You are on page 1of 18

Heart Failure Reviews (2021) 26:1231–1248

https://doi.org/10.1007/s10741-020-09953-9

Extracellular matrix–based biomaterials for cardiac


regeneration and repair
Haotong Li 1 & Minghui Bao 2 & Yu Nie 1

Published online: 18 April 2020


# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
The spectrum of ischemic heart diseases, encompassing acute myocardial infarction to heart failure, represents the leading cause
of death worldwide. Although extensive progress in cardiovascular diagnoses and therapy has been made, the prevalence of the
disease continues to increase. Cardiac regeneration has a promising perspective for the therapy of heart failure. Recently,
extracellular matrix (ECM) has been shown to play an important role in cardiac regeneration and repair after cardiac injury.
There is also evidence that the ECM could be directly used as a drug to promote cardiomyocyte proliferation and cardiac
regeneration. Increasing evidence supports that applying ECM biomaterials to maintain heart function recovery is an important
approach to apply the concept of cardiac regenerative medicine to clinical practice in the future. Here, we will introduce the
essential role of cardiac ECM in cardiac regeneration and summarize the approaches of delivering ECM biomaterials to promote
cardiac repair in this review.

Keywords Cardiac regeneration . Extracellular matrix . Cardiac injury . Myocardial infarction . Heart failure . Tissue engineering

Introduction treatment for heart failure besides left ventricular assist device
and heart transplantation. Promoting cardiomyocyte prolifer-
Ischemic heart disease is the leading cause of death and more ation and heart regeneration will be a promising approach to
than 8.9 million individuals died of ischemic heart disease intervene heart failure and improve the prognosis of ischemic
globally in 2015 [1]. The rate of death of ischemic heart dis- heart disease in the future. Recently, with the role of extracel-
ease has steadily declined since 1960s due to great attention to lular matrix (ECM)–oncogenic signalling emerging [4], the
primary prevention and significant advances in medical, sur- clinical operating systems for heart regeneration and cardiac
gical, and interventional therapies [2]. However, the heart repair begin to turn to the “Matrix Medicine” [5].
function of most cardiovascular disease patients is impaired The ECM is essential for cells to connect and communicate
with cardiomyocyte loss and cardiac injury, which is the fun- with each other. Previous studies have emphasized the func-
damental cause of heart failure [3]. There is no effective tional role of the ECM primarily in cell junction and tissue
structure organization; nevertheless, emerging studies have
revealed that the ECM can actively participate in intercellular
signalling [6, 7]. Tissue repair and regeneration after injury are
* Yu Nie highly influenced by cytokines, proteases, and communica-
nieyu@fuwaihospital.org tion between different kinds of cells. The ECM contains many
Haotong Li signalling molecules that are essential for cell behaviors, in-
lihaotongdoc@163.com cluding growth, polarity, migration, differentiation, and pro-
Minghui Bao liferation [4, 8]. Under physiological conditions, the ECM
baominghui@mail.ccmu.edu.cn components in mammals have been proven to be well con-
served while the proportion of each component changes with
1
State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, development [9]. Once the tissue is injured, for example, myo-
National Center for Cardiovascular Disease, Chinese Academy of
Medical Sciences and Peking Union Medical College, cardial infarction (MI) or heart apical resection, the cardiac
Beijing 100037, China ECM is likely to undergo temporal and spatial dynamic
2
Department of Cardiology, Peking University First Hospital, changes, and as a result, substantial distinctions may occur
Beijing, China compared with an uninjured heart [10]. ECM can affect cell
1232 Heart Fail Rev (2021) 26:1231–1248

function; in turn, the phenotypic and functional changes of the periostin is required for maturation of the heart and stabiliza-
cell will enhance or attenuate the synthesis and secretion pro- tion of the cardiac ECM. The majority of periostin knockout
cedures of some ECM components and result in ECM remod- mice survived well into adulthood whereas approximately
elling. The bidirectional information flow between the ECM 14% died before weaning due to cardiac architectural and
and the cell within them indicates that the ECM can be valvular anomalies [15].
regarded as an information storage medium. Therefore, a
growing number of ECM reprogramming strategies have been Cardiac ECM in post-infarction conditions
applied to tissue engineering and regenerative medicine which
could be used to treat multiple disorders, such as MI. ECM changes dynamically after MI which plays a critical role
In this review, we introduce the composition of cardiac in cardiac repair and regeneration [16–18]. Compared with
ECM and focus on the topic of ECM in cardiac regeneration those in healthy heart tissue, many components of the cardiac
and repair. We also summarize the delivery strategies of ECM- ECM of the infarction zone, particularly collagens, increase
based biomaterials for tissue engineering, which is essential with the progress of cardiac remodelling and lead to functional
for the development of ECM-based biomaterials in the medi- adaptation [16]. The remodelling of the ECM is described
cine of heart regeneration and cardiac repair. below in chronological order after MI.
Myocardial injury causes rapid activation of neutrophil and
macrophage infiltrate into the heart during the first days after
Cardiac ECM MI. Inflammatory mediators stimulate neutrophils and macro-
phages to produce and secrete matrix metalloproteinases
Cardiac ECM in physiological conditions (MMPs), which subsequently play enzymatic roles in
degrading substrates [19]. For example, MMP9, the most
The cardiac ECM is made up of proteins and polysaccharides widely studied MMP, can proteolyze ECM components such
which can be further divided into three components: glyco- as collagen, fibronectin, osteopontin, tenascin C, and
proteins, proteoglycans, and glycosaminoglycans. The com- thrombospondin 1 [19]. After MI, VEGF increases vascular
ponents of these groups are all proteins decorated by glycosyl permeability and leads to extravasation of plasma fibrinogen
groups. Glycoproteins include fiber proteins, fibronectin, lam- and fibronectin, forming a complex dynamic fibrin matrix
inin, and prototypical matricellular proteins, such as periostin. network [20]. In addition to its hemostatic role, the provisional
Different components play different roles in the cardiac ECM. ECM may act as a scaffold for migrating inflammatory cells to
Two of the fiber proteins, collagen and elastin, are important support the proliferation of endothelial and fibroblast cells
for the architectural structure of cardiac ECM [11]. Fibrillar [10]. The provisional ECM may also serve as a reservoir for
collagen accounts for the largest proportion of proteins in the cytokines and growth factors and play an important role in the
cardiac ECM. Cardiac collagens predominantly consist of transition from the cardiac repair to the proliferative phase.
type I and type III collagens. Type I collagens are the major Then, plasma-derived ECM is phagocytosed by recruited
structural component of the cardiac interstitium, accounting macrophages and replaced by fibronectin, hyaluronan, lami-
for more than 80% of the collagenous matrix, while type III nin, and matricellular macromolecules secreted by fibroblasts
collagens constitute less than 10%, and type VI collagens and macrophages that make up cell-derived ECM [20].
comprise approximately 5% [12]. Compared with type I and ECM remodelling is beneficial for structural repair, func-
type III collagens, which are highly prevalent in the epimysi- tional adaptation, and preventing ventricular rupture.
um and endomysium, respectively, type VI collagens and fi- However, excessive deposition of cell-derived ECM causes
bronectin surround cells in the terminal aspects of the network impaired cardiac function [21]. Therefore, the balance be-
[12, 13]. Fibronectin can also support the ECM structure and tween ECM degradation and deposition is critical for cardiac
plays non-structural roles; for example, it is strongly associat- repair. In the later phases of cardiac repair, many collagen
ed with cell adhesion and migration, which are essential for fibers are deposited in the infarcted area and scar maturation
cell behaviors in embryogenesis and wound healing. occurs as a consequence of ECM cross-linking [22].
Knockout of the fibronectin (Fn1) gene resulted in early em- Additionally, some ECM components may not provide
bryonic lethality due to failure to form a heart tube [14]. structural support, but instead impact cellular phenotypes
Importantly, fibronectin can specifically bind to many cellular and functions by regulating cytokines and growth factors
or ECM molecules. This special function allows bidirectional [23–25]. Osteopontin can prevent ventricular aneurysm and
communication between cells and cardiac ECM. While the ventricular dilatation by promoting ECM deposition [23].
role of matrix networks in maintaining ventricular morpholo- SPARC has been shown to prevent heart rupture and heart
gy, providing structural support, and facilitating force delivery failure after infarction by activating growth factor signalling
is intuitive, it can also transduce critical signals necessary for to promote collagen maturation [24]. Selective endogenous
cardiac cell function. Previous studies have shown that matricellular protein thrombospondin 1 (TSP-1) expression
Heart Fail Rev (2021) 26:1231–1248 1233

in the border zone of the MI may act as a barrier to protect into the injured region, there is no evidence that fibronectin
non-infarcted myocardium from fibrotic remodelling by ac- could promote cardiomyocyte proliferation [30]. Co-culture
tivating anti-inflammatory signalling pathways [25]. After of mammalian embryonic cardiac fibroblasts and
various changes in ECM remodelling, upregulated collage- cardiomyocytes can promote cardiomyocyte proliferation,
nous proteins and matricellular proteins ultimately lead to and fibronectin secreted by embryonic mouse cardiac fibro-
extensive myocardial fibrosis. blasts plays a pro-proliferative role in this process [31]. In
contrast, knockdown of fibronectin expression in embryonic
The relationship between ECM and cardiac cardiac fibroblasts impaired cardiomyocyte proliferation [31].
regeneration The mechanism of cardiomyocyte proliferation promoted by
fibronectin is still unclear but could be partially attributed to
Lower vertebrates such as salamander or zebrafish, upon in- the impacts of β1 integrin signalling and fibronectin signal-
jury (amputation or apical resection), are able to achieve com- ling on epicardial cell function [31].
plete regeneration, namely, the reconstruction of normal mor- To further determine whether ECM is the key for cardiac
phological structure and function [26, 27]. However, mamma- regeneration, Chen et al. [17] used decellularized zebrafish
lian hearts cannot regenerate in adults but can regenerate cardiac ECM to cure adult mice after MI by intramyocardial
completely with cardiomyocyte proliferation in neonates after injection. The results showed that zebrafish ECM could facil-
cardiac injury [28]. It is generally accepted that the mamma- itate adult mammalian cardiomyocyte proliferation in ische-
lian heart regenerative capacity is gradually lost with postnatal mic myocardium and thus contribute to improved cardiac
development. Therefore, in addition to learning from the re- function [17]. In addition to lower vertebrates, neonatal mam-
generation processes of lower vertebrates, the research strate- mals could be an important model to elucidate these process-
gy in the field of heart regeneration is to uncover the differ- es. A recent study showed that the cell-free ECM derived from
ences between neonates and adults, and to analyze the contri- postnatal day 1 (P1) mouse hearts could induce P1 and P7
bution of differentially expressed elements. One study com- cardiomyocyte proliferation in vitro [4]. By using liquid
pared the composition and proportion of fetal, neonatal, and chromatography-mass spectrometry, Bassat et al. [32] com-
adult rat cardiac ECM by using liquid chromatography– pared the cardiac ECM fragments of P1 and P7 and identified
tandem mass spectrometry. The results showed that collagen agrin, a large heparin sulfate proteoglycan, that was enriched
I, collagen III, and laminin increased gradually, while fibro- in neonatal but not in juvenile or adult hearts. Agrin promotes
nectin and periostin decreased with development [29]. These cardiomyocyte proliferation and reduce infarct size after MI in
findings suggested that ECM components change during heart both juvenile and adult hearts by promoting disassembly of
development processes and may play essential roles in the the dystrophin-glycoprotein complex (DGC) through binding
regulation of the regenerative capacity of cardiomyocytes to its receptor dystroglycan 1 (DAG1). This phenomenon
(Fig. 1). One notable question is whether ECM is the key for could subsequently result in myofiber disassembly which is
cardiac regeneration. crucial for cardiomyocyte dedifferentiation. The regenerative
Fibronectin, a core ECM component, is predominantly process from terminally differentiated cardiomyocytes to new
synthesized and secreted by epicardial cells dynamically after cardiomyocytes is achieved in a “dedifferentiation-prolifera-
cardiac injury in zebrafish and plays an important role in car- tion-redifferentiation” manner.
diac regeneration [30]. Although loss-of-function experiments Periostin, another important ECM component, is barely
in zebrafish showed that fibronectin is necessary for cardiac detectable in the healthy adult heart but is highly expressed
regeneration by mobilizing and integrating cardiomyocytes in the neonatal heart [15]. Periostin can switch differentiated

Fig. 1 The role of the


extracellular matrix in heart
regeneration. The schematic
shows the significantly increased
or decreased extracellular matrix
components with the
development of mouse
1234 Heart Fail Rev (2021) 26:1231–1248

cardiomyocytes into the cell cycle and induce cardiac regen- Sources of ECM-based biomaterials
eration with improved myocardial function [33], although this
conclusion remains controversial in adult mice [34]. As the ECM is specifically secreted by native cells of each
Furthermore, a recent study has shown that periostin can pro- tissue [43], it is naturally a complex and bioactive scaffold,
mote myocardial proliferation in neonatal mice by the whose structural and molecular components enable commu-
PI3K/GSK3b/cyclin D1 signalling pathway [35]. Overall, nication between native cells as well as the biological linkage
these studies suggest that ECM components are essential for between native cells and the external environment and dictate
cardiac regeneration and repair. the fate of the cell, such as migration, differentiation, and
In addition to quantitative changes in ECM components, apoptosis [10]. Over the years, various attempts have been
mechanical properties influence cardiac regeneration and made to recreate or simulate ECM by using either naturally
function. Mechanotransduction can profoundly affect the derived ECM components or synthetic polymers to facilitate
synthesis and secretion processes of various ECM molecules cardiac regeneration and repair [44–46]. The predominant
through cell-matrix interactions [36]. The transmembrane sources of ECM-based biomaterials are reviewed below
receptor integrins, as mechanotransducers for contracting (Table 1).
muscle cells, connect the ECM to the cytoskeleton and play
important roles in translating extracellular mechanical infor- Naturally derived ECM biomaterials
mation to intracellular biochemical information. With heart
postnatal development, the stiffness of the ECM in myocar- Purified ECM components
dium increases progressively, which is correlated with car-
diomyocyte cell cycle arrest. Cell experiments showed that Cardiac regeneration and repair efforts mainly rely on the use
reduced matrix rigidity promotes neonatal cardiomyocyte of naturally occurring polymers such as collagen, chitosan,
dedifferentiation and proliferation [37]. In vitro, Akhyari fibrin, gelatin, and alginate [81]. As naturally derived bioma-
et al. found that cardiomyocyte proliferation could be en- terials, these materials exhibit biocompatible, non-immuno-
hanced by loading a 14-day treatment of 20% cyclic stretch genic, and biodegradable characteristics and serve as natural
at 80 cycles/minute on human neonatal cardiomyocytes scaffolds for cellular attachment, proliferation, and differenti-
[38]. However, 10% cyclic stretch at 60 cycles/minute for ation [10, 82]. Collagen, collagen-glycosaminoglycan, fibrin,
1 day had no pro-proliferative effect on the neonatal gelatin, and alginate have been extensively investigated for
cardiomyocytes [39]. In vivo, unloading the human heart myocardial tissue engineering [83, 84]. These ECM mole-
after LVAD implantation increased cardiomyocyte prolifera- cules are isolated and purified from natural tissues and then
tion, which was reflected by added phosphorylated histone applied to impaired hearts as injectable hydrogels by
H3-positive, and Aurora B-positive cardiomyocytes [40]. intramyocardial injection or intracoronary perfusion, or as col-
The physical properties of the ECM can also influence the lagen patches directly attached to the surface of the infarcted
efficiency of cardiac reprogramming, which is closely linked myocardium with or without co-delivery of cells and bioactive
to cardiac repair. The elasticities of the ECM differ from the molecules, either by a single component or mixtures of vari-
developing myocardium to pathologic myocardium and ous components [83, 85].
have different effects on cardiac reprogramming. Kong
et al. [41] tested 0.2, 2, 20, and 200 kPa elasticities of Decellularized ECM
collagen I on polyacrylamide substrates and found that the
reprogramming efficiency peaked at 20 kPa and decreased Although purified ECM components are promising for clini-
thereafter. According to a series of experiments, they created cal introduction to ischemic heart disease, their applications
a model to predict cardiac reprogramming outcomes on dif- are sometimes compromised by the lack of structural integrity
ferent biomaterials based on their mechanobiology. In the and biological cues compared with those of native cardiac
field of heart regeneration, stem cell therapy is controversial ECM. Consequently, by removing cellular and nuclear sub-
because of its lack of efficacy, reproducibility, and integra- stances from tissues and leaving the mixture of structural and
tion. Recently, an important study revealed that an acute functional proteins, decellularized ECM has become a prefer-
immune response that results in reduced ECM content of able biomaterial for repairing cardiac tissue due to its ability to
the border zone and enhanced mechanical properties of the provide 3D fibrous and porous topographies [86, 87], and
injured area is the main mechanism of stem cell therapy for thus, recapitulate the complexity of matrix components of
ischemic heart disease [42]. The mechanical properties of natural ECM and provide biological cues for cardiac regener-
ECM are crucial for cardiac regeneration and repair, and ation and repair after ischemic injury [88, 89].
not only the biochemical but also the physical characteristics Many approaches, such as physical and chemical as well as
of ECM components should be taken into consideration for enzymatic methods, have been used to remove cells from the
basic research and clinical application. ECM [87, 90]. Decellularization strategies are required to
Heart Fail Rev (2021) 26:1231–1248 1235

Table 1 ECM-based biomaterials for cardiac regeneration and repair

Advantages Disadvantages Functional roles References

Naturally derived ECM biomaterials


Purified ECM components Biocompatible; biodegradable; Poor mechanical Can be applied as injectable [45,
non-immunogenic properties; uncontrolled hydrogels or as collagen 47–57]
degradation process patches with or without
co-delivery of cells and bioac-
tive molecules
Decellularized ECM Biocompatible; biodegradable; Lack of standardized Provide 3D structure to [58–64]
non-immunogenic; retained 3D decellularization process; recapitulate the matrix
fibrous and porous topogra- decellularization processes may components of natural ECM
phies as well as macrostructures potentially damage and provide biological cues for
such ultrastructure of ECM; cardiac regeneration after
as the vasculature inflammatory responses due to ischemic injury
incomplete decellularization,
endotoxin residues, as well as
detergent residues
Cell culture–derived bio- Natural ECM with bioactivity, Limited thickness; poor Can be adhered to the heart surface [65–69]
materials biocompatibility and mechanical properties spontaneously without sutures
biodegradability during epicardial implantation
to promote cardiac regeneration
Biohybrid ECM-based biomaterials
Biohybrid ECM-based Desirable mechanical properties; Foreign body reactions; Can be applied as injectable [52,
biomaterials able to mimic the biological compromised degradability hydrogels or as patches with or 70–74]
functions of native ECM, while without co-delivery of cells and
maintaining good control over bioactive molecules
multiple physical properties
ECM-based biomimetic materials
ECM-based biomimetic Preferable physical and Lack of comprehensive Can be applied as scaffold [75–80]
materials mechanical properties; understanding of natural ECM; materials or constructs to
can be precisely characterized foreign body reactions deliver cells or drugs
and fabricated with good
control over physical and
chemical properties

ECM extracellular matrix

efficiently remove cells with minimal adverse effects on the isopropylacrylamide, and secrete their own ECM [92]. The
components, biological activity, and mechanical integrity of dishes were hydrophobic and cell adhesive at 37 °C. When
the remaining ECM. Several factors may affect the composi- the cells are confluent on dishes, the culture temperature is
tion and ultrastructure of decellularized ECM and further in- decreased to 32 °C, which could lead to the detachment of
fluence the host response to the implants as well as the func- the monolayer cultured cells along with the ECM and attach-
tional performances of the implants. Inflammatory responses ment proteins as intact, free-floating 2D cell sheets. Once the
may occur due to altered ECM ultrastructure, incomplete 2D cell layers are piled, they can quickly attach and form cell-
decellularization, endotoxin residues, and detergent residues to-cell connections [93, 94]. Further, multiple cell sheets can
[87]. In recent years, decellularized ECM-based biomaterials be stacked into a thicker 3D tissue that can survive for longer
have been used in clinical management of cardiovascular dis- time either in culture or in living heart tissues [95–97] and can
eases limited to heart valve disease and congenital heart dis- spontaneously adhere to the heart surface without sutures dur-
ease, whereas only two main decellularized ECM products ing epicardial implantation. Furthermore, cell sheets exhibit
(VentriGel and CorMatrix) are being evaluated in clinical test- low immunogenicity after transplantation due to their non-
ing for ischemic heart disease [91]. scaffold nature [98].

Cell culture–derived biomaterials Biohybrid ECM-based biomaterials

Cell culture–derived biomaterials are another type of ECM- Although naturally derived ECM-based biomaterials have
based biomaterial. The cells are initially cultured on dishes been widely applied to the fields of cardiac regeneration and
coated with a temperature-sensitive polymer, poly-N- repair and are considered the biomaterials that best resemble
1236 Heart Fail Rev (2021) 26:1231–1248

the biocompatibility, biodegradability, and bioactivity of na- Functionalized ECM-based biomaterials


tive cardiac tissue, some ever-present limitations such as un- and delivery systems
desirable mechanical strength may hinder their clinical trans-
lation. Given the requirement for optimizing multiple proper- With the rapid development of tissue engineering, various
ties of ECM-based biomaterials, biohybrid materials are de- types of functionalized ECM-based biomaterials have been
veloped to address these challenges. By cross-linking natural- devised for repair of injured myocardium either to serve as
ly derived ECM-based biomaterials with natural [99] or syn- structural supports or constructs that efficiently deliver cells
thetic polymers [100], hybrid ECM-based biomaterials allow and/or bioactive molecules to injured myocardial tissue [106,
increased control of their components and physical features, 107]. Multiple biomaterials have been used for co-delivery of
such as stiffness, contractibility, conductivity, and porosity cell sources to promote cell retention and improve the thera-
[101]. More importantly, they exhibit considerable potential peutic outcomes of cell therapy [44, 108]. Primary types of
to mimic the native ECM in biological functions, while main- functionalized ECM-based biomaterials are reviewed in both
tain a variety of physical properties at the same time [102, cell-loaded and cell-free manners (Fig. 2).
103]. Therefore, biohybrid ECM-based biomaterials have
been introduced to the clinical applications of ischemic heart
diseases [85, 104]. Acellular patches

Cell-free ECM-based biomaterials have long been a hot re-


ECM-based biomimetic materials search area for their simplicity. Acellular patches, as well as
decellularized injectable hydrogels, are among the most in-
The natural ECM is considered as a natural scaffold that vestigated strategies in cardiac regeneration medicine.
provides essential biological cues and mechanical sup- Engineered cell-free ECM biomaterials are produced to offer
ports for native cells [105]. As the ultimate aim of car- mechanical support and deliver cells, drugs, or other bioac-
diac regeneration and repair is to repair injured myocar- tive factors to the injured myocardium, promoting the regen-
dium by using biomaterials and further promote endoge- eration of cardiomyocytes and the formation of vasculature
nous regeneration with cardiomyocyte proliferation, an [46]. Naturally derived ECM components (such as collagen,
emerging strategy is to fabricate biomaterials that mimic fibrin, and chitosan), natural polymers (such as alginate), and
the biological and mechanical characteristics of the na- synthetic polymers (such as polycaprolactone [PCL] and
tive cardiac ECM. Over the past few decades, extensive polyglycerol sebacate [PGS]) are applied to fabricate acellu-
research has illustrated the structural and functional roles lar ECM-based biomaterials in either a solo or combined
of natural ECM. Simulating the natural ECM to optimize manner [109, 110].
the biological and mechanical performances of biomate- Acellular cardiac patches are designed predominantly to
rials is a promising approach to achieve cardiac regener- provide mechanical support for impaired cardiac tissues and
ation and repair. offer microenvironments for native cell integration and

Fig. 2 Functionalized ECM-


based biomaterials and delivery
approaches. ECM extracellular
matrix
Heart Fail Rev (2021) 26:1231–1248 1237

vascularization post-MI with limited immune reaction and maintained cardiac function without inducing arrhythmias.
relatively low cost. As collagens account for a large propor- The researchers also tested the ability of this hydrogel to be
tion of the cardiac ECM contents, extensive research has been delivered percutaneously by catheter in large animal model. In
conducted to develop collagen patches and explore their ap- a porcine model, the hydrogel was delivered using the
plications in MI models. After grafting 3D collagen I patches NOGA-guided MyoStar catheter and the material retention
onto the injured myocardium of rats with MI, researchers ob- was assessed using a histological strategy. The results demon-
served new vessel formation and reduced ventricular remod- strated the feasibility of transendocardial catheter injection in a
elling after implantation [111]. Some physical processing ap- large animal model, suggesting that it is a promising novel
proaches have also been adopted to optimize the mechanical therapy for treating MI [127].
properties of collagen patches and result in preferable cell
integration, vessel formation, and cardiac function with Cell sheets with ECM-based biomaterials
inhibited ventricular dilation [112]. Moreover, some hybrid
collagen-based patches made by incorporating collagens with Another approach to generate ECM-based biomaterials is cell
other natural or synthetic polymers have been produced to sheets. Cells are instructed to produce their own ECM under
modify their mechanical characteristics to provide improved defined in vitro culture conditions to form a scaffold-free
structural supports [113]. However, other ECM components ECM-based biomaterial. In recent years, cell sheets have been
and polymers are commonly used to fabricate injectable ma- successfully applied to clinical settings of regenerative medi-
terials in gel formats based on their biological, physical, and cine for the heart [128], cornea [129], cartilage [130], and
chemical properties. esophagus [131]. In cardiac regeneration, human iPSC–
derived cardiomyocytes were also cultivated as cell sheets
Decellularized injectable hydrogels and implanted into MI pig models. The cell sheets could con-
duct action potentials [132] and beat spontaneously [133].
Hydrogels are one of the most promising biomaterials be- When the cell sheets were piled in layers, they became elec-
cause of their high water content, biocompatibility, and easy tronically coupled [134, 135] and were associated with im-
tunability for mimicking the properties of the ECM [114]. To proved cardiac function, vascularity, and fibrosis after trans-
date, various types of ECM-based hydrogels with optimized plantation [136].
biological properties have been devised for cardiac regenera- Attempts to transplant these materials into patients with
tion and repair [115]. Injectable hydrogels for cardiac engi- cardiovascular diseases have been made and have shown pos-
neering can be fabricated from natural materials, synthetic itive effects. In 2012, a cell sheet–based cardiac patch was
materials, or a hybrid of the two [116]. The most commonly reported to be used in a 56-year-old male who suffered from
used natural hydrogels include collagen, hyaluronic acid, gel- dilated cardiomyopathy. Skeletal muscle pieces were excised
atin, chitosan, alginate, and chondroitin sulfate [117], while from this patient and the autologous myoblasts derived from
synthetic polymers include poly(ethylene glycol) (PEG), this muscle were cultured and grown on temperature-
poly(N-isopropylacrylamide) (PNIPAm), poly(glycolic acid) responsive culture dishes. After 48 h, they formed cell sheets
(PGA), and poly(lactic-co-glycolic) acid (PLGA) [118, 119]. with diameters of approximately 4 cm. Researchers implanted
To improve the biological and physical properties of inject- 4-layered myoblast sheets on one site and a total of 20 myo-
able hydrogels, researchers have developed synthetic blast sheets on five sites to the anterior to the lateral surface of
hydrogels such as chitosan-PEG, collagen-PNIPAm, and chi- the dilated heart through a left lateral thoracotomy, observed
tosan-poly(vinyl alcohol) (PVA) [120, 121]. In recent years, markedly improved left ventricular ejection fraction without
hydrogels derived from decellularized materials have drawn arrhythmia, and could remove the left ventricular assist system
much attention due to their ability to recapitulate the physical [128]. Generally, the findings above may indicate that the cell
and biological characteristics of native ECM and minimize sheet strategy may be an effective alternative to heart trans-
the immunogenic response [122]. For ischemic heart dis- plantation in the near future.
eases, hydrogels can serve as constructs for cells [123, 124],
drugs, and other bioactive molecules [125, 126] by directly
injected into the injured myocardium or delivered through Delivery approaches
catheter-based systems.
To evaluate the use of an injectable hydrogel derived from The administration of biomaterials is an important clinical
cardiac ECM for treating MI, Singelyn et al. developed a procedure. Various routes can be used to deliver biomaterials
myocardial-specific decellularized ventricular ECM hydrogel to the heart. These approaches include catheter-based mini-
and injected the hydrogel 2 weeks post-MI. The researchers mally invasive delivery approaches, including intracoronary
found that injection of the material in the rat MI model in- perfusion, endocardial delivery, transvenous delivery, and
creased endogenous cardiomyocytes in the infarct area and open-chest surgery–based invasive delivery approaches (Fig.
1238 Heart Fail Rev (2021) 26:1231–1248

2). The choice of ideal delivery approaches predominantly thoracotomy and a pericardial incision to access the epicardi-
depends on the characteristics of biomaterials, the specific um [139]. Future development of minimally invasive or even
type of heart disease, the disease intervention approach, and non-invasive delivery approaches for non-injectable biomate-
the availability of multiple delivery systems. rials may significantly facilitate the clinical translation of such
cardiac repair strategies.
Catheter-based minimally invasive delivery
approaches
ECM-based biomaterial requirements
The development of injectable hydrogels based on stimuli- for tissue engineering and cardiac
sensitive systems enables the application of catheter technol- regeneration
ogy. Such hydrogels are administered in a liquid state and
become a gel only when they have reached the myocardium, Biomaterials, either used to retain ventricular geometry, or
without blocking catheters and vessels [137]. used as scaffolds to generate tissue patches, should be func-
With catheter-based approaches, intracoronary injection tionalized materials that meet several basic requirements, in-
can be accessed with a catheter via a percutaneous translumi- cluding biocompatibility, non-allergenicity, non-toxicity, bio-
nal coronary angioplasty (PTCA) balloon. After injection, the degradability, adequate mechanical strength, conductivity, and
leaky vasculature allows the transported materials to enter the injectability. Some pathological conditions, typically heart
infarcted zone, and therefore increases the number of cells failure and arrhythmia, may occur if biological substitutes
delivered to the injured area. Endocardial injection obtains for lost or failing tissues do not possess identical or approxi-
access to the myocardium from the interior of the ventricle. mate biochemical and biomechanical characteristics as the
After puncture of the radial artery or femoral artery, catheters native cardiac tissue.
are introduced to the arterial system and penetrate through the Since one of the essential roles of biomaterials is serving as
aortic valve. Combined with imaging technology, endocardial a suitable environment or construct for transplanted or native
delivery could achieve targeted injection towards ischemic cells, tissue engineering strategies can be adopted to repair
sites. The transvenous delivery system approaches the myo- injured myocardium by carrying signalling molecules to stim-
cardium via the epicardial route. Catheters are placed through ulate endogenous cell adhesion processes [47, 140]. The
the femoral veins and then pass through one of the coronary ECM-based biomaterials should exhibit biocompatible prop-
veins. Under the assistance of intravascular ultrasound erties with native myocardial tissue and remain bioactive after
(IVUS), the coronary vein is punctured and the catheter passes implantation, consequently leading to improved functional
through the puncture site to the epicardial wall. outcomes of impaired myocardium [49]. Biocompatible
ECM-based materials are required to maintain an appropriate
Open-chest surgery–based invasive delivery host response instead of stimulating severe immune responses
approaches [141]. However, some beneficial inflammatory and immune
responses could be retained due to their tissue reparative na-
Direct injection of injectable biomaterials ture [142]. Biocompatibility also lies in resistance to
thrombogenesis as well as bacterial colonization, which could
A direct injection strategy is extensively applied in small an- guarantee the functioning and survival of biomaterials both
imal models (such as rats, mouse, and rabbits) in which inject- in vivo and in vitro. Over the past decades, ECM-based natu-
able hydrogels are assessed. The hydrogel is injected directly rally occurring polymers such as collagen, chitosan, fibrin,
into the desired location. This method has the most location gelatin, and alginate have been extensively investigated in
control for delivery but requires invasive surgery [138]. The myocardial tissue engineering [45, 47, 48, 143]. As natural
advantage of intramyocardial injection is its ability to achieve biomaterials, they can provide a natural substrate for cell ad-
precise administration of the biomaterials to the target region hesion, proliferation, and differentiation in a relatively native
under direct visualization. state without causing immune reactions [49, 144]. Various
forms of ECM-derived biomaterials have been commercially
Implantation of non-injectable biomaterials used as scaffolds across many clinical settings for their
bioinductive properties [49].
For prefabricated patches and cell sheets, there is no available Biodegradability indicates the ability of an implanted ma-
catheter-based approach that can be used either experimental- terial to break down through various mechanisms, such as
ly or clinically. To date, cardiac patches and cell sheets have hydrolytic mechanisms (bioerosion), cellular activity mecha-
been delivered to the epicardium via open-chest surgery. nisms (bioresorption), and enzymatic mechanisms (biodegra-
However, this invasive delivery approach could utilize dation). Equally important is that the degradation products
transapical delivery approaches by undertaking a mini- should also meet the requirements of both biocompatibility
Heart Fail Rev (2021) 26:1231–1248 1239

and biodegradability because incompletely degradable mate- as natural cardiac tissue. Given that natural ECM-based ma-
rial or toxic degraded products cannot be used for clinical terials are likely to have weak mechanical properties and are
application [145]. ECM-based biomaterials, such as collagen prone to degradation, functional chemical groups have been
and fibrin, can perfectly meet the requirements above because added to them to obtain mechanically controllable materials
they possess the natural molecular components required for that are more suitable for clinical application [152]. As the
cell attachment and survival and are able to degrade into bio- heart is a constantly contracting organ driven by spontaneous
compatible and nontoxic products through the enzymatic ef- electrical activities, the electrophysiological and conductive
fect of cells [146]. Benefitting from these merits, several performances of the biomaterials should match the physical
ECM-based biomaterials have been commercialized and ap- needs of the beating heart in addition to barely withstanding
plied to clinical use [147]. However, as biomaterials degrade, the biomechanical forces of the cardiac contractile cycle
the degradation rate and survival time of ECM-based bioma- [145]. Given that bioengineered ECM-based materials offer-
terials have emerged as other issues of concern. By enzymatic ing a three-dimensional (3D) combination of cell sources, tis-
digestion, ECM-derived materials degrade considerably post- sue growth factors, and scaffolds could result in improved
implantation. Therefore, controlling the degradation rate of tissue reconstruction [153, 154], attempts have been made
biomaterials to match the specific rate requirements of new by implanting cardiomyocytes, cardiac progenitors, skeletal
tissue formation and ensuring the biomaterials exert the de- muscle cells, and mesenchymal stem cells (MSCs) together
sired function remains a priority in biomaterial design [148]. and cultured as a sheet format on bioscaffolds and these pro-
Some chemical groups, such as glutaraldehyde and ethyl cesses ultimately resulted in promoted cell connections, func-
carbodiimide, have been used to form cross-links with tional integration, and synchronic contraction [155].
ECM-based materials to obtain a controlled degradation rate
[149]. Additionally, photolabile chemical strategies have also
been used to form these cross-links [150]. Limitations of the ECM-based biomaterials
The mechanical properties of ECM-based materials are an- and challenges of the delivery approaches
other important concern of tissue engineering and cardiac re-
generation. The mechanical requirements for ECM-based bio- Limitations of the ECM-based biomaterials
materials differed based on the specific types of tissue engi-
neering usage. The engineered tissue, especially patch-like In the past few decades, studies involving the development
materials, should harbor the proper mechanical properties to of biomaterials for tissue engineering and cardiac regenera-
withstand the consistent contraction and dilation of the heart tion have significantly progressed by continuously improv-
[44]. Generally, a healthy adult heart contracts approximately ing material properties to mimic native cardiac tissue.
100,000 times a day and 3 × 109 times in a lifetime. The cyclic Currently, researchers can recapitulate the structural, molec-
hemodynamic changes in the circulatory system require heart ular, and mechanical properties of the native cardiac ECM to
tissue to have enormous strength, flexibility, and durability. mimic the native cellular environment to a large extent
When in vivo tissue engineering is used, the biomaterial has [156]. However, some problems involving ECM-based de-
to function as an alternative to injured tissue. Their mechanical livery need to be addressed.
properties need to be precisely tuned to ensure that they can Although natural tissue–derived biomaterials are ideal can-
withstand the mechanical demands that will be placed upon didates for cardiac repair because they offer suitable substrates
them immediately after application. Additionally, the im- for cellular attachment, proliferation, and differentiation in the
planted material should not hinder the surrounding tissues to native state [157], the poor mechanical properties and ease of
exert their normal mechanical functions. In a cardiac contrac- degradation of these materials remain a major obstacle for
tion cycle, the pressure load of the left ventricle ranges from clinical application. Some efforts have been made to modulate
1.3 to 16 kPa, while the myofiber stress along the myofiber these mechanical properties. Through generation of compos-
axis ranges from 5 to 50 kPa [151]. ites of two or more types of natural biomaterials, the mechan-
If the ECM-based biomaterial is intended to be used to ical strength and elasticity can be improved without
strengthen the ventricle wall or to maintain ventricular integ- compromising the compatibility [158]. Glycosaminoglycans
rity, the mechanical property of the implanted material should (GAGs) can also be used to cross-link with collagen fibers to
achieve the upper limit of the mechanical range of the native enhance the mechanical properties of collagen gels [159].
ventricle. However, if the biomaterial is designed to be used as Therefore, the development of hybrid biomaterials containing
an injectable hydrogel or a temporary scaffold to deliver cells natural ECM with biodegradable synthetic materials may offer
or initiate adhesion processes of endogenous cells, a stiffness practical solutions for the applications of biomaterials in car-
reaching the low-end of the mechanical range of native tissue diac repair from bench to bedside.
might be enough, since the seeded cells are able to remodel the The electrical integration of these materials to develop con-
ECM-derived scaffold and form a mechanically similar tissue ductive and contractile cardiac constructs is another important
1240 Heart Fail Rev (2021) 26:1231–1248

concern. The contractile activity of cardiomyocyte sheets is been carried out to explore the feasibility and optimize ap-
expected to contribute directly to myocardial performance. proaches to catheter-based biomaterial delivery.
However, the transplanted sheet may remain electronically To investigate whether biomaterials can be delivered effec-
isolated from the native myocardium. Although cell sheets tively into the infarcted myocardium by intracoronary injec-
have been applied to cardiac regeneration engineering and tion, Leor et al. [166] prepared a calcium cross-linked alginate
displayed beneficial effects on cardiac function after trans- solution that undergoes liquid to gel phase transition after
plantation into the infarcted myocardium in animal models deposition in the infarcted myocardium. In a porcine model,
[130], the implanted cells cannot electro-physiologically cou- the alginate solution was injected 4 days after MI. The results
ple to the native cardiomyocytes, which may lead to an in- showed that intracoronary injection of alginate reversed left
creased risk for ventricular arrhythmia after transplantation. ventricular enlargement at 60 days. This attempt demonstrated
However, this limitation may hamper the clinical application the feasibility and effectiveness of delivering injectable
of cell sheets. Evidence has shown that electrical stimulation hydrogels using a catheter system for the first time. Singelyn
could induce synchronous contractions of cultured cardiac et al. [127] evaluated the use of an in situ gelling injectable
constructs and significant ultrastructural organization charac- hydrogel derived from ventricular ECM for treating MI and its
terized by an increased density of intercalated discs, gap junc- ability to be delivered by catheter system in a porcine model.
tions, and t-tubules [160]. Additionally, physiological load The hydrogel self-assembles could lead to increased endoge-
and electrical stimulation have been adopted in the construc- nous cardiomyocytes in the infarct area and maintain cardiac
tion of 3D myocardial tissue in vitro and result in the genera- function without inducing arrhythmias, which demonstrated
tion of pump function [161], which provides a possibility for that the in situ gelling decellularized ECM hydrogel was com-
the regeneration of the whole functional heart [45]. patible with catheter delivery.
To mimic the natural ECM of tissues under physiological Moreover, to adapt the hydrogels to be delivered by a range
conditions, constructs are required to maintain structural in- of commercially available catheters, Martens et al. investigat-
tegrity and support cell adhesion and tissue growth without ed the polymerization kinetics of fibrin hydrogels and identi-
activating foreign bodies or harmful immune responses [162, fied the ranges of concentrations compatible with catheter
163]. Therefore, using natural ECM components or cell-free delivery in a nude rat model of MI [167]. To achieve a soft
ECM scaffolds may be optimal choices for tissue engineering. hydrogel with stiffening to enhance the therapeutic efficacy of
Ideally, the decellularized ECM is predominantly composed preventing left ventricular remodelling, Rodell et al. devel-
of collagen and elastin [164]. Since cellular antigens are elim- oped a catheter-deliverable hyaluronic acid hydrogel by using
inated, the decellularization process could theoretically result a tandem cross-linking approach where the first cross-linking
in a non-immunogenic acellular scaffold with a native intact (guest-host) enabled injection and localized retention of a soft
structure for new tissue regeneration. However, harmful im- hydrogel and a second cross-linking reaction (dual-cross-
mune responses can also be observed after implantation for linking) stiffened the hydrogel after injection. In an ovine
cellular components that have not been completely eliminated. MI model, hydrogels with increased stiffness were found to
Thus, to minimize the immune response, novel strategies need be most effective at ameliorating left ventricular remodelling
to be developed and adopted to achieve efficient and complete and preserving function [168].
decellularization. Considering the layered and hierarchical The intramyocardial injection approach requires open-
structure of myocardium tissue, it is important for ECM- chest surgery, and this invasive approach may limit its appli-
based materials to offer exact spatiotemporal delivery of mo- cation. Moreover, solutions other than saline are needed to
lecular and biophysical cues to facilitate the processes of cell prevent hydrogels from leaking into the ventricle under con-
adhesion, differentiation, migration, new ECM synthesis, and stant contraction and dilation in cardiac cycles [169, 170].
remodelling [165]. Therefore, optimizing the timing and ad-
ministration procedures of biomaterial delivery remains a
daunting task for cardiac engineering. Clinical trials and perspectives

Clinical trials for ECM-based biomaterials in cardiac


Challenges of the delivery approaches repair

Although catheter-based delivery approaches have been ex- Although various biomaterials have been shown to be appli-
tensively used in humans to deliver cell sources without con- cable matrices for cardiac regeneration, only a few of these
structs derived from natural or synthetic biomaterials, existing biomaterials would be assessed in clinical settings. To date,
“on the shelf” catheters are not suitable to deliver injectable several clinical trials have been carried out to test the effec-
biomaterials because of some hurdles such as rapid gelation tiveness and safety of some ECM-based biomaterials
kinetics and high viscosity. Therefore, various studies have (Table 2).
Table 2 Clinical trials for ECM-based biomaterials in cardiac repair

Trial name Trial identifier Substrate Administration strategy Sample size Cardiac injury Study phase Recruitment states and References
Heart Fail Rev (2021) 26:1231–1248

outcomes

PRESERVATION-1 NCT01226563 Sodium alginate Intracoronary 303 Recent STEMI NA Recruitment completed; no [171, 172]
calcium gluconate functional improvement;
no safety concerns
AUGMENT-HF NCT01311791 Calcium alginate Intramyocardial 58 Congestive heart failure; Phase 2, Recruitment completed; [173]
EF < 35% phase 3 mildly improved peak
VO2; mildly improved
6-min walk test
AUGMENT-HF II NCT03082508 Calcium alginate Intramyocardial > 200 planned Congestive heart failure; NA Not yet recruiting NA
EF < 35%
VentriGel NCT02305602 Porcine ECM Transendocardial 15 Remote STEMI Phase 1 Recruitment completed; no NA
results published
EIR NCT02887768 CorMatrix-extracellular Surgically applied 8 STEMI within 6 weeks Early phase 1 Recruitment completed; no [174, 175]
matrix to the epicardial surface and underwent CABG human results published
ESCORT NCT02057900 Human ESC–derived Surgically applied 10 Congestive heart failure; Phase 1 Recruitment completed; [176]
progenitors embed- to the epicardial surface EF < 35% short-term and
ded into a fibrin patch medium-term safety;
symptomatically im-
proved with increased
systolic motion of the
cell-treated segments
CARDIOMESH NCT03746938 Adipose-derived stem Surgically applied 10 Ischemic heart disease Phase 1 Recruiting NA
cells seeded into a to the epicardial surface and left ventricular
collagen membrane dysfunction, EF < 35%

PRESERVATION-1 IK-5001 for the prevention of remodelling of the ventricle and congestive heart failure after acute myocardial infarction, AUGMENT-HF a randomized, controlled study to evaluate
Algisyl-LVR™ as a method of left ventricular augmentation for heart failure, AUGMENT-HF II a pivotal trial to establish the efficacy and safety of Algisyl in patients with moderate to severe heart failure,
VentriGel a study of ventrigel in post-mi patients, EIR epicardial infarct repair using CorMatrix®-ECM: Clinical Feasibility Study, ESCORT transplantation of human embryonic stem cell-derived
progenitors in severe heart failure, CARDIOMESH first in humans to evaluate collagen patches with stem cells in patients with ischemic left ventricular dysfunction, NA not applicable
1241
1242 Heart Fail Rev (2021) 26:1231–1248

Two types of alginate-based injectable hydrogels have been ongoing trials involving the clinical application of ECM-
developed and assessed in two clinical trials (PRESERVATION- based biomaterials have not been completed with sufficient
I trial: NCT01226563 [171, 172]; AUGMENT-HF trial: promising results, there is still much hope in the field of
NCT01311791 [173]). The hydrogel used in PRESERVATION- cardiac tissue engineering aiming to improve regenerative
I is an injectable bioabsorbable cardiac matrix, composed of an therapies for ischemic heart disease and heart failure.
aqueous mixture of 1% sodium alginate and 0.3% calcium glu- Through optimization of the mechanical, degradation,
conate (IK-5001). When exposed to excess ionized calcium and bioactivity properties, biomaterials may exert prefera-
present in infarcted myocardium, it assembles to form a flexible ble functions in providing mechanical support for injured
gel, structurally resembling ECM, which provides a temporary myocardium after MI. Moreover, precisely modifying the
bioabsorbable cardiac scaffold. IK-5001 was delivered by a immune and regenerative signalling characteristics, realiz-
catheter to the infarct-related coronary artery within 7 days ing spatiotemporally controlled delivery of biomaterials,
post-MI. The effectiveness and safety endpoints were evaluated and rationally designing electrically conductive scaffolds
after a 6-month follow-up. However, intracoronary deployment could also largely benefit the development of functional
of IK-5001 failed to reduce adverse left ventricular remodelling engineered cardiac tissue and enhance the progress of car-
or cardiac clinical events at 6 months, although no device-related diac regenerative medicine.
adverse events, serious arrhythmias, blood abnormalities, or Due to the increasing demand for clinical translation, the
death were noted. development of injectable biomaterials has entered a new
The AUGMENT-HF trial is aimed at assessing the effect of generation and some novel strategies have been included
biomaterials and superior to standard medical therapy (SMT) to improve the bioactivity of biomaterials. Increasing evi-
for improving the functional capacity and clinical outcomes of dence has shown that actively secreted membrane vesicles,
patients with advanced heart failure. The injectable hydrogel especially exosomes, may serve as new candidates with im-
(Algisyl) consists of sodium alginate solution in 4.6% manni- portant roles in the repair mechanisms after MI [177]. A
tol and calcium alginate particles suspended in 4.6% mannitol. recent study has shown that hydrogel patches slowly releas-
Algisyl was injected into the myocardium under direct visu- ing extracellular vesicles secreted from cardiomyocytes de-
alization during the surgical procedure. After a 1-year follow- rived from induced pluripotent stem (iPS) cells could reduce
up, Algisyl in addition to SMT was more effective than SMT arrhythmic burden, promote ejection fraction, decrease car-
alone for improving exercise capacity, symptoms, and clinical diomyocyte apoptosis 24 h after infarction, and reduce in-
status for patients, which supports larger clinical evaluations farct size and cell hypertrophy 4 weeks post-infarction when
of this novel therapy. Given the promising results identified by implanted onto infarcted rat hearts [178]. Other attempts
AUGMENT-HF (NCT03082508), AUGMENT-HF II was es- have also been established by adding cell-sourced products
timated to start in August 2017 and enroll 240 participants to injectable hydrogels to improve the bioactivity of bioma-
with dilated cardiomyopathy. Algisyl will be injected into terials. Some secreted ECM modulatory proteins [179],
the myocardium under direct visualization during the surgical cardiac-specific miRNAs [180], and PLGA-based micropar-
procedure. The purpose of this study is to investigate Algisyl ticles containing oxygen-releasing moieties [181] or mixed
employed as a method of left ventricular augmentation and with stem cell membrane fragments [182] have been fused
restoration in patients with dilated cardiomyopathy. However, with injectable biomaterials and were demonstrated to im-
the current stage of AUGMENT-HF II is “not yet recruiting.” prove the functional outcomes of MI animal models.
Apart from alginate-based injectable hydrogels, porcine- Although these novel biomaterial and cell therapy strate-
derived materials have also been applied and assessed in a gies employed for constructing and delivering engineered car-
clinical setting. By trans-endocardially delivering VentriGel diac tissues will likely play key roles in improving the func-
(a porcine-derived acellular hydrogel), the VentriGel trial tional integration of engrafted materials, the reproducibility
(NCT02305602) aimed to investigate the effectiveness, safety, and long-term viability of these strategies remain unknown.
and feasibility of VentriGel in patients who have experienced Therefore, further investigation is needed to verify the efficacy
ST-elevation MI (STEMI) treated by PCI within the past and safety of these emerging strategies in either animal models
3 years and have evidence of left ventricular remodelling. or clinical settings. Moreover, the reproducibility of the pro-
This trial is estimated to be completed by December 2018; duction process and the stability of the products are also some
however, no results have been released thus far. of the factors that should be standardized to achieve valid
comparisons among all research outcomes in the field and
Perspectives facilitate collaboration between groups and future develop-
ments of cardiac repair.
In recent years, the importance of ECM in restoring tissue
organization and functionality to the heart after myocardial Author contributions Conceptualization: Y.N.; Writing: H.L., M.B., and
injury has been well established. Although some of the Y.N.; Editing: H.L. and Y.N.
Heart Fail Rev (2021) 26:1231–1248 1243

Funding information This research was funded by Innovation Fund for 14. Astrof S, Kirby A, Lindblad-Toh K, Daly M, Hynes RO (2007)
Medical Sciences (CIFMS, 2016-I2M-1-015); National Natural Science Heart development in fibronectin-null mice is governed by a ge-
Foundation of China (NSFC, 81770308, 81500239); the National Key netic modifier on chromosome four. Mech Dev 124(7-8):551–
Research and Development Project of China (2019YFA0801500); and 558. https://doi.org/10.1016/j.mod.2007.05.004
CAMS Beijing Natural Science Foundation (7172183). 15. Snider P, Hinton RB, Moreno-Rodriguez RA, Wang J, Rogers R,
Lindsley A et al (2008) Periostin is required for maturation and
extracellular matrix stabilization of noncardiomyocyte lineages of
Compliance with ethical standards the heart. Circ Res 102(7):752–760. https://doi.org/10.1161/
circresaha.107.159517
Conflict of interest The authors declare that they have no conflicts of 16. Sullivan KE, Quinn KP, Tang KM, Georgakoudi I, Black LD 3rd.
interest. (2014) Extracellular matrix remodeling following myocardial in-
farction influences the therapeutic potential of mesenchymal stem
cells. Stem Cell Res Ther 5(1):14. https://doi.org/10.1186/scrt403
References 17. Chen WC, Wang Z, Missinato MA, Park DW, Long DW, Liu HJ
et al (2016) Decellularized zebrafish cardiac extracellular matrix
induces mammalian heart regeneration. Sci Adv 2(11):e1600844.
1. Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K
https://doi.org/10.1126/sciadv.1600844
et al (2017) Reducing the global burden of cardiovascular disease,
18. Mercer SE, Odelberg SJ, Simon HG (2013) A dynamic spatiotem-
part 1: the epidemiology and risk factors. Circ Res 121(6):677–
poral extracellular matrix facilitates epicardial-mediated vertebrate
694. https://doi.org/10.1161/circresaha.117.308903
heart regeneration. Dev Biol 382(2):457–469. https://doi.org/10.
2. Nabel EG, Braunwald E (2012) A tale of coronary artery disease
1016/j.ydbio.2013.08.002
and myocardial infarction. N Engl J Med 366(1):54–63. https://
19. Lindsey ML (2018) Assigning matrix metalloproteinase roles in
doi.org/10.1056/NEJMra1112570
ischaemic cardiac remodelling. Nat Rev Cardiol 15(8):471–479.
3. Xin M, Olson EN, Bassel-Duby R (2013) Mending broken hearts:
https://doi.org/10.1038/s41569-018-0022-z
cardiac development as a basis for adult heart regeneration and
repair. Nat Rev Mol Cell Biol 14(8):529–541. https://doi.org/10. 20. Dobaczewski M, Bujak M, Zymek P, Ren G, Entman ML,
1038/nrm3619 Frangogiannis NG (2006) Extracellular matrix remodeling in ca-
nine and mouse myocardial infarcts. Cell Tissue Res 324(3):475–
4. Bassat E, Mutlak YE, Genzelinakh A, Shadrin IY, Baruch
488. https://doi.org/10.1007/s00441-005-0144-6
Umansky K, Yifa O et al (2017) The extracellular matrix protein
agrin promotes heart regeneration in mice. Nature 547(7662): 21. Li L, Zhao Q, Kong W (2018) Extracellular matrix remodeling
179–184. https://doi.org/10.1038/nature22978 and cardiac fibrosis. Matrix Biol 68-69:490–506. https://doi.org/
5. Eroglu E, Chien KR (2017) Heart regeneration 4.0: Matrix 10.1016/j.matbio.2018.01.013
Medicine. Dev Cell 42(1):7–8. https://doi.org/10.1016/j.devcel. 22. Ren G, Michael LH, Entman ML, Frangogiannis NG (2002)
2017.06.017 Morphological characteristics of the microvasculature in healing
6. Shiro Yui LA, Maimets M, Pedersen MT, Fordham RP, Hansen myocardial infarcts. J Histochem Cytochem 50(1):71–79. https://
SL, Larsen HL, Guiu J, Alves MRP, Rundsten CF, Johansen JV, Li doi.org/10.1177/002215540205000108
Y, Madsen CD, Nakamura T, Watanabe M, Nielsen OH, 23. Trueblood NA, Xie Z, Communal C, Sam F, Ngoy S, Liaw L et al
Schweiger PJ, Piccolo S, Jensen KB (2018) YAP/TAZ- (2001) Exaggerated left ventricular dilation and reduced collagen
dependent reprogramming of colonic epithelium links ECM re- deposition after myocardial infarction in mice lacking osteopontin.
modeling to tissue regeneration. Cell Stem Cell 22(1):35–49 e7 Circ Res 88(10):1080–1087. https://doi.org/10.1161/hh1001.
7. Fry CS, Kirby TJ, Kosmac K, McCarthy JJ, Peterson CA (2017) 090842
Myogenic progenitor cells control extracellular matrix production 24. Schellings MW, Vanhoutte D, Swinnen M, Cleutjens JP, Debets J,
by fibroblasts during skeletal muscle hypertrophy. Cell Stem Cell van Leeuwen RE et al (2009) Absence of SPARC results in in-
20(1):56–59. https://doi.org/10.1016/j.stem.2016.09.010 creased cardiac rupture and dysfunction after acute myocardial
8. Kutys ML, Yamada KM (2014) An extracellular-matrix-specific infarction. J Exp Med 206(1):113–123. https://doi.org/10.1084/
GEF-GAP interaction regulates Rho GTPase crosstalk for 3D col- jem.20081244
lagen migration. Nat Cell Biol 16(9):909–917. https://doi.org/10. 25. Frangogiannis NG, Ren G, Dewald O, Zymek P, Haudek S,
1038/ncb3026 Koerting A et al (2005) Critical role of endogenous
9. Rozario T, DeSimone DW (2010) The extracellular matrix in de- thrombospondin-1 in preventing expansion of healing myocardial
velopment and morphogenesis: a dynamic view. Dev Biol 341(1): infarcts. Circulation. 111(22):2935–2942. https://doi.org/10.1161/
126–140. https://doi.org/10.1016/j.ydbio.2009.10.026 circulationaha.104.510354
10. Frangogiannis NG (2017) The extracellular matrix in myocardial 26. Brockes JP (1997) Amphibian limb regeneration: rebuilding a
injury, repair, and remodeling. J Clin Invest 127(5):1600–1612. complex structure. Science (New York, NY) 276(5309):81–87
https://doi.org/10.1172/jci87491 27. Poss KD, Wilson LG, Keating MT (2002) Heart regeneration in
11. Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen zebrafish. Science (New York, NY) 298(5601):2188–2190.
MJ, Manon-Jensen T et al (2017) The good and the bad collagens https://doi.org/10.1126/science.1077857
of fibrosis - their role in signaling and organ function. Adv Drug 28. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA,
Deliv Rev 121:43–56. https://doi.org/10.1016/j.addr.2017.07.014 Olson EN et al (2011) Transient regenerative potential of the neo-
12. Bashey RI, Martinez-Hernandez A, Jimenez SA (1992) Isolation, natal mouse heart. Science (New York, NY) 331(6020):1078–
characterization, and localization of cardiac collagen type VI. 1080. https://doi.org/10.1126/science.1200708
Associations with other extracellular matrix components. Circ 29. Williams C, Quinn KP, Georgakoudi I, Black LD 3rd. (2014)
Res 70(5):1006–1017 Young developmental age cardiac extracellular matrix promotes
13. Jugdutt BI (2003) Ventricular remodeling after infarction and the the expansion of neonatal cardiomyocytes in vitro. Acta Biomater
extracellular collagen matrix: when is enough enough? 10(1):194–204. https://doi.org/10.1016/j.actbio.2013.08.037
Circulation. 108(11):1395–1403. https://doi.org/10.1161/01.cir. 30. Wang J, Karra R, Dickson AL, Poss KD (2013) Fibronectin is
0000085658.98621.49 deposited by injury-activated epicardial cells and is necessary for
1244 Heart Fail Rev (2021) 26:1231–1248

zebrafish heart regeneration. Dev Biol 382(2):427–435. https:// 47. Christman KL, Lee RJ (2006) Biomaterials for the treatment of
doi.org/10.1016/j.ydbio.2013.08.012 myocardial infarction. J Am Coll Cardiol 48(5):907–913. https://
31. Ieda M, Tsuchihashi T, Ivey KN, Ross RS, Hong TT, Shaw RM doi.org/10.1016/j.jacc.2006.06.005
et al (2009) Cardiac fibroblasts regulate myocardial proliferation 48. Zimmermann WH, Melnychenko I, Eschenhagen T (2004)
through beta1 integrin signaling. Dev Cell 16(2):233–244. https:// Engineered heart tissue for regeneration of diseased hearts.
doi.org/10.1016/j.devcel.2008.12.007 Biomaterials. 25(9):1639–1647
32. Wang WE, Li L, Xia X, Fu W, Liao Q, Lan C et al (2017) 49. Reis LA, Chiu LL, Feric N, Fu L, Radisic M (2016) Biomaterials
Dedifferentiation, proliferation, and redifferentiation of adult in myocardial tissue engineering. J Tissue Eng Regen Med 10(1):
mammalian cardiomyocytes after ischemic injury. Circulation. 11–28. https://doi.org/10.1002/term.1944
136(9):834–848. https://doi.org/10.1161/circulationaha.116. 50. Johnson TD, Braden RL, Christman KL (2014) Injectable ECM
024307 scaffolds for cardiac repair. Methods Mol Biol (Clifton, NJ) 1181:
33. Kuhn B, del Monte F, Hajjar RJ, Chang YS, Lebeche D, Arab S 109–120. https://doi.org/10.1007/978-1-4939-1047-2_10
et al (2007) Periostin induces proliferation of differentiated 51. Duan Y, Liu Z, O'Neill J, Wan LQ, Freytes DO, Vunjak-
cardiomyocytes and promotes cardiac repair. Nat Med 13(8): Novakovic G (2011) Hybrid gel composed of native heart matrix
962–969. https://doi.org/10.1038/nm1619 and collagen induces cardiac differentiation of human embryonic
34. Lorts A, Schwanekamp JA, Elrod JW, Sargent MA, Molkentin JD stem cells without supplemental growth factors. J Cardiovasc
(2009) Genetic manipulation of periostin expression in the heart Transl Res 4(5):605–615. https://doi.org/10.1007/s12265-011-
does not affect myocyte content, cell cycle activity, or cardiac 9304-0
repair. Circ Res 104(1):e1–e7. https://doi.org/10.1161/circresaha. 52. Segers VF, Lee RT (2011) Biomaterials to enhance stem cell func-
108.188649 tion in the heart. Circ Res 109(8):910–922. https://doi.org/10.
35. Chen Z, Xie J, Hao H, Lin H, Wang L, Zhang Y et al (2017) 1161/circresaha.111.249052
Ablation of periostin inhibits post-infarction myocardial regener- 53. Tiburcy M, Hudson JE, Balfanz P, Schlick S, Meyer T, Chang
ation in neonatal mice mediated by the phosphatidylinositol 3 Liao ML et al (2017) Defined engineered human myocardium
kinase/glycogen synthase kinase 3beta/cyclin D1 signalling path- with advanced maturation for applications in heart failure model-
way. Cardiovasc Res 113(6):620–632. https://doi.org/10.1093/ ing and repair. Circulation. 135(19):1832–1847. https://doi.org/
cvr/cvx001 10.1161/circulationaha.116.024145
36. Gupta V, Grande-Allen KJ (2006) Effects of static and cyclic
54. Hirt MN, Boeddinghaus J, Mitchell A, Schaaf S, Bornchen C,
loading in regulating extracellular matrix synthesis by cardiovas-
Muller C et al (2014) Functional improvement and maturation of
cular cells. Cardiovasc Res 72(3):375–383. https://doi.org/10.
rat and human engineered heart tissue by chronic electrical stim-
1016/j.cardiores.2006.08.017
ulation. J Mol Cell Cardiol 74:151–161. https://doi.org/10.1016/j.
37. Yahalom-Ronen Y, Rajchman D, Sarig R, Geiger B, Tzahor E
yjmcc.2014.05.009
(2015) Reduced matrix rigidity promotes neonatal cardiomyocyte
55. Nawroth JC, Scudder LL, Halvorson RT, Tresback J, Ferrier JP,
dedifferentiation, proliferation and clonal expansion. eLife. 4.
Sheehy SP et al (2018) Automated fabrication of photopatterned
https://doi.org/10.7554/eLife.07455
gelatin hydrogels for organ-on-chips applications. Biofabrication.
38. Akhyari P, Fedak PW, Weisel RD, Lee TY, Verma S, Mickle DA
10(2):025004. https://doi.org/10.1088/1758-5090/aa96de
et al (2002) Mechanical stretch regimen enhances the formation of
bioengineered autologous cardiac muscle grafts. Circulation. 56. Shin SR, Jung SM, Zalabany M, Kim K, Zorlutuna P, Kim SB et al
106(12 Suppl 1):I137–I142 (2013) Carbon-nanotube-embedded hydrogel sheets for engineer-
39. Birla RK, Huang YC, Dennis RG (2007) Development of a novel ing cardiac constructs and bioactuators. ACS Nano 7(3):2369–
bioreactor for the mechanical loading of tissue-engineered heart 2380. https://doi.org/10.1021/nn305559j
muscle. Tissue Eng 13(9):2239–2248. https://doi.org/10.1089/ten. 57. Gaetani R, Feyen DA, Verhage V, Slaats R, Messina E, Christman
2006.0359 KL et al (2015) Epicardial application of cardiac progenitor cells
40. Canseco DC, Kimura W, Garg S, Mukherjee S, Bhattacharya S, in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac
Abdisalaam S et al (2015) Human ventricular unloading induces function after myocardial infarction. Biomaterials. 61:339–348.
cardiomyocyte proliferation. J Am Coll Cardiol 65(9):892–900. https://doi.org/10.1016/j.biomaterials.2015.05.005
https://doi.org/10.1016/j.jacc.2014.12.027 58. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren SM, Netoff TI
41. Kong YP, Rioja AY, Xue X, Sun Y, Fu J, Putnam AJ (2018) A et al (2008) Perfusion-decellularized matrix: using nature’s plat-
systems mechanobiology model to predict cardiac reprogramming form to engineer a bioartificial heart. Nat Med 14(2):213–221.
outcomes on different biomaterials. Biomaterials. 181:280–292. https://doi.org/10.1038/nm1684
https://doi.org/10.1016/j.biomaterials.2018.07.036 59. Robertson MJ, Dries-Devlin JL, Kren SM, Burchfield JS, Taylor
42. Vagnozzi RJ, Maillet M, Sargent MA, Khalil H, Johansen AKZ, DA (2014) Optimizing recellularization of whole decellularized
Schwanekamp JA et al (2020) An acute immune response under- heart extracellular matrix. PLoS One 9(2):e90406. https://doi.
lies the benefit of cardiac stem cell therapy. Nature. 577(7790): org/10.1371/journal.pone.0090406
405–409. https://doi.org/10.1038/s41586-019-1802-2 60. Tondreau MY, Laterreur V, Gauvin R, Vallieres K, Bourget JM,
43. Bissell MJ, Aggeler J (1987) Dynamic reciprocity: how do extra- Lacroix D et al (2015) Mechanical properties of endothelialized
cellular matrix and hormones direct gene expression? Prog Clin fibroblast-derived vascular scaffolds stimulated in a bioreactor.
Biol Res 249:251–262 Acta Biomater 18:176–185. https://doi.org/10.1016/j.actbio.
44. Pomeroy JE, Helfer A, Bursac N (2019) Biomaterializing the 2015.02.026
promise of cardiac tissue engineering. Biotechnol Adv. https:// 61. Wang Z, Long DW, Huang Y, Chen WCW, Kim K, Wang Y
doi.org/10.1016/j.biotechadv.2019.02.009 (2019) Decellularized neonatal cardiac extracellular matrix pre-
45. Wang F, Guan J (2010) Cellular cardiomyoplasty and cardiac tis- vents widespread ventricular remodeling in adult mammals after
sue engineering for myocardial therapy. Adv Drug Deliv Rev myocardial infarction. Acta Biomater 87:140–151. https://doi.org/
62(7-8):784–797. https://doi.org/10.1016/j.addr.2010.03.001 10.1016/j.actbio.2019.01.062
46. Radisic M, Christman KL (2013) Materials science and tissue 62. Tang-Quan KR, Mehta NA, Sampaio LC, Taylor DA (2018)
engineering: repairing the heart. Mayo Clin Proc 88(8):884–898. Whole cardiac tissue bioscaffolds. Adv Exp Med Biol 1098:85–
https://doi.org/10.1016/j.mayocp.2013.05.003 114. https://doi.org/10.1007/978-3-319-97421-7_5
Heart Fail Rev (2021) 26:1231–1248 1245

63. Daley MC, Fenn SL, Black LD 3rd. (2018) Applications of car- 78. Frydrych M, Roman S, MacNeil S, Chen B (2015) Biomimetic
diac extracellular matrix in tissue engineering and regenerative poly(glycerol sebacate)/poly(l-lactic acid) blend scaffolds for adi-
medicine. Adv Exp Med Biol 1098:59–83. https://doi.org/10. pose tissue engineering. Acta Biomater 18:40–49. https://doi.org/
1007/978-3-319-97421-7_4 10.1016/j.actbio.2015.03.004
64. Seo Y, Jung Y, Kim SH (2018) Decellularized heart ECM hydro- 79. Neal RA, McClugage SG, Link MC, Sefcik LS, Ogle RC,
gel using supercritical carbon dioxide for improved angiogenesis. Botchwey EA (2009) Laminin nanofiber meshes that mimic mor-
Acta Biomater 67:270–281. https://doi.org/10.1016/j.actbio.2017. phological properties and bioactivity of basement membranes.
11.046 Tissue Eng C Methods 15(1):11–21. https://doi.org/10.1089/ten.
65. Shudo Y, Cohen JE, MacArthur JW, Goldstone AB, Otsuru S, tec.2007.0366
Trubelja A et al (2015) A tissue-engineered chondrocyte cell sheet 80. Migliorini E, Thakar D, Sadir R, Pleiner T, Baleux F, Lortat-Jacob
induces extracellular matrix modification to enhance ventricular H et al (2014) Well-defined biomimetic surfaces to characterize
biomechanics and attenuate myocardial stiffness in ischemic car- glycosaminoglycan-mediated interactions on the molecular, su-
diomyopathy. Tissue Eng A 21(19-20):2515–2525. https://doi. pramolecular and cellular levels. Biomaterials. 35(32):8903–
org/10.1089/ten.TEA.2014.0155 8915. https://doi.org/10.1016/j.biomaterials.2014.07.017
66. Lee KM, Kim H, Nemeno JG, Yang W, Yoon J, Lee S et al (2015) 81. Hematti P (2018) Role of extracellular matrix in cardiac cellular
Natural cardiac extracellular matrix sheet as a biomaterial for car- therapies. Adv Exp Med Biol 1098:173–188. https://doi.org/10.
diomyocyte transplantation. Transplant Proc 47(3):751–756. 1007/978-3-319-97421-7_9
https://doi.org/10.1016/j.transproceed.2014.12.030 82. Singelyn JM, DeQuach JA, Seif-Naraghi SB, Littlefield RB,
67. Ishii M, Shibata R, Shimizu Y, Yamamoto T, Kondo K, Inoue Y Schup-Magoffin PJ, Christman KL (2009) Naturally derived
et al (2014) Multilayered adipose-derived regenerative cell sheets myocardial matrix as an injectable scaffold for cardiac tissue en-
created by a novel magnetite tissue engineering method for myo- gineering. Biomaterials. 30(29):5409–5416. https://doi.org/10.
cardial infarction. Int J Cardiol 175(3):545–553. https://doi.org/ 1016/j.biomaterials.2009.06.045
10.1016/j.ijcard.2014.06.034 83. Wu WQ, Peng S, Song ZY, Lin S (2019) Collagen biomaterial for
68. Yeh YC, Lee WY, Yu CL, Hwang SM, Chung MF, Hsu LW et al the treatment of myocardial infarction: an update on cardiac tissue
(2010) Cardiac repair with injectable cell sheet fragments of hu- engineering and myocardial regeneration. Drug Deliv Transl Res.
man amniotic fluid stem cells in an immune-suppressed rat model. https://doi.org/10.1007/s13346-019-00627-0
Biomaterials. 31(25):6444–6453. https://doi.org/10.1016/j. 84. Roura S, Galvez-Monton C, Bayes-Genis A (2017) Fibrin, the
biomaterials.2010.04.069 preferred scaffold for cell transplantation after myocardial infarc-
69. Wang CC, Chen CH, Lin WW, Hwang SM, Hsieh PC, Lai PH et al tion? An old molecule with a new life. J Tissue Eng Regen Med
(2008) Direct intramyocardial injection of mesenchymal stem cell 11(8):2304–2313. https://doi.org/10.1002/term.2129
sheet fragments improves cardiac functions after infarction. 85. Rufaihah AJ, Seliktar D (2016) Hydrogels for therapeutic cardio-
Cardiovasc Res 77(3):515–524. https://doi.org/10.1093/cvr/ vascular angiogenesis. Adv Drug Deliv Rev 96:31–39. https://doi.
cvm046 org/10.1016/j.addr.2015.07.003
70. Bracaglia LG, Winston S, Powell DA, Fisher JP (2019) Synthetic 86. Arenas-Herrera JE, Ko IK, Atala A, Yoo JJ (2013)
polymer coatings diminish chronic inflammation risk in large Decellularization for whole organ bioengineering. Biomed
ECM-based materials. J Biomed Mater Res A 107(3):494–504. Mater (Bristol, England) 8(1):014106. https://doi.org/10.1088/
https://doi.org/10.1002/jbm.a.36564 1748-6041/8/1/014106
71. Guan J, Wang F, Li Z, Chen J, Guo X, Liao J et al (2011) The 87. Keane TJ, Swinehart IT, Badylak SF (2015) Methods of tissue
stimulation of the cardiac differentiation of mesenchymal stem decellularization used for preparation of biologic scaffolds and
cells in tissue constructs that mimic myocardium structure and in vivo relevance. Methods (San Diego, Calif) 84:25–34. https://
biomechanics. Biomaterials. 32(24):5568–5580. https://doi.org/ doi.org/10.1016/j.ymeth.2015.03.005
10.1016/j.biomaterials.2011.04.038 88. Taylor DA, Sampaio LC, Ferdous Z, Gobin AS, Taite LJ (2018)
72. Loke WK, Khor E, Wee A, Teoh SH, Chian KS (1996) Hybrid Decellularized matrices in regenerative medicine. Acta Biomater
biomaterials based on the interaction of polyurethane oligomers 74:74–89. https://doi.org/10.1016/j.actbio.2018.04.044
with porcine pericardium. Biomaterials. 17(22):2163–2172 89. Spang MT, Christman KL (2018) Extracellular matrix hydrogel
73. Yang MC, Wang SS, Chou NK, Chi NH, Huang YY, Chang YL therapies: in vivo applications and development. Acta Biomater
et al (2009) The cardiomyogenic differentiation of rat mesenchy- 68:1–14. https://doi.org/10.1016/j.actbio.2017.12.019
mal stem cells on silk fibroin-polysaccharide cardiac patches 90. Badylak SF, Taylor D, Uygun K (2011) Whole-organ tissue engi-
in vitro. Biomaterials. 30(22):3757–3765. https://doi.org/10. neering: decellularization and recellularization of three-
1016/j.biomaterials.2009.03.057 dimensional matrix scaffolds. Annu Rev Biomed Eng 13:27–53.
74. Zhang B, Xiao Y, Hsieh A, Thavandiran N, Radisic M (2011) https://doi.org/10.1146/annurev-bioeng-071910-124743
Micro- and nanotechnology in cardiovascular tissue engineering. 91. Bejleri D, Davis ME (2019) Decellularized extracellular matrix
Nanotechnology. 22(49):494003. https://doi.org/10.1088/0957- materials for cardiac repair and regeneration. Adv Healthc Mater
4484/22/49/494003 8(5):e1801217. https://doi.org/10.1002/adhm.201801217
75. Kim TG, Shin H, Lim DW (2012) Biomimetic scaffolds for tissue 92. Schenke-Layland K, Rofail F, Heydarkhan S, Gluck JM, Ingle NP,
engineering. Adv Funct Mater 22(12):2446–2468. https://doi.org/ Angelis E et al (2009) The use of three-dimensional nanostruc-
10.1002/adfm.201103083 tures to instruct cells to produce extracellular matrix for regenera-
76. Boffito M, Di Meglio F, Mozetic P, Giannitelli SM, Carmagnola I, tive medicine strategies. Biomaterials. 30(27):4665–4675. https://
Castaldo C et al (2018) Surface functionalization of polyurethane doi.org/10.1016/j.biomaterials.2009.05.033
scaffolds mimicking the myocardial microenvironment to support 93. Alrefai MT, Murali D, Paul A, Ridwan KM, Connell JM, Shum-
cardiac primitive cells. PLoS One 13(7):e0199896. https://doi.org/ Tim D (2015) Cardiac tissue engineering and regeneration using
10.1371/journal.pone.0199896 cell-based therapy. Stem Cells Cloning 8:81–101. https://doi.org/
77. Kyburz KA, Anseth KS (2015) Synthetic mimics of the extracel- 10.2147/sccaa.s54204
lular matrix: how simple is complex enough? Ann Biomed Eng 94. Sakaguchi K, Shimizu T, Okano T (2015) Construction of three-
43(3):489–500. https://doi.org/10.1007/s10439-015-1297-4 dimensional vascularized cardiac tissue with cell sheet
1246 Heart Fail Rev (2021) 26:1231–1248

engineering. J Control Release 205:83–88. https://doi.org/10. for cardiac repair in a rat infarct model. Tissue Eng A 20(7-8):
1016/j.jconrel.2014.12.016 1325–1335. https://doi.org/10.1089/ten.TEA.2013.0312
95. Cyranoski D (2018) ‘Reprogrammed’ stem cells approved to 111. Gaballa MA, Sunkomat JN, Thai H, Morkin E, Ewy G, Goldman
mend human hearts for the first time. Nature. 557(7707):619– S (2006) Grafting an acellular 3-dimensional collagen scaffold
620. https://doi.org/10.1038/d41586-018-05278-8 onto a non-transmural infarcted myocardium induces neo-
96. Kawamura M, Miyagawa S, Fukushima S, Saito A, Miki K, angiogenesis and reduces cardiac remodeling. J Heart Lung
Funakoshi S et al (2017) Enhanced therapeutic effects of human Transplant 25(8):946–954. https://doi.org/10.1016/j.healun.2006.
iPS cell derived-cardiomyocyte by combined cell-sheets with 04.008
omental flap technique in porcine ischemic cardiomyopathy mod- 112. Serpooshan V, Zhao M, Metzler SA, Wei K, Shah PB, Wang A
el. Sci Rep 7(1):8824. https://doi.org/10.1038/s41598-017-08869- et al (2013) The effect of bioengineered acellular collagen patch on
z cardiac remodeling and ventricular function post myocardial in-
97. Shimizu T, Sekine H, Yamato M, Okano T (2009) Cell sheet- farction. Biomaterials. 34(36):9048–9055. https://doi.org/10.
based myocardial tissue engineering: new hope for damaged heart 1016/j.biomaterials.2013.08.017
rescue. Curr Pharm Des 15(24):2807–2814 113. Deng C, Zhang P, Vulesevic B, Kuraitis D, Li F, Yang AF et al
98. Matsuura K, Utoh R, Nagase K, Okano T (2014) Cell sheet ap- (2010) A collagen-chitosan hydrogel for endothelial differentia-
proach for tissue engineering and regenerative medicine. J Control tion and angiogenesis. Tissue Eng A 16(10):3099–3109. https://
Release 190:228–239. https://doi.org/10.1016/j.jconrel.2014.05. doi.org/10.1089/ten.tea.2009.0504
024 114. Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A,
99. Efraim Y, Sarig H, Cohen Anavy N, Sarig U, de Berardinis E, Peppas NA (2009) Hydrogels in regenerative medicine. Adv
Chaw SY et al (2017) Biohybrid cardiac ECM-based hydrogels Mater (Deerfield Beach, Fla) 21(32-33):3307–3329. https://doi.
improve long term cardiac function post myocardial infarction. org/10.1002/adma.200802106
Acta Biomater 50:220–233. https://doi.org/10.1016/j.actbio. 115. Phelps EA, Enemchukwu NO, Fiore VF, Sy JC, Murthy N,
2016.12.015 Sulchek TA et al (2012) Maleimide cross-linked bioactive PEG
100. Klouda L (2015) Thermoresponsive hydrogels in biomedical ap- hydrogel exhibits improved reaction kinetics and cross-linking for
plications: a seven-year update. Eur J Pharm Biopharm 97(Pt B): cell encapsulation and in situ delivery. Adv Mater (Deerfield
338–349. https://doi.org/10.1016/j.ejpb.2015.05.017 Beach, Fla) 24(1):64–70, 2. https://doi.org/10.1002/adma.
101. Zhu J, Marchant RE (2011) Design properties of hydrogel tissue- 201103574
engineering scaffolds. Expert Rev Med Dev 8(5):607–626. https://
116. Saludas L, Pascual-Gil S, Prosper F, Garbayo E, Blanco-Prieto M
doi.org/10.1586/erd.11.27
(2017) Hydrogel based approaches for cardiac tissue engineering.
102. Ravichandran R, Venugopal JR, Sundarrajan S, Mukherjee S,
Int J Pharm 523(2):454–475. https://doi.org/10.1016/j.ijpharm.
Ramakrishna S (2012) Minimally invasive cell-seeded biomaterial 2016.10.061
systems for injectable/epicardial implantation in ischemic heart
117. Annabi N, Tamayol A, Uquillas JA, Akbari M, Bertassoni LE,
disease. Int J Nanomedicine 7:5969–5994. https://doi.org/10.
Cha C et al (2014) 25th anniversary article: Rational design and
2147/ijn.s37575
applications of hydrogels in regenerative medicine. Adv Mater
103. Lau HK, Kiick KL (2015) Opportunities for multicomponent hy-
(Deerfield Beach, Fla) 26(1):85–123
brid hydrogels in biomedical applications. Biomacromolecules.
118. Shi K, Wang YL, Qu Y, Liao JF, Chu BY, Zhang HP et al (2016)
16(1):28–42. https://doi.org/10.1021/bm501361c
Synthesis, characterization, and application of reversible PDLLA-
104. Rufaihah AJ, Vaibavi SR, Plotkin M, Shen J, Nithya V, Wang J
PEG-PDLLA copolymer thermogels in vitro and in vivo. Sci Rep
et al (2013) Enhanced infarct stabilization and neovascularization
6:19077. https://doi.org/10.1038/srep19077
mediated by VEGF-loaded PEGylated fibrinogen hydrogel in a
rodent myocardial infarction model. Biomaterials. 34(33):8195– 119. Vo TN, Ekenseair AK, Spicer PP, Watson BM, Tzouanas SN, Roh
8202. https://doi.org/10.1016/j.biomaterials.2013.07.031 TT et al (2015) In vitro and in vivo evaluation of self-
105. Toba H, Lindsey ML (2019) Extracellular matrix roles in mineralization and biocompatibility of injectable, dual-gelling
cardiorenal fibrosis: potential therapeutic targets for CVD and hydrogels for bone tissue engineering. J Control Release 205:
CKD in the elderly. Pharmacol Ther 193:99–120. https://doi.org/ 25–34. https://doi.org/10.1016/j.jconrel.2014.11.028
10.1016/j.pharmthera.2018.08.014 120. Barnes AL, Genever PG, Rimmer S, Coles MC (2016) Collagen-
106. Yanamandala M, Zhu W, Garry DJ, Kamp TJ, Hare JM, Jun HW poly(N-isopropylacrylamide) hydrogels with tunable properties.
et al (2017) Overcoming the roadblocks to cardiac cell therapy Biomacromolecules. 17(3):723–734. https://doi.org/10.1021/acs.
using tissue engineering. J Am Coll Cardiol 70(6):766–775. biomac.5b01251
https://doi.org/10.1016/j.jacc.2017.06.012 121. Das D, Ghosh P, Ghosh A, Haldar C, Dhara S, Panda AB et al
107. Zhu Y, Matsumura Y, Wagner WR (2017) Ventricular wall bioma- (2015) Stimulus-responsive, biodegradable, biocompatible, cova-
terial injection therapy after myocardial infarction: advances in lently cross-linked hydrogel based on dextrin and poly(N-
material design, mechanistic insight and early clinical experiences. isopropylacrylamide) for in vitro/in vivo controlled drug release.
Biomaterials. 129:37–53. https://doi.org/10.1016/j.biomaterials. ACS Appl Mater Interfaces 7(26):14338–14351. https://doi.org/
2017.02.032 10.1021/acsami.5b02975
108. Chien KR, Frisen J, Fritsche-Danielson R, Melton DA, Murry CE, 122. Chen FM, Liu X (2016) Advancing biomaterials of human origin
Weissman IL (2019) Regenerating the field of cardiovascular cell for tissue engineering. Prog Polym Sci 53:86–168. https://doi.org/
therapy. Nat Biotechnol 37(3):232–237. https://doi.org/10.1038/ 10.1016/j.progpolymsci.2015.02.004
s41587-019-0042-1 123. Hastings CL, Roche ET, Ruiz-Hernandez E, Schenke-Layland K,
109. Kai D, Wang QL, Wang HJ, Prabhakaran MP, Zhang Y, Tan YZ Walsh CJ, Duffy GP (2015) Drug and cell delivery for cardiac
et al (2014) Stem cell-loaded nanofibrous patch promotes the re- regeneration. Adv Drug Deliv Rev 84:85–106. https://doi.org/10.
generation of infarcted myocardium with functional improvement 1016/j.addr.2014.08.006
in rat model. Acta Biomater 10(6):2727–2738. https://doi.org/10. 124. Wang H, Zhou J, Liu Z, Wang C (2010) Injectable cardiac tissue
1016/j.actbio.2014.02.030 engineering for the treatment of myocardial infarction. J Cell Mol
110. Wendel JS, Ye L, Zhang P, Tranquillo RT, Zhang JJ (2014) Med 14(5):1044–1055. https://doi.org/10.1111/j.1582-4934.2010.
Functional consequences of a tissue-engineered myocardial patch 01046.x
Heart Fail Rev (2021) 26:1231–1248 1247

125. Ungerleider JL, Johnson TD, Rao N, Christman KL (2015) heart muscle. J Thorac Cardiovasc Surg 128(4):571–578. https://
Fabrication and characterization of injectable hydrogels derived doi.org/10.1016/j.jtcvs.2004.05.021
from decellularized skeletal and cardiac muscle. Methods (San 139. Iyisoy A, Ozturk C, Celik T, Demirkol S, Cingoz F, Unlu M et al
Diego, Calif) 84:53–59. https://doi.org/10.1016/j.ymeth.2015.03. (2015) Percutaneous transapical closure of cardiac apex with an
024 ADO-II device after successful transapical transcatheter prosthetic
126. Shu Y, Hao T, Yao F, Qian Y, Wang Y, Yang B et al (2015) RoY mitral paravalvular leak closure. Int J Cardiol 189:289–292.
peptide-modified chitosan-based hydrogel to improve angiogene- https://doi.org/10.1016/j.ijcard.2015.04.076
sis and cardiac repair under hypoxia. ACS Appl Mater Interfaces 140. Wu KH, Liu YL, Zhou B, Han ZC (2006) Cellular therapy and
7(12):6505–6517. https://doi.org/10.1021/acsami.5b01234 myocardial tissue engineering: the role of adult stem and progen-
127. Singelyn JM, Sundaramurthy P, Johnson TD, Schup-Magoffin PJ, itor cells. Eur J Cardiothorac Surg 30(5):770–781. https://doi.org/
Hu DP, Faulk DM et al (2012) Catheter-deliverable hydrogel de- 10.1016/j.ejcts.2006.08.003
rived from decellularized ventricular extracellular matrix increases 141. Aamodt JM, Grainger DW (2016) Extracellular matrix-based bio-
endogenous cardiomyocytes and preserves cardiac function post- material scaffolds and the host response. Biomaterials. 86:68–82.
myocardial infarction. J Am Coll Cardiol 59(8):751–763. https:// https://doi.org/10.1016/j.biomaterials.2016.02.003
doi.org/10.1016/j.jacc.2011.10.888 142. Alvarez MM, Liu JC, Trujillo-de Santiago G, Cha BH,
128. Sawa Y, Miyagawa S, Sakaguchi T, Fujita T, Matsuyama A, Saito Vishwakarma A, Ghaemmaghami AM et al (2016) Delivery strat-
A et al (2012) Tissue engineered myoblast sheets improved cardi- egies to control inflammatory response: modulating M1-M2 po-
ac function sufficiently to discontinue LVAS in a patient with larization in tissue engineering applications. J Control Release
DCM: report of a case. Surg Today 42(2):181–184. https://doi. 240:349–363. https://doi.org/10.1016/j.jconrel.2016.01.026
org/10.1007/s00595-011-0106-4 143. Du C, Cui FZ, Zhu XD, de Groot K (1999) Three-dimensional
129. Nishida K, Yamato M, Hayashida Y, Watanabe K, Yamamoto K, nano-HAp/collagen matrix loading with osteogenic cells in organ
Adachi E et al (2004) Corneal reconstruction with tissue- culture. J Biomed Mater Res 44(4):407–415
engineered cell sheets composed of autologous oral mucosal epi- 144. Badylak SF (2007) The extracellular matrix as a biologic scaffold
thelium. N Engl J Med 351(12):1187–1196. https://doi.org/10. material. Biomaterials. 28(25):3587–3593. https://doi.org/10.
1056/NEJMoa040455 1016/j.biomaterials.2007.04.043
130. Sato M, Yamato M, Hamahashi K, Okano T, Mochida J (2014) 145. Bouten CV, Dankers PY, Driessen-Mol A, Pedron S, Brizard AM,
Articular cartilage regeneration using cell sheet technology. Anat Baaijens FP (2011) Substrates for cardiovascular tissue engineer-
Rec (Hoboken, NJ : 2007) 297(1):36–43. https://doi.org/10.1002/ ing. Adv Drug Deliv Rev 63(4-5):221–241. https://doi.org/10.
ar.22829 1016/j.addr.2011.01.007
131. Ohki T, Yamato M, Murakami D, Takagi R, Yang J, Namiki H et al 146. Bracaglia LG, Fisher JP (2015) Extracellular matrix-based
(2006) Treatment of oesophageal ulcerations using endoscopic biohybrid materials for engineering compliant, matrix-dense tis-
transplantation of tissue-engineered autologous oral mucosal epi- sues. Adv Healthc Mater 4(16):2475–2487. https://doi.org/10.
thelial cell sheets in a canine model. Gut. 55(12):1704–1710. 1002/adhm.201500236
https://doi.org/10.1136/gut.2005.088518 147. Mathur A, Ma Z, Loskill P, Jeeawoody S, Healy KE (2016)
132. Itabashi Y, Miyoshi S, Yuasa S, Fujita J, Shimizu T, Okano T et al In vitro cardiac tissue models: current status and future prospects.
(2005) Analysis of the electrophysiological properties and arrhyth- Adv Drug Deliv Rev 96:203–213. https://doi.org/10.1016/j.addr.
mias in directly contacted skeletal and cardiac muscle cell sheets. 2015.09.011
Cardiovasc Res 67(3):561–570. https://doi.org/10.1016/j. 148. Leor J, Amsalem Y, Cohen S (2005) Cells, scaffolds, and mole-
cardiores.2005.03.014 cules for myocardial tissue engineering. Pharmacol Ther 105(2):
133. Itabashi Y, Miyoshi S, Kawaguchi H, Yuasa S, Tanimoto K, 151–163. https://doi.org/10.1016/j.pharmthera.2004.10.003
Furuta A et al (2005) A new method for manufacturing cardiac 149. Berglund JD, Galis ZS (2003) Designer blood vessels and thera-
cell sheets using fibrin-coated dishes and its electrophysiological peutic revascularization. Br J Pharmacol 140(4):627–636. https://
studies by optical mapping. Artif Organs 29(2):95–103. https:// doi.org/10.1038/sj.bjp.0705457
doi.org/10.1111/j.1525-1594.2005.29020.x 150. Tedder ME, Liao J, Weed B, Stabler C, Zhang H, Simionescu A
134. Matsuura K, Wada M, Shimizu T, Haraguchi Y, Sato F, Sugiyama et al (2009) Stabilized collagen scaffolds for heart valve tissue
K et al (2012) Creation of human cardiac cell sheets using plurip- engineering. Tissue Eng A 15(6):1257–1268. https://doi.org/10.
otent stem cells. Biochem Biophys Res Commun 425(2):321– 1089/ten.tea.2008.0263
327. https://doi.org/10.1016/j.bbrc.2012.07.089 151. Arts T, Bovendeerd PH, Prinzen FW, Reneman RS (1991)
135. Lee P, Klos M, Bollensdorff C, Hou L, Ewart P, Kamp TJ et al Relation between left ventricular cavity pressure and volume
(2012) Simultaneous voltage and calcium mapping of genetically and systolic fiber stress and strain in the wall. Biophys J 59(1):
purified human induced pluripotent stem cell-derived cardiac 93–102. https://doi.org/10.1016/s0006-3495(91)82201-9
myocyte monolayers. Circ Res 110(12):1556–1563. https://doi. 152. Mol A, van Lieshout MI, Dam-de Veen CG, Neuenschwander S,
org/10.1161/circresaha.111.262535 Hoerstrup SP, Baaijens FP et al (2005) Fibrin as a cell carrier in
136. Fujita J, Itabashi Y, Seki T, Tohyama S, Tamura Y, Sano M et al cardiovascular tissue engineering applications. Biomaterials.
(2012) Myocardial cell sheet therapy and cardiac function. Am J 26(16):3113–3121. https://doi.org/10.1016/j.biomaterials.2004.
Physiol Heart Circ Physiol 303(10):H1169–H1182. https://doi. 08.007
org/10.1152/ajpheart.00376.2012 153. Langer R, Vacanti JP (1993) Tissue engineering. Science.
137. Galaup A, Gomez E, Souktani R, Durand M, Cazes A, Monnot C 260(5110):920–926
et al (2012) Protection against myocardial infarction and no- 154. Fuchs JR, Nasseri BA, Vacanti JP (2001) Tissue engineering: a
reflow through preservation of vascular integrity by 21st century solution to surgical reconstruction. Ann Thorac Surg
angiopoietin-like 4. Circulation. 125(1):140–149. https://doi.org/ 72(2):577–591
10.1161/circulationaha.111.049072 155. Ishii O, Shin M, Sueda T, Vacanti JP (2005) In vitro tissue engi-
138. Kofidis T, de Bruin JL, Hoyt G, Lebl DR, Tanaka M, Yamane T neering of a cardiac graft using a degradable scaffold with an
et al (2004) Injectable bioartificial myocardial tissue for large- extracellular matrix-like topography. J Thorac Cardiovasc Surg
scale intramural cell transfer and functional recovery of injured 130(5):1358–1363. https://doi.org/10.1016/j.jtcvs.2005.05.048
1248 Heart Fail Rev (2021) 26:1231–1248

156. Freytes DO, Wan LQ, Vunjak-Novakovic G (2009) Geometry and to evaluate the safety and effectiveness of intracoronary applica-
force control of cell function. J Cell Biochem 108(5):1047–1058. tion of a novel bioabsorbable cardiac matrix for the prevention of
https://doi.org/10.1002/jcb.22355 ventricular remodeling after large ST-segment elevation myocar-
157. Adams JC, Watt FM (1993) Regulation of development and dif- dial infarction: Rationale and design of the PRESERVATION I
ferentiation by the extracellular matrix. Development (Cambridge, trial. Am Heart J 170(5):929–937. https://doi.org/10.1016/j.ahj.
England) 117(4):1183–1198 2015.08.017
158. Brown RE, Butler JP, Rogers RA, Leith DE (1994) Mechanical 172. Rao SV, Zeymer U, Douglas PS, Al-Khalidi H, White JA, Liu J
connections between elastin and collagen. Connect Tissue Res et al (2016) Bioabsorbable intracoronary matrix for prevention of
30(4):295–308 ventricular remodeling after myocardial infarction. J Am Coll
159. Duffy GP, McFadden TM, Byrne EM, Gill SL, Farrell E, O'Brien Cardiol 68(7):715–723. https://doi.org/10.1016/j.jacc.2016.05.
FJ (2011) Towards in vitro vascularisation of collagen-GAG scaf- 053
folds. Eur Cells Mater 21:15–30 173. Mann DL, Lee RJ, Coats AJ, Neagoe G, Dragomir D, Pusineri E
160. Radisic M, Park H, Shing H, Consi T, Schoen FJ, Langer R et al et al (2016) One-year follow-up results from AUGMENT-HF: a
(2004) Functional assembly of engineered myocardium by elec- multicentre randomized controlled clinical trial of the efficacy of
trical stimulation of cardiac myocytes cultured on scaffolds. Proc left ventricular augmentation with Algisyl in the treatment of heart
Natl Acad Sci U S A 101(52):18129–18134. https://doi.org/10. failure. Eur J Heart Fail 18(3):314–325. https://doi.org/10.1002/
1073/pnas.0407817101 ejhf.449
161. Wei HJ, Chen CH, Lee WY, Chiu I, Hwang SM, Lin WW et al 174. Mewhort HE, Turnbull JD, Satriano A, Chow K, Flewitt JA,
(2008) Bioengineered cardiac patch constructed from multilayered Andrei AC et al (2016) Epicardial infarct repair with bioinductive
mesenchymal stem cells for myocardial repair. Biomaterials. extracellular matrix promotes vasculogenesis and myocardial re-
29(26):3547–3556. https://doi.org/10.1016/j.biomaterials.2008. covery. J Heart Lung Transplant 35(5):661–670. https://doi.org/
05.009 10.1016/j.healun.2016.01.012
162. Huebsch N, Mooney DJ (2009) Inspiration and application in the 175. Mewhort HE, Turnbull JD, Meijndert HC, Ngu JM, Fedak PW
evolution of biomaterials. Nature. 462(7272):426–432. https://doi. (2014) Epicardial infarct repair with basic fibroblast growth factor-
org/10.1038/nature08601 enhanced CorMatrix-ECM biomaterial attenuates postischemic
163. Williams DF (2009) On the nature of biomaterials. Biomaterials. cardiac remodeling. J Thorac Cardiovasc Surg 147(5):1650–
30(30):5897–5909. https://doi.org/10.1016/j.biomaterials.2009. 1659. https://doi.org/10.1016/j.jtcvs.2013.08.005
07.027 176. Menasche P, Vanneaux V, Hagege A, Bel A, Cholley B,
164. Wainwright JM, Czajka CA, Patel UB, Freytes DO, Tobita K, Parouchev A et al (2018) Transplantation of human embryonic
Gilbert TW et al (2010) Preparation of cardiac extracellular matrix stem cell-derived cardiovascular progenitors for severe ischemic
from an intact porcine heart. Tissue Eng C Methods 16(3):525– left ventricular dysfunction. J Am Coll Cardiol 71(4):429–438.
532. https://doi.org/10.1089/ten.TEC.2009.0392 https://doi.org/10.1016/j.jacc.2017.11.047
165. Ismagilov RF, Maharbiz MM (2007) Can we build synthetic, mul- 177. Sahoo S, Losordo DW (2014) Exosomes and cardiac repair after
ticellular systems by controlling developmental signaling in space myocardial infarction. Circ Res 114(2):333–344. https://doi.org/
and time? Curr Opin Chem Biol 11(6):604–611. https://doi.org/ 10.1161/circresaha.114.300639
10.1016/j.cbpa.2007.10.003
178. Liu B, Lee BW, Nakanishi K, Villasante A, Williamson R, Metz J
166. Leor J, Tuvia S, Guetta V, Manczur F, Castel D, Willenz U et al
et al (2018) Cardiac recovery via extended cell-free delivery of
(2009) Intracoronary injection of in situ forming alginate hydrogel
extracellular vesicles secreted by cardiomyocytes derived from
reverses left ventricular remodeling after myocardial infarction in
induced pluripotent stem cells. Nat Biomed Eng 2(5):293–303.
Swine. J Am Coll Cardiol 54(11):1014–1023. https://doi.org/10.
https://doi.org/10.1038/s41551-018-0229-7
1016/j.jacc.2009.06.010
179. Purcell BP, Lobb D, Charati MB, Dorsey SM, Wade RJ, Zellars
167. Martens TP, Godier AF, Parks JJ, Wan LQ, Koeckert MS, Eng
KN et al (2014) Injectable and bioresponsive hydrogels for on-
GM et al (2009) Percutaneous cell delivery into the heart using
demand matrix metalloproteinase inhibition. Nat Mater 13(6):
hydrogels polymerizing in situ. Cell Transplant 18(3):297–304.
653–661. https://doi.org/10.1038/nmat3922
https://doi.org/10.3727/096368909788534915
180. Wang LL, Liu Y, Chung JJ, Wang T, Gaffey AC, Lu M et al (2017)
168. Rodell CB, Lee ME, Wang H, Takebayashi S, Takayama T,
Local and sustained miRNA delivery from an injectable hydrogel
Kawamura T et al (2016) Injectable shear-thinning hydrogels for
promotes cardiomyocyte proliferation and functional regeneration
minimally invasive delivery to infarcted myocardium to limit left
ventricular remodeling. Circ Cardiovasc Interv 9(10). https://doi. after ischemic injury. Nat Biomed Eng 1:983–992. https://doi.org/
10.1038/s41551-017-0157-y
org/10.1161/circinterventions.116.004058
169. Johnson TD, Christman KL (2013) Injectable hydrogel therapies 181. Fan Z, Xu Z, Niu H, Gao N, Guan Y, Li C et al (2018) An
and their delivery strategies for treating myocardial infarction. injectable oxygen release system to augment cell survival and
Expert Opin Drug Deliv 10(1):59–72. https://doi.org/10.1517/ promote cardiac repair following myocardial infarction. Sci Rep
17425247.2013.739156 8(1):1371. https://doi.org/10.1038/s41598-018-19906-w
170. Dib N, Campbell A, Jacoby DB, Zawadzka A, Ratliff J, 182. Tang J, Shen D, Caranasos TG, Wang Z, Vandergriff AC, Allen
Miedzybrocki BM et al (2006) Safety and feasibility of percuta- TA et al (2017) Therapeutic microparticles functionalized with
neous autologous skeletal myoblast transplantation in the coil- biomimetic cardiac stem cell membranes and secretome. Nat
infarcted swine myocardium. J Pharmacol Toxicol Methods Commun 8:13724. https://doi.org/10.1038/ncomms13724
54(1):71–77. https://doi.org/10.1016/j.vascn.2005.12.002
171. Rao SV, Zeymer U, Douglas PS, Al-Khalidi H, Liu J, Gibson CM Publisher’s note Springer Nature remains neutral with regard to jurisdic-
et al (2015) A randomized, double-blind, placebo-controlled trial tional claims in published maps and institutional affiliations.

You might also like