and products have been approved by the U.S. further fueled by nanotechnology-based delivery strategy that allows
Food and Drug Administration (FDA) [7]. For delivery purposes, nano­ targeted delivery of payloads to pathological areas. Besides, these
medicine has multiple advantageous properties such as drug protection pathophysiological alterations that distinguish diseases and healthy
from systemic degradation, controlled and sustained release of payloads, parts can serve as biological stimuli to “turn on” nanomedicine in the
tissue targeting and reduced secondary effects [8]. However, despite body so that the nanomedicine can only provide its therapeutic effect on

* Corresponding authors.
E-mail addresses: gaohuile@scu.edu.cn (H. Gao), ouyangliang@scu.edu.cn (L. Ouyang).

https://doi.org/10.1016/j.jconrel.2021.08.002
Received 24 May 2021; Received in revised form 2 August 2021; Accepted 2 August 2021
Available online 4 August 2021
0168-3659/© 2021 Elsevier B.V. All rights reserved.
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

the pathological site. ischemia/reperfusion injury [18]. In this study, a prolonged circulation
In this review, we focus on the advances in cardiac nanomedicine time of PEGylated liposomal formulation was observed by labeling
oriented to the pathophysiological characteristics of HF. The review adenosine with radioisotope. However, if the PEGylated formulation is
begins by discussing the key biological barriers of nanomedicine for HF, repeatedly injected during long-term treatment, the prolonged circula­
and the directions of nanocarrier design are highlighted. On this basis, tion effect may disappear caused by the production of anti-PEG IgM
the recent advances in the development of cardiac nanomedicine that [19,20]. Researchers have been endeavor to overcome this challenging
target the key pathophysiological properties of HF are reviewed and dilemma of PEGylation, such as pre-infusion with high molecular weight
finally the major challenges are proposed. Through such a compact re­ free PEG [21]. McSweeney et al. explored infusion of free PEG to satu­
view, we anticipate to afford a meaningful platform for better HF rate anti-PEG antibodies (APA) in mice. They found that the circulation
treatment and further nanomedicine research. time is effectively restored by infusion of 40 kDa free PEG without
causing excess humoral anti-PEG responses and toxicity. Alternatively,
2. Key biological barriers of cardiac nanomedicine the reaction of MPS to the surface charge of nanocarrier also offers cues
to extend residence time of nanocarrier in circulation [22]. Zwitterionic
Taking the advantage of nanotechnology, therapeutic agent can be materials such as poly (carboxybetaine) and poly (sulfobetaine) are
delivered to the injured heart in a targeted manner. It is imperative to considered as substitutes of PEG because of their strong hydration ability
accumulate predominant biomolecules/drugs within the target site for a and strong resistance to nonspecific protein adsorption [23]. Another
successful targeted delivery system [10,11]. In this regard, the pur­ robust top-down approach is cellular membrane coating. Red blood
posefully designed nanosystems for HF have to overcome a complex cells, leukocytes cells or platelets, as carriers of long circulation in na­
series of biological barriers on the journey from the circulatory system to ture, provide a platform for the design of the next generation of nano­
the heart. Mononuclear phagocyte system (MPS) opsonization, hemor­ carriers [24–26].
heological limitations, contractile forces extravasation, and endosomal/ After escaping the clearance of MPS, nanomedicine needs to be
lysosomal degradation are the main biological barriers for cardiac effectively attached to the damaged heart. At this time, the fluid dy­
nanomedicine. In Fig. 1, the four key biological barriers and the po­ namics in blood vessels becomes the main factor affecting the possibility
tential strategies to overcome these barriers were highlighted. of nanocarriers reaching the heart. The dynamics of a nanocarrier in
As an important part of the immune system, MPS is the first obstacle blood vessels is mainly relied on its size and geometry. As reported,
to nanomedicine. Through the phagocytosis of MPS, nanoparticles can nanocarriers with diameters >200 nm retained readily in the liver and
be directly removed, thereby significantly reducing the number of spleen, while those <5 nm are filtered by the kidney [27]. On this basis,
nanoparticles reaching the target. To reduce the recognition rate of MPS nanocarriers may be prepared with diameters ranging from 5 to 200 nm,
and prolong the blood residence time, the most common solution is making them ideal for long-lasting in circulation. Before extravasation,
coating the carrier with water-soluble polymers [12–14]. Polyethylene- the nanoparticles have to first move to the wall of endothelial cells. That
glycol (PEG) is a well-developed coating polymer for nanocarriers, is to say, margination is the premise. Notably, as a muscular power
conferring good stealth against the MPS and low immunogenicity organ for pumping blood, the heart is the source of blood circulation
[15–17]. PEGylated liposomal adenosine, for example, was prepared for power. The speed of the heart pumping blood is amazing. It ejects blood

Fig. 1. Framework of biological barriers that nanomedicines encounter and the potential strategies to overcome the barrier.

495
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

at a speed of 8 m per second. Therefore, margination dynamics of membrane destabilization have been proposed. Deepen the underlying
nanoparticles to endothelial walls is a specifically essential consider­ mechanism of endosomal escape and ultimately engineering nano­
ation for the design of cardiac nanomedicine. Recent studies revealed particles capable of escape the endosomes are necessary.
that the attachment rate of nanoparticles is related to the shear stress of
endothelial wall and geometry of circulatory system [28,29]. It also has 3. Development of cardiac nanomedicine based on HF
been reported that physical characteristics of nanoparticles such as the pathophysiology
size, shape and density affect their margination towards endothelium
[30]. Take the shape for example, researchers have shown that the Notably, either ischemic or nonischemic HF is accompanied by a
attachment of discoidal nanocarriers is 5-fold higher than spherical ones series of pathophysiological responses including activated oxidative
[31]. For nonspherical nanocarriers, their lateral drifting velocity is stress, aberrant extracellular matrix (ECM), overloaded intracellular
proportional to their aspect ratio, and they exhibit unique tumbling and calcium, and activated immune system. Under the guidance of the above
rolling activity. However, for spherical nanocarriers, they tend to flow mentioned design principles, further development of nanomedicine
parallel to the vessel wall. As such, reimagining of nanoparticle geom­ based on these specific pathophysiological characteristics may be a
etry departure from conventional spherical shapes is a promising promising direction for preventing the progresses of HF. In this section,
effective strategy for nanomedicine. the key pathophysiological changes of HF are summarized and the latest
Subsequently, the cardiac nanomedicine should cross the endocar­ examples of nanotechnology-based delivery strategies targeting these
dium to further accumulate and remain in the injured heart. In contrast pathophysiological characteristics are reviewed (Fig. 2).
with cancer, the window of the enhanced permeability and retention
(EPR) effect in HF is short-lived [32]. Additionally, the issues of EPR 3.1. Nanomedicine targeting activated oxidative stress
effect including heterogeneity, lack of cellular specificity and limited
patient data have also attracted the researchers' attention. It is note­ Since ischemia is the most common cause of HF where blood flow
worthy that subjected to the contractile forces, cardiomyocytes envi­ and oxygen supply interrupted, the damaged heart tends to be exposed
ronment tends to expel injected drugs, which comes to be another key to a hypoxic condition [42]. As a consequence, the injured tissues/cells
barrier for the retention of cardiac nanomedicines. These findings cast show elevated oxidative stress. Oxidative stress is identified as an
doubt on the application of the EPR effect in nanomedicines of HF. extreme imbalance of “redox state” [43]. Under pathological condition,
Therefore, alternative and complementary strategies to enhance the high reactive oxygen species (ROS) are produced via various sources
permeability and retention of cardiac nanomedicines are explored. One including mitochondria, xanthine oxidase, NADPH oxidases and
of the most attractive strategies to enhance the permeability and uncoupled nitric oxide synthase (NOS) [44]. The excessive ROS can
retention of cardiac nanomedicines is active targeting. Active targeting activate apoptosis pathway and damage cell membrane and etc., thereby
simply referred as “ligand-receptor interaction”, can enhance the ag­ affecting the growth, proliferation and metabolism of cardiomyocytes in
gregation of nanoparticles at HF sites by cooperating with EPR effect. a variety of ways [45]. Therefore, strategies to minimize ROS of HF
The target can be used for HF targeted therapy should unique expressed patients are clinically important.
or at least overexpressed (e.g. CD13 and AT1) at the injured heart Endogenous/exogenous antioxidants are the first choice to deplete
relative to other part of body. From this perspective, identify more the excessive ROS in HF. However, they are either “ too nonspecific, too
specific receptors or proteins that can be employed in nanotherapy little, too late”, or they are related to the possible side effects [46–48].
targeting HF is urgently needed. Another controllable and intuitive To address these limitations, researchers have developed various
method is to design nanomedicines with adjustable shapes or sizes, nanovactors including liposomes, micelles, polyamidoamine (PAMAM)
which have recently proven their superiority in enhancing permeability dendrimers and polymer nanoparticles to deliver these ROS scavengers
and retention [33]. Typically, enzyme, pH, redox, temperature and light (Table 1). It is worth noting that the hypoxic condition in HF patients
are utilized to trigger the transformation of these formulations [34]. enhances vascular permeability through the production of angiogenic
Regarding transformable nanomedicines, the triggering factor that ini­ factors and the migration of endothelial cells [49]. As such, EPR effect, a
tiates the transformation process is the dominant factor and should be well-known concept in tumor nanotherapy, is also the most common
carefully designed. To ensure the conversion occurs at where it is principle in the research field of HF nanomedicines [50]. Taking the
needed, the distribution and quantity of the specific trigger need to be advantage of the disrupted endothelia wall, the nanoparticles can
sufficient evaluated. Besides, the in vivo fate of the transformable for­ passively pass through the blood vessels and endocardium, so that
mulations also should be closely monitored. relatively large doses of antioxidants are delivered to the damaged
Finally, the nanomedicine is expected to enter the damaged car­ myocardium [51]. Then exhibited their cardioprotective effects by in­
diomyocytes and escape from the endosome/lysosome, thereby hibition of inflammation or triggering the cascades involved in the
releasing the cargo into the cytoplasm. Normally, the cellular membrane antioxidant defense system. In this way, previously discarded antioxi­
traversal of nanoparticles is through endocytosis pathway [35]. The dants for HF therapy may still prove useful and be worthy of re-
exact endocytosis pathway varies with the size and surface modification examination.
of nanoparticles [36]. For example, the endocytosis of cellular receptor Since mitochondria are the main source of ROS, studies for the
modified nanoparticles is achieved through the clathrin-mediated elimination of mitochondrial ROS in cardiomyocytes is drawing more
endocytosis, while the caveolae-receptor modified nanoparticles inter­ attention for HF treatment in recent years. Taking the advantage of
nalize cells through the caveolae-mediated endocytosis pathway mitochondria-targeting materials, such as small mitochondria-targeting
[37–39]. The endocytosis of nanoparticles begins with engulfment in cationic peptides, triphenylphosphonium(TPP) and fusogenic lipid,
membrane invagination, and then forms intracellular vesicles (endo­ mitochondrial drug delivery can be achieved [52]. For instance, a smart
somes). The early endosome matures into a late endosomal compart­ dual-shell polymeric nanoparticle (MCTD-NPs) using multistage
ment and finally fuses with lysosomes [40]. During this process, the pH continuous targeted strategy to mitochondrial delivery of resveratrol
decreases from ~pH 6.5 in early endosome, down to ~pH 6.0 in late was fabricated (Fig. 3) [53]. In the study, ischemic myocardium targeted
endosome. In lysosome, the pH even down to ~pH 5.0, with abundant peptide (IMTP) was conjugated with poly(lactic-co-glycolic acid)
enzyme to degrade the cargo [41]. That means, if the nanoparticles (PLGA) via a long pH-responsive PEG chain to form outshell of the
failure to escape from the endo/lysosomal pathway, they will entrap and nanoparticle, which reduced the recognition rate of MPS and prolonged
degrade in lysosome, thus preventing the successful transport of the the blood residence time to enhance the accumulation in the ischemic
cargo into the cytoplasm. Up to now, several endosomal escape mech­ myocardium. At the same time, a positively charged polypeptide SS-31
anisms including membrane fusion, proton sponge effect, swelling and was also modified on the nanoparticles to form an inner-shell, which

496
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

Fig. 2. A summary of key pathophysiological changes during HF, proposed therapeutic strategies and representative nanoparticle architectures for treatment of HF.

Table 1
Examples of nanomedicine targeting oxygen stress.
Nanovector Load drug Model of use Key result Ref

Liposome CoQ10 New Zealand white rabbits with Improved the intracellular delivery of CoQ10 and reduced the [57]
experimental MI infarction size
Guar gum nanoparticles Selenium Ischemic buffer induced H9c2 Protected H9c2 cells from I/R injury through improving the [58]
cells efficiency of mitochondrial electron transport chain
Pluronic F-127 micelle Resveratrol- Doxorubicin-induced Swiss Reduced the doxorubicin-induced ROS damage [59]
quercetin Webster mice
G4 PAMAM dendrimers S-nitroso-N- No-flow I/R ex vivo rat hearts Regulated the nitric oxide release and limited I/R injury [60]
acetylpenicillamine
Carboxymethyl chitosan Curcumin I/R injury rats Inhibited cardiomyocyte apoptosis and cardiac hypertrophy in rats, [61]
nanoparticles-peptide and improved the bioavailability
PLGA nanoparticle Quercetin Antimycin A challenged H9c2 Preservation of mitochondrial function and ATP synthesis, [62]
cells suppressed oxidative stress
PLA nanoparticle Curcumin-nisin Guinea pigs with isoproterenol Significant cardioprotection effect with no toxicity [63]
induced MI

could strongly bind to mitochondria and promote the nanoparticles to around 5 nm, which have the ideal size to overcome the above
escape lysosome. When entrapped in the acidic lysosomes, the hydrazine mentioned limitations of hemorheology. After its treatment, the serum
bond on outshell of the nanoparticle broke rapidly, and then SS-31 is cardiac markers including CK-MB, LDH, AST and ALT in isoproterenol-
exposed. In the mitochondria, resveratrol was released, showing induced myocardial infarction (MI) rats were noticeably decreased,
significantly reduction of fraction size in the myocardial ischemia- while the antioxidant enzymes level including catalase and superoxide
reperfusion injury (MI/RI) rats. The precise delivery of antioxidants to dismutase were increased [55]. Although the above mentioned study
the ROS generation organelles provides a promising direction for scav­ provide a positive outcome for HF treatment, safety profiles of utilizing
enging excess ROS. this kind of metal oxide nanoparticle still need to be assessed.
Apart from encapsulating antioxidant in nanovectors, nanovector ROS-responsive nanomedicine is another effective approach to
therapy is also a way to regulate the excess ROS. Some of the nano­ reduce the damage caused by oxidative stress. For instance, a H2O2-
materials per se exhibiting antioxidant properties, like CeO2, have been triggered bubble-generating antioxidant polymeric nanoparticle for
employed in HF targeted nanotherapy. In CeO2, Ce is Ce3+ and Ce4+ hepatic I/R injury has been developed [56]. In the study, peroxalate
[54]. Under the mixed valences states, cerium particles can easily esters bond that can rapidly oxidize by H2O2 was introduced to form the
change its electronic design and make it most suitable for the sur­ polymeric prodrug, poly(vanillin oxalate) (PVO). Once exposed to H2O2,
rounding environment through the rapid and convenient transformation vanillyl alcohol was released from the PVO nanoparticles to exert its
between Ce3+ and Ce4+. Shereen et al. synthesized a CeO2 nanoparticle anti-apoptotic and anti-inflammatory activities. Also as a H2O2-

497
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

Fig. 3. (A) Schematic image of precise treatment of MI/RI through multistage mitochondrial delivery of resveratrol by using MCTD-NPs; (B) Experimental protocols
for the in vivo rat MI/RI model; (C) Sections from left ventricle regions indicated in the artist's rendition were subjected to ex vivo distribution research; (D) Ex vivo
distribution of cy7.5 labeled PLGA-NPs and cy7.5 labeled MCTD-NPs in sham and MI/RI heart; (E) Ex vivo distribution of cy7.5 labeled PLGA-NPs and cy7.5 labeled
MCTD-NPs in four sections of MI/RI heart. Nanomedicine: nanotechnology, biology, and medicine. 2019;16:236–49. © Elsevier Inc.

associated disease, this PVO nanoparticle may be translated to treat HF. where they discrete into network-like scaffolds. However, affected by
As an attractive and viable direction, other ROS-responsive materials or contractile force, the injected drugs tend to be expelled by the car­
structure, such as thioester, phenylboronic ester and polypropylene diomyocyte environment. By this morphological transition strategy
sulfide may also be explored in designing ROS-triggered cardiac nano­ based on MMPs response, the nanoparticles can remain in the injured
medicines [33]. heart even be observed at day 28 post-injection (Fig. 4). Critically, the
study provided a useful platform with targeted accumulation and pro­
3.2. Nanomedicine targeting aberrant ECM longed retention for MI therapy.
Encapsulate drugs/biomolecular in MMPs-responsive nanovectors
The myocardial ECM is a dynamically changing environment, which may achieve “on-demand” and localized drug release by utilizing MMPs
is fundamental for the physiological homeostasis and structural of the hyperactivation as a trigger. A dual functional MMPs-responsive
heart. In HF patients under oxidative stress, the ECM homeostasis is hydrogel was developed for basic fibroblast growth factor (bFGF)
disturbed by highly expressed extracellular matrix metalloproteinase localized release and MMPs inhibition to promote the recovery of MI
inducer (EMMPRIN) [64]. EMMPRIN not only active matrix metal­ [72]. Although this hydrogel ameliorated myocardium remodeling and
loproteinases (MMPs), but also increase MMPs expression [65,66]. In enhanced vascularization by targeting upregulated MMPs, it is unlikely
fact, the MMPs also exist under physiological conditions, but in an that patients with weakened MI wall receiving intramyocardial admin­
inactive form. Once myocardial injury occurred, they become activated. istration given the safety concerns. Further studies to exploit systemi­
The activation and overexpression of MMPs results in collagen degra­ cally nanoparticle formulations targeting the upregulated MMPs are still
dation and thereby induces fibroblast proliferation [67]. Therefore, needed.
inhibit the activation of EMMPRIN and MMPs to minimize the degra­
dation of cardiac ECM is a promising resolution to ameliorate HF 3.3. Nanomedicine targeting overloaded intracellular calcium
[68,69].
As mentioned above, EMMPRIN inhibition may point to a new Ca2+ is essential for the activation of myofilaments and electrical
approach to migrate HF injury. Under this concept, Cervadoro A et al. activity that enable contraction of heart. However, Ca2+ overload occurs
conjugated the paramagnetic/fluorescent micellar nanoparticles with in patients with HF due to ischemia. Ischemia induced acidosis directly
AP-9 peptide (NAP9), which can specifically bind with EMMPRIN. By increases the accumulation of Na+ through Na+/H+ exchanger (NHE),
this active targeting strategy, the aggregation of NAP9 at HF sites was and then transports Ca2+ into cells by reversing the function of Na+/
enhanced and in turn prevented the MMP-mediated ECM degradation Ca2+ exchanger [73]. In addition, under oxidative stress, excess O2−
and reduced the infarct size in mice [70]. The abnormal ECM can not interacts with NO to form peroxynitrite, which triggers protein oxida­
only be used as a therapeutic target, but also as an effective stimulus for tion that will further damage the excitation-contraction coupling [74].
the design of HF targeted nanoparticles. A MMPs response morphology In the case of excitation contraction coupling damaging, L-type calcium
transformable nanoparticle was fabricated using brush peptide-polymer channels, potassium channels, sodium channels and the Na+/Ca2+
amphiphiles (PPAs) [71]. Interestingly, the peptide sequences PLGLAG exchanger are involved, directly resulting in Ca2+ overload in the
in PPAs can be specifically recognized by the upregulated MMPs, MMP-2 injured heart. Meanwhile, the sarcoplasmic reticulum Ca2+-adenosine
and MMP-9. After intravenous injection, the spherical enzyme- triphosphatase (SERCA) is altered and the calcium sensitivity of the
responsive nanoparticles can circulate freely in the bloodstream until myofilaments is reduced [75,76]. Accordingly, HF patients may benefit
they extravasate the leaky vascular system and reach the infarct site, from targeting therapies of calcium mishandling.

498
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

Fig. 4. (A) Diagram of a dye-labeled brush peptide-polymer amphiphile (PPA) containing an MMP-2 and MMP-9 specific recognition sequence, shown underlined.
PPAs self-assemble into nanoparticles through hydrophobic-hydrophilic interactions when dialyzed into aqueous buffer. (B) Schematic of nanoparticles freely
circulating in the bloodstream (not to scale) upon systemic delivery. Nanoparticles enter the infarct tissue through the leaky acute MI vasculature, and upregulated
MMPs within the infarct induce the formation of an aggregate-like scaffold. (C) In vitro, responsive (top) and nonresponsive (bottom) nanoparticles are monodisperse
micelles with diameters of 15–20 nm, and (D) upon activation, only responsive nanoparticles form an aggregate-like scaffold. (E) Corresponding images of nano­
particle solutions following activation. (F) Dynamic light scattering of nanoparticles before and after MMP activation. (G) Retention of IV delivered nanoparticles
with the infarct. H&E images are shown in A, and neighboring fluorescent sections are shown in B. Particles are shown in red and myocardium. Scale bar: 100 μm.
Adv Mater. 2015 Oct 7;27(37):5547–52. © WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. (For interpretation of the references to colour in this figure legend, the
reader is referred to the web version of this article.)

At present, the efficacy of calcium channel antagonists including L- development for HF therapy. Table 2 summarizes representative ex­
type Ca2+ channel antagonists, mitochondrial Ca2+ channel antagonists amples of the present nanotherapy strategies targeting intracellular
and sarcolemma Ca2+ channel antagonists is often limited due to the calcium overload. Additionally, a number of Ca2+ channel blockers such
lack of solubility, bioavailability or specificity [77]. Using nanovactors as amiodarone, diltiazem hydrochloride and nitrendipine have
to target deliver the modulators of Ca2+, either to cardiac calcium benefited from nanotechnology [18,78–81]. Take the class III antiar­
channels, or specifically to mitochondria, could be an exciting rhythmic drug amiodarone as an example. It has low solubility and acts

Table 2
Examples of nanomedicine targeting intracellular calcium overload.
Nanovector Load drug Model of use Key result Ref

Solid-lipid nanoparticle Nitrendipine In vitro Improved the effective bioavailability up to 2.81–5.35-folds greater [90]
than nitrendipine suspension
PEGylated liposome Amiodarone I/R injury rats Prolonged blood residence time and reduced the infarction size and [18,79]
mortality
Chitosan-coated Fe3O4 Omecamtive Mecarbil Male Wistar rats Improved the cardiac function with only 4% necessary dose of OM [84]
nanoparticle amount
GlcNAc-polyketal S100A1 Sprague-Dawley rats with Decreased the irregular calcium release through “two-hit” effect on [88]
nanoparticles nonischemic HF modulation of calcium regulation proteins
PLGA nanoparticle Cyclosporine A I/R injury mice Increased accumulation of cyclosporine A in myocardium [91]
mitochondria and limited HF progression via preventing mPTP-
opening
Polymeric nanoparticle alpha-interacting domain I/R ex vivo rat hearts Decreased oxidative stress and superoxide production in cardiac [92]
peptide- curcumin myocytes, reduced muscle damage
CaP nanoparticle mimetic peptide Rodent models of diabetic Improved myocardial contraction and in turn restored cardiac [6]
cardiomyopathy (mice, rats, pigs) function via inhalation of this peptide-loaded nanoparticles

499
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

on many ion channels of myocardium and vascular smooth muscle [82]. the anti-inflammatory cytokine IL-10 and transforming growth factor β
To improve its solubility and reduce its acute toxicity, a nanosuspension (TGFβ). Therapy strategies to judiciously regulate the polarization of
of amiodarone was prepared [78]. PEGylated liposomes of amiodarone macrophages and cytokines production may help alleviate damage of
also have been developed [18,79]. In the I/R rats, the liposomal amio­ HF. In recent years, nanoimmunotherapy has been an increasingly
darone with prolonged blood residence time significantly reduced the popular approach in nanomedicine. Using nanomedicine or surface
infarct size (29.5 ± 6.5%) compared to the control (53.2 ± 3.5%), and functionalized nanomaterials to judiciously regulate the polarization of
reduced the mortality due to the negative hemodynamic and lethal macrophages and cytokines production may help alleviate damage of
arrhythmia caused by adenosine. HF.
Instead of reducing the calcium flux, improving the calcium sensi­ Modulation of macrophage polarization to a reparative state is a
tivity of myofilament is also a practical strategy for the treatment of HF. promising approach to elicit infarct repair and promote resolution of
Omecamtive Mecarbil (OM) is a novel Ca2+ sensitizing positive inotrope inflammation. Notably, nanoparticles composed of polyurethane (PU)
for the treatment of systolic HF [83]. Dose-dependent side effects and were found to induce the polarization of macrophage to M2, which
repeated administration are the major downside for the use of OM. To suppress the activation of inflammasome signals and NF-κB, and in turn
tackle these problems, a chitosan-coated magnetic Fe3O4 nanoparticle reduce the production of inflammatory cytokines like IL-1β and TNF-α
was developed for OM heart targeting delivery in a very recent study [97]. Owing to its excellent elastic properties, the biodegradable PU is
[84]. The cationic nature and hydrophilicity of chitosan helped to avoid gaining attention in recent years. Exploring more nanomaterials like PU
opsonization of the MPS and increased the colloidal stability of the nanoparticle can be a potential therapeutic nanovector for HF
Fe3O4 nanoparticles. Fe3O4 nanoparticles, also referred as surperpar­ immunomodulation.
amagnetic iron oxide nanoparticles (SPIONs), not only conferred In another case, phosphatidylserine (PS)-presenting liposomes were
biocompatibility and safety of the nanoparticles but also the therapeutic intravenously injected to modulate cardiac macrophages and improve
effect on patients with low-risk MI [85]. A single dose injection con­ infract repair of MI rats [98]. The strategy was designed based on the
taining 510 mg SPIONs significantly lowered end systolic volume (ESV) underlying principle of anti-inflammatory effects of apoptotic cells. As
and increased the ejection fraction (EF) from 53 to 59%. In addition, known, PS is identified as a “death signal” of cells, which can be
SPIONs also take part in increasing the formation of mitochondrial ATP recognized by macrophages via the PS receptor. PS presenting on the
and improving the survival of cardiomyocytes. As a result, by this tar­ surface of liposomes skillfully mimicked the anti-inflammatory effects of
geting strategy, the OM dose was reduced to 4% of the traditional dose, apoptotic cells. After the “mimic apoptotic cells” uptake by macro­
and EF was improved by 18%. This study highlights the prospective of phages, anti-inflammatory cytokines TGFβ and IL-10 were secreted,
developing SPIONs for HF treatment in which synergistic effects can be while the pro-inflammatory markers such as TNFα and CD86 were
achieved. downregulated. As a consequent, this strategy promoted the preserva­
To further improve targeting efficacy, the specific targeting moieties tion of small scars and angiogenesis, and prevented ventricular remod­
was also introduced into the design of nanotherapy targeting intracel­ eling. This ingenious strategy provides a candidate platform for immune
lular calcium overload. N-Acetylglucosamine (GlcNAc), a sugar with response modulating and HF nanoimmunotherapy.
high affinity for cardiomyocytes has aroused the interest of researchers Another possible immunomodulation strategy is to directly regulate
[86]. Decoration of GlcNAc on the surfaces of nanoparticles increased the production of cytokines. Among the cytokines of HF, the key pro-
their uptake by cardiomyocytes. Furthermore, in cardiomyocytes, O- inflammatory marker, TNFα has been extensively studied and is found
GlcNAc transferase catalyzes the GlcNAc attachment to threonine or to be a potential therapeutic target. As such, deoxyribozyme (DNAzyme)
serine residues of key intracellular Ca2+ regulating proteins such as functionalized gold nanoparticle (AuNPs) was fabricated to catalytically
SERCA2a, STIM1, PLB and CaMKII [87]. In a recent study, GlcNAc was silence TNFα for MI [99]. After local injection of this DNAzyme AuNPs in
conjugated on the surface of biodegradable polyketal nanoparticles for the myocardium of MI rats, the TNFα knockdown efficiency achieved as
S100A1 delivery [88]. S100A1 is a Ca2+-binding protein, which also can high as 50%, which subsequently resulted in noticeable improvement in
interact with the calcium regulation proteins like SERCA2a to modulate acute cardiac function. The study represents an example of the use of
myocardial contraction [89]. This GlcNAc nanoparticles carrying DNAzyme AuNP conjugates for gene regulation and viable delivery.
S100A1 protein demonstrated a “two-hit” effect on modulation of cal­ Similar approaches in regulating the immune environment and
cium regulation proteins, contributing for the reduction of abnormal improving the treatment of HF will be an exciting development.
calcium release and the recovery of sarcomere function. This study of­
fers new insights into the development of bioactive nanovectors to 4. Major challenges of cardiac nanomedicine
optimize therapies.
Despite the encouraging results, there are still many challenges to be
3.4. Nanomedicine targeting immune activation solved and further optimized before cardiac nanomedicine can be
transformed into viable clinical products. In this section, the major
As a defense mechanism, innate immune system activation is a challenges including the formation of the protein corona, off-target ef­
protective response to myocardial injury, in which the inflammatory fect, safety warnings and potential adverse effects are listed and
cells and mediators work in tandem. Generally, the very early healing discussed.
processes are initiated by leucocytes infiltrating to remove irreparably One of the central challenges in targeted nanomedicine is the for­
damaged cells. Other inflammatory cells including neutrophils and mation of the protein corona. Once the nanocarrier enters the circula­
macrophages are also considered to be essential components for infarct tory system, they are wrapped in proteins in a process called
area repair [93]. Neutrophils orchestrate the healing process through opsonization, marking that they are phagocytized by MPS then cleared
polarizing macrophages from M1 to M2 [94]. It has been validated that from the body. In particularly, the protein mentioned here has been
M1 macrophages active the pro-inflammatory pathway, while in extensively studied for a decade, which is termed as protein corona
contrast, M2 macrophages induce anti-inflammatory pathways and [100]. As reported, when in contact with biological fluids, different
exhibit cardiac repair ability [95]. Apart from cellular immune response, types of proteins are adsorbed on nanoparticles to form protein corona.
recent evidence also highlights the potential role of inflammatory me­ The protein corona can substantially affect the biological fate of nano­
diators and their interactions in driving HF progress [96]. In HF, carriers, such as increasing uptake by the MPS [101–103]. In addition to
accelerated concentrations of pro-inflammatory cytokines including mediating the uptake of MPS, protein corona has also been shown to be
tumor necrosis factor α (TNFα), interleukin 1 (IL-1) and 6 (IL-6) are detrimental to active targeting strategies [104,105]. The specificity of
observed. The production of TNFα, IL-1 and -6 can be downregulated by the targeted ligand will be significantly reduced due to the covering of

500
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

the attached biological corona [106]. For example, it was demonstrated nanomedicine goes beyond the concept of “magic particulate bullet”
that transferrin-modified silica nanoparticles lost the targeting ability [119]. Nowadays, cardiac nanomedicine is not limited to nanoparticles
after incubation with plasma [107]. Although researchers have made and the technology to evaluate their application in the body, but also
numerous efforts, few effective strategies have been proposed to mini­ involves the design of new scaffolds (i.e. nanorods) to enhance thera­
mize the mistargeting caused by corona. Among which, coating zwit­ peutic cells' engraftment in myocardial tissue and surfaces for engi­
terionic compounds on the surface of nanoparticles is validated to be a neering sensors to identify HF biomarkers (i.e. nanosheets) or
potential strategy. For instance, corona-free Au nanoparticles have been implantable devices to aid in the regeneration of tissues (i.e. carbon
fabricated by a range of zwitterionic coatings [108]. nanotubes) [120–123]. Although many of these concepts are still in the
Cardiac nanomedicine is designed to specifically deliver the drug/ early stages of development, the application of nanotechnology has
biomolecule to injured heart and enhance their accumulation. However, brought great benefits to the significant increase in bioavailability and
the off-target effect always occurs, which attenuates their efficacy. On specificity. Taking the advantage of nanotechnology, therapies by
the one hand, this is because HF is a progressive disease with complexed addressing pathological mechanisms of HF are being the most promising
and changeable pathological state. For example, following MI, the heart approaches to prevent the progression of HF.
first subjects to an early inflammatory phase (phase 1) with pro- Although nanotechnology has made great progress in the treatment
inflammatory cytokines such as IL-1β and TNF-α released, when turns of HF, the application of cardiac nanomedicine in patients is still in its
to inflammation resolution stage (phase 2), the anti-inflammatory cy­ infancy. By settling each of the existing challenges in turn and
tokines IL-10 and TNF-β are secreted. In this regard, time selection has combining innovative design functions reasonably, effective nano­
become a non-negligible factor in the selection of different targeting medicine for HF will definitely be developed, thereby realizing a para­
strategies for the treatment of HF. On the other hand, as mentioned digm shift of therapy delivery based on nanoparticles. For future study,
previously, ligand-receptor binding is the most effective and commonly researchers should carefully learn from the lessons of cancer nano­
used targeting strategy in nanotherapy to help target damaged car­ technology, such as by reporting the basic information required (e.g.,
diomyocytes. However, the intrinsic structure of ligands may be physicochemical properties of nanoparticles, detailed information on
destroyed by chemical modification, resulting in the decrease of binding the biological interface of nanoparticles, biological properties of nano­
efficiency [109]. The emerging of bio-orthogonal reactions provides a particles, and complete information about biological systems). Addi­
potential resolution to avoid the off-target effects, in which the second tionally, development of cardiac nanomedicine requires coordinating
tag can specifically identify the labeled cells by labeling the target cells and connecting multiple fields, including molecular and cellular
with a primary exogenous tag first [110–112]. However, the related biology, chemistry and biochemistry, cardiology and materials sciences,
research only stays at the cellular level, and further efforts are needed to among others. In this regard, build a strong bridge between experts in
make it better applied in vivo. the field of cardiac nanotechnology is essential importance.
Last but not least, safety warnings and potential adverse effects are
also the main concerns for the application of nanomedicine in cardiac Declaration of Competing Interest
disease. For instance, recent studies have shown that iron compounds
can induce ferroptosis in the presence of ROS, a newly discovered pro­ The authors have no conflict of interest to declare.
grammed cell death form associated with I/R injury [113–115]. Fenton
reaction (i.e., the process of converting hydrogen peroxide to hydroxyl Acknowledgements
radicals)/iron metabolism leads to excessive production of ROS,
resulting in regulatory cell death [116,117]. These findings challenge The authors gratefully acknowledge the financial support from
the use of iron-based nanoparticles and remind experts to be more Natural Science Foundation of Heilongjiang Province of China (No.
cautious in choosing the composition of nanoparticles. In addition, it is YQ2019H004), Post-Doctor Research Project of West China Hospital of
essential to evaluate the biological fate and safety profiles of nonde­ Sichuan University (No. 2020HXBH172), 111 project (No. B18035) and
gradable inorganic nanoparticles for long-term use. Recent studies have Excellent Yong Talents Funding of College of Pharmacy of Harbin
shown that polymeric-coated Au nanoparticles could be degraded by the Medical University (No. 2019YQ07).
liver proteolytic enzymes [118]. This suggests that the physicochemical
properties of the nanoparticles may alteration in vivo, resulting in un­ References
necessary or unpredictable toxicity, aggregation or immune activation.
The changes of physical and chemical properties of nanoparticles in­ [1] E. Tanai, S. Frantz, Pathophysiology of heart failure, Comprehensive Physiol.
(2015) 187–214.
crease the complexity of host immune system inflammatory response, [2] M. Iafisco, A. Alogna, M. Miragoli, D. Catalucci, Cardiovascular nanomedicine:
which may take along more adverse effects on HF process than expected. the route ahead, Nanomedicine (London) 14 (2019) 2391–2394.
Therefore, it is necessary to study the biological fate of nanoparticles [3] H. Dokainish, K. Teo, J. Zhu, A. Roy, K.F. AlHabib, A. ElSayed, et al., Global
mortality variations in patients with heart failure: results from the international
more carefully. congestive heart failure (INTER-CHF) prospective cohort study, Lancet Glob.
Health 5 (2017) e665-e72.
5. Conclusions and future perspectives [4] S.S. Virani, A. Alonso, E.J. Benjamin, M.S. Bittencourt, C.W. Callaway, A.
P. Carson, et al., Heart disease and stroke statistics-2020 update: a report from the
American heart association, Circulation. 141 (2020) e139–e596.
In decades, a number of studies have been carried out to limit the [5] D. Snipelisky, S.P. Chaudhry, G.C. Stewart, The many faces of heart failure,
damage of HF by addressing their pathological mechanisms. Indeed, the Cardiac electrophysiology clinics. 11 (2019) 11–20.
[6] M. Miragoli, P. Ceriotti, M. Iafisco, M. Vacchiano, N. Salvarani, A. Alogna, et al.,
therapies have shown their potential in animal models. However, many Inhalation of peptide-loaded nanoparticles improves heart failure, Sci. Transl.
of them have limited bioavailability and poor specificity, which leads to Med. 10 (2018).
their failure in clinical trials. As shown in this review, cardiac nano­ [7] F. Farjadian, A. Ghasemi, O. Gohari, A. Roointan, M. Karimi, M.R. Hamblin,
Nanopharmaceuticals and nanomedicines currently on the market: challenges
medicines have circumvented many limitations of traditional therapies
and opportunities, Nanomedicine (London) 14 (2019) 93–126.
by improving drug accumulation in injured cardiomyocytes, and [8] V.M. Martin Gimenez, D.E. Kassuha, W. Manucha, Nanomedicine applied to
reducing unwanted side effects and systemic dosage. In particular, in cardiovascular diseases: latest developments, Ther. Adv. Cardiovasc. Dis. 11
(2017) 133–142.
recent years, intelligent and on-demand delivery nanomedicine have
[9] G.M. Felker, L.K. Shaw, C.M. O’Connor, A standardized definition of ischemic
further been developed according to the unique pathological environ­ cardiomyopathy for use in clinical research, J. Am. Coll. Cardiol. 39 (2002)
ment of HF to achieve accurate local drug release. With the application 210–218.
of nanotechnology in the control of biological systems, diagnosis,
treatment and monitoring of diseases, the development of cardiac

501
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

[10] L.C. Ou, S. Zhong, J.S. Ou, J.W. Tian, Application of targeted therapy strategies [40] Y.M. Bae, Y.I. Park, S.H. Nam, J.H. Kim, K. Lee, H.M. Kim, et al., Endocytosis,
with nanomedicine delivery for atherosclerosis, Acta Pharmacol. Sin. 42 (1) intracellular transport, and exocytosis of lanthanide-doped upconverting
(2021) 10–17. nanoparticles in single living cells, Biomaterials. 33 (2012) 9080–9086.
[11] F. Shakeel, Recent advances in liposomal drug delivery system for drug targeting, [41] G.K. Such, Y. Yan, A.P. Johnston, S.T. Gunawan, F. Caruso, Interfacing materials
Current drug delivery. 17 (2020) 824–825. science and biology for drug carrier design, Adv. Mater. 27 (2015) 2278–2297.
[12] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific [42] M. Gunata, H. Parlakpinar, A review of myocardial ischaemia/reperfusion injury:
nanoparticles: theory to practice, Pharmacol. Rev. 53 (2001) 283–318. pathophysiology, experimental models, biomarkers, genetics and
[13] V.P. Torchilin, Polymer-coated long-circulating microparticulate pharmacological treatment, Cell Biochem. Funct. 39 (2) (2020) 190–217.
pharmaceuticals, J. Microencapsul. 15 (1998) 1–19. [43] H. Tsutsui, S. Kinugawa, S. Matsushima, Oxidative stress and heart failure, Am. J.
[14] Y. Sheng, C. Liu, Y. Yuan, X. Tao, F. Yang, X. Shan, et al., Long-circulating Physiol. Heart Circ. Physiol. 301 (2011) H2181–H2190.
polymeric nanoparticles bearing a combinatorial coating of PEG and water- [44] A. van der Pol, W.H. van Gilst, A.A. Voors, P. van der Meer, Treating oxidative
soluble chitosan, Biomaterials. 30 (2009) 2340–2348. stress in heart failure: past, present and future, Eur. J. Heart Fail. 21 (2019)
[15] J.V. Jokerst, T. Lobovkina, R.N. Zare, S.S. Gambhir, Nanoparticle PEGylation for 425–435.
imaging and therapy, Nanomedicine (London) 6 (2011) 715–728. [45] M.G. Perrelli, P. Pagliaro, C. Penna, Ischemia/reperfusion injury and
[16] M. Kanamala, B.D. Palmer, H. Ghandehari, W.R. Wilson, Z. Wu, PEG- cardioprotective mechanisms: role of mitochondria and reactive oxygen species,
benzaldehyde-hydrazone-lipid based PEG-dheddable pH-sensitive liposomes: World J. Cardiol. 3 (2011) 186–200.
abilities for endosomal escape and long circulation, Pharm. Res. 35 (2018) 154. [46] A.K. Jain, N.K. Mehra, N.K. Swarnakar, Role of antioxidants for the treatment of
[17] S. Mustafa, V.K. Devi, R.S. Pai, Effect of PEG and water-soluble chitosan coating cardiovascular diseases: challenges and opportunities, Curr. Pharm. Des. 21
on moxifloxacin-loaded PLGA long-circulating nanoparticles, Drug delivery and (2015) 4441–4455.
translational research. 7 (2017) 27–36. [47] J.G. Farias, V.M. Molina, R.A. Carrasco, A.B. Zepeda, E. Figueroa, P. Letelier, et
[18] H. Takahama, T. Minamino, H. Asanuma, M. Fujita, T. Asai, M. Wakeno, et al., al., Antioxidant therapeutic strategies for cardiovascular conditions associated
Prolonged targeting of ischemic/reperfused myocardium by liposomal adenosine with oxidative stress, Nutrients. 9 (9) (2017) 966.
augments cardioprotection in rats, J. Am. Coll. Cardiol. 53 (2009) 709–717. [48] O. Pechanova, E. Dayar, M. Cebova, Therapeutic potential of polyphenols-loaded
[19] H. Xu, K.Q. Wang, W.W. Huang, Y.H. Deng, D.W. Chen, Recent advances in the polymeric nanoparticles in cardiovascular system, Molecules. 25 (15) (2020)
study of accelerated blood clearance phenomenon of PEGylated liposomes, Acta 3322.
Pharm. Sin. 45 (2010) 677–683. [49] J. Kim, L. Cao, D. Shvartsman, E.A. Silva, D.J. Mooney, Targeted delivery of
[20] M.M. El Sayed, H. Takata, T. Shimizu, Y. Kawaguchi, A.S. Abu Lila, N.E. Elsadek, nanoparticles to ischemic muscle for imaging and therapeutic angiogenesis, Nano
et al., Hepatosplenic phagocytic cells indirectly contribute to anti-PEG IgM Lett. 11 (2011) 694–700.
production in the accelerated blood clearance (ABC) phenomenon against [50] S. Taurin, H. Nehoff, K. Greish, Anticancer nanomedicine and tumor vascular
PEGylated liposomes: appearance of an unexplained mechanism in the ABC permeability; where is the missing link? J. Control. Release 164 (2012) 265–275.
phenomenon, J. Control. Release 323 (2020) 102–109. [51] G.U. Ruiz-Esparza, J.H. Flores-Arredondo, V. Segura-Ibarra, G. Torre-Amione,
[21] M.D. McSweeney, L.S.L. Price, T. Wessler, E.C. Ciociola, L.B. Herity, J. M. Ferrari, E. Blanco, et al., The physiology of cardiovascular disease and
A. Piscitelli, et al., Overcoming anti-PEG antibody mediated accelerated blood innovative liposomal platforms for therapy, Int. J. Nanomedicine 8 (2013)
clearance of PEGylated liposomes by pre-infusion with high molecular weight 629–640.
free PEG, J. Control. Release 311–312 (2019) 138–146. [52] F. Forini, P. Canale, G. Nicolini, G. Iervasi, Mitochondria-targeted drug delivery
[22] S. Jiang, Z. Cao, Ultralow-fouling, functionalizable, and hydrolyzable zwitterionic in cardiovascular disease: a long road to nano-cardio medicine, Pharmaceutics.
materials and their derivatives for biological applications, Adv. Mater. 22 (2010) 12 (11) (2020) 1122.
920–932. [53] Y. Cheng, D.Z. Liu, C.X. Zhang, H. Cui, M. Liu, B.L. Zhang, et al., Mitochondria-
[23] W. Yang, L. Zhang, S. Wang, A.D. White, S. Jiang, Functionalizable and ultra targeted antioxidant delivery for precise treatment of myocardial ischemia-
stable nanoparticles coated with zwitterionic poly(carboxybetaine) in undiluted reperfusion injury through a multistage continuous targeted strategy, Nanomed.
blood serum, Biomaterials. 30 (2009) 5617–5621. Nanotechnol. Biol. Med. 16 (2019) 236–249.
[24] J.O. Martinez, R. Molinaro, K.A. Hartman, C. Boada, R. Sukhovershin, E. De Rosa, [54] N.V. Skorodumova, S.I. Simak, B.I. Lundqvist, I.A. Abrikosov, B. Johansson,
et al., Biomimetic nanoparticles with enhanced affinity towards activated Quantum origin of the oxygen storage capability of ceria, Phys. Rev. Lett. 89
endothelium as versatile tools for theranostic drug delivery, Theranostics. 8 (2002) 166601.
(2018) 1131–1145. [55] S.S. El Shaer, T.A. Salaheldin, N.M. Saied, S.M. Abdelazim, In vivo ameliorative
[25] C.M. Hu, L. Zhang, S. Aryal, C. Cheung, R.H. Fang, L. Zhang, Erythrocyte effect of cerium oxide nanoparticles in isoproterenol-induced cardiac toxicity,
membrane-camouflaged polymeric nanoparticles as a biomimetic delivery Exp. Toxicol. Pathol. 69 (2017) 435–441.
platform, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 10980–10985. [56] C. Kang, W. Cho, M. Park, J. Kim, S. Park, D. Shin, et al., H2O2-triggered bubble
[26] C.M. Hu, R.H. Fang, K.C. Wang, B.T. Luk, S. Thamphiwatana, D. Dehaini, et al., generating antioxidant polymeric nanoparticles as ischemia/reperfusion targeted
Nanoparticle biointerfacing by platelet membrane cloaking, Nature. 526 (2015) nanotheranostics, Biomaterials. 85 (2016) 195–203.
118–121. [57] D.D. Verma, W.C. Hartner, V. Thakkar, T.S. Levchenko, V.P. Torchilin, Protective
[27] M. Longmire, P.L. Choyke, H. Kobayashi, Clearance properties of nano-sized effect of coenzyme Q10-loaded liposomes on the myocardium in rabbits with an
particles and molecules as imaging agents: considerations and caveats, acute experimental myocardial infarction, Pharm. Res. 24 (2007) 2131–2137.
Nanomedicine (London) 3 (2008) 703–717. [58] R.S. Soumya, A. Prathapan, P.S. Raj, V.P. Vineetha, K.G. Raghu, Selenium
[28] S.S. Hossain, Y. Zhang, X. Liang, F. Hussain, M. Ferrari, T.J. Hughes, et al., In incorporated guar gum nanoparticles safeguard mitochondrial bioenergetics
silico vascular modeling for personalized nanoparticle delivery, Nanomedicine during ischemia reperfusion injury in H9c2 cardiac cells, Int. J. Biol. Macromol.
(London) 8 (2013) 343–357. 107 (2018) 254–260.
[29] J. Tan, A. Thomas, Y. Liu, Influence of red blood cells on nanoparticle targeted [59] B. Cote, L.J. Carlson, D.A. Rao, A.W.G. Alani, Combinatorial resveratrol and
delivery in microcirculation, Soft Matter 8 (2011) 1934–1946. quercetin polymeric micelles mitigate doxorubicin induced cardiotoxicity in vitro
[30] R. Toy, E. Hayden, C. Shoup, H. Baskaran, E. Karathanasis, The effects of particle and in vivo, J. Control. Release 213 (2015) 128–133.
size, density and shape on margination of nanoparticles in microcirculation, [60] T.A. Johnson, N.A. Stasko, J.L. Matthews, W.E. Cascio, E.L. Holmuhamedov, C.
Nanotechnology. 22 (2011) 115101. B. Johnson, et al., Reduced ischemia/reperfusion injury via glutathione-initiated
[31] F. Gentile, C. Chiappini, D. Fine, R.C. Bhavane, M.S. Peluccio, M.M. Cheng, et al., nitric oxide-releasing dendrimers, Nitric Oxide Biol. Chem. 22 (2010) 30–36.
The effect of shape on the margination dynamics of non-neutrally buoyant [61] A. Ray, S. Rana, D. Banerjee, A. Mitra, R. Datta, S. Naskar, et al., Improved
particles in two-dimensional shear flows, J. Biomech. 41 (2008) 2312–2318. bioavailability of targeted Curcumin delivery efficiently regressed cardiac
[32] T. Dvir, M. Bauer, A. Schroeder, J.H. Tsui, D.G. Anderson, R. Langer, et al., hypertrophy by modulating apoptotic load within cardiac microenvironment,
Nanoparticles targeting the infarcted heart, Nano Lett. 11 (2011) 4411–4414. Toxicol. Appl. Pharmacol. 290 (2016) 54–65.
[33] W. Yu, R. Liu, Y. Zhou, H. Gao, Size-tunable strategies for a tumor targeted drug [62] O. Lozano, A. Lazaro-Alfaro, C. Silva-Platas, Y. Oropeza-Almazan, A. Torres-
delivery system, ACS central science. 6 (2020) 100–116. Quintanilla, J. Bernal-Ramirez, et al., Nanoencapsulated quercetin improves
[34] Q. Hu, Q. Chen, Z. Gu, Advances in transformable drug delivery systems, cardioprotection during hypoxia-reoxygenation injury through preservation of
Biomaterials. 178 (2018) 546–558. mitochondrial function, Oxidative Med. Cell. Longev. 2019 (2019) 7683051.
[35] I. Canton, G. Battaglia, Endocytosis at the nanoscale, Chem. Soc. Rev. 41 (2012) [63] W.E.E. Nabofa, O.O. Alashe, O.T. Oyeyemi, A.F. Attah, A.A. Oyagbemi,
2718–2739. Omobowale TO, et al., Cardioprotective effects of curcumin-nisin based poly
[36] C. Le Roy, J.L. Wrana, Clathrin- and non-clathrin-mediated endocytic regulation lactic acid nanoparticle on myocardial infarction in guinea pigs, Sci. Rep. 8
of cell signalling, Nat. Rev. Mol. Cell Biol. 6 (2005) 112–126. (2018) 16649.
[37] S.A. Smith, L.I. Selby, A.P.R. Johnston, G.K. Such, The endosomal escape of [64] H.M. Sanders, G.J. Strijkers, W.J. Mulder, H.P. Huinink, S.J. Erich, O.C. Adan, et
nanoparticles: toward more efficient cellular delivery, Bioconjug. Chem. 30 al., Morphology, binding behavior and MR-properties of paramagnetic collagen-
(2019) 263–272. binding liposomes, Contrast media & molecular imaging. 4 (2009) 81–88.
[38] F. Benyettou, R. Rezgui, F. Ravaux, T. Jaber, K. Blumer, M. Jouiad, et al., [65] K.Y. DeLeon-Pennell, C.A. Meschiari, M. Jung, M.L. Lindsey, Matrix
Synthesis of silver nanoparticles for the dual delivery of doxorubicin and metalloproteinases in myocardial infarction and heart failure, Prog. Mol. Biol.
alendronate to cancer cells, J. Mater. Chem. B 3 (2015) 7237–7245. Transl. Sci. 147 (2017) 75–100.
[39] C. Liu, W. Yu, Z. Chen, J. Zhang, N. Zhang, Enhanced gene transfection efficiency [66] R. Schmidt, A. Bultmann, M. Ungerer, N. Joghetaei, O. Bulbul, S. Thieme, et al.,
in CD13-positive vascular endothelial cells with targeted poly(lactic acid)-poly Extracellular matrix metalloproteinase inducer regulates matrix
(ethylene glycol) nanoparticles through caveolae-mediated endocytosis, metalloproteinase activity in cardiovascular cells: implications in acute
J. Control. Release 151 (2011) 162–175. myocardial infarction, Circulation. 113 (2006) 834–841.

502
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

[67] A. Ducharme, S. Frantz, M. Aikawa, E. Rabkin, M. Lindsey, L.E. Rohde, et al., [94] M. Horckmans, L. Ring, J. Duchene, D. Santovito, M.J. Schloss, M. Drechsler, et
Targeted deletion of matrix metalloproteinase-9 attenuates left ventricular al., Neutrophils orchestrate post-myocardial infarction healing by polarizing
enlargement and collagen accumulation after experimental myocardial macrophages towards a reparative phenotype, Eur. Heart J. 38 (2017) 187–197.
infarction, J. Clin. Invest. 106 (2000) 55–62. [95] A. Mantovani, A. Sica, S. Sozzani, P. Allavena, A. Vecchi, M. Locati, The
[68] Z. Fan, M. Fu, Z. Xu, B. Zhang, Z. Li, H. Li, et al., Sustained release of a peptide- chemokine system in diverse forms of macrophage activation and polarization,
based matrix metalloproteinase-2 inhibitor to attenuate adverse cardiac Trends Immunol. 25 (2004) 677–686.
remodeling and improve cardiac function following myocardial infarction, [96] P.C. Westman, M.J. Lipinski, D. Luger, R. Waksman, R.O. Bonow, E. Wu, et al.,
Biomacromolecules. 18 (2017) 2820–2829. Inflammation as a driver of adverse left ventricular remodeling after acute
[69] A. Krzywonos-Zawadzka, A. Franczak, A. Olejnik, M. Radomski, J.F. Gilmer, myocardial infarction, J. Am. Coll. Cardiol. 67 (2016) 2050–2060.
G. Sawicki, et al., Cardioprotective effect of MMP-2-inhibitor-NO-donor hybrid [97] Y.J. Huang, K.C. Hung, H.S. Hung, S.H. Hsu, Modulation of macrophage
against ischaemia/reperfusion injury, J. Cell. Mol. Med. 23 (2019) 2836–2848. phenotype by biodegradable polyurethane nanoparticles: possible relation
[70] I. Cuadrado, M.J. Piedras, I. Herruzo, C. Turpin Mdel, B. Castejon, P. Reventun, et between macrophage polarization and immune response of nanoparticles, ACS
al., EMMPRIN-targeted magnetic nanoparticles for in vivo visualization and Appl. Mater. Interfaces 10 (2018) 19436–19448.
regression of acute myocardial infarction, Theranostics. 6 (2016) 545–557. [98] T. Harel-Adar, T. Ben Mordechai, Y. Amsalem, M.S. Feinberg, J. Leor, S. Cohen,
[71] M.M. Nguyen, A.S. Carlini, M.P. Chien, S. Sonnenberg, C. Luo, R.L. Braden, et al., Modulation of cardiac macrophages by phosphatidylserine-presenting liposomes
Enzyme-responsive nanoparticles for targeted accumulation and prolonged improves infarct repair, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 1827–1832.
retention in heart tissue after myocardial infarction, Adv. Mater. 27 (2015) [99] I. Somasuntharam, K. Yehl, S.L. Carroll, J.T. Maxwell, M.D. Martinez, P.L. Che, et
5547–5552. al., Knockdown of TNF-alpha by DNAzyme gold nanoparticles as an anti-
[72] C. Fan, J. Shi, Y. Zhuang, L. Zhang, L. Huang, W. Yang, et al., Myocardial- inflammatory therapy for myocardial infarction, Biomaterials. 83 (2016) 12–22.
infarction-responsive smart hydrogels targeting matrix metalloproteinase for on- [100] M. Mahmoudi, I. Lynch, M.R. Ejtehadi, M.P. Monopoli, F.B. Bombelli, S. Laurent,
demand growth factor delivery, Adv. Mater. 31 (2019), e1902900. Protein-nanoparticle interactions: opportunities and challenges, Chem. Rev. 111
[73] T. Kalogeris, C.P. Baines, M. Krenz, R.J. Korthuis, Cell biology of ischemia/ (2011) 5610–5637.
reperfusion injury, Int. Rev. Cell Mol. Biol. 298 (2012) 229–317. [101] M.P. Monopoli, D. Walczyk, A. Campbell, G. Elia, I. Lynch, F.B. Bombelli, et al.,
[74] R.L. Winslow, J. Rice, S. Jafri, E. Marban, B. O’Rourke, Mechanisms of altered Physical-chemical aspects of protein corona: relevance to in vitro and in vivo
excitation-contraction coupling in canine tachycardia-induced heart failure, II: biological impacts of nanoparticles, J. Am. Chem. Soc. 133 (2011) 2525–2534.
model studies, Circ. Res. 84 (1999) 571–586. [102] R. Cai, C. Chen, The crown and the scepter: roles of the protein corona in
[75] T.J. Samuel, R.P. Rosenberry, S. Lee, Z. Pan, Correcting calcium dysregulation in nanomedicine, Adv. Mater. 31 (2019), e1805740.
chronic heart failure using SERCA2a gene therapy, Int. J. Mol. Sci. 19 (4) (2018) [103] W. Xiao, H. Gao, The impact of protein corona on the behavior and targeting
1086. capability of nanoparticle-based delivery system, Int. J. Pharm. 552 (2018)
[76] P.A. Gorski, D.K. Ceholski, R.J. Hajjar, Altered myocardial calcium cycling and 328–339.
energetics in heart failure–a rational approach for disease treatment, Cell Metab. [104] N. Bertrand, P. Grenier, M. Mahmoudi, E.M. Lima, E.A. Appel, F. Dormont, et al.,
21 (2015) 183–194. Mechanistic understanding of in vivo protein corona formation on polymeric
[77] H.M. Viola, W.A. Macdonald, H. Tang, L.C. Hool, The L-type Ca(2+) channel as a nanoparticles and impact on pharmacokinetics, Nat. Commun. 8 (2017) 777.
therapeutic target in heart disease, Curr. Med. Chem. 16 (2009) 3341–3358. [105] M.P. Monopoli, C. Aberg, A. Salvati, K.A. Dawson, Biomolecular coronas provide
[78] E.L. Barle, M. Cerne, L. Peternel, M. Homar, Reduced intravenous toxicity of the biological identity of nanosized materials, Nat. Nanotechnol. 7 (2012)
amiodarone nanosuspension in mice and rats, Drug Chem. Toxicol. 36 (2013) 779–786.
263–269. [106] V. Mirshafiee, M. Mahmoudi, K. Lou, J. Cheng, M.L. Kraft, Protein corona
[79] H. Takahama, H. Shigematsu, T. Asai, T. Matsuzaki, S. Sanada, H.Y. Fu, et al., significantly reduces active targeting yield, Chem. Commun. 49 (2013)
Liposomal amiodarone augments anti-arrhythmic effects and reduces 2557–2559.
hemodynamic adverse effects in an ischemia/reperfusion rat model, Cardiovasc. [107] A. Salvati, A.S. Pitek, M.P. Monopoli, K. Prapainop, F.B. Bombelli, D.R. Hristov, et
Drugs Ther. 27 (2013) 125–132. al., Transferrin-functionalized nanoparticles lose their targeting capabilities when
[80] S.R. Abulateefeh, M.O. Taha, Enhanced drug encapsulation and extended release a biomolecule corona adsorbs on the surface, Nat. Nanotechnol. 8 (2013)
profiles of calcium-alginate nanoparticles by using tannic acid as a bridging cross- 137–143.
linking agent, J. Microencapsul. 32 (2015) 96–105. [108] D.F. Moyano, K. Saha, G. Prakash, B. Yan, H. Kong, M. Yazdani, et al., Fabrication
[81] K. Manjunath, V. Venkateswarlu, A. Hussain, Preparation and characterization of of corona-free nanoparticles with tunable hydrophobicity, ACS Nano 8 (2014)
nitrendipine solid lipid nanoparticles, Die Pharmazie. 66 (2011) 178–186. 6748–6755.
[82] M.D. Freedman, J.C. Somberg, Pharmacology and pharmacokinetics of [109] B.L. Oliveira, Z. Guo, G.J.L. Bernardes, Inverse electron demand Diels-Alder
amiodarone, J. Clin. Pharmacol. 31 (1991) 1061–1069. reactions in chemical biology, Chem. Soc. Rev. 46 (2017) 4895–4950.
[83] L. Nagy, A. Kovacs, B. Bodi, E.T. Pasztor, G.A. Fulop, A. Toth, et al., The novel [110] N.K. Devaraj, The future of bioorthogonal chemistry, ACS central science. 4
cardiac myosin activator omecamtiv mecarbil increases the calcium sensitivity of (2018) 952–959.
force production in isolated cardiomyocytes and skeletal muscle fibres of the rat, [111] E.J.L. Steen, P.E. Edem, K. Norregaard, J.T. Jorgensen, V. Shalgunov, A. Kjaer, et
Br. J. Pharmacol. 172 (2015) 4506–4518. al., Pretargeting in nuclear imaging and radionuclide therapy: improving efficacy
[84] N. Kiaie, S.H. Emami, S. Rabbani, R.M. Aghdam, H.A. Tafti, Targeted and of theranostics and nanomedicines, Biomaterials. 179 (2018) 209–245.
controlled drug delivery to a rat model of heart failure through a magnetic [112] K. Westerlund, A. Vorobyeva, B. Mitran, A. Orlova, V. Tolmachev, A.E. Karlstrom,
nanocomposite, Ann. Biomed. Eng. 48 (2020) 709–721. et al., Site-specific conjugation of recognition tags to trastuzumab for peptide
[85] A. Florian, A. Ludwig, S. Rosch, H. Yildiz, S. Klumpp, U. Sechtem, et al., Positive nucleic acid-mediated radionuclide HER2 pretargeting, Biomaterials. 203 (2019)
effect of intravenous iron-oxide administration on left ventricular remodelling in 73–85.
patients with acute ST-elevation myocardial infarction - a cardiovascular [113] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason,
magnetic resonance (CMR) study, Int. J. Cardiol. 173 (2014) 184–189. et al., Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell. 149
[86] W.D. Gray, P. Che, M. Brown, X. Ning, N. Murthy, M.E. Davis, N- (2012) 1060–1072.
acetylglucosamine conjugated to nanoparticles enhances myocyte uptake and [114] W.S. Yang, B.R. Stockwell, Ferroptosis: death by lipid peroxidation, Trends Cell
improves delivery of a small molecule p38 inhibitor for post-infarct healing, Biol. 26 (2016) 165–176.
J. Cardiovasc. Transl. Res. 4 (2011) 631–643. [115] Y. Xie, W. Hou, X. Song, Y. Yu, J. Huang, X. Sun, et al., Ferroptosis: process and
[87] S.A. Marsh, H.E. Collins, J.C. Chatham, Protein O-GlcNAcylation and function, Cell Death Differ. 23 (2016) 369–379.
cardiovascular (patho)physiology, J. Biol. Chem. 289 (2014) 34449–34456. [116] S.J. Dixon, B.R. Stockwell, The role of iron and reactive oxygen species in cell
[88] J.T. Maxwell, I. Somasuntharam, W.D. Gray, M. Shen, J.M. Singer, B. Wang, et al., death, Nat. Chem. Biol. 10 (2014) 9–17.
Bioactive nanoparticles improve calcium handling in failing cardiac myocytes, [117] H. Bulluck, S. Rosmini, A. Abdel-Gadir, S.K. White, A.N. Bhuva, T.A. Treibel, et
Nanomedicine (London) 10 (2015) 3343–3357. al., Residual myocardial iron following intramyocardial hemorrhage during the
[89] P. Most, J. Bernotat, P. Ehlermann, S.T. Pleger, M. Reppel, M. Borries, et al., convalescent phase of reperfused ST-segment-elevation myocardial infarction and
S100A1: a regulator of myocardial contractility, Proc. Natl. Acad. Sci. U. S. A. 98 adverse left ventricular remodeling, Circulation cardiovascular imaging. 9 (10)
(2001) 13889–13894. (2016), e004940.
[90] K. Manjunath, V. Venkateswarlu, Pharmacokinetics, tissue distribution and [118] W.G. Kreyling, A.M. Abdelmonem, Z. Ali, F. Alves, M. Geiser, N. Haberl, et al., In
bioavailability of nitrendipine solid lipid nanoparticles after intravenous and vivo integrity of polymer-coated gold nanoparticles, Nat. Nanotechnol. 10 (2015)
intraduodenal administration, J. Drug Target. 14 (2006) 632–645. 619–623.
[91] G. Ikeda, T. Matoba, Y. Nakano, K. Nagaoka, A. Ishikita, K. Nakano, et al., [119] B. Pelaz, C. Alexiou, R.A. Alvarez-Puebla, F. Alves, A.M. Andrews, S. Ashraf, et al.,
Nanoparticle-mediated targeting of cyclosporine a enhances cardioprotection Diverse applications of nanomedicine, ACS Nano 11 (2017) 2313–2381.
against ischemia-reperfusion injury through inhibition of mitochondrial [120] M. Malki, S. Fleischer, A. Shapira, T. Dvir, Gold Nanorod-based engineered
permeability transition pore opening, Sci. Rep. 6 (2016) 20467. cardiac patch for suture-free engraftment by near IR, Nano Lett. 18 (2018)
[92] N. Hardy, H.M. Viola, V.P. Johnstone, T.D. Clemons, H. Cserne Szappanos, 4069–4073.
R. Singh, et al., Nanoparticle-mediated dual delivery of an antioxidant and a
peptide against the L-type Ca2+ channel enables simultaneous reduction of
cardiac ischemia-reperfusion injury, ACS Nano 9 (2015) 279–289.
[93] S.C. Latet, V.Y. Hoymans, P.L. Van Herck, C.J. Vrints, The cellular immune system
in the post-myocardial infarction repair process, Int. J. Cardiol. 179 (2015)
240–247.

503
C. Lin et al. Journal of Controlled Release 337 (2021) 494–504

[121] S.W. Lee, N. Yabuuchi, B.M. Gallant, S. Chen, B.S. Kim, P.T. Hammond, et al., nanorods and a SWCNT layer for detection of cardiac troponin T protein, J. Mater.
High-power lithium batteries from functionalized carbon-nanotube electrodes, Chem. B 8 (2020) 18–26.
Nat. Nanotechnol. 5 (2010) 531–537. [123] M. Adeel, M.M. Rahman, J.J. Lee, Label-free aptasensor for the detection of
[122] S.G. Surya, S.M. Majhi, D.K. Agarwal, A.A. Lahcen, S. Yuvaraja, K.N. Chappanda, cardiac biomarker myoglobin based on gold nanoparticles decorated boron
et al., A label-free aptasensor FET based on au nanoparticle decorated Co3O4 nitride nanosheets, Biosens. Bioelectron. 126 (2019) 143–150.

504

You might also like