Вы находитесь на странице: 1из 13

Biomedical Materials

SPECIAL SECTION TOPICAL REVIEW Related content


- Fabrication of arbitrary 3D components in
Current progress in 3D printing for cardiovascular cardiac surgery: from macro-, micro- to
nanoscale
tissue engineering Ranjith Kumar Kankala, Kai Zhu, Jun Li et
al.

- Research and development of 3D printed


To cite this article: Bobak Mosadegh et al 2015 Biomed. Mater. 10 034002 vasculature constructs
Xinda Li, Libiao Liu, Xinzhi Zhang et al.

- Integrating three-dimensional printing and


nanotechnology for musculoskeletal
regeneration
View the article online for updates and enhancements. Margaret Nowicki, Nathan J Castro, Raj
Rao et al.

Recent citations
- Benjamin W. Streeter and Michael E.
Davis

- Highly stretchable sensors for wearable


biomedical applications
Qinwu Gao et al

- Janarthanan Gopinathan and Insup Noh

This content was downloaded from IP address 202.170.60.252 on 16/02/2019 at 11:04


Biomed. Mater. 10 (2015) 034002 doi:10.1088/1748-6041/10/3/034002

Special SECTION TOPICAL REVIEW

Current progress in 3D printing for cardiovascular tissue


engineering
received
16 December 2014
re vised
28 January 2015
accep ted for publication
Bobak Mosadegh, Guanglei Xiong, Simon Dunham and James K Min
9 February 2015 Dalio Institute of Cardiovascular Imaging, Department of Radiology, Weill Cornell Medical College, New York, NY 10021, USA
published Email: bom2008@med.cornell.edu
16 March 2015
Keywords: 3D printing, bioprinting, cardiovascular, tissue engineering, cardiology

Abstract
3D printing is a technology that allows the fabrication of structures with arbitrary geometries and
heterogeneous material properties. The application of this technology to biological structures that
match the complexity of native tissue is of great interest to researchers. This mini-review highlights
the current progress of 3D printing for fabricating artificial tissues of the cardiovascular system,
specifically the myocardium, heart valves, and coronary arteries. In addition, how 3D printed sensors
and actuators can play a role in tissue engineering is discussed. To date, all the work with building 3D
cardiac tissues have been proof-of-principle demonstrations, and in most cases, yielded products less
effective than other traditional tissue engineering strategies. However, this technology is in its infancy
and therefore there is much promise that through collaboration between biologists, engineers and
material scientists, 3D bioprinting can make a significant impact on the field of cardiovascular tissue
engineering.

1. Introduction a­ ccommodate biopolymers, which are typically used


for providing the scaffolding and/or ECM for tissue
Tissue engineering comprises the optimization of three engineering applications: (i) selective laser sintering
primary components: (i) the type or types of cells be- (SLS) [5], (ii) stereolithography (SLA) [6], (iii) fused
ing implanted (e.g. somatic, embryonic stem cells, adult deposition modeling (FDM) [7], and (iv) pressure-
stem cells, or induced-pluripotent stem cells) [1], (ii) based extrusion (PBE) [8, 9].
type of scaffolds supporting the cells (i.e. the mechani- SLS uses a CO2 laser to locally raise the tempera-
cal cues provided to the cells), and (iii) type of drugs, ture of a thin layer of powder so the particles fuse to
extra-cellular matrix (ECM), and growth factors con- form a solid object (figure 1(a)). SLA uses UV or vis-
ditioning the cells, (i.e. the chemical cues provided to ible light to polymerize a thin layer of a solution con-
the cells). In addition, the conditions (e.g. fluid flow, taining a photocrosslinkable resin (figure 1(b)). FDM
oxygenation, temperature) in which the construct is melts and extrudes a polymer through a nozzle onto a
cultured can have a significant impact on its matura- flat substrate to build up a 3D structure (figure 1(c)).
tion, making the development of novel bioreactors a PBE uses a differential pressure, generated by a pres-
major part of tissue engineering [2]. Typically these sure reservoir upstream or a syringe pump, to drive the
constructs are fabricated using manual procedures material through a nozzle (figure 1(c)). While SLS and
and are therefore limited in the complexity by which SLA are typically faster than FDM and PBE, they both
materials of varying properties and dimensions can be require lasers and optics. SLS is ideal for materials such
interfaced. 3D printing provides a means to meet this as ceramics or metals that might require higher tem-
technological challenge by automating the fabrication peratures, but the temperatures required for printing
process of these materials. 3D printing is an additive can be prohibitive. SLA can be both fast and straight-
manufacturing technique that builds structures, based forward, however materials must be available in photo-
on a computer-aided design (CAD), by depositing curable chemistries (typically plastics). Both FDM and
material in a layer-by-layer manner. There are several PBE can be much simpler systems, However, they can be
methods to accomplish the layer-by-layer fabrication quite slow compared to other methods. FDM is limited
depending on the type of material printed [3, 4]. There to thermally extruded materials (typically thermoplas-
are four methods of 3D printing that most readily tics), but can be simple, high resolution and produce

© 2015 IOP Publishing Ltd


Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

tissues with limited cellular involvement (e.g. bone)


[14, 15]. This outcome is reasonable since 3D printing
is inherently a technique developed for structural fab-
rication of rigid materials. Furthermore, the complex
cues needed to facilitate desired cellular phenotypes
are still not well understood. The promise of 3D print-
ing, however, is that once those cues are discovered, this
technology will enable such a microenvironment to
be built. Already, 3D printing has been shown to build
structures at both the nano-scale [16] and macro-scale
[17]. There currently, however, is not a single system
that can build artificial tissues that span the full range
of hierarchical structures observed in vivo. For example,
muscle comprises protein filaments, myofibrils, muscle
fibers, and fascicles, all of which are well organized and
span several orders of magnitude in length. To bridge
this gap, novel methods of 3D printing and new classes
of bioinks are being developed [3, 4, 18, 19]. In general,
bioinks require three characteristics to be useable: (i)
biocompatible (i.e. will not induce harm to the host
after implantation), (ii) non-cytotoxic (i.e. will not kill
or damage cells during printing due to chemical, ther-
mal, or mechanical issues) (iii) printable (i.e. has the
proper rheological properties to be extruded or assem-
bled effectively during printing), and (iv) robust (i.e.
Figure 1.  Schematic illustration of various modes of 3D able to withstand the physical forces of the environment
printing: (a) SLS: focused CO2 laser heats loose powder of into which it is applied) [3, 4].
print material to sinter into solid traces, (b) SLA: focused UV
According to the Centers for Disease Control and
light induces chemical change from liquid to solid in bath
of precursor liquid, (c) extrusion methods: both heating of Prevention (CDC) and World Health Organization,
molten solids extruded via FDM or pressure based extrusion cardiovascular disease (CVD) is the number one cause
of viscous liquids. of death for both men and women, and is responsible
for killing nearly 30% of people world-wide [20, 21].
dense parts. Finally, PBE offers the advantage that most CVD is a broad term that refers to disorders of the
materials with appropriate viscocities for printing can heart and blood vessels, including coronary heart dis-
be achieved including hydrogels and direct printing of ease, peripheral arterial disease, rheumatic heart dis-
cells, however, like FDM, it can be quite slow. We refer ease, heart failure, arrhythmia, myocardial infarction,
readers to other reviews regarding which 3D printing and even stroke [22]. Often these diseases are linked
methods are useful for different biopolymers [8, 10]. to atherosclerosis, a disease in which a build-up of
Here, we focus on the use of these techniques specifi- plaque causes inflammation, but other congestive and
cally for 3D printing for cardiac tissue engineering. congenital diseases exist as well [22]. Medications can
For tissue engineering applications, 3D printing be effective if administered early, but if the disease
serves two main purposes: (i) scaffolds of complex progresses, surgical intervention that aims to repair or
geometries are directly fabricated using biocompat- replace the damaged tissue is often required. Due to a
ible materials, which may or may not be seeded with lack of organ donors and adequate synthetic solutions,
cells, and (ii) artificial tissues are directly fabricated tissue-engineering approaches are being developed.
with cells encapsulated within the ink, or ‘bioink’, dur- This review focuses on the use of 3D printing to fab-
ing the printing process [10, 11]. The first purpose of ricate tissue-engineered cardiac tissue, but we refer the
3D printing uses routine methods for industrial appli- reader to the following reviews that survey cardiac tissue
cations, thus allowing the use of commercial printers engineering using other fabrication methods [23–28].
without any modifications. The second purpose of 3D For cardiovascular applications, 3D printing has
printing for tissue engineering, however, requires the already been used in a clinical setting to visualize ana-
use of materials not currently used by most industrial tomical structures for surgeons [29–31]. For this rea-
users, and therefore custom printers are typically built. son, the NIH has already begun a public resource for
Two commercially available bioprinters and services the exchange of CAD files, medical images, tutorials
are the 3D-Bioplotter from envision TEC [12] (http:// and software [32]. These surgical models are made of
envisiontec.com/3d-printers/3d-bioplotter/) and the solid plastic that are not directly useful for tissue engi-
NovoGen MMX Bioprinter from Organovo [13]. The neering purposes. Bioinks, which typically comprise of
impact of 3D printing on the field of tissue engineering cells suspended in hydrogels that serve as the ECM, are
has been most prevalent in applications involving hard beginning to be developed and printed directly to form

2
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

tissues that can be implanted for cardiovascular tissue


engineering [8, 10, 17, 33–35], specifically in the form
of myocardial tissue, heart valves, and coronary arteries.
Furthermore, 3D printing has the ability to integrate
electronics into tissue-engineered constructs to provide
additional functionality, such as sensing and actuation.
Although currently there is no clinical impact of 3D
printed tissues, as techniques are refined and the tech-
nology improved in resolution and scale, this technol-
ogy should yield an unprecedented ability to integrate
artificial tissues with native cardiac tissue seamlessly
with respect to geometry, mechanical properties, elec-
trical properties, and ability to remodel.

2.  3D printing for myocardial tissue

When heart tissue undergoes damage, the heart pumps


blood inefficiently due to the loss of contractile muscle
tissue (primarily cardiomyocytes) and the formation
of stiff scar tissue (due to activated fibroblasts) [36].
The resulting decrease in cardiac output often results
in ischemia that can lead to death [37]. One approach Figure 2.  SLS fabricated scaffolds. (a) CAD drawing of the
unit cell and final geometry of the scaffold made from PCL.
to repair the heart is to implant cells at the site of the (b) SEM image (scale bar: 100 µm) and optical image of the
damaged tissue, a treatment option referred to as cel- PCL scaffold (scale bar: 4 mm). (c) Merged fluorescence
lular therapy [1]. The effectiveness of cellular therapy image of DAPI (blue) and F-actin (red) expression of C2C12
cells 6 d after seeding in the PCL scaffold (scale bar: 100 µm).
depends on the ability of the cells to survive the im-
plantation and properly integrate into the heart tissue
in a manner that increases cardiac output. Due to the ­ etabolic requirements of tissues at those high densi-
m
demanding constraints of the heart, cell therapy alone ties, and therefore will result in significant cell death,
has not been able to cause sufficient regeneration, and and (ii) cardiomyocytes do not proliferate, so the use of
therefore techniques of tissue engineering are being stem cells is needed if sufficient densities of cardiomyo-
explored [37]. There are many combinations of strate- cytes are not seeded initially.
gies being explored for cardiac tissue engineering, and 3D printing holds promise to overcome the above
we refer the readers to the following reviews [36–40]. limitation by enabling the direct fabrication of thick
One of the main limitations to survival of the 3D constructs of a defined geometry of arbitrary com-
implanted cells is the immediate availability of oxy- plexity, with uniformly dispersed cells. For example,
gen [41]. During in vitro culture, the effective trans- Yeong et al used the additive manufacturing technique
port of oxygen to cells is facilitated by either, (i) limit- of SLS to produce structures that were porous on both
ing the thickness of the construct of cells to thin slices the micron and millimeter scale, ensuring efficient
(<200 µm) [38], and/or, (ii) integrating open pores or mass transport throughout the structure (figure 2).
channels to allow oxygen to more easily reach the mid- SLS uses a CO2 laser to sinter a specific pattern of a thin
dle regions of the construct either by diffusion or con- layer of power, based on a CAD model, and similar to
vective mass transport. The disadvantage of limiting other 3D printing methods, builds up the structure
the thickness of the construct in vitro, is that multiple layer-by-layer. The power used in this study was poly-
constructs will need to be implanted in vivo, a process caprolactone (PCL), which yields sintered structures
that requires repeated surgeries to obtain useful clini- with ~48% porosity, ~34 µm surface roughness, a ten-
cal outcomes since proper vascularization by the host sile stiffness of ~0.43 MPa, and a yield strain of ~89%.
takes days [42]. Currently, strategies to build thicker Viable C2C12 cells were maintained for three weeks in
multi-layered tissues are being pursued by integrating culture and formed multi-nucleated myotubes after
a vascular system prior to implantation. So far, only tis- 11 d. The authors did not conduct any further studies
sues on the order of 100 µm thick have been produced, to characterize the performance of this construct for
and still suffer from cell death [43]. The second method therapeutic use. This method of additive manufactur-
of integrating pores or channels into the scaffold allows ing also prevents the direct printing of cells due to the
larger constructs to be built in vitro, but currently all high temperatures needed to sinter the power.
methods use cellular densities at least 100 ×  less than Gaetani et al used 3D bioprinting (using pressure-
that of in vivo tissue, which yields limited improve- based extrusion) to create a construct containing
ment in heart function [44]. Simply increasing the cell human cardiac-derived cardiomyocyte progenitor
density, however, is not trivial since (i) current meth- cells (hCMPCs) and RGD-modified sodium alginate
ods to provide proper vascularization have not met the as the ECM (figure 4) [45]. This construct was shown to

3
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

maintain the viability of the hCMPCs in vitro, promote


the differentiation of hCMPCs into cardiomyocyte-like
cells, and allow the migration of the hCMPCs out of
the construct when cultured in a Matrigel migration
assay. The use of hCMPCs as a cell source is attractive
since they are already committed to the cardiac line-
age, able to differentiate into cardiomyocytes, and able
to proliferate in vitro. The authors show that hCMPCs
have a greater potential to differentiate in the 3D bio-
printed construct than when cultured in 2D, as shown
by the increased expression of the transcription factors
Nkx2.5, GATA-4, Mef-2c, and TnT. The hCMPCs, how-
ever, did not display the striated phenotype of cardio-
myocytes, suggesting that additional factors (i.e. chemi-
cal, mechanical) are needed to fully differentiate this
population of cells in vitro. The authors, however, do
show that these progenitor cells are able to migrate from
the construct in a Matrigel invasion assay, suggesting
the therapeutic potential of this population of cells to
integrate into damaged heart tissue if implanted.
Another avenue in 3D bioprinting being pursued
by the Cho group is the use of bioinks generated from
decellularized ECM (dECM) for extrusion-based
printing [19]. dECM of whole organs have been shown
to provide instructional cues to cells that better main-
tain a proper phenotype [46–50]. A drawback of the
Figure 3.  Decellulrized ECM bioprinting. (a) Images that
top-down approach of decellularized organs is that pat-
compare native heart tissue to decellularized ECM in bulk
terning of multiple types of cells is difficult, and not all and histological form (scale bar: 100 µm). (b) Schematic of
applications of tissue engineering require whole organ extrusion printing of the dECM bioink into a 3D porous
replacement. Using similar methods for decellularizing tissue construct (scale bar: 5 mm). (c) Microscopic images of
bioprinted construct (scale bar: 400 µm).
whole organs, tissue from the heart, fat, and cartilage
were made into bioinks and printed to form porous
tissue-like structures, similar to those by Gaetani et ­ recisely patterned and oriented on scales ranging three
p
al (figure 3(b)). Due to the limited sources of human orders of magnitude (microns to millimeters), and (iii)
cardiomyocytes, the Cho group encapsulated rat myo- materials that induce and maintain proper phenotype
blast cells into the heart-derived bioink. As compared of the cells and that do not illicit a negative reaction
to collagen constructs, the bioink increased expression from the host.
cardiac-specific genes (Myh6 and Actn1) after 4 d in
culture, and the increased expression was maintained 3.  3D printing for heart valves
for at least 14 d. Furthermore, the bioink constructs
induced higher expression of cardiac myosin heavy Currently, two options currently exist for heart valve
chain (β-MHC) than the collagen construct. replacement surgery: (i) using a mechanical heart valve,
Although 3D bioprinting is useful for building and (ii) using a biological heart valve [51]. The advan-
porous constructs that can maintain the viability of tage of prosthetic valves is that they are mechanically
cells, as shown in the described examples, to date this robust and have a long lifetime before a subsequent
technology has not successfully printed tissue that has replacement surgery is needed (relative to biological
clinical relevance. Similar to standard tissue engineer- valves) [52]. Their disadvantage is they require the pa-
ing approaches, 3D printing of myocardial tissue is pri- tient to take anticoagulants for life. Biological valves,
marily limited by: (i) the source of human cardiac cells made from either an allographic or xenographic source
able to be implanted, and (ii) the limited resolution by of tissue, do not require the patient to take anticoagu-
which complex structures can be built. The promise lants for life, but have a significantly shorter lifetime
of iPS cells and cardiac progenitor cells will hopefully (10–20 years) than prosthetic valves [53].
address the issues associated with the first limitation, a Tissue engineered heart valves have the promise
feat more likely to be solved by biologists. Tissue engi- of using a patient’s own cells, mitigating the need for
neers, however, have the responsibility to address the long-term drug use, improving hemocapability of
second limitation by building tissues that, at a mini- the replacement valve, and adding the capacity for the
mum, possess the following features: (i) a functional replacement heart valve to repair itself, and even grow
vascular system that can easily be integrated with a with the patient (a major attribute for younger patients)
host, (ii) a hierarchical structure that enables cells to be [51, 52, 54, 55]. The desired qualities for any heart valve

4
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

are that they: (i) have minimal regurgitation of blood


upstream, (ii) have a low transvalvular pressure gradi-
ent, (iii) have minimal thrombogenetic response, and
(iv) have a high capacity to repair damage [56].
3D printing has the potential to improve the clinical
outcome of tissue engineered heart valves by allowing
their fabrication to better match: (i) the patient-­specific
geometries of the heart valve, (ii) the spatial heterogene-
ity of the mechanical properties of the heart valve, and
(iii) the spatial heterogeneity of the cell types seeded
within and on the surface of the heart valve.
Currently, Jonathon Butcher’s lab has been lead-
ing the front for 3D printing of tissue engineered heart
valves [56–58]. In 2012 the Butcher group developed an
extrusion-based 3D printer, which deposited two types
of photocrosslinkable hydrogels, one rigid (~75 kPa)
hydrogel for the root and another soft (~5 kPa) hydro-
gel for the leaflets (figure 4) [56]. This artificial valve
demonstrated that 3D printing easily allowed arbitrary
geometries, which ranged between 12 mm (average
size of a 6 month old heart valve) and 22 mm (aver-
age size of an adult) in diameter. The heart valves were
3D printed within 45 min and with an accuracy up to
93% (when compared to the desired geometries). The
ability to print two materials simultaneously with dis-
tinct mechanical properties allowed the fabrication of
heart valves that better mimicked the varying stiffness
in native heart valves between the leaflets and the root
(figure 4(b)). Since this work was a proof of principle
for the use of 3D printing for the fabrication of tissue
engineered heart valves, no functional testing was per-
formed. The authors did, however, demonstrate that Figure 4.  3D printed heart valve. (a) Schematic of 3D
porcine aortic valve interstitial cells (PAVIC) could be printer system, including two extrusion systems with
seeded into the 3D printed heart valves and survive for integrated UV light source to crosslink the hydrogel.
(b) CAD model and image of a heart valve with a geometry
up to three weeks (figure 4(c)). In another study, the obtained from microCT images. The red regions are the
Butcher group used 3D printing to directly fabricate leaflets and the blue regions are the roof of the heart valve.
a heart valve with two types of cells, aortic root sinus (c) Microscopic images of the PAVIC cells in the root/
annulus of the heart valve after being stained with a live
smooth muscle cells (SMCs) and aortic valve leaflet (green) and dead (red) stain (scale bar: 2 mm).
interstitial cells (VICs), encapsulated into the root and
leaflet regions, respectively. The results showed that the
cells maintained the mechanical properties of the algi- and (iii) have a relatively lower contribution of cellular
nate/gelatin mixture over 7 d but that an acellular heart activity, such that complex differentiation pathways
valve of the same material would lose its mechanical and need for highly ordered vascular networks are not
properties [57]. Most recently, the Butcher group dem- required for the successful development of a heart valve.
onstrated that methacrylated hydrogels (mixtures of With that said, thorough testing of 3D printed tissue
gelatin and hyaluorinc acid) could be printed with sus- engineered heart valves has yet to be done, and only by
pensions of human aortic valvular interstitial (HAVIC) doing so can their improved functionality be tested.
cells [58]. The HAVIC cells could remodel the printed
hydrogels by depositing their own ECM (collagen and 4.  3D printing for coronary arteries
glycosaminoglycans), and furthermore would undergo
changes in their phenotype based on the stiffness of the Despite improvements in therapies targeted at reduc-
printed hydrogels (a softer hydrogel better maintained ing disease burden, coronary artery disease continues
the fibroblastic behavior of the HAVIC cells). to afflict more than 16 million US adults, accounting
As shown by the work of the Butcher group, 3D for more than 1/3 of all deaths and is responsible for
printing is particularly suited for developing heart approximately 1.2 million hospitalizations annually.
valves because: (i) they are of a sufficiently complex Depending on the stage of coronary artery disease,
geometry that normal fabrication methods ca not eas- its symptoms are managed through lifestyle modi-
ily address, (ii) require heterogeneity in stiffness, that is fications, medical therapy, coronary angioplasty, or
easily achieved by incorporating multiple print heads, coronary artery bypass grafting (CABG) surgery. For

5
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

complex multivessel coronary artery disease, CABG vitro tissue development, and (ii) cell-sheet-based, in
is often recommended in clinical practice. This pro- which a monolayer of cells is cultured and then rolled
cedure involves diverting blood around a section of on a mandrel to produce a tubular conduit mimick-
severely narrowed or blocked artery using grafts har- ing the media and adventitia of an artery. Despite these
vested from other parts of the body. About 400 000 recent efforts, few artificial coronary bypass grafts have
CABG operations are performed annually in the US. matched the long-term performance of autologous
In a typical CABG surgery, one or more conduits, grafts and thus none is commercially available for clini-
typically internal thoracic arteries, radial arteries, or cal use to date.
saphenous veins harvested from the rest of the body, 3D printing is a promising tool to facilitate in both
are placed to ­supply additional blood flow to the distal tissue engineering approaches by: (i) generation of
coronary arteries blocked by stenosis. While CABG can scaffolds, whose paths conform to the patient-specific
alleviate the symptoms and improve the survival rate geometry and printing using synthetic biomaterials,
of patients with coronary artery disease, approximately and (ii) directly printing using differentiated endothe-
30% of patients are not eligible for this procedure due lial cells, fibroblasts and SMCs, or mesenchymal and
to the lack of suitable autologous vessels. Even if one of hematopoietic stem cells in biocompatible materials
these vessels is available for harvest, a number of draw- (e.g. hydrogel). However, 3D printing has not been
backs are associated with CABG, including accidental directly applied to the creation of patient-specific coro-
graft damage during harvest, relatively poor long-term nary bypass graft. Current research has been focused on
patency, and morbidity at donor sites after surgery [59]. the in-vitro generation of vascular models and lining
Consequently, it has long been hoped that artificial with endothelial cells in the inner surface, particularly
coronary bypass grafts can satisfy this unmet clinical for the formation of microvascular networks to study
need. The ideal vascular graft should be biocompatible, angiogenesis and thrombosis or supply of nutrients and
anti-thrombogenic, durable, and have similar compli- oxygen to engineered tissue.
ance and density to native vessels [60]. Early attempts Wu et al demonstrated omnidirectional printing of
to synthesize coronary grafts used expanded polyte- 3D biomimetic microvascular networks by direct-write
trafluoroethylene (ePTFE, also known as Gortex) and assembly of fugitive ink filaments within a photocur-
woven polyethylene terephthalate (PET, also known able gel reservoir (figure 5). This method broadens the
as Dacron)—both of which are frequently used for network design space and removes the need for tradi-
aorta and peripheral vascular territories. The results of tional layer-by-layer patterning [65]. The ink can be
using these conventional materials for making small- subsequently removed by liquefaction under a mod-
diameter low-flow coronary grafts are, however, disap- est vacuum to yield the desired microvascular network
pointing. At least two major limitations are attributed within the solidified gel matrix. However, this study
to the poor outcome of these synthesized grafts: (i) does not discuss the integration of cells into the con-
improper mechanical characteristics and (ii) undesir- structed networks.
able biochemical properties. In spite of their strength, Miller et al [66] printed rigid 3D filament networks
both ePTFE and PET are stiff and lack the flexibility for rapid casting of patterned vascular networks in
to stay in the proximity of the heart with its complex engineered tissues using extrusion through a syringe
geometry and cyclic deformation. In addition, inflam- mounted on a custom-modified RepRap Mendel 3D
matory responses are activated by exposure of these printer (figure 6). For this purpose, they developed a
materials to the blood and causes poor patency due biocompatible sacrificial material based on carbohy-
to thrombogenesis. A shift in research has been made drates. To show the flexibility of the approach, they pat-
to favor the exploration of more compliant materials terned vascular channels in the presence of living cells
as well as the combination with pharmaceutical and in a wide range of natural and synthetic ECM mate-
tissue-engineered modifications [61]. Polyurethane rials. In particular, they also demonstrated three key
grafts, although more compliant than ePTFE grafts, compartments of vascularized solid tissues (i.e. lumen,
have yielded higher rates of thrombosis and infection endothelial cells, and the matrix and cells residing in the
[62] and aneurysm formation [63]. With the promise interstitial zone).
of creating living conduits for restoring hemodynamic Li et al fabricated a hybrid cell/hydrogel vascular-
function, vascular tissue engineering has been hoped like network using a double-nozzle assembling tech-
to become a new avenue to make artificial coronary nique [67]. Adipose-derived stromal cells (ADSCs)
bypass grafts. The ideal tissue-engineered graft should were combined within a gelatin/alginate/fibrinogen
be endothelialized, non-thrombogenic, and have com- hydrogel to form the network construct and hepato-
parable biomechanical properties to the native blood cytes combined gelatin/alginate/chitosan were placed
vessel. around it. After assembly, the printed construct was
There are two main approaches in engineering stabilized and the ADSCs were induced to differenti-
biomimetic blood vessels [64]: (i) scaffold-guided, in ate into endothelial-like cells with endothelial growth
which scaffolds using natural, synthetic biomaterials, factor. Interestingly, the ADSCs at the periphery of the
or decellularized matrix are developed to support cell vascular-like network demonstrated some endothelial-
attachment, infiltration and proliferation during the in like cell properties.

6
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

Figure 5.  Omnidirectional printing of 3D microvascular networks. (a) Deposition of a fugitive ink into a physical gel reservoir
to pattern hierarchical, branching networks. (b) Photopolymerization of the reservoir yields a chemically cross-linked, hydrogel
matrix. (c) After the ink is liquefied, it is removed by a vacuum to expose the microvascular channels. (d) Fluorescent image of a 3D
microvascular network fabricated via omnidirectional printing of a fugitive ink (dyed red) within the photopolymerized matrix.
Reprinted from [65].

Figure 6.  3D printing of filament networks with carbohydrate glass and generation cylindrical channels lined with endothelial
cells (expressing mCherry, shown as red) and perfused with blood to support 10T1/2 cells (expressing enhanced green fluorescent
protein, shown as green) in the interstitial space. (a) Top view (scale bar: 1 mm) of printed multiscale carbohydrate-glass lattice
and magnified interfilament melt fusions in side view (scale bar: 200 µm). (b) Multilayered lattices are fabricated with precise
lateral and axial positioning resolution (scale bar: 1 mm). (c) After 9 d in culture, cross-section imaging showed the endothelial
monolayer lining the vascular lumen became surrounded by 10T1/2 cells (scale bar: 200 µm). D) Endothelial cells formed single and
multicellular sprouts (arrowheads) from patterned vasculature. Reprinted from [66].

Cui and Boland simultaneously deposited human aqueous processes that were shown to induce ­minimal
microvascular endothelial cells (HMVECs) and fibrin damage to cells. When printing HMVECs in con-
to form the microvasculature using a modified ther- junction with the fibrin, the cells were found to align
mal inkjet printer [68]. This printing technique used ­themselves inside the channels, and proliferated within

7
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

7 d to form a confluent lining. The authors concluded


that simultaneous cell and scaffold printing promoted
HMVEC proliferation and microvasculature forma-
tion. Despite the promising results, the risk of exposing
cells and materials to thermal and mechanical stress,
low droplet directionality, imprecise droplet size and
shape, frequent clogging of the nozzle and unreliable
cell encapsulation pose considerable challenges for the
use of ink-jet printers.
These important results have demonstrated the
potential to generate tubular structures in small scales
using 3D printing methods. In the meantime, it is clear
that several challenges still exist to translate the early
successes in microvascular 3D printing to the ultimate
goal of making an ideal artificial coronary bypass graft.
First, autologous blood vessels have three layers and
each has different cellular components, stiffness, and
functions. It is difficult for scaffold printing and sub-
sequent cell seeding to mimic such layered structures,
given the current limitations of resolution and scale.
Second, almost all of tissue-engineered vasculature by
3D printing is microscale. It remains unclear how to Figure 7.  (a) Image of an array of stretchable sensors on
a Langendorff-perfused rabbit heart. Sensors can detect,
seed cells of multiple types in the printed tubular scaf- electrocardiogram, pH, temperature, and strain. µLEDs are
folds of a few millimeters in diameter. Finally, the cells also integrated to allow for optical mapping. Scale bar: 6 mm.
within the coronary bypass graft should be kept alive Reprinted from [82]. (b) Image of a strain and pressure
sensor, comprising three layers of 3D printed conductive
by continuous supply of oxygen and other nutrients. ink in a silicone elastomer. Scale bar: 10 mm. Reprinted
In normal coronary arteries, nutrients come from the from [99]. (c) Stretchable display of organic LEDs folded
vasa vasorum (capillaries in the vessel wall). Therefore, in half. Reprinted from [101]. (d) Image of a 3  ×  4 array of
capacitive sensors connected to electrochemical transistors
future tissue-engineered grafts may require creation that control an electrochromic display. Reprinted from [88].
of both microvascular and macrovascular structures;
further emphasizing the need to build systems that can
span the large range of scales associated with biological e­ lectronic sensing [73–77], artificial skins [78–81],
tissues. heart shaped membranes [82, 83], and balloon
catheters [84]. These devices suggest the potential
5.  3D printing for cardiac sensors and for cardiovascular implants or surgical tools that
electrodes implement multifunctional sensing and actuation.
A critical aspect of all of these devices is the need
Whether fabricated from synthetic material or biologi- for geometries, form factors and material properties
cal tissue, a wide variety of cardiovascular implants and (especially stiffness and biocompatibility) that can
surgical tools either require or benefit greatly from the match and allow for seamless integration or interfac-
incorporation of sensing or actuation elements that al- ing with biological tissue.
low the implant or tool to collect information about the 3D printing is an optimal technology for these
surrounding conditions and apply a response. Today, types of devices. It provides the ability to create geom-
a variety of clinically relevant surgical and diagnostic etries that are highly complex and patient specific
tools utilize individual sensor elements, made from (based on conventional imaging techniques) [83]. Xu
small but complex rigid elements mounted on cath- et al, demonstrated this by creating a patient-specific
eters or guidewires. Guidewires with embedded flow thin flexible membrane to surround the heart with
[69] and pressure sensors [70] as well as catheters with embedded electronic sensors and actuators to moni-
complex imaging functionality such as optical coher- tor cardiac function based on a 3D printed mold [82].
ence tomography and ultrasound are common tools in Figure 7 shows an image of this device conforming to
interventional medicine [71, 72]. These are robust tech- a rabbit heart. Here, microtransfer printing was used
nologies and reviews of them can be found elsewhere, to fabricate electronics on this complex 3D mem-
however, they demonstrate the existing utility of these brane based on electronics fabricated on a 2D wafer
types of sensors. (via traditional microfabrication). This suggests a
Recently, new classes of devices that incorpo- general methodology for fabricating 3D patient spe-
rate arrays of similar (although in most cases, much cific sensing devices based on fabrication of soft and
simpler) sensing or actuation elements into mate- flexible 2D arrays or sensors and actuators which
rials and surfaces have been seamlessly integrated can then be transferred or bonded to complex 3D
with biological systems or surgical tools to create implants or tools.

8
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

To date, there are several approaches that allow for to overcome. While these types of integrated sensing
precision printing of 2D patterns of these soft func- implant and tools are still an emerging technology, they
tional materials. Examples include inkjet printing provide great potential to solve a wide variety of clinical
[85], ejet printing [86], roll-to-roll techniques [87], challenges.
and flexible printed circuit board technologies. A few
noteworthy examples include flexible arrays of pres- 6.  Future perspective
sure sensors and flexible displays shown in figures 7(b)
and (c). These approaches typically (but not always) 3D printing holds much promise for the development
utilize conducting, semiconducting, or piezoelectric of cardiac tissues because of its inherent ability to
polymers, which are flexible and, when utilized prop- build structures with arbitrary geometry and material
erly, can impart a wide range of sensing modalities. properties. Despite its promise, it appears that most
For cardiovascular applications, those which can examples of 3D printed tissues for cardiac applications
measure pressure [75, 76, 80, 88] and flow [74, 89] are are behind those of traditional tissue engineering ap-
particularly useful. Furthermore, for devices that aim proaches. Several limitations prevent this technology
to mimic biological systems (pumps, valves, etc), sen- from reaching fruition since the promise of arbitrary
sors and actuators are particularly important to create features is not actually achievable with current print-
or detect mechanical motion [90–94] and electrical ing systems and bioinks. Although nanoscale struc-
signals [95]. Other potentially useful sensors include tures can be built using two-photon lithography, this
temperature [74, 80], pH [73] and chemical [96] or high resolution 3D printing system does not trans-
biological sensors [97], which can detect particular late into tissue engineering since cells are not easily
markers of various CVDs or dysfunctions. Also note- incorporated into these systems, and if they were, they
worthy are approaches that allow for 2D arrays of still are not able to build structures large enough to
functional inorganic components to be assembled and be useful for tissue engineering purposes. Therefore,
integrated with soft substrates, whereby flexible and a major need for 3D printing is the development of
stretchable configurations can be achieved with the Hierarchical structures that can span the entire range
excellent performance of inorganic materials [77, 98]. of scales associated with tissues (i.e. nearly 7 orders of
For applications where limited flexibility is sufficient, magnitude). In addition, development of printers that
simple solutions such as flex printed circuit board can easily accommodate multiple materials within a
technology can be sufficient. When true stretchabil- given job need to be built, so that a sufficient variety
ity is required, approaches that utilize microtransfer of cell types and ECM proteins can be used to build
printing and advanced geometries with serpentine complex tissues.
interconnects and neutral mechanical plane designs An ideal bioprinter could switch between laying
should be used. down ECM at the nanoscale resolution, then switch
To date, there are very few demonstrations of direct to printing single cells at the microscale, in a seamless
3D printing of soft functional sensors or actuators, fashion that allows truly arbitrary features to be printed.
however several examples exist that utilize printing of For example, the formation of a basement membrane
conductive liquids or greases that can be utilized for in both planar and cylindrical geometries would allow
basic strain sensors [99, 100]. Figure 7(d) shows a 3D printed endothelial and epithelial cells to be printed
flexible trace of conductive carbon grease, which was with a proper orientation to form vasculature and tis-
printed in a flexible elastomer matrix. Development sue boundaries, respectively. Furthermore, the ability
of materials and methods for direct printing of 3D to print ECM with an orientation would allow polarity
sensors and actuators opens the door for cardiovascu- to be given to cells, such as cardiomyocytes, as is done
lar implants with even greater complexity that could with electrospinning. An ideal bioprinter would also
mimic the functionality observed in many existing integrate components that allow actuation, electrical
biological systems. However, this is challenging: tech- stimulation, and sensing to of the tissue to (i) opti-
niques common in 2D printing of such as solvent based mize maturation by providing additional mechano/
deposition of functional polymers are limited by the eletrical cues, (ii) provide quantitative data to assess
need to create viscous spanning and supportive features performance pre-implantation, (iii) provide quantita-
during 3D complex drying dynamics make it difficult tive data to monitor performance of the tissue post-
to control morphology and resulting properties. New implantation.
innovations are required. Despite the limitations of current 3D printers, which
3D printing, as a template or mold for integrated will undoubtedly improve, some of the major limita-
electronic sensing or for direct printing of sensors, tions of cardiac tissue engineering are not related to
opens the door for a wide range of implants and surgi- fabrication but are rooted in much more fundamentals
cal tools such as balloon catheters or stents with inte- issues, such as sources of human stem cells, protocols
grated sensing capabilities or grafts, valves and artificial for proper differentiation, and methodologies for effi-
hearts that integrate mechanical pumping and/or sens- cient conditioning of the formed tissues. Nonetheless,
ing and feedback. For all of these devices, biocompat- it is likely that advances in all these areas of research will
ibility and/or encapsulation will be critical challenges synergistically improve the entire field, suggesting that

9
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

direct communication and collaboration between biol- [24] Lee A Y, Mahler N, Best C, Lee Y-U and Breuer C K 2014
Regenerative implants for cardiovascular tissue engineering
ogists, engineers, and material scientists will expedite
Translational Res. 163 321–41
such progress. [25] Udelsman B V, Maxfield M W and Breuer C K 2013 Tissue
engineering of blood vessels in cardiovascular disease: moving
towards clinical translation Heart 99 454–60
References [26] Bouten C V C, Dankers P Y W, Driessen-Mol A, Pedron S,
Brizard A M A and Baaijens F P T 2011 Substrates for
[1] Cho G S, Fernandez L and Kwon C 2014 Regenerative medicine cardiovascular tissue engineering Adv. Drug Deliv. Rev.
for the heart: perspectives on stem-cell therapy Antioxidants 63 221–41
Redox Signaling 21 2018–31 [27] Klopsch C and Steinhoff G 2012 Tissue-engineered
[2] Emmert M Y, Hitchcock R W and Hoerstrup S P 2014 Cell devices in cardiovascular surgery Eur. Surgical Res.
therapy, 3D culture systems and tissue engineering for cardiac 49 44–52
regeneration Adv. Drug Deliv. Rev. 69 254–69 [28] Zhang B, Xiao Y, Hsieh A, Thavandiran N and Radisic M
[3] Chung J H Y et al 2013 Bio-ink properties and printability 2011 Micro- and nanotechnology in cardiovascular tissue
for extrusion printing living cells Biomater. Sci. engineering Nanotechnology 22 494003
1 763–73 [29] Markert M, Weber S, Lueth T C and IEEE 2007 A beating heart
[4] Ferris C J, Gilmore K J, Beirne S, McCallum D, Wallace G G and model 3D printed from specific patient data Annual Int. Conf.
Panhuis M I H 2013 Bio-ink for on-demand printing of living IEEE Engineering in Medicine and Biology Society vol 1-162007,
cells Biomater. Sci. 1 224–30 pp 4472–5
[5] Yeong W Y et al 2010 Porous polycaprolactone scaffold for [30] Weber S, Sodian R, Markert M, Reichart B, Daebritz S and
cardiac tissue engineering fabricated by selective laser sintering Lueth T C 2007 3D printing of anatomical heart models for
Acta Biomater. 6 2028–34 surgical planning in cardiac surgery Int. J. Comput. Assist.
[6] Melchels F P W, Feijen J and Grijpma D W 2010 A review Radiol. Surg. 2 S171–3
on stereolithography and its applications in biomedical [31] Shiraishi I, Kurosaki K, Kanzaki S and Ichikawa H 2014
engineering Biomaterials 31 6121–30 Development of suepr flexible replica of congenital heart
[7] Candiotti H et al 2013 Fused deposition modeling bioprinter disease with stereolithography 3D printing for simulation
39th Annual Northeast Bioengineering Conf. (NEBEC 2013) surgery and medical education J. Cardiac. Failure 20 S180–1
pp 177–8 [32] Coakley M F et al 2014 The NIH 3D print exchange: a public
[8] Li X M et al 2014 3D-printed biopolymers for tissue resource for bioscientific and biomedical 3D prints 3D Print.
engineering application Int. J. Polym. Sci. 2014 829145 Additive Manuf. 1 137–40
[9] Mironov V, Visconti R P, Kasyanov V, Forgacs G, Drake C J [33] Mironov V, Boland T, Trusk T, Forgacs G and Markwald R R
and Markwald RR 2009 Organ printing: tissue spheroids as 2003 Organ printing: computer-aided jet-based 3D tissue
building blocks Biomaterials 30 2164–74 engineering Trends Biotechnol. 21 157–61
[10] Richards D J, Tan Y, Jia J, Yao H and Mei Y 2013 3D printing for [34] Ferris C J, Gilmore K G, Wallace G G and Panhuis M I H
tissue engineering Isr. J. Chem. 53 805–14 2013 Biofabrication: an overview of the approaches used for
[11] Seliktar D, Dikovsky D and Napadensky E 2013 Bioprinting printing of living cells Appl. Microbiol. Biotechnol.
and tissue engineering: recent advances and future 97 4243–58
perspectives Isr. J. Chem. 53 795–804 [35] Shapira A, Kim D H and Dvir T 2014 Advanced micro-
[12] http://envisiontec.com/3d-printers/3d-bioplotter/ and nanofabrication technologies for tissue engineering
[13] http://www.invetech.com.au/portfolio/life-sciences/3d- Biofabrication 6 020301
bioprinter-world-first-print-human-tissue/ [36] Cohen S and Leor J 2004 Rebuilding broken hearts Sci. Am.
[14] Gibbs D M R, Vaezi M, Yang S F and Oreffo R O C 2014 Hope 291 44–51
versus hype: what can additive manufacturing realistically [37] Silvestri A, Boffito M, Sartori S and Ciardelli G 2013
offer trauma and orthopedic surgery? Regenerative Med. Biomimetic materials and scaffolds for myocardial tissue
9 535–49 regeneration Macromol. Biosci. 13 984–1019
[15] Derby B 2012 Printing and prototyping of tissues and scaffolds [38] Curtis M W and Russell B 2009 Cardiac tissue engineering
Science 338 921–6 J. Cardiovasc. Nursing 24 87–92
[16] Kawata S, Sun H B, Tanaka T and Takada K 2001 Finer features [39] Georgiadis V, Knight R A, Jayasinghe S N and Stephanou A
for functional microdevices—micromachines can be created 2014 Cardiac tissue engineering: renewing the arsenal for the
with higher resolution using two-photon absorption Nature battle against heart disease Integrative Biol. 6 111–26
412 697–8 [40] Hirt M N, Hansen A and Eschenhagen T 2014 Cardiac tissue
[17] Xu T, Reyna-Soriano D, Rodriguez-Devora J I, Bhuyan M and engineering state of the art Circ. Res. 114 354–67
Boland T 2014 Organ printing Biomaterials and Regenerative [41] Radisic M, Malda J, Epping E, Geng W L, Langer R and
Medicine (Cambridge: Cambridge University Press) pp 332–73 Vunjak-Novakovic G 2006 Oxygen gradients correlate with
[18] Guo Q Q, Cai X B, Wang X L and Yang J 2013 ‘Paintable’ 3D cell density and cell viability in engineered cardiac tissue
printed structures via a post-ATRP process with antimicrobial Biotechnol. Bioeng. 93 332–43
function for biomedical applications J. Mater. Chem. B [42] Shimizu T et al 2006 Polysurgery of cell sheet grafts overcomes
1 6644–9 diffusion limits to produce thick, vascularized myocardial
[19] Pati F et al 2014 Printing 3D tissue analogues with tissues FASEB J. 20 708–10
decellularized extracellular matrix bioink Nat. Commun. [43] Sakaguchi K et al 2013 In vitro engineering of vascularized
5 3935 tissue surrogates Sci. Rep. 3 1316
[20] Alwan A 2010 Global status report on noncommunicable [44] Miller J S 2014 The billion cell construct: Will 3D printing get
diseases: Introduction (Geneva: WHO Press) us there? PLOS Biol. 12 e1001882
[21] Murphy S L, Xu J and Kochanek K D 2013 Deaths: final data [45] Gaetani R et al 2012 Cardiac tissue engineering using tissue
for 2010 National Vital Statistics Reports Centers for Disease printing technology and human cardiac progenitor cells
Control and Prevention, National Center for Health Statistics, Biomaterials 33 1782–90
National Vital Statistics System vol 61, pp 1–117 [46] Ott H C et al 2008 Perfusion-decellularized matrix: using
[22] Mendis S, Puska P, Norrving B, World Health O, World Heart F nature’s platform to engineer a bioartificial heart Nat. Med.
and World Stroke O 2011 Global atlas on cardiovascular 14 213–21
disease prevention and control (Geneva: WHO Press) [47] Flynn LE 2010 The use of decellularized adipose tissue to
[23] Haraguchi Y, Shimizu T, Yamato M and Okano T 2012 Concise provide an inductive microenvironment for the adipogenic
review: cell therapy and tissue engineering for cardiovascular differentiation of human adipose-derived stem cells
disease Stem Cells Translational Med. 1 136–41 Biomaterials 31 4715–24

10
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

[48] Ott H C et al 2010 Regeneration and orthotopic [71] Mintz G S 2014 Clinical utility of intravascular imaging and
transplantation of a bioartificial lung Nat. Med. physiology in coronary artery disease J. Am. Coll. Cardiol.
16 927–U131 64 207–22
[49] Petersen T H et al 2010 Tissue-engineered lungs for in vivo [72] Ferrante G, Presbitero P, Whitbourn R and Barlis P 2013
implantation Science 329 538–41 Current applications of optical coherence tomography for
[50] Uygun B E et al 2010 Organ reengineering through coronary intervention Int. J. Cardiol. 165 7–16
development of a transplantable recellularized liver graft using [73] Chung H-J et al 2014 Stretchable, multiplexed ph sensors with
decellularized liver matrix Nat. Med. 16 814–U120 demonstrations on rabbit and human hearts undergoing
[51] Jana S, Tefft B J, Spoon D B and Simari R D 2014 Scaffolds for ischemia Adv. Healthc. Mater. 3 59–68
tissue engineering of cardiac valves Acta Biomater. 10 2877–93 [74] Webb R C et al 2013 Ultrathin conformal devices for precise
[52] Schoen F J 2011 Heart valve tissue engineering: quo vadis? and continuous thermal characterization of human skin
Curr. Opin. Biotechnol. 22 698–705 Nat. Mater. 12 938–44
[53] Chambers J 2014 Prosthetic heart valves Int. J. Clin. Pract. [75] Maeda R, Tsaur J J, Lee S H and Ichiki M 2004 Piezoelectric
68 1227–30 microactuator devices J. Electroceramics 12 89–100
[54] Lueders C, Jastram B, Hetzer R and Schwandt H 2014 Rapid [76] Dagdeviren C et al 2014 Conformable amplified lead zirconate
manufacturing techniques for the tissue engineering of human titanate sensors with enhanced piezoelectric response for
heart valves Eur. J. Cardio-Thoracic Surg. 46 593–601 cutaneous pressure monitoring Nat. Commun. 5 4496
[55] Masoumi N et al 2014 Tri-layered elastomeric scaffolds for [77] Kim D-H, Ghaffari R, Lu N and Rogers J A 2012 Flexible and
engineering heart valve leaflets Biomaterials 35 7774–85 stretchable electronics for biointegrated devices Ann. Rev.
[56] Hockaday L A et al 2012 Rapid 3D printing of anatomically Biomed. Eng. 14 113–28
accurate and mechanically heterogeneous aortic valve [78] Someya T, Sekitani T, Iba S, Kato Y, Kawaguchi H and Sakurai T
hydrogel scaffolds Biofabrication 4 035005 2004 A large-area, flexible pressure sensor matrix with organic
[57] Duan B, Hockaday L A, Kang K H and Butcher J T 2013 3D field-effect transistors for artificial skin applications Proc. Natl
bioprinting of heterogeneous aortic valve conduits with Acad. Sci. USA 101 9966–70
alginate/gelatin hydrogels J. Biomed. Mater. Res. A 101 1255–64 [79] Yeo W-H et al 2013 Multifunctional epidermal electronics
[58] Duan B, Kapetanovic E, Hockaday L A and Butcher J T 2014 printed directly onto the skin Adv. Mater. 25 2773–8
3D printed trileaflet valve conduits using biological [80] Someya T et al 2005 Conformable, flexible, large-area networks
hydrogels and human valve interstitial cells Acta Biomater. of pressure and thermal sensors with organic transistor active
10 1836–46 matrixes Proc. Natl Acad. Sci. USA 102 12321–5
[59] Hillis L D et al 2011 ACCF/AHA Guideline for Coronary [81] Hammock M L, Chortos A, Tee B C K, Tok J B H and Bao Z
Artery Bypass Graft Surgery. A report of the American College 2013 25th anniversary article: the evolution of electronic skin
of Cardiology Foundation/American Heart Association Task (E-Skin): a brief history, design considerations, and recent
Force on Practice Guidelines. Developed in collaboration with progress Adv. Mater. 25 5997–6038
the American Association for Thoracic J. Am. Coll. Cardiol. [82] Xu L et al 2014 3D multifunctional integumentary membranes
58 e123–210 for spatiotemporal cardiac measurements and stimulation
[60] Desai M, Seifalian A M and Hamilton G 2011 Role of across the entire epicardium Nat. Commun. 5 3329
prosthetic conduits in coronary artery bypass grafting Eur. J. [83] Gutbrod S R, Sulkin M S, Rogers J A and Efimov I R 2014
Cardio-Thoracic Surg. 40 394–8 Patient-specific flexible and stretchable devices for cardiac
[61] Byrom M J, Ng M K C and Bannon P G 2013 Biomechanics diagnostics and therapy Prog. Biophys. Mol. Biol.
and biocompatibility of the perfect conduit—can we build 115 244–51
one? Ann. Cardiothoracic. Surg. 2 435–43 [84] Dae-Hyeong K et al 2011 Materials for multifunctional balloon
[62] Nakagawa Y, Ota K, Sato Y, Fuchinoue S, Teraoka S and catheters with capabilities in cardiac electrophysiological
Agishi T 1994 Complications in blood access for hemodialysis mapping and ablation therapy Nat. Mater. 10 316–23
Artif. Organs 18 283–8 [85] Singh M, Haverinen H M, Dhagat P and Jabbour G E 2010
[63] Brothers T E, Stanley J C, Burkel W E and Graham L M 1990 Inkjet printing—process and its applications Adv. Mater.
Small-caliber polyurethane and polytetrafluoroethylene 22 673–85
grafts: a comparative study in a canine aortoiliac model [86] Park J-U et al 2007 High-resolution electrohydrodynamic jet
J. Biomed. Mater. Res. 24 761–71 printing Nat. Mater. 6 782–9
[64] Seifu D G, Purnama A, Mequanint K and Mantovani D 2013 [87] Manceau M, Angmo D, Jørgensen M and Krebs F C 2011 ITO-
Small-diameter vascular tissue engineering Nat. Rev. Cardiol. free flexible polymer solar cells: from small model devices to
10 410–21 roll-to-roll processed large modules Org. Electron.
[65] Wu W, DeConinck A and Lewis J A 2011 Omnidirectional 12 566–74
printing of 3D microvascular networks Adv. Mater. 23 [88] Zirkl M et al 2011 An all-printed ferroelectric active matrix
H178–83 sensor network based on only five functional materials
[66] Miller J S et al 2012 Rapid casting of patterned vascular forming a touchless control interface Adv. Mater.
networks for perfusable engineered 3D tissues Nat. Mater. 23 2069–74
11 768–74 [89] van der Horst A, van der Voort D, Mimoun B, Rutten M, van
[67] Li S, Xiong Z, Wang X, Yan Y, Liu H and Zhang R 2009 Direct de Vosse F and Dekker R 2013 A novel flexible thermoelectric
fabrication of a hybrid cell/hydrogel construct by a double- sensor for intravascular flow assessment IEEE Sensors J.
nozzle assembling technology J. Bioactive Compatible Polym. 13 3883–91
24 249–65 [90] Chandrana C, Talman J, Pan T, Roy S and Fleischman A
[68] Cui X and Boland T 2009 Human microvasculature 2010 Design and analysis of MEMS based PVDF ultrasonic
fabrication using thermal inkjet printing technology transducers for vascular imaging Sensors 10 8740–50
Biomaterials 30 6221–7 [91] Xia F, Tadigadapa S and Zhang Q M 2006 Electroactive
[69] Dib N, Bajwa T, Shalev Y, Nesto R and Schmidt D H 1998 polymer based microfluidic pump Sensors Actuators A
Validation of Doppler FloWire for measurement of coronary 125 346–52
flow reserve in humans Catheterization Cardiovascular Diagn. [92] Sang Min W et al 2011 Piezoresistive strain sensors and
45 382–5 multiplexed arrays using assemblies of single-crystalline
[70] De Bruyne B, Paulus W J, Vantrimpont P J, Sys S U, silicon nanoribbons on plastic substrates IEEE Trans. Electron
Heyndrickx G R and Pijls N H J 1993 Transstenotic coronary Devices 58 4074–8
pressure gradient measurement in humans: in vitro and in vivo [93] Lu N, Lu C, Yang S and Rogers J 2012 Highly sensitive skin-
evaluation of a new pressure monitoring angioplasty guide mountable strain gauges based entirely on elastomers Adv.
wire J. Am. Coll. Cardiol. 22 119–26 Funct. Mater. 22 4044–50

11
Biomed. Mater. 10 (2015) 034002 B Mosadegh et al

[94] Yu-Jen H, Jia Z and Kymissis I 2011 A locally amplified strain [98] Kim D-H et al 2008 Stretchable and foldable silicon
sensor based on a piezoelectric polymer and organic field- integrated circuits Science 320 507–11
effect transistors IEEE Trans. Electron Devices 58 910–7 [99] Muth J T et al 2014 Embedded 3D printing of strain sensors
[95] Béduer A, Joris P, Mosser S, Delattre V, Fraering P C and within highly stretchable elastomers Adv. Mater.
Renaud P 2014 Accurate resistivity mouse brain mapping 26 6307–12
using microelectrode arrays Biosens. Bioelectron. 60 143–53 [100] Ladd C, So J-H, Muth J and Dickey M D 2013 3D printing of
[96] Lin P and Yan F 2012 Organic thin-film transistors for free standing liquid metal microstructures Adv. Mater.
chemical and biological sensing Adv. Mater. 24 34–51 25 5081–5
[97] Magliulo M et al 2013 Electrolyte-gated organic field- [101] Sekitani T et al 2009 Stretchable active-matrix organic light-
effect transistor sensors based on supported biotinylated emitting diode display using printable elastic conductors
phospholipid bilayer Adv. Mater. 25 2090–4 Nat. Mater. 8 494–9

12

Вам также может понравиться