You are on page 1of 14

View Article Online

View Journal

Lab on a Chip
Devices and applications at the micro- and nanoscale

Accepted Manuscript

This article can be cited before page numbers have been issued, to do this please use: C. Richard, A.
Neild and V. Cadarso, Lab Chip, 2020, DOI: 10.1039/C9LC01184F.
Volume 17
Number 1
7 January 2017
This is an Accepted Manuscript, which has been through the
Pages 1-200
Royal Society of Chemistry peer review process and has been
accepted for publication.
Lab on a Chip
Devices and applications at the micro- and nanoscale Accepted Manuscripts are published online shortly after acceptance,
rsc.li/loc

before technical editing, formatting and proof reading. Using this free
service, authors can make their results available to the community, in
citable form, before we publish the edited article. We will replace this
Accepted Manuscript with the edited and formatted Advance Article as
soon as it is available.

You can find more information about Accepted Manuscripts in the


Information for Authors.

Please note that technical editing may introduce minor changes to the
text and/or graphics, which may alter content. The journal’s standard
ISSN 1473-0197 Terms & Conditions and the Ethical guidelines still apply. In no event
PAPER
Chengzhi Hu, Bradley J. Nelson et al.
Charactization of size-dependent mechanical properties of
shall the Royal Society of Chemistry be held responsible for any errors
tip-growing cells using a lab-on-a-chip device

or omissions in this Accepted Manuscript or any consequences arising


from the use of any information it contains.

rsc.li/loc
Page 1 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

Jour
nalName

The Emerging Role of Microfluidics in Multi-Material 3D Bioprinting


Cynthia Richard, a,b Adrian Neild, a Victor J. Cadarso b,c

Lab on a Chip Accepted Manuscript


To assist the transition of 3D bioprinting technology from simple lab-based tissue fabrication, to
fully functional and implantable organs, the technology must not only provide shape control, but
also functional control. This can be accomplished by replicating the cellular composition of the
native tissue at the microscale, such that cell types interact to provide the desired function. There is
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

therefore a need for precise, controllable, multi-material printing that could allow for high, possibly
even single cell, resolution. This paper aims to draw attention to technological advancements made
in 3D bioprinting that target the lack of multi-material, and/or multi cell-type, printing capabilities
of most current devices. Unlike other reviews in the field, which largely focus on variations in
single-material 3D bioprinting involving the standard methods of extrusion-based, droplet-based,
laser-based, or stereolithographic methods; this review concentrates on sophisticated multi-material
3D bioprinting using multi-cartridge printheads, co-axial nozzles and microfluidic-enhanced printing
nozzles.

Research into the development of new tissue engineering tech-


nologies may solve the under supply of transplant organs 1–3 . The approach; firstly by expression of genetic information on a cellu-
U.S. Department of Health and Human Services estimates that a lar level, followed by proliferation, differentiation and lastly by
patient is added to the national transplant waiting list every ten the self-assembly of various cell types with different functions to
minutes. Unfortunately, whilst donor numbers are slowly rising, form specific tissues, which then come together to form organs 9 .
the demand is too high to be matched by donors and alterna- Bottom-up biofabrication processes aim to produce macroscopic
tives need to be developed 4 . Tissue engineering has the potential biological tissues by attempting to imitate and utilize the self-
to address this deficiency in organ donation through the fabrica- assembly of biomaterials, and particularly of cells. In tissue en-
tion of artificial organs, which, if constructed from patient derived gineering, the self-assembling process involves a system where
cells, could also lead to a reduction of adverse immune response spontaneous cell organization occurs, relying on the cell’s abil-
upon implantation. However, implantation is not the only moti- ity to produce extracellular matrix (ECM) naturally 10 . Currently
vation for complex tissue engineering, there are also highly com- however, the construction of tissues require the use of a scaffold,
pelling in vitro physiological applications. For example, engineer- meaning that cell seeding/adherence is needed to encourage the
ing physiological or pathological tissue models could, eventually, formation of the ECM.
eliminate the use of animals in pharmacology and pharmaceuti- Researchers are working to realise the full potential of the op-
cal research for drug efficacy, toxicity, delivery and disease de- portunities in tissue engineering through advancing biofabrica-
tection 5–7 . Furthermore, the efficient development of complex tion technologies (Fig 1. Among other methods, bioprinting has
tissues made from patient samples will have a positive impact on emerged as an ideal candidate to manufacture complex 3D tissues
personalized medicine, including predictive drug screening, and and organs 11 . Much like polymer or metallic 3D printing 10,11 ,
is expected to lead to advances in regenerative therapies 8 . 3D bioprinting is a form of additive manufacturing where 3D
In order to create complex tissues and even fully functional or- structures are produced in a layer-by-layer procedure with the
gans, emphasis must be placed on the fabrication of biomimetic use of computer-aided transfer (e.g. reading in data from CAD
tissues. In nature, living organisms are assembled in a bottom-up drawings) and build-up processes 5,12 . 3D printing is currently al-
ready used in a variety of applications in the medical industry, in-
cluding surgical instruments, prostheses, anatomical models, and
a
Laboratory for Micro Systems, Department of Mechanical and Aerospace Engineering, customized implants using inert materials (metals, ceramics, or
Monash University, Clayton, VIC 3800, Australia. Email: adrian.neild@monash.edu polymers) for medical devices like dental implants and hearing
b
Applied Micro- and Nanotechnology Laboratory, Department of Mechanical and aids 13,14 , as well as for other components for instance in me-
Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia. Email: vic-
chanical, aerospace and structural engineering 15 . However, 3D
tor.cadarso@monash.edu
c
The Melbourne Centre for Nanofabrication, Australian National Fabrication Facility bioprinting involves the use of biological components (e.g. pro-
– Victorian Node, Clayton, Victoria 3800, Australia teins, peptides, DNA, cells, hormones, etc.) together with bioma-

J
our
nal
Name,
[yea
r][
,vol
.
],1–13 | 1
Lab on a Chip Page 2 of 13
View Article Online
DOI: 10.1039/C9LC01184F

Lab on a Chip Accepted Manuscript


Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

Fig. 1 The current biofabrication techniques that have been developed (top: 3D Bioprinting and bottom: Microfluidic biofabrication) along with a
new method currently under development (middle: Microfluidic-enhanced 3D bioprinting). 3D cell-laden hydrogel scaffolds have been 3D bioprinted
using single-material printheads to print with a single cell-type (SCT3D – single cell-type 3D bioprinting) for simple tissue fabrication or multi-cartridge
printheads for multi-material printing (MCT3D – multi cell-type 3D bioprinting) for slightly more complex hydrogel scaffolds. Examples of such
tissues can be seen in Fig 2 and Fig 3. Microfluidic biofabrication (MFBF) has been used for organ-on-a-chip applications and to create biological
constructs such as organoids. Organ-on-a-chip methods are used for cell culture within microlfuidic chips and are not used in MF3D. Methods such
as multiple-phase microfluidics, t-junctions and emulsions, or double emulsions are used to create cells in droplets, cell-laden filaments and multi-layer
cell droplets/filaments, respectively. Such microfluidic capabilities can be combined with 3D bioprinting methods allowing for increased fluid and cell
handling capabilities outside of the chip and can offer enhanced 3D bioprinting methods (MF3D – microfluidic-enhanced 3D printing) for the fabrication
of highly organized, multi-cell-type/multi-material tissue constructs. The methods that can be incorporated into microfluidic-enhanced bioprinting
nozzles include utilizing the laminar flow regime, flow-focusing, or co-axial flow focusing, fluid mixing, as well as the previously mentioned methods
for the fabrication of organoids. Organoid fabrication in MFBF methods are produced within the chip and would be extruded in MF3D methods to
produce 3D constructs of multi-layered organoids containing different cell types in order to construct complex, organized tissues. Examples of tissues
fabricated using these methods can be seen in Fig 4, Fig 5, and Fig 6. Further examples are tabulated in Table 1.

terials, such as hydrogels, to construct living tissue 3 . 3D bioprint- ditive manufacturing used for the effective fabrication of com-
ing has the potential to generate specific structures and shapes plex nanocomposites 15 , is now also being used to construct tis-
with materials that can be tailored to deliver the desired physi- sues from cell-laden scaffolds in 3D bioprinting. A more detailed
cal, chemical and biological properties 5,12,16 , hence developing overview of the capabilities of these different single cell-type 3D
and mastering these techniques has become a major focus in the bioprinting methods, we will term this SCT3D, can be seen in Ta-
tissue engineering community. ble 1 of the SI. For a detailed overview of the use of standard
Most of the current 3D bioprinting technologies available use additive manufacturing processes for SCT3D, and of their use in
standard 3D printing platforms to deposit biological material printing tissues and organs, please refer to the review paper by
(Fig 1). As such, 3D bioprinting can be divided into the same Vijayavenkataraman et al. 3 . There are also numerous other arti-
four main categories as 3D printing: laser-based methods, stere- cles offering detailed reviews of the full range of SCT3D Bioprint-
olithography, extrusion-based (EB) methods and droplet-based ing methods and their applications in tissue engineering 3,18–23 ,
(DB) (also known as printer-based) methods. For instance inkjet as well as method specific reviews 24–27 , and tissue specific re-
printing (IJP), one of the main printing methods for the addi- views 7,11,16,28–30 , just to name a few.
tive manufacturing of ceramics 15 , is also a commonly used sin- The progress of bioprinting processes has allowed tissues and
gle cell-type 3D bioprinting method, known as droplet-based 3D anatomical structures, from several of the eleven organ systems
bioprinting. Stereolithography, one of the earliest methods of ad- present in the human body, to be printed and tested 3 . These

2| J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 3 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

eleven organ systems consist of the skeletal 31–34 , muscular 35–37 , lenges, each organ system pose tissue-specific issues which must
nervous 38–40 , lymphatic 41 , endocrine 42,43 , reproductive 44 , in- be addressed 3 .
tegumentary 45–47 , respiratory 48 , digestive 49,50 , urinary 51 , and The loss of microscale precision during mesoscale fabrication
circulatory systems 52–54 . Nonetheless, there is still a long way hinders the arrangement of different cell types within a tissue
to go before 3D bioprinting can be moved from lab-based ex- assembly. But, it is the microenvironment created by signalling
periments to full-scale manufacturing of functional tissues and between neighbouring cells that can influence primary cell differ-
organs 55 . The anatomical arrangement of human tissues at the entiation, and lead to spontaneous morphogenesis 56 . Thus, lack
micro-, meso-, and macro-scales to form complex 3D hierarchi- of microscale accuracy placement in turn affects the macroscale
cal structures is integral to the proper operation of the individual organoid morphology and therefore limits the reproducibility and
tissues and organs and thus to the organism as a whole 3 . But, application of artificial tissues 56 . In order to fabricate artificial
one of the major limiting factors of current 3D bioprinting tech- biological tissues that function just as well as natural ones, it
nologies is the lack of microscale precision and regulation when it is crucial to position specific cell types in multiscale structures
comes to cellular composition or arrangement using multiple cell with hierarchical features that replicate the natural microscale tis-

Lab on a Chip Accepted Manuscript


types, or single cell resolution, during the printing process. Ex- sue environment 9 . For that reason, a multi-material, bottom-up
isting technologies only have control over the macroscale geom- biofabrication approach would be highly beneficial. Researchers
etry of cell-laden bioinks and hydrogel scaffolds. However, print- have therefore begun addressing these issues by combining the
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

ing multiple materials, such as heterogeneous cell populations, cell and fluid handling capabilities offered by microfluidics with
growth factors and nutrients, in a well-timed and orderly manner, the biofabrication potential of 3D bioprinting.
or recreating micro-scale structure by controlling the placement In order to improve microscale precision, microfluidic biofab-
of individual cells, is still under development. Beyond these chal- rication techniques utilise the offered cell and biomaterial han-

Fig. 2 Multi-Cartridge 3D Bioprinting of human-scale tissues. (a) The ITOP system. (b) Mandible bone construct where (i) shows the 3D architecture
using CAM software developed by Kang et al. (the lines of red, blue and green show the dispensing paths of the cell-laden hydrogel, Pluronic F-127
and PCL respectively), (ii) demonstrates the patterning of one layer of the 3D printing process, (iii) is a photograph of the 3D printed bone defect
construct and (iv) shows the osteogenic differentiation of hAFSCs in the printed construct by Alizarin Red S staining, indicating the deposition of
calcium. (c) Calvarial bone construct where the dispensing paths of the PCL/TCP mixture and the cell-laden hydrogel are shown. (d) (i) photographs
of the printed bone implanted at day 0 (top) and at 5 months (bottom), and (ii – iv) show the histological and immunohistological images of the
nontreated construct (top) and the hAFSCs-printed construct after 5 months of implantation (bottom). (iii) H&E staining, (iv) modified tetrachrome
staining (Tetrachrome staining: red, mature bone: blue), and (v) vWF immunostaining (blood vessels: red). In the bottom images, the bioprinted
construct shows newly formed vascularized bone tissue throughout the implant, including in the central region, whereas in the untreated implant (top),
there is minimal bone tissue formation around the periphery of the implant. NB: new bone, PCL/TCP: remaining scaffold. (Reprinted with permission
from 17 )

J
our
nal
Name,
[yea
r][
,vol
.
],1–13 | 3
Lab on a Chip Page 4 of 13
View Article Online
DOI: 10.1039/C9LC01184F

dling and manipulation capabilities (Fig 1). Since microfluidic field from shape control to functional control. Meaning that the
devices offer the geometrical constraint of fluids in the micro- juxtaposition of different cell types causes interactions to occur
scale, where lower dimensional forces such as viscous forces and which deliver the desired function, for example, in the case of
surface tension dominate, the laminar flow can be utilized to gen- heart tissue the appropriate location of cardiomyocytes and car-
erate well-defined patterns by controlling the fluids containing diac pacemaker cells is required.
biomolecules, cells, organism, or chemical agents 57 . For instance,
different fluidic mechanisms such as focusing 58–61 , concentrat- Advanced Multiple Cell-type 3D Bioprinting by Multi-
ing 62–64 , mixing 65–68 or separating can be utilized to produce cartridge
various constructs, within the microfluidic device, such as cells in
droplets, microfibers, or cell sheets 9,69,70 . These constructs can Tissues normally have complex architectures, with distinct layers
be composed of different biomaterials or cell-laden bioinks. In having different cell densities and mechanical properties, often
addition, the microfluidic environment with multiple inlets could composed of different cell types 3 . This is of critical relevance for
allow for growth factors and nutrients to be provided during the bioprinting as cell behaviour and function are strongly affected

Lab on a Chip Accepted Manuscript


printing process, increasing cell viability and guiding primary cell by their neighbouring cells. Having different cells within close
differentiation. Furthermore, a seamless transition between ma- proximity to one another in an uncontrolled way could lead to
terials is possible. We will term microfluidic biofabrication as differentiation into an undesired cell type or to negatively af-
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

MFBF. These systems do not include bioprinting, rather the con- fecting proliferation. For example, bone tissue has a solid outer
structs are internal to the chip. However, MFBF techniques could part but an inner porous structure and would thus require both a
be easily combined with current 3D bioprinting platforms by mod- permeable scaffold structure and cell-laden hydrogels during the
ifying the nozzles to incorporate microfluidic chips, allowing for printing process. Other tissues in the human body, such as lymph
multi-material printing with greater control over material con- nodes, are intricate structures with highly organized stromal and
centration, positioning and composition. Microfluidic-enhanced lymphoid structures with a large variety of cell types, functional
3D bioprinting (MF3D) could be used to print various organoids skin also has many specialized cell types including sweat glands,
in a 3D manner that would better represent the structural and sweat ducts, hair follicles, and a multitude of other skin cells
functional integrity of natural tissues. For example, in order to in the epidermis and dermis 3 . So to fabricate biomimetic tis-
achieve a 3D multi-material organized tissue construct as demon- sues, with zone-specific heterogeneity, multi-material and multi
strated in the (Fig 1) MF3D, cell-laden multi-layer filaments could cell-type bioprinting with control of the exact cell-type in each
be used to print vascularized hollow tissue to serve as blood ves- location is crucial. To this end, some bioprinters have incorpo-
sel, interlaced with cardiac cells printed as cells in droplets along rated multiple cartridges to allow for material switching during
with a hydrogel to serve as a scaffold to achieve functional cardiac the printing process, while others use co-axial extruders which
tissue. allow for a bioink and a crosslinking agent to be printed simulta-
There have been several recent review papers that have neously.
focused solely on compiling and describing the differences A simple approach for multi-material bioprinting is to mod-
between traditional 3D bioprinting technologies (laser-based, ify current extrusion-based or droplet-based 3D bioprinting plat-
droplet-based, extrusion-based and stereolithographic methods) forms to incorporate multiple cartridges where controlled switch-
and their applications in tissue engineering and regenerative ing between materials is possible. Kang et al. used an inte-
medicine 3,5,7,14,18–21,71 . These reviews focus on printing of sin- grated tissue-organ printing (ITOP) extrusion-based system with
gle cell types, SCT3D. The review of Sodupe-Ortega et al. cov- a multi-cartridge module to simultaneously deliver different cell
ers multi-cartridge 3D bioprinting 72 , lets call this MCT3D – or types embedded in hydrogels, reinforcing polymers and sacri-
multiple cell-type 3D bioprinting, whilst that of Costantini et al. ficial hydrogels to fabricate human-scale tissue constructs such
includes co-axial 3D bioprinting 73 . The use of microfluidics to as mandible and calvarial bone, cartilage and skeletal muscle 17 .
enhance microstructural control has been reviewed by Ma et al. Material dispensing and switching is accomplished via XYZ stage
in 2018, but the emphasis is on printing cells within microfluidic movements and air pressure actuation which is computer con-
systems to form organ-on-a-chip constructs and layer-based bio- trolled (Fig. 2(a)). A mandible bone fragment was fabricated
printing methods that form built-in microchannels 70 , rather than in a size and shape that could be suitable for facial reconstruc-
on the use of microfluidic technologies to form structures external tion using human amniotic fluid-derived stem cells (hAFSCs) in
to the chip. a bioink (a mixture of poly(ε-caprolactone) (PCL) and trical-
In this review we cover the recent technological advancements cium phosphate (TCP)), a composite hydrogel (Gelatin, fibrino-
made in 3D bioprinting that address the need for multi cell- gen, hyaluronic acid (HA) and glycerol), and a Pluronic F127
type and/or multi-material bioprinting. Emphasis is placed on temporary support (Fig. 2(b)i). After 28 days of osteogenic dif-
3D bioprinting using multi-cartridge printheads and microfluidic- ferentiation, calcium deposition was observed through staining of
enhanced printing nozzles which transfer the sample and cell the 3D bone structure using Alizarin Red S, in the hAFSC-laden
handling control offered by microfluidics to complex tissue print- hydrogel (Fig. 2(b)iv). The team also fabricated a rat calvarial
ing (microfluidic 3D bioprinting or MF3D). As such, the focus bone construct (Fig. 2(c)) to study in vivo maturation of the bio-
is on the technological developments enabling replication of the printed bone 17 . The artificial bone was cultured in osteogenic
type of microscale cellular compositions that are transitioning the medio for 10 days and then implanted. Following 5 months of

4| J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 5 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

implantation, the bioprinted construct was analysed and showed


newly formed vascularized bone tissue throughout the implant
(Fig. 2(d)iii-v upper) and in the central portion. The untreated
defect without cells (only the scaffold) showed fibrotic tissue in-
growth and minimal bone tissues formation (Fig. 2(d)iii-v lower)
only along the periphery of the implant. In a more recent work,
Seol et al. used the same ITOP system for the 3D bioprinting
of a skin substitute combined with a wound dressing layer for
facial skin reconstruction 74 . A custom ‘BioMask’ was fabricated
by depositing three layers; a porous polyurathan (PU) layer, a
keratincyte-laden hydrogel layer, and a fibroblast-laden hydrogel
layer. The researchers were able to develop patient-specific fa-
cial skin composed of two different skin cell types for the epi-

Lab on a Chip Accepted Manuscript


dermis and dermis layers. After 14 days of implantation onto a
face-shaped construct, the BioMask showed promising skin tissue
regeneration and integration into the surround tissue.
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

Using a more multifaceted bioprinting platform, with a valve-


based inkjet and an extrusion-based printing system incorporat-
ing a crosslinking laser unit, Horváth et al. engineered an in
vitro human alveolar air-blood barrier 48 . The two cell-layer bar-
rier system was printed over two days with extracellular matrix
(ECM) being printed first on a porous membrane (serving merely
as a support for the printing process), followed by a layer of
EA.hy926 endothelial cells suspended in cell culture medium on
MatrigelTM. On the second day, a second ECM layer was printed
on the endothelial cells, subsequently a layer of A549 epithelial
cells suspended in cell culture medium was printed. The printed
construct was designed to attain close physiological resemblance
to the microenvironment of the native tissue. The bioprinted tis-
sue was compared to manually seeded cultures to demonstrate
the benefit of precise cell and biomaterial positioning on the de-
velopment of biomimetic tissue. The cellular morphology be-
tween the manually seeded and the bioprinted methods were
compared using laser scanning microscopy (LMS) and the results
are shown in Fig 3 (a) - (d), where the nuclei are depicted in
white, and the F-actin of immunostained cells are in green. It can
be seen that the cells seeded manually typically grow in patches,
and often in multi-layered clusters, whereas the bioprinted cells
spread uniformly over the available surface to form thin mono-
Fig. 3 Characterization and comparison of manually seeded and bio- layers. This was seen for both the endothelial (EA.hy926) and
printed cultures by LSM and brightfield microscopy for manually seeded epithelial (A549) cells. Similar results were obtained for the two
(left) and bioprinted (right) tissue. (a) – (d) Immunofluoresnce labelling
cell-layers, as shown in Fig 3 (e) and (f), and in the brightfield
of Factin cytoskeleton (green), nuclei (gray) and labelling of endothelial
cells with PECAM-1 (blue), in mono-cultures. (e) & (f) Laser scanning images obtained in Figures (g) and (h). This demonstrates the
micrographs of the two-layer co-cultures after 3 days. Cells stained for need for organized cell placement for successful tissue growth.
F-actin (red) and nuclei (white). A549 cells are in green and endothelial Merceron et al. utilized a four-cartridge bioprinter for the as-
cells are labelled with VE-cadherin (pink). Yellow arrow heads in (e)
sembly of different components into a single integrated muscle-
shows the thick MatrigelTM layer. (g) & (h) Brightfield micrographs
of embedded histological cross sections stained with Masson-Goldner tendon unit (MTU) construct 35 . For the formation of muscle tis-
trichrome coloration. Cytoplasm is red, collagen fibers of the ECM Ma- sue thermoplastic polyurethane (PU) was co-printed on one side
trigelTM are green and cell nuclei are brown. It is seen that the cells of the construct with C2C12 cell-laden hydrogel-based bioink,
seeded manually (left) typically grow in patches, and often in multi- while poly(ε-caprolactone) (PCL) was cp-printed with NIH/3T3
layered clusters, whereas the bioprinted cells (right) spread uniformly
cell-laden hydrogel-based bioink on the other side for tendon de-
over the available surface to form thin monolayers. This is observed for
both cell types and for single cell and and two cell layers. Scale bars (e velopment. The overall construct had >80 % cell viability 7 days
– f) 50 µm, 10 µm for the close-up, (g – h) 100 µm. (Reprinted with after printing and demonstrated early stage tissue development,
permission from 48 ) cell differentiation, and interaction of the different cell types at
the junction.
For high resolution bioprinting (approximately 50 µm) of

J
our
nal
Name,
[yea
r][
,vol
.
],1–13 | 5
Lab on a Chip Page 6 of 13
View Article Online
DOI: 10.1039/C9LC01184F

Lab on a Chip Accepted Manuscript


Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

Fig. 4 (a) Schematic of the co-axial printing nozzle where the bioink is delivered from the core and the crosslinking agent (CaCl2 ) is delivered from the
outer sheath. (b) Diagram showing the two-step crosslinking process, alginate is first crosslinked during the printing process by CaCl2 (left), followed
by crosslinking the GelMA by UV illumination. (c) Confocal fluorescence micrograph showing the distribution of HUVECs in a single printed microfiber
at day 14. (d) Confocal fluorescence micrograph showing the GFP-HUVECs in a single fibre for CD31, GFP, and nuclei. (e) Schematic of the scaffold
seeded with the cardiomyocytes. (f) F-actin (green) staining showing the location where two microfiber layers intersect with the cardiomyocytes can
be seen on the surface, this expression is an indicator of proper contractile and intercellular conductive function. (Reprinted with permission from 75 )

multi-component hydrogels, Zimmermann et al. used an liquid confined to microchannels allows for fine-tuning of phys-
inkjet printer with a dual printhead composed of piezoelectric ical and chemical compositions of the tissues being synthesized
pipettes 76 . The platform allowed for two reactive hydrogel pre- by allowing for chemical reactions to be triggered at specific lo-
cursor solutions to be dispensed simultaneously to create spatially cations, for instance through the use of controlled mixing (Fig 1),
structured multi-component hydrogels as well as cell-laden hy- with precise timing, inside the fluidic channel during the printing
drogel scaffolds using human bone-marrow derived mesenchymal process 9 .
stem cells (hMSCs). Whilst MCT3D can print a range of individual cell types, mi-
Multi-cartridge 3D bioprinting platforms have had many suc- crofluidic biofabrication (MFBF) methods can be used to produce
cesses in the past few years by offering the capability of simul- small constructs often referred to as microtissues or organoids.
taneously dispensing different cell-laden hydrogels, reinforcing Once created these micrometer-scale cellular building blocks 59
polymers and sacrificial elements 77 . The method also allows con- these microtissues can be assembled into off-chip 3D constructs.
tinuous, smooth, and rapid switching between different material Microtissues can be categorized into point-, line-, and plane-
types 78 . The result is a technology which researchers have used shaped and they provide high cell-density monolayer sheets,
to print complex tissue constructs with regional differences in cell spheroids, or cell-laden hydrogel blocks which are then spatially
types, while also providing structural integrity 35,77 . An outcome organized to produce heterogeneous macrotissues, to replicate
which is not viable with single-material 3D bioprinting devices. those produced naturally 72 .
Different fluidic mechanisms have been applied for the genera-
Microfluidic Biofabrication tion of these different shaped-microtissues (Fig 1). For example,
Microfluidics provide accurate control of fluids carrying in order to generate line-shape (fibrous) organoids flow-focusing
biomolecules, cells, organisms or chemical agents and are ideal or multiple-phase flows are commonly used. Whereas for point-
for performing a multitude of tasks and are thus highly benefi- shaped microtissues ( droplets) T-junctions or emulsions (Fig 1)
cial for integration into modified 3D bioprinting nozzles. Fur- are needed 9 . Microfluidic biofabrication methods have been used
thermore, the laminar flow-regime offered by microfluidic devices to prepare wide range of biological building blocks, including li-
also allows for multiple materials to be input simultaneously with- posomes 86,87 , cell-laden microfibers 88–90 , hydrogel sheets for co-
out mixing, or provides a seamless transition between these dif- culture 91 , and protein microbeads 92 . In addition, in their recent
ferent materials (Fig 1). The regulation of minute amounts of work Kang et al. explored the use of microfluidics and acoustics

6| J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 7 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

to create 3D cell patterns to construct extrudable functional blood ents 97–100 , switching between flow regimes 101,102 and changing
vessels 93 , a key aspect required for all tissues. For further read- suspension medium 103,104 . The inclusion of microfluidics within
ing, reviews of bottom-up microfluidic biofabrication techniques a bioprinter’s nozzle opens this range of controls over the fluidic
are given by Nie et al. 9 and Wang et al. 69 . Such MFBF methods environment and the suspended matter. One clever example, util-
can be used to generate the initial building blocks that can then ising ultrasonic methods of manipulating suspended matter, by
be bioprinted. Collino et al., addressed both the issues with nozzle clogging and
multi-material bioprinting 105 . They incorporated an acoustically-
Microfluidic-Enhanced 3D Bioprinting excited microfluidic printing nozzle where acoustic focusing was
used to deposit a two-phase ordered structure using a single noz-
The major limiting factors of current bioprinting techniques are
zle 105 . Microfluidic devices have been integrated into bioprinting
the lack of multi-material printing and regional material control,
nozzles to provide improved regulation and precision for multi-
damage imparted to cells during the printing process and noz-
material, or multi cell-type printing.
zle clogging due to viscous bioinks. There is therefore a need

Lab on a Chip Accepted Manuscript


to incorporate more appropriate biomaterial and cell handling In comparison to standard printing nozzles, microfluidic mod-
methods into current devices to address and eliminate these is- ified printheads would not only provide access to multiple bio-
sues. Examples of microfluidic suspended cell handling meth- materials or different cell-laden bioinks simultaneously, but they
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

ods include systems using forcefields generated from magnetic, would also provide a micro/nano-scale reaction chamber for
electrical, optical and acoustical effects, which can operate even increased structural complexity and micro-architecture control.
down to the single cell level if required. In addition, a range Furthermore, the microfluidic environment can help to protect
of methods have been described for fluid control in the form cells from damage induced by the printing process by hydrody-
of, for example, mixing 94–96 , generating concentration gradi- namically focusing cells in the centre of the channel; shear-stress

Table 1 Overview of Microfluidic-enhanced 3D bioprinting technologies refereing to the standard bioprinting platform used as well as the general
microfluidic method employed.

Bioprinting Method Microfluidic Component Application Advantages Reference


Extrusion-based & Stere- Laminar flow (Fig. 1) Heterogeneous cellularized • Multi-material
olithography constructs (Fig. 7) • Fast switching 79
• High spatial resolution
Extrusion-based • Laminar flow (Fig. 1) Complex heterogenous cellu- • Multi-material
• Flow-focusing (Fig. 1) larized constructs • Vary ratio between core 80
• Herringbone micro-mixer and sheath materials
(Fig. 1) • Printing of reactive com-
• T-junction (Fig. 1) ponents
• Cross-flow-filter for cell • Cells in droplets
concentration • Removes excess liquid
right before dispensing
Extrusion-based Multi-axial channel Bi- and tri-layered hollow • Multi-material
channel constructs of differ- • Seamless transition 81
ent cell types (endothelial • Hollow channel constructs
cell and fibroblasts)
Co-axial Laminar flow (Fig. 1) Hydrogel fibres laden with • Multi-material
muscle precursor cells • On the fly crosslinking 82

Extrusion-based Laminar flow (Fig. 1) Hetero-cellular model of hu- • Multi-material


man liver • Print cell-laden bioinks 83
and structural materials
simultaneously
Co-axial Serpentine-based mixing Osteochondral tissue (Fig. 5) • Layers or continuous
(Fig. 1) gradients of chemical, 84
mechanical and biological
cues
• Simultaneous fabrication
of scaffolds
Co-axial Laminar flow (Fig. 1) Glioblastoma tumor model • Multi-material
using fibrin-based bioink for • On the fly crosslinking 85
drug screening (Fig. 6) • Minimizes shear-stress

J
our
nal
Name,
[yea
r][
,vol
.
],1–13 | 7
Lab on a Chip Page 8 of 13
View Article Online
DOI: 10.1039/C9LC01184F

Lab on a Chip Accepted Manuscript


Fig. 5 (a) Microfluidic extrusion system composed of (b) the microfluidic printing head and (c) the co-axial adapter. (d) Zonal structure of the
osteochondral (OC) interface and the 3D bioprinted design of the layered scaffold for the regeneration of the cartilaginous part of the OC region. (e)
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

Hematoxylin and eosin staining with insets showing higher magnification. (i) Shows the 3D bioprinted structure where the black arrows in the inset
point to cartilage cell differentiation in the trochlea of the knee implant. In the control implant (ii) this differentiation is missing and a white region
is seen. (Reprinted with permission from 84 )

is at a minimum at the centre of the flow profile. Microfluidic- bladder smooth muscle cells in a single step using a multi-channel
enhanced 3D bioprinting (MF3D) methods have thus become a co-axial extrusion system 107 . In their setup, the crosslinking
new focus in tissue printing applications, with a multitude of pa- agent (CaCl2 ) was delivered from the core of the printing needle,
pers being published in recent years (Table 1). whereas two bioinks were simultaneously extruded from concen-
tric rings.
In a commonly used approach, co-axial wet-spinning extrusion
3D bioprinting delivers a bioink from the core of the printing In their recent paper, Idaszek et al. developed a microfluidic
nozzle and an ionic crosslinking solution is sheathed on the side printing head with a serpentine-based passive mixer with two-
(Fig 4(a)), allowing the crosslinking to occur only at the noz- inlets and a co-axial adapter for precise material crosslinking
zle tip. The solutions can also be inversed so that hollow hy- (Fig. 5(a-c)) 84 . Their device was designed to address the problem
drogel fibres are produced. An advantage of this method is that of long-term stability in regenerated osteochondral (OC) tissue.
it permits for lower viscosity bioinks to be used which is benefi- By allowing for multiple bioinks to be delivered either individ-
cial for reducing nozzle clogging 73,106 . For instance, Zhang et al. ually, or at the same time and rapidly mixed, it enables layers
generated endothelialized myocardium for blood vessels through or continuous gradients of chemical, mechanical and biological
the use of co-axial printing technology 75 . They first generated cues to be extruded such that scaffolds with very high shape fi-
multilayer hydrogel microfibrous scaffolds, where the bioink was delity and cell viability can be fabricated to mimic the compart-
composed of a mixture of hydrogel precursors (alginate, GelMA, mentalized zonal microstructure and composition of OC tissue
and a photo-initiator) and encapsulating human umbilical vein (Fig. 5(d)). In comparison to the SCT3D methods, the microflu-
endothelial cells (HUVECs). The crosslinking agent (CaCl2 ) was idic component provided additional control of chemical reactions
simultaneously dispensed through the outer sheath flow of the and provided higher resolution due to a better control over cell
printing needle to crosslink the alginate component in the bioink type positioning. Furthermore, they were able to develop bioinks
(Fig 4(b) left). A second chemical crosslinking step through UV that mimic the composition of different zones that are present
illumination was then used to crosslink the GelMA component in the native articular cartilage and the 3D bioprinted construct
(Fig 4(b) right). Following two weeks of culture, the HUVECs in demonstrated heterogenous differentiation of stem cells, show-
the bioprinted scaffold migrated to the peripheries of the microfi- ing promise for the regeneration of the interfacial tissue in vitro
bres leading to the formation of a pattern similar to blood vessel and in vivo. The presence of differentiating cartilaginous cells at
walls (Fig 4(c & d)). The cardiomyocytes could then be seeded the implantation site in the knees, was shown through H and E
into the endothelialized scaffold structure to form engineered en- staining (Fig. 5(e) i). The differentiation was not present in the
dothelialized myocardium that structurally resembles native my- control group (Fig. 5(e) ii).
ocardium (Fig 4(e & f)). They found that the cardiomyocytes that
Lee et al. utilized a multi-inlet microfluidic printhead with a
grew on the bioprinted microfibrous scaffold strongly expressed
co-axial flow-focusing extruder giving them the choice of three
proteins required for proper contractile function as well as proper
different cell-types, or biomaterials, during the printing process,
inter-cellular conductive function. Not only did the microfluidic
and crosslinking of the bioink at the nozzle tip (Fig. 6(a)) 85 . They
nozzle allow for the structure of the native tissue to be replicated,
used this technology to fabricate a fibrin-based 3D printed model
it demonstrated the possibility of becoming a functional tissue.
of glioblastoma multiforme (GBM), a deadly primary brain can-
In a more recent work, Pi et al. produced tubular urothelial cer, for cellular characterization and drug screening. Over a 12-
tissues composed of inner human muscle cells and outer human day culture period, the 3D bioprinted glioblastoma cells displayed

8| J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 9 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

high levels of cell viability and demonstrated the ability to spon-


taneously form increasingly large and dense spheroids (Fig. 6(b))
with high levels of protein markers associated with cancer stem
cells and metastatic invasiveness. The cells ability to naturally
form spheroids provides accurate in vivo tumor propagation char-
acteristics that can be used to study cell-cell interactions within
tumors, as well as drug penetration, response and resistance.
In order to package multiple cell types into heterotypic arrays,
Snyder et al. developed an improved multi-material bioprinter by
incorporating a microfluidic printing nozzle with multiple inlets
and one outlet to exploit the laminar flow to print several ma-
terials without mixing 83 . Their droplet-based MF3D platformed

Lab on a Chip Accepted Manuscript


allowed for packaging of multiple cell types to be printed into
200 nl droplets to model human liver cells for pharmacokinetic
studies. They were also able to operate their device in extrusion
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

mode to fabricate 3D hetero-cellular scaffolds. Miri et al. created Fig. 6 Microfluidic-enhanced co-axial bioprinter nozzle. (a) Depicts the
a unique microfluidic chip for the consecutive printing of different microfluidic chip and the reaction between the bioink and cross-linker.
bioinks in a digital micromirror device (DMD) -based projection (b) Is the 3D bioprinted glioblastoma cells at day 0 (top) and at day 12
(bottom) of culture. Over time they observed a spontaneous formation of
bioprinting setup (Fig. 7(a)) 79 . An elastomeric membrane was
increasingly large and dense spheroids. The formation of such spheroids
also included in the channel design to seal the microfluidic cham- within the scaffolds demonstrates not only high cell viability but also the
ber during the exchange of bioinks and the transitional channel cells ability to naturally form spheroids despite being printed as single
washing steps. The device was used to create a variety of multi cells. (Reprinted with permission from 85 )
cell-type models, for example a tumour model was fabricated by
printing scattered breast cancer cells (MCF7) in GelMA and the
structure was further seeded with HUVECS (Fig. 7(b)). A muscle Conclusions and Outlook
model and a tendon-to-bone insertion model were also carried
out to demonstrate the diversity and success of applications using 3D bioprinting is a rapidly growing technology for the develop-
this bioprinting platform (Fig. 7(a)). The post-printing cell viabil- ment of engineered tissues and organs. In this review, we summa-
ity for all models indicated all cell types maintained satisfactory rized the development of multi-material 3D bioprinting methods
proliferation and metabolic activity after seven days of culture. with an emphasis on multi-cartridge systems, and more specif-
ically on microfluidic-enhanced printing nozzles. Despite many
In a very detailed work, Serex et al. demonstrated how mi- advancements made in recent years, there are still many limita-
crofluidics can be used to increase the functionality of current tions and hurdles that need to be overcome in order to create
extrusion-based 3D printers by integrating various microfluidic completely functional, biomimetic tissues and organs that can be
components into the printhead 80 . Multi-inlet microfluidic chips used for human implantation and for reliable drug modelling and
were designed for material switching, hydrodynamic focusing for testing.
controlled material thickness, a herringbone micro-mixer and a One of the major enduring restrictions is the inability to pro-
concentrator using a cross-flow filter. The implementation of hy- duce tissue constructs with highly complex microarchitectures
drodynamic focusing introduces core width adjustability during with precise control over local cellular composition. Being able
the printing process by simply varying the ratio between the core to precisely place individual cells at specific locations to perfectly
and lateral flows. This allows for increased resolution of the de- mimic the micro-scale hierarchical structures of the natural tis-
sired material at specific locations in printed structure. sue is the next stage that researchers should focus on to improve
The Herringbone micro-mixer allows even very viscous materi- the current technology. In order for this to be achieved, a plat-
als (glycerol with a viscosity of approximately 1.412 Pa s 108 ) to be form allowing for multiple cell types to be printed in a controlled,
mixed right before being extruded. This represents an advance on and patterned way, is critical. Furthermore, a built-in method to
a previous work in which Serex et al. had used a meander-based provide nutrients, growth factors, or buffer changes, during the
static micro-mixer to perform mixing of slow reacting materials printing process would be advantageous. In addition, since most
for the 3D printing of carboxymethylcellulose-based cryogels 109 . current methods use devices with nozzles, strategies to avoid noz-
Lastly, the concentrator uses a crossflow filter to remove excess zle clogging and to reduce shear stress on cell populations during
fluid while keep particles/cells within the centre of the channel. the printing process are of great importance.
Concentrating the particles in a composite material right before Microfluidic-enhanced bioprinting methods are a promising ap-
printing makes it possible to work with diluted solutions, which proach that may allow the development of hybrid 3D bioprinting
have lower viscosities, right until the printing happens, reducing platforms capable of printing multiple materials, and/or multiple
the risk of clogging. This simplifies the printing process but still cell types, and to create constructs with controlled heterogenous
produces constructs with the desired composition and reduces the cell populations in more of a bottom-up approach. Some first re-
amount of unnecessary fluid. sults regarding these advantages have been reviewed here; for

J
our
nal
Name,
[yea
r][
,vol
.
],1–13 | 9
Lab on a Chip Page 10 of 13
View Article Online
DOI: 10.1039/C9LC01184F

Lab on a Chip Accepted Manuscript


Fig. 7 Microfluidic-enhanced multi-material bioprinter. Schematic showing the bioprinter setup with the microfluidic device (left) with different tissue
models showing, in order, for the tumor (a) and muscle (b) models the schematic of the tissue model, the mask for printing, the bioprinted construct,
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

and lastly the bright-field optical stained images depicting the cells present in the construct. In the case of the tendon model (c), the schematic of the
tissue model, the mask for printing, the bright-field optical image showing the different cells present, and lastly a measurement of the cell proliferation.
The MCF7 cells are stained in blue and the HUVECs in green for the tumor model in (a). For the muscle model in (b), the C2C12 cells are red and the
fibroblasts are blue. In the tendon model in (c), the osteoblasts are blue, the MSCs red, and the fibroblasts in green; the inset shows a magnified image
of the region hosting fibroblasts, where the cells were stained for f-actin (green) and nuclei (blue). All cell types maintained satisfactory proliferation
and metabolic activity post-bioprinting. (Reprinted with permission from 79 )

example in the successful fabrication of osteochondral (OC) tis- microfluidic capabilities, in which external force fields, including
sue by Idaszek et al., where microlfuidics allowed for the com- but not limited to, electric, acoustic, or magnetic approaches, of-
partmentalized zonal microstructure and composition of the OC fer further control of fluid and cell motion within the microfluidic
tissue to be reconstructed. Additionally, the integration of mi- printer head, furthering our ability to accurately mimic natural
crofluidic devices allows for quick and easy medium change as tissues in both microstructure and microenvironment.
well as the delivery of growth factors and other nutrients during
the printing process. This would be extremely beneficial to min- Conflicts of interest
imize cell damage and increase cell survival. For example, Miri There are no conflicts to declare.
et al. utilized a pneumatic valve in their microchannel to change
between materials, however this technology could be utilized to Acknowledgements
provide nutrients in a controlled and timely manner. This could
The authors are grateful for their support through the Monash -
not be incorporated into the traditional printing nozzles but re-
Warwick Alliance Accelerator scheme.
quires the addition of a microfluidic component. Another advan-
tage of a multi-material microfluidic-enhanced printer rather than Notes and references
a multi-cartridge system, is that the transition between materials
1 H. Cui, S. Miao, T. Esworthy, X. Zhou, S.-J. Lee, C. Liu, Z.-X.
in the latter is not smooth and rather slow, whereas microfluidic
Yu, J. P. Fisher, M. Mohiuddin and L. G. Zhang, Advanced
printing would allow for a seamless transition.
drug delivery reviews, 2018, 132, 252–269.
Although there has been creditable progress in developing new
2 R. Cortesini, Transplant Immunology, 2005, 15, 81–89.
multi-material 3D bioprinting platforms which have been success-
fully used to fabricate tissue constructs, taking the technology 3 S. Vijayavenkataraman, W.-C. Yan, W. F. Lu, C.-H. Wang and
from the laboratory bench to real world applications still requires J. Y. H. Fuh, Advanced Drug Delivery Reviews, 2018, 132,
focused efforts, specifically towards achieving functional form. 296–332.
For example current research demonstrates cell growth and dif- 4 H. Resources and S. Administration, Organ Dona-
ferentiation over a series of days in the 3D bioprinted constructs tion Statistics, 2019, https://www.organdonor.gov/
however, the published works do not provide data about cell func- statistics-stories/statistics.html.
tionality. Researchers will therefore not only need to work on ad- 5 P. Kačarević, P. M. Rider, S. Alkildani, S. Retnasingh,
vancing the technology, but also on insuring that the cells have R. Smeets, O. Jung, Z. Ivanišević and M. Barbeck, Materi-
not undergone any functional changes during the printing pro- als (Basel, Switzerland), 2018, 11, 2199.
cess, which would negatively impact the fabricated tissue. 6 G. Zhang, Organogenesis, 2012, 8, 101–102.
Significant advantages have already been gained through the 7 S. Vijayavenkataraman, W. F. Lu and J. Y. H. Fuh, Biofabri-
use of passive microfluidic elements within 3D bioprinting, the cation, 2016, 8, 032001.
control of the microenvironment and the delivery of multiple cell 8 L. Ferroni, A. Della Puppa, D. D’Avella, M. Isola, R. Scienza,
types has led to the fabrication of a range of functional tissues. We C. Gardin and B. Zavan, Artificial Organs, 2015, 39, E114–
expect that future developments will include further use of active E126.

10 | J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 11 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

9 M. Nie and S. Takeuchi, Biofabrication, 2018, 10, 044103. 34 X. Cui, G. Gao, T. Yonezawa and G. Dai, JoVE, 2014, e51294.
10 K. Athanasiou, E. A., H. Rajalakshmanan, H. Pasha and J. C. 35 T. K. Merceron, M. Burt, Y.-J. Seol, H.-W. Kang, S. J. Lee,
Hu, Annu Rev Biomed Eng, 2013, 15, 115–136. J. J. Yoo and A. Atala, Biofabrication, 2015, 7, 035003.
11 S. Wüst, R. Müller and S. Hofmann, J Funct Biomater, 2011, 36 J. A. Phillippi, E. Miller, L. Weiss, J. Huard, A. Waggoner and
2, 119–154. P. Campbell, STEM CELLS, 2008, 26, 127–134.
12 E. Hoch, G. E. Tovar and K. Borchers, European Journal of 37 S. Levenberg, J. Rouwkema, M. Macdonald, E. S. Garfein,
Cardio-Thoracic Surgery, 2014, 46, 767–778. D. S. Kohane, D. C. Darland, R. Marini, C. A. van Blitter-
13 C. Y. Liaw and M. Guvendiren, Biofabrication, 2017, 9, swijk, R. C. Mulligan, P. A. D’Amore and R. Langer, Nature
024102. Biotechnology, 2005, 23, 879–884.
14 C. Balletti, M. Ballarin and F. Guerra, 3D printing: State of 38 F.-Y. Hsieh, H.-H. Lin and S.-h. Hsu, Biomaterials, 2015, 71,
the art and future perspectives, 2017. 48–57.
15 T. D. Ngo, A. Kashani, G. Imbalzano, K. T. Q. Nguyen and 39 S. England, A. Rajaram, D. J. Schreyer and X. Chen, Bioprint-

Lab on a Chip Accepted Manuscript


D. Hui, Composites Part B: Engineering, 2018, 143, 172–196. ing, 2017, 5, 1–9.
16 G. Cidonio, M. Glinka, J. I. Dawson and R. O. C. Oreffo, 40 Y.-B. Lee, S. Polio, W. Lee, G. Dai, L. Menon, R. S. Carroll
Biomaterials, 2019, 209, 10–24. and S.-S. Yoo, Experimental Neurology, 2010, 223, 645–652.
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

17 H.-W. Kang, S. J. Lee, I. K. Ko, C. Kengla, J. J. Yoo and 41 L. Gibot, T. Galbraith, B. Kloos, S. Das, D. A. Lacroix, F. A.
A. Atala, Nature Biotechnology, 2016, 34, 312. Auger and M. Skobe, Biomaterials, 2016, 78, 129–139.
18 E. S. Bishop, S. Mostafa, M. Pakvasa, M. J. Luu, Hue 42 E. A. Bulanova, E. V. Koudan, J. Degosserie, C. Heymans,
H.and Lee, J. M. Wolf, G. A. Ameer, T.-C. He and R. R. Reid, F. D. A. S. Pereira, V. A. Parfenov, Y. Sun, Q. Wang, S. A.
Genes Diseases, 2017, 4, 185–195. Akhmedova, I. K. Sviridova, N. S. Sergeeva, G. A. Frank, Y. D.
19 S. V. Murphy and A. Atala, Nature Biotechnology, 2014, 32, Khesuani, C. E. Pierreux and V. A. Mironov, Biofabrication,
773. 2017, 9, 034105.
20 F. P. W. Melchels, M. A. N. Domingos, T. J. Klein, J. Malda, 43 J. Song and J. R. Millman, Biofabrication, 2016, 9, 015002.
P. J. Bartolo and D. W. Hutmacher, Progress in Polymer Sci- 44 C.-Y. Kuo, H. Baker, M. H. Fries, J. J. Yoo, P. C. W. Kim and
ence, 2012, 37, 1079–1104. J. P. Fisher, Tissue Engineering Part B: Reviews, 2016, 23,
21 S. Derakhshanfar, R. Mbeleck, K. Xu, X. Zhang, W. Zhong 294–306.
and M. Xing, Bioactive Materials, 2018, 3, 144–156. 45 W. Lee, J. C. Debasitis, V. K. Lee, J.-H. Lee, K. Fischer, K. Ed-
22 B. K. Gu, D. J. Choi, S. J. Park, M. S. Kim, C. M. Kang and minster, J.-K. Park and S.-S. Yoo, Biomaterials, 2009, 30,
C. H. Kim, Biomater Res, 2016, 20, 12. 1587–1595.
23 B. Zhang, L. Gao, L. Ma, Y. Luo, H. Yang and Z. Cui, Engi- 46 M. Rimann, E. Bono, H. Annaheim, M. Bleisch and U. Graf-
neering, 2019, 5, 777–794. Hausner, Journal of Laboratory Automation, 2015, 21, 496–
24 H. Gudapati, M. Dey and I. Ozbolat, Biomaterials, 2016, 102, 509.
20–42. 47 N. Cubo, M. Garcia, J. F. del Cañizo, D. Velasco and J. L.
25 I. T. Ozbolat and M. Hospodiuk, Biomaterials, 2016, 76, Jorcano, Biofabrication, 2016, 9, 015006.
321–343. 48 L. Horváth, Y. Umehara, C. Jud, F. Blank, A. Petri-Fink and
26 S. Azizi Machekposhti, S. Mohaved and R. J. Narayan, Expert B. Rothen-Rutishauser, Scientific Reports, 2015, 5, 7974.
Opinion on Drug Discovery, 2019, 14, 101–113. 49 A. Griffith, Linda G.and Wells and D. B. Stolz, Hepatology,
27 T. Jiang, J. G. Munguia-Lopez, S. Flores-Torres, J. Kort- 2014, 60, 1426–1434.
Mascort and J. M. Kinsella, Applied Physics Reviews, 2019, 50 A. Faulkner-Jones, C. Fyfe, D.-J. Cornelissen, J. Gardner,
6, 011310. J. King, A. Courtney and W. Shu, Biofabrication, 2015, 7,
28 W. Luo, H. Liu, C. Wang, Y. Qin, Q. Liu and J. Wang, Ad- 044102.
vanced Engineering Materials, 2019, 21, 1900019. 51 V. Kasyanov, K. Brakke, T. Vilbrandt, R. Moreno-Rodriguez,
29 P. Datta, B. Ayan and I. T. Ozbolat, Acta Biomaterialia, 2017, A. Nagy-Mehesz, R. Visconti, R. Markwald, I. Ozolanta, R. A.
51, 1–20. Rezende, A. L. Lixandrão Filho, P. Inforçati Neto, F. D. A. S.
30 S. Lopa, C. Mondadori, V. L. Mainardi, G. Talo, M. Costan- Pereira, D. T. Kemmoku, J. V. L. da Silva and V. Mironov,
tini, C. Candrian, W. Swieszkowski and M. Moretti, Stem Virtual and Physical Prototyping, 2011, 6, 197–213.
Cells Int, 2018, 2018, 6594841. 52 B. Duan, L. A. Hockaday, K. H. Kang and J. T. Butcher, Jour-
31 D. W. Hutmacher, Biomaterials, 2000, 21, 2529–2543. nal of Biomedical Materials Research Part A, 2013, 101A,
1255–1264.
32 M. Neufurth, X. Wang, H. C. Schröder, Q. Feng, B. Diehl-
Seifert, T. Ziebart, R. Steffen, S. Wang and W. E. G. Müller, 53 R. Gaebel, N. Ma, J. Liu, J. Guan, L. Koch, C. Klop-
Biomaterials, 2014, 35, 8810–8819. sch, M. Gruene, A. Toelk, W. Wang, P. Mark, F. Wang,
B. Chichkov, W. Li and G. Steinhoff, Biomaterials, 2011, 32,
33 S. Rhee, J. L. Puetzer, B. N. Mason, C. A. Reinhart-King and
9218–9230.
L. J. Bonassar, ACS Biomaterials Science Engineering, 2016,
2, 1800–1805. 54 R. Gaetani, P. A. Doevendans, C. H. G. Metz, J. Alblas,

J
our
nal
Name,
[yea
r][
,vol
.
],
1–13 | 11
Lab on a Chip Page 12 of 13
View Article Online
DOI: 10.1039/C9LC01184F

E. Messina, A. Giacomello and J. P. G. Sluijter, Biomaterials, 045008.


2012, 33, 1782–1790. 77 C. Arrigoni, M. Gilardi, S. Bersini, C. Candrian and
55 C. Wu, B. Wang, C. Zhang, R. A. Wysk and Y.-W. Chen, Crit- M. Moretti, Stem Cell Rev Rep, 2017, 13, 407–417.
ical Reviews in Biotechnology, 2017, 37, 333–354. 78 W. Liu, Y. S. Zhang, M. A. Heinrich, F. De Ferrari, H. L. Jang,
56 C. R. Marti-Figueroa and R. S. Ashton, Acta biomaterialia, S. M. Bakht, M. M. Alvarez, J. Yang, Y. C. Li, G. Trujillo-de
2017, 54, 35–44. Santiago, A. K. Miri, K. Zhu, P. Khoshakhlagh, G. Prakash,
57 D. Mark, S. Haeberle, G. Roth, F. von Stetten and H. Cheng, X. Guan, Z. Zhong, J. Ju, G. H. Zhu, X. Jin, S. R.
R. Zengerle, Chem. Soc. Rev., 2010, 39, 1153–1182. Shin, M. R. Dokmeci and A. Khademhosseini, Adv Mater,
58 J. M. R. Rickel, A. J. Dixon, A. L. Klibanov and J. A. Hossack, 2017, 29, year.
Lab on a chip, 2018, 18, 2653–2664. 79 A. K. Miri, D. Nieto, L. Iglesias, H. Goodarzi Hosseinabadi,
59 V. Gnyawali, M. Saremi, M. C. Kolios and S. S. H. Tsai, Biomi- S. Maharjan, G. U. Ruiz-Esparza, P. Khoshakhlagh, A. Man-
crofluidics, 2017, 11, 034104. bachi, M. R. Dokmeci, S. Chen, S. R. Shin, Y. S. Zhang and

Lab on a Chip Accepted Manuscript


60 S. Hong, P.-H. Tsou, C.-K. Chou, H. Yamaguchi, C. B. Su, M.- A. Khademhosseini, Adv. Mater., 2018, 30, 1800242.
C. Hung and J. Kameoka, Biomicrofluidics, 2012, 6, 24132– 80 L. Serex, A. Bertsch and P. Renaud, Micromachines, 2018, 9,
24132. 11.
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

61 A. Yaghmur, A. Ghazal, R. Ghazal, M. Dimaki and W. E. 81 R. Attalla, E. Puersten, N. Jain and P. R. Selvaganapathy,
Svendsen, Physical Chemistry Chemical Physics, 2019, 21, Biofabrication, 2018, 11, 015012.
13005–13013. 82 M. Costantini, S. Testa, P. Mozetic, A. Barbetta, C. Fuoco,
62 J. M. Martel, K. C. Smith, M. Dlamini, K. Pletcher, J. Yang, E. Fornetti, F. Tamiro, S. Bernardini, J. Jaroszewicz,
M. Karabacak, D. A. Haber, R. Kapur and M. Toner, Scientific W. Świ˛eszkowski, M. Trombetta, L. Castagnoli, D. Seliktar,
Reports, 2015, 5, 11300. P. Garstecki, G. Cesareni, S. Cannata, A. Rainer and C. Gar-
63 X. Wang, Z. Liu and Y. Pang, RSC Advances, 2017, 7, 29966– gioli, Biomaterials, 2017, 131, 98–110.
29984. 83 J. Snyder, A. R. Son, Q. Hamid, H. Wu and W. Sun, Biofabri-
64 C. Zhao, Z. Ge and C. Yang, Micromachines, 2017, 8, 28. cation, 2016, 8, 015002.
65 T. J. Johnson, D. Ross and L. E. Locascio, Anal Chem, 2002, 84 J. Idaszek, M. Costantini, T. A. Karlsen, J. Jaroszewicz,
74, 45–51. C. Colosi, S. Testa, E. Fornetti, S. Bernardini, M. Seta,
66 J. Melin, G. Gimenez, N. Roxhed, W. van der Wijngaart and K. Kasarełło, R. Wrzesień, S. Cannata, A. Barbetta, C. Gar-
G. Stemme, Lab on a chip, 2004, 4, 214–9. gioli, J. E. Brinchman and W. Świ˛eszkowski, Biofabrication,
2019, 11, 044101.
67 S. H. Wong, M. C. L. Ward and C. W. Wharton, Sensors and
Actuators B: Chemical, 2004, 100, 359–379. 85 C. Lee, E. Abelseth, L. de la Vega and S. M. Willerth, Materi-
als Today Chemistry, 2019, 12, 78–84.
68 V. Mengeaud, J. Josserand and H. H. Girault, Anal Chem,
2002, 74, 4279–86. 86 E. Kastner, R. Kaur, D. Lowry, B. Moghaddam, A. Wilkinson
and Y. Perrie, International Journal of Pharmaceutics, 2014,
69 X. Wang, J. Liu, P. Wang, A. deMello, L. Feng, X. Zhu,
477, 361–368.
W. Wen, R. Kodzius and X. Gong, Genes (Basel), 2018, 9,
283. 87 S. Deshpande, Y. Caspi, A. E. Meijering and C. Dekker, Nat
Commun, 2016, 7, 10447.
70 J. Ma, Y. Wang and J. Liu, RSC Advances, 2018, 8, 21712–
21727. 88 E. Kang, G. S. Jeong, Y. Y. Choi, K. H. Lee, A. Khademhosseini
and S.-H. Lee, Nature Materials, 2011, 10, 877.
71 C. Mandrycky, Z. Wang, K. Kim and D.-H. Kim, Biotechnol
Adv, 2016, 34, 422–434. 89 K. H. Lee, S. J. Shin, Y. Park and S.-H. Lee, Small, 2009, 5,
1264–1268.
72 E. Sodupe-Ortega, A. Sanz-Garcia, A. Pernia-Espinoza and
C. Escobedo-Lucea, Materials (Basel), 2018, 11, year. 90 M. Yamada, R. Utoh, K. Ohashi, K. Tatsumi, M. Yamato,
T. Okano and M. Seki, Biomaterials, 2012, 33, 8304–8315.
73 M. Costantini, C. Colosi, W. Świ˛eszkowski and A. Barbetta,
Biofabrication, 2018, 11, 012001. 91 A. Kobayashi, K. Yamakoshi, Y. Yajima, R. Utoh, M. Yamada
and M. Seki, Journal of Bioscience and Bioengineering, 2013,
74 Y.-J. Seol, H. Lee, J. S. Copus, H.-W. Kang, D.-W. Cho,
116, 761–767.
A. Atala, S. J. Lee and J. J. Yoo, Bioprinting, 2018, 10,
e00028. 92 A. Sheikhi, J. de Rutte, R. Haghniaz, O. Akouissi, A. Sohrabi,
D. Di Carlo and A. Khademhosseini, Biomaterials, 2019, 192,
75 Y. S. Zhang, A. Arneri, S. Bersini, S.-R. Shin, K. Zhu, Z. Goli-
560–568.
Malekabadi, J. Aleman, C. Colosi, F. Busignani, V. Dell’Erba,
C. Bishop, T. Shupe, D. Demarchi, M. Moretti, M. Rasponi, 93 B. Kang, J. Shin, H.-J. Park, C. Rhyou, D. Kang, S.-J. Lee,
M. R. Dokmeci, A. Atala and A. Khademhosseini, Biomateri- Y.-s. Yoon, S.-W. Cho and H. Lee, Nature Communications,
als, 2016, 110, 45–59. 2018, 9, 5402.
76 R. Zimmermann, C. Hentschel, F. Schrön, D. Moedder, 94 H. Song, M. R. Bringer, J. D. Tice, C. J. Gerdts and R. F.
T. Büttner, P. Atallah, T. Wegener, T. Gehring, S. Howitz, Ismagilov, Applied physics letters, 2003, 83, 4664–4666.
U. Freudenberg and C. Werner, Biofabrication, 2019, 11, 95 J. Ou, G. R. Moss and J. P. Rothstein, Phys Rev E Stat Nonlin

12 | J
our
nal
Name,
[yea
r][
,vol
.
],1–13
Page 13 of 13 Lab on a Chip
View Article Online
DOI: 10.1039/C9LC01184F

Soft Matter Phys, 2007, 76, 016304. S. R. Quake, Analytical Chemistry, 2007, 79, 8557–8563.
96 S. Sivashankar, S. Agambayev, Y. Mashraei, E. Q. Li, S. T. 104 A. Au, H. Lai, B. Utela and A. Folch, Micromachines, 2011, 2,
Thoroddsen and K. N. Salama, Biomicrofluidics, 2016, 10, 179–220.
034120. 105 R. R. Collino, T. R. Ray, R. C. Fleming, J. D. Cornell, B. G.
97 W. Saadi, S. W. Rhee, F. Lin, B. Vahidi, B. G. Chung and N. L. Compton and M. R. Begley, Extreme Mechanics Letters, 2016,
Jeon, Biomed Microdevices, 2007, 9, 627–35. 8, 96–106.
98 C. Joanne Wang, X. Li, B. Lin, S. Shim, G. L. Ming and 106 C. Colosi, S. R. Shin, V. Manoharan, S. Massa, M. Costantini,
A. Levchenko, Lab Chip, 2008, 8, 227–37. A. Barbetta, M. R. Dokmeci, M. Dentini and A. Khademhos-
99 N. Zaari, P. Rajagopalan, S. Kim, A. Engler and J. Wong, seini, Adv Mater, 2016, 28, 677–84.
Advanced Materials, 2004, 16, 2133–2137. 107 Q. Pi, Y. Zhang, X. Hou, F. Robledo-Padilla, X. Liu, S. Ma-
100 C. G. Sip, N. Bhattacharjee and A. Folch, Biomicrofluidics, harjan, R. Parra, S. Hassan, N. Hu, A. M. van Genderen and
2011, 5, 22210. A. Khademhosseini, Adv. Mater., 2018.

Lab on a Chip Accepted Manuscript


101 C. S. Niman, J. P. Beech, J. O. Tegenfeldt, P. M. G. Curmi, 108 A. G. M. Ferreira, A. P. V. Egas, I. M. A. Fonseca, A. C. Costa,
D. N. Woolfson, N. R. Forde and H. Linke, Lab on a Chip, D. C. Abreu and L. Q. Lobo, The Journal of Chemical Thermo-
2013, 13, 2389–2396. dynamics, 2017, 113, 162–182.
Published on 18 May 2020. Downloaded on 5/19/2020 7:38:13 AM.

102 R. R. Collino, N. Reilly-Shapiro, B. Foresman, K. Xu, M. Utz, 109 L. Serex, T. Braschler, A. Filippova, A. Rochat, A. Béduer,
J. P. Landers and M. R. Begley, Lab Chip, 2013, 13, 3668–74. A. Bertsch and P. Renaud, Advanced Materials Technologies,
103 R. Gómez-Sjöberg, A. A. Leyrat, D. M. Pirone, C. S. Chen and 2018, 3, 1700340.

J
our
nal
Name,
[yea
r][
,vol
.
],
1–13 | 13

You might also like