You are on page 1of 21

Review

www.advmat.de

Bioprinting of Complex Multicellular Organs with


Advanced Functionality—Recent Progress
and Challenges Ahead
Luiz E. Bertassoni

1. Introduction
Bioprinting has emerged as one of the most promising strategies for
fabrication of functional organs in the lab as an alternative to transplant
The field of biofabrication has emerged
organs. While progress in the field has mostly been restricted to a few in recent years to encompass a number of
miniaturized tissues with minimal biological functionality until a few years advanced engineering strategies, geared
ago, recent progress has advanced the concept of building three-dimensional toward building biomimetic cellular tissue
multicellular organ complexity remarkably. This review discusses a series constructs.[1–5] Bioprinting is one, and
arguably the most prominent, of these
of milestones that have paved the way for bioprinting of tissue constructs
methods, and has generated enormous
that have advanced levels of biological and architectural functionality. attention in the past decade.[6–10] The broad
Critical materials, engineering and biological challenges that are key to definition of biofabrication converges con-
addressing the desirable function of engineered organs are presented. These cepts that were independently developed
are discussed in light of the many difficulties to replicate the heterotypic for fields such as regenerative medicine,
organization of multicellular solid organs, the nanoscale precision of the developmental biology, and process engi-
neering, among other themes.[3] Broadly
extracellular microenvironment in hierarchical tissues, as well as the
speaking, each of these areas evolved at
advantages and limitations of existing bioprinting methods to adequately their own pace to address different needs
overcome these barriers. In summary, the advances of the field toward in their respective industries. Only rela-
realistic manufacturing of functional organs have never been so extensive, tively recently have these areas converged
and this manuscript serves as a road map for some of the recent progress into what is now a mainstream method in
biomedical and regenerative engineering.
and the challenges ahead.
From a chronological perspective,
it is straightforward to trace back the
convergence of the themes that have
evolved into what defines contemporary
bioprinting. Early in the development of conventional office
L. E. Bertassoni printers, biomedical researchers were already contemplating
Division of Biomaterials and Biomechanics the prospects of dispensing cells out of conventional inkjet
School of Dentistry cartridges (cytoscribing), proposed by Klebe et  al. back in the
Oregon Health and Science University late 80s.[11] Obviously, the process at the time did not intend
Portland, OR 97201, USA
E-mail: bertasso@ohsu.edu to fabricate functional tissues with viable cells; but arguably
L. E. Bertassoni that paved the way for far more advanced methods of cell and
Department of Biomedical Engineering tissue printing that are utilized nowadays, and which will
School of Medicine be discussed more broadly in this review. The emergence of
Oregon Health and Science University widespread methods to isolate, expand, and differentiate cells
Portland, OR 97239, USA
in the lab,[12] have contributed greatly to the biological devel-
L. E. Bertassoni
Center for Regenerative Medicine
opments that enabled the establishment of bioprinting early
Oregon Health and Science University on.[13] Similarly, the ability to synthesize and manipulate the
Portland, OR 97239, USA structural, physical, and compositional properties of natural
L. E. Bertassoni and synthetic biomolecules, have also contributed dramatically
Cancer Early Detection Advanced Research (CEDAR) to the evolution of the field.[4,14,15] Another pillar, obviously,
Knight Cancer Institute has been the evolution of methods to dispense materials, both
Oregon Health and Science University
Portland, OR 97239, USA synthetic and natural in 3D, irrespective of the presence of
The ORCID identification number(s) for the author(s) of this article
cells;[16] but arguably, it is the intersection of these constitu-
can be found under https://doi.org/10.1002/adma.202101321. ents and processes, rather than an isolated development, that
catapulted the field of bioprinting into a mainstream line of
DOI: 10.1002/adma.202101321 science and technology.

Adv. Mater. 2022, 34, 2101321 2101321  (1 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 1.  A) General view of high-density cell-laden hydrogel and tissue spheroid printing in air and fluid. A) Reproduced with permission.[26] Copyright
2009, Elsevier. B) Aspiration assisted bioprinting showing the transfer of cell spheroids to fabricate high-density tubular construct in a granular microgel
supporting bath. B) Reproduced under the terms of the CC-BY Creative Commons Attribution 4.0 International license (https://creativecommons.org/
licenses/by/4.0).[109] Copyright 2021, The Authors, published by Springer Nature. C) Generation of iPSC-derived kidney organoids by extrusion-based
bioprinting. Bright-field (day 7 + 7) and whole-mount immunofluorescence (day 7 + 14) images of manual and bioprinted kidney organoids generated
simultaneously from the same batch of iPSC-derived intermediate mesoderm showing patterning and segmented nephrons. C) Reproduced with
permission.[73] Copyright 2020, The Authors, published by Springer Nature. D) i) Schematic diagram showing cortico-striatal projections in the devel-
oping brain, and in vitro modeling of cortico-striatal projections using human cortical spheroid with human striatal spheroid to form multicellular
assembloids derived from hiPS cells; ii) immunostaining of cortico-striatal at day 83 in low (scale bar: 500 μm) and high magnification (scale bar:
100 μm) showing intertwined axonal projections interconnecting the two different regions. D) Reproduced with permission.[38] Copyright 2020, The
Authors, published by Springer Nature.

For a long time regenerative medicine relied on the use by embryology and developmental cell biology,[25,26] led to the
of biocompatible materials that could be implanted in the ability of manipulating high-density cell aggregates as “control-
body as a supporting scaffold for the invasion and repopula- lable biologic materials”, such as in the form of cell spheroids/
tion of cells from the host, as originally proposed by Langer aggregates,[27–30] embryoid bodies,[31–33] and more recently orga-
and Vacanti in some seminal papers in the 90s.[12,17] With the noids,[34] or even assembloids.[35–38] These latter developments
popularization of densely cellularized materials in the form are largely credited to the early work of Forgacs, Mironov,
of cell-laden hydrogels,[14,18–22] again with substantial contribu- and other pioneers in the area of scaffold-free cell printing
tions from the Langer lab and other active groups in the field (Figure 1a).[25,26] Similarly, this was enabled by a rapid revolu-
in the early-mid 2000s, the concept of “fabricating with bio- tion that the field of 3D printing experienced through the first
logical living matter” became far more attainable. Similarly, and second decades of the current century, mostly driven by
an improved understanding of mechanisms of cell morpho- the expiration of protective patents that held the technology to
genesis[23,24] and the development of methods for manipula- a few pioneering companies profiting from the concept.[39] With
tion of the process of cell and tissue assembly, mostly inspired the widespread popularization of 3D printing technology for

Adv. Mater. 2022, 34, 2101321 2101321  (2 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

polymer plastics,[40] metals,[41] viscous liquids,[42–44] and many in reproducing the microscale complexity of human biology
other synthetic materials, including hydrogels,[45–48] it was only are also presented, focusing mostly on biologic and materials
a question of time until the field of biomedical engineering questions, and less so on hardware challenges. Additionally,
adapted many of the well-developed scaffold materials and the limited ability of existing methods to control the nanoscale
cell-manipulating techniques to enable controlled dispensing heterogeneous and hierarchical organization of the human
of these materials in biologically meaningful structures, and native extracellular micro­ environment is revisited, together
in the presence of living biologic organisms. Remarkable con- with recent solutions to integrate and feed multiple tissue
tributions by Anthony Atala’s group, especially in translating types via an efficient circulatory and vascular system based on
some of these early 3D printing technologies from the bench bioprinting methods. Lastly, this review covers existing chal-
to the clinic, played a major role in facilitating the translation lenges and presents possible solutions that may shine light on
and progress of many of the technologies used today by many potential transformative outcomes for the field of organ and
in the field, especially (but not only) on extrusion bioprinting of tissue engineering via bioprinting.
biocompatible materials for patient implantation.[9] One relevant aspect that is worth pointing out is that despite
Currently, there are many modalities of bioprinting that years of discussions, there is still controversy around the defi-
are available, and existing methods have been combined to nition of bioprinting.[3,57,58] On one hand, there is a school of
result in an ever-increasing list of novel bioprinting strate- thought that defends the perspective that bioprinting pertains
gies; we encourage the reader to refer to recent reviews of to the dispensing of biological material, be it nonliving bio-
current bioprinting methodologies for further details of such logical molecules (proteins, cytokines, etc.) or live components
methods.[7,8,49–51] In a brief summary, from a fundamental tech- (mammalian, bacterial cells, viruses, etc.). On the other hand,
nological perspective, most printers can be linked to one of the there is the competing perspective that bioprinting should refer
four methods: extrusion, light, inkjet, and laser bioprinting. In only to the deposition of biomaterials composed of, at least
principle, extrusion relies on dispensing of fibers in X–Y while in part, living biological organisms, such as the ones referred
the coordinated motion of a build platform or the dispensing above. One agreement that appears to be drawn between these
head moves in Z.[46,52] In light-based bioprinting, cells are competing definitions is that inert synthetic materials that are
loaded to a photocrosslinkable biomaterial, and the exposure of printed for applications in medicine or biology, but are not con-
the photo-monomers to light, either via rastering or a planar stituted of any form of biological matter, should not fall under
photomask, generates an XY pattern of photo­ polymerized the true umbrella of “bio”-printing. This may be seen as a ques-
material, while the build platform moves in Z for a sequenced tion of lower relevance, however, one may argue that the com-
deposition of layers.[53,54] On inkjet bioprinting cells and mate- plexity of the processes and techniques that are associated with
rials of low viscosity are dispensed using a jetting system onto manipulating living biological systems, far exceed those that are
a platform that coordinates the XYZ movement of the pat- associated with printing of non-living matter. Additionally it is
terned substrate.[55] Laser bioprinters, on the other hand, use worth pointing out that these distinct definitions of bioprinting,
a different approach where cells or materials are loaded to a considering cell-rich versus cell-free approaches, speak to dif-
laser-sensitive tape, and as the rastering of such a laser on the ferent objectives of the fabrication process. While cell-free
surface of the cell-covered tape occurs, cells are transferred onto printing requires post-processing of the printed part by cells to
an underlying substrate; the coordinated motion of the laser result in meaningful biological function, bioprinting with cellu-
light with the Z movement of the underlying platform ensures larized materials refers to the goal of direct fabrication of living
the three-dimensionality of the construct.[56] As described ear- tissues and organs, which is, perhaps, more closely aligned
lier, currently there are many variations and combinations of with the overarching goal of controlled organ fabrication. For
these original methods, and these general descriptions have these reasons, this review will use the definition of bioprinting
become far more complex. that considers living cells as a bioink, whereas methods that
While the field has evolved at an increasingly faster pace, 3D print with non-living biological matter will be referred to
our ability to generate truly meaningful biological substitutes simply as 3D printing (without bio), for clarity.
with clinical relevance and size via bioprinting has remained
limited to a few tissues, and these still rely mostly on the ability
of the body to remodel cell-laden tissue constructs.[9] And 2. The Complexity of Bioprinting Heterotypic
while it has been suggested that, perhaps, biomimicry may
Organs with Microscale Resolution
reach a point where increased complexity no longer improves
functional outcomes,[9] replicating tissue microstructure and 2.1. The Complex Cellular Architecture of Solid Organs—The
cellular organization remains a critical challenge in the field. Liver as a Model
Here we discuss the myriad cell, micro­ environment, engi-
neering and materials challenges that, we argue, will require One of the key parameters that define human biology is the
some attention before bioprinting can become an efficient tool complexity of the composition and the biological processes
for fabrication of sizeable complex tissues and organs for both that regulate the human body. Although connective tissues can
clinical and research use. The specific challenges and pos- be perceived as simplified mixtures of proteins and cells that
sible solutions that will enable the manufacturing of hetero- perform relatively simple and repetitive functions of structural
typic tissue constructs with the desired interactions between support, specialized tissues and organs, especially those that are
the multiple populations of cells in an organ are discussed; the solid and cellularly dense, are characterized by their increased
engineering challenges and limitations of existing methods architectural intricacy.

Adv. Mater. 2022, 34, 2101321 2101321  (3 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

For instance, one of the tissues that have received extensive 2.2. Recent Efforts toward Bioprinting of Complex Heterotypic
attention in the bioprinting community, both in academic[59–63] Tissues and Organs
and commercial settings,[64,65] is the liver. So we will use this
organ as a core example to illustrate the inherent challenges of It is critical to acknowledge that the field of bioprinting, and
mimicking the complexity of vital solid organs of comparable most of the other biofabrication methods available, have ena-
difficulty on the microscale and in 3D. bled the engineering of many complexly structured tissue- and
The liver is both subjected to irreparable damage due to the organ-like constructs.[7,9,76,77] However, generally speaking,
improper interaction with drugs and exogenous compounds, we are only beginning to understand and reproduce the true
and is also difficult to transplant and find matching donors. complexity that is endowed by the diverse cell interactions in
Taking the liver as an example, it is easy to identify many of engineered tissue constructs. How bioprinting may enable this
the biological challenges that may be associated with repro- set of complex interactions remains relatively poorly explored.
ducing the native complexity of human organs. The arche- For instance, very early on, we have enabled the extrusion of
typical building block of the liver is the hepatic lobule.[66] In hepatocyte-laden methacrylated gelatin (GelMA) hydrogels with
a single lobular unit, there is a very carefully structured, minimal phenotypic function, determined by the secretion of
three-dimensionally organized layer of a few cells, which are trace levels of albumin by bioprinted liver cells.[59] Others have
closely connected to one another in palisades of hepatocytes. continued in this direction to illustrate the ability of simple bio-
These cells polarize individually, and form cell–cell commu- printed hepatocyte aggregates embedded in gelatin methacry-
nication to enable the formation of biliary canaliculi between loyl hydrogels to metabolize different drugs.[78] In both cases
cells, which will drain the liver metabolites into an adjacent no biomimetic structure or architecture were attempted to be
biliary duct.[67,68] These laterally layered hepatocytes, which reproduced, owning to the relatively low resolution of the extru-
are confined to an area comprising of only a few cells spaced sion printing system that was utilized. In a far more elegant
transversally between sinusoids, are bordered by a plasma-rich approach, Ma et  al. developed a series of light based polym-
region, which is known as the space of Disse.[69] This region is erization bioprinting methods to reproduce the bear-minimal
populated by a low concentration of stellate and dendritic cells, structure of planar lobule-like organization.[79] In more recent
which participate in responding to liver injury and inducing efforts, advanced extrusion systems were developed to dispense
fibrosis, also playing a critical role in development and regen- fiber-like structures with a pre-set liver lobule-like structure.[80,81]
eration.[69] Adjacent to that, there are endothelial sinusoidal On a separate strategy, others have used a scaffold-free extru-
cells, which coat the lumen of the liver circulatory system sion bioprinting approach to dispense high-density aggregates
(sinusoids), and harbor Kupfer cells, connecting the central of liver cells that self-organize to have some liver-like activity.[82]
hepatic vein to the hepatic portal vein, and artery branches.[66] Irrespective of the strategy in question, most, if not all of these
Despite this intricate organization, this actually represents methods, do not reproduce the true complexity that is inherent
only a simplified view of one single liver lobule at a planar to the actual organ, as explained in the earlier paragraphs.
dissecting view, and does not account for the complex three- These bioprinted tissues have been typically composed of 2–3
dimensionality of these same cell–cell and cell–matrix interac- cells types or less, as opposed to five or more, and the extent
tions, that repeat themselves over 1 million times (estimated of structural and biological interactions that are required to
number of individual lobules) in a healthy adult liver. More- ensure adequate organ function remain recognizably difficult
over, this is only restricted to an area of ≈200 µm in diameter to reproduce.[8] This has prevented these bioprinted tissues
(average size of a single liver lobule). Therefore, from a reverse from achieving any meaningful success in reproducing full
engineering standpoint, there is hardly anything more compli- organ functionality, despite their early success in detecting drug
cated in tissue engineering than recreating the architectural toxicity in vitro[78] and replicating some key organ structures.
organization of a solid organ,[70] like the liver. The questions that remain then are, what are the critical bio-
In the example above, there are at least five different pop- logical and engineering challenges that need to be addressed to
ulations of specialized cells that are organized with single enable the deposition of structures that can actually recapitulate
cell precision in space, and it is well established that their the level of complexity seen in a native organ?
expected function depends almost exclusively on the presence
and interaction of the adjacent cell type,[70] as well as the dis-
tance from one cell to the next.[71] Therefore, the challenges 2.3. Culturing Bioprinted Multilineage Tissue Constructs
associated with reproducing this highly intricate set of cell–
cell and cell–matrix interactions are many. And while recent In the context of building the biological complexity described
organoid 3D printing efforts[72–75] have focused on developing above, one solution is the development of methods for rapid
methods to coax stem cells into self-organizing into mature deposition of cells in 3D, at single cell resolution, which we
organ-like cell-aggregates, without the need for positioning discuss further throughout this section and in Section 4 below.
cells with single-cell precision, the concept of 3D bioprinting Associated with this solution, there are challenges related with
is geared toward exactly that—putting cells at their exact place. the basic biology of orchestrating adequate cell–cell commu-
Therefore, a critical question that comes up is how much bio- nication in heterotypic engineered tissues. In other words, if
printing precision is the minimum necessary to reproduce one is to bioprint tissues as biologically complex as the liver,
this set of interactions in a manner that the final outcome is or any other solid organ with multiple cell types, it is vital that
a well-functioning, phenotypically accurate and reproducible we understand the conditions at which these cells can prop-
organ system? erly communicate and function in synergy. For instance, it is

Adv. Mater. 2022, 34, 2101321 2101321  (4 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

well known in tissue engineering that co-cultures of tissues the engineering ones, since reproducing adequate tissue and
with more than 2 cell types can be a challenge if these cells organ function is invariably dependent on biology, and less so
require a different set of nutrients, vitamins, and growth fac- on engineering advances.
tors to perform their desired functions.[83–85] Tissues of high
cellular diversity could require many different compositions
of cell medium, at obviously different ratios, to account for the 2.4. Bioprinting of Organ Constructs toward Advanced
different concentrations of each cell type. The simple optimiza- Functionality
tion of this medium composition poses a significant challenge,
which is not unique to bioprinted constructs, but since printing Although bioprinting of complex multi-lineage hetero-
should facilitate the fabrication of multi-component tissues, typic tissue constructs, like the ones exemplified above,
this becomes an inherent problem in the field. There have been remain highly challenging, bioprinting of relatively sim-
efforts to develop and test universal media, and some of these pler tissue structures, that also have highly intricate func-
alternatives have been developed.[86] But still, this is a critical tion, have been very successfully reported in the literature
biological challenge to enable effective and predictable func- in recent years. For instance, a highly successful report by
tion, as opposed to solely dispensing cells of different lineages the Atala group showed for the first time the bioprinting
next to one another. Similarly, the optimal timing and ratio of of cellularized tissue structures with dimensions and func-
these cell interactions remain poorly understood. One approach tion remarkably similar to that in humans[77] (Figure 2a).
is to bioprint terminally differentiated cell types at their end The work focused primarily on muskuloskeletal tissues, with
stage of maturation, which has been the mainstream strategy advanced examples of cartilage, bone and muscle, and these
in the bioprinting field so far.[7,8] Another strategy is to bioprint examples certainly point to the possibilities of bioprinting for
stem cells enriched with the correct cues[45–48,87,88] that can then more complex organs. Using an extrusion bioprinter equipped
stimulate cells to evolve into different lineages and communi- with cell-laden hydrogels, sacrificial inks (pluronic) and a
cate during such process.[45,46] supporting ink scaffold composed of PLGA, the authors were
Similarly, with the explosion of organoid technologies in able to replicate the architecture of an entire human ear of infant
recent years,[89–100] and many successful results obtained with size.[77] Replicating the structure of the ear has been attempted
simpler methods for controlled assembly of engineered orga- and reported several times before by various groups,[110–118]
noids—commonly referred to as assembloids,[35–38,101,102] it however, what is remarkable about this achievement was the
is natural to expect that far more complex organoid assembly ability of the bioprinted tissue to develop and mature in vivo,
processes can benefit from existing bioprinting methods, such and virtually match a series of functional parameters from the
as those utilized for scaffold free bioprinting in the early days native counterpart, including microstructure and mechanical
of the technology[5,26,103–106] (Figure 1a) and currently mar- function. The deposition of collagen, glycosaminoglycans, the
keted by Organovo, as well as more recent spheroid assembly histological characteristics, and even many of the mechanical
processes.[107–109] Such recent processes have taken advantage of properties at a tissue level, were virtually the same as those
the ability of cell spheroids to be aspirated at the end of a small from the native tissue.[77] This is an important achievement,
diameter pipette without entering the pipette channel, such that especially considering that scalability has been a great chal-
the spheroids are attached like a hanging microtissue that can lenge in the field. The authors had considerable success by
be transferred from one place to another. This allows tissues taking advantage of a set of techniques that were developed
to be built up one spheroid at a time to result in heterotypic independently, and then integrated into this method, which
complexity of the engineered constructs[108,109] (Figure  1b,c). points to future directions in the field. For instance, tissue con-
Between the referred methods of: a) organoid or undifferenti- structs were fabricated by combining a supporting PLGA scaf-
ated stem-cell printing that is induced to self-organize into a fold of higher rigidity, which was extruded adjacent to cell-laden
functional organ, and b) printing of terminally differentiated hydrogels with two cells types, while microchannels that were
cells that establish cell–cell communication to enable function, analogous to a circulatory system were fabricated by printing a
it remains to be seen what the better approach may be. Recent sacrificial template material. Each one of these techniques was
efforts on bioprinting of scaffold free kidney organoids point to developed independently in previous works, showing the com-
relevant and successful directions in this area,[72,75] and in that plexity of creating a successful multicomponent system.
of bioprinted assembloids (Figure  1c). As it has been the case The far majority of bioprinting methods depend on the depo-
for other successful examples of bioprinted tissues in the field, sition of living biological matter onto a static and stable sub-
it may be advantageous to design techniques that combine strate that prevents cells from being flushed away during the
bioprinting of terminally differentiated cells, such as cells in printing process. And without being able to rapidly immerse
the vasculature, in connection with organoid-like approaches, cells into a medium, the tendency for cells to undergo cell-
where undifferentiated cells can self-organize and differentiate death is typically high.[59,119] Not only that, but convention-
to give rise to more advanced tissue morphologies. ally, the bigger the cellularized construct, the lower the cell
Finally, although many of the efforts to develop improved viability will be.[59] This trend is consistent for virtually all
bioprinting strategies have concentrated on the many engi- bioprinting methods. Many strategies attempted to overcome
neering questions surrounding the method, it appears that this limitation, such as the bioprinting of biomaterials con-
bioprinting creates a set of new biological questions and chal- nected to syringe pumps that increased the hydration of a
lenges that are only beginning to be understood. One may cell-laden hydrogels while also promoting biomaterial crossl
argue that the biological challenges may be as consequential as inking,[47,120–123] or the use of humidifiers onto the bioprinted

Adv. Mater. 2022, 34, 2101321 2101321  (5 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 2.  A) An integrated tissue–organ printer (ITOP) for ear cartilage reconstruction. i) A 3D CAD model of human ear developed from medical
image data and ii) a visualized motion program was used to print the 3D architecture of a human ear. iii) Green, blue, and red lines indicate dispensing
paths of PCL, Pluronic F-127, and cell-laden hydrogel to achieve layer-by-layer 3D printing of ear cartilage. vi) Gross appearance, Safranin-O staining,
and collagen type II immunostaining of the retrieved ear construct after 2 months post-implantation demonstrates vii) the increased GAG and col-
lagen contents of the bioprinted ear cartilage tissues post-implantation. A) Reproduced with permission.[77] Copyright 2016, Springer Nature. B) FRESH
3D-bioprinting of heart tissue using dispensed collagen. i) Schematic of collagen solution injected into the FRESH support bath (pH 7.4) where col-
lagen undergoes fibrillogenesis. The FRESH v2.0 method can print collagen filaments of 20 to 200 µm in diameter. ii) A 3D model of derived from a
magnetic resonance image of an infant human heart (left) and FRESH-printed collagen heart (right). iii) A dual-material FRESH printing method using
a collagen ink and a high-concentration cardiac cell ink was used to build a ventricle model (right) with uniform cell distribution that achieved spon-
taneous, directional calcium wave propagation with adequate conduction velocity. B) Reproduced with permission.[134] Copyright 2019, The Authors,
published by American Association for the Advancement of Science (AAAS). C) Intravital 3D bioprinting schematic showing two-photon crosslinking
of HCC–hydrogel into dermis across the epidermis and skeletal muscle across epimysium. A 3D-volume reconstruction showing HCC–8-arm PEG
structure fabricated between undamaged myofibres and epimysium of skeletal muscle in GFP+ mice. C) Reproduced with permission.[127] Copyright
2020, The Authors, published by Springer Nature.

structure,[124,125] to ensure immediate hydration and mainte- decade,[128–130] and so have been other methods of transdermal
nance of temperature and oxygenation. Even the integration hydrogel photopolymerization in vivo.[131–133]
of an entire incubating system onto commercial bioprinters, Despite the emerging methods of intra-vital 3D bio-
equipped with UV light, temperature, and humidity control, printing,[127] the seemingly simple problem of printing cells in
has been reported.[126] One very interesting recent solution to a liquid bath was only truly overcome with the development of
this problem, has been the development of methods of intra- printing methods that enabled the omnidirectional extrusion of
vital bioprinting. To that end, Urciuolo et al. injected cell-laden viscous fluids or pre-solidified granular materials onto a non-
photosensitive polymer hydrogels that were subsequently cross- Newtonian liquid bath.[109,134–141] The development of printing
linked using a bio-orthogonal two-photon cycloaddition poly­ methods that allowed biomaterials and cells to be dispensed
merization in live mice, inside tissues including skeletal muscle, within a liquid bath is certainly a stepping-stone in the field
brain, and skin (Figure 2c).[127] Such an important milestone in toward fabrication of complexly organized tissues and organs.
the field was mostly enabled by the identification of 7-hydroxy- These methods, which were originally developed for fabrica-
coumarin-3-carboxylic acid as a molecule that, when functional- tion of cell-free biomimetic structures[139,142] typically work
ized into poly(ethylene glycol) or gelatin hydrogels, allowed for with two components: a bath of granular microgels which are
deeper penetration of light and cross-linking of the hydrogel immersed in a precise range of temperature, pH or chemical
pre-polymers at specific wavelengths. This enabled both in vivo composition; and an extruder, which dispenses a biomaterial—
hydrogel patterning, as well as desirable cell viability. This has either with or without cells—which will undergo rapid gelation
been only the first method to 3D bioprinting cells in vivo using upon contact with the supporting bath. The granular material
light-based technologies and through the epithelium, however allows the print-head to travel freely in X, Y, and Z, and to dis-
the concept of in situ bioprinting has been around for nearly a pense cells in arbitrary locations without the need to fabricate

Adv. Mater. 2022, 34, 2101321 2101321  (6 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

a supporting structure, which has been standard practice in significant challenge, the intercommunication of such cells in a
the field. In other words, before these developments, in order bioprinted 3D constructs appears to be something that is more
to extrusion-print the shape of tissues with complex undercuts advanced in the field. Vascularization is the most basic require-
and hollowed architectures, one would have to come up with ment for tissue survival in the human body. A functional vascu-
an architectural strategy to prevent the unsupported structures lature guarantees adequate tissue oxygenation, nutrient delivery,
from collapsing. With these recently developed strategies, the and removal of waste products.[144] Hence a blood capillary must
granular bath provides the structural support in any direction, be ultimately located within at least 200  µm of virtually every
while the tissue is printed, and upon hardening of the bioink, cell in the body. Printing of the human vasculature has been a
the engineered tissue can be retrieved by sacrificing the gran- consistent focus of research in biofabrication for a number of
ular supporting medium. A number of reports have used these years, and to date, there is a variety of methods and printing-
technologies,[109,134–141] and perhaps the most impacting example based strategies that enable the fabrication of cellularized tissue
of use of this technology was reported on the FRESH-enabled constructs with a remarkably high degree of success. A couple
bioprinting of autonomous beating hearts with human-like fea- of review papers and book chapters are noteworthy in this topic.
tures (Figure  2b).[134] In that paper, the authors demonstrated The Miller group has authored a comprehensive review of
the precise imaging, slicing, code-generation, and subsequent existing methods for printing of microchannels and vasculature
extrusion printing of a high-density (cell-free) collagen material in a recent report in Lab on a Chip,[145] whereas our group dis-
in the shape of a tricuspid heart valve, and also a full length cussed a number of biological challenges associated with repli-
neonatal heart, with vessels, chambers, and architectural com- cating the true function of vascular capillaries that have been 3D
plexity of the native tissue.[134] They then went on to demon- printed on a recent book chapter.[146] Several other reviews have
strate that structurally simpler samples filled with high-density been published on this subject, and the reader is encouraged to
cardiac cell inks could actually be manufactured to respond refer to these publications for more details on these topics.
with synchronous beating, with or without stimulation, which One aspect that is noteworthy regarding 3D printing of the
is a remarkable advance to the field. In this particular method, human vasculature is that many methods have relied on an
the authors took advantage of the controllable dissolution of indirect printing approach, where the capillary microchannels
gelatin microbeads that can be dissolved simply by increasing and vessels are first created and subsequently seeded with cul-
the liquid temperature. The gelatin beads were immersed in tures of endothelial cells (ECs) or mixtures of ECs and other
a solution of pH 7.4, which allowed extruded fibers of high- supporting perivascular cell types. One of the first methods
density collagen to gel as the print-head was moved in X–Y–Z. developed to generate bifurcating 3D capillaries in cell-laden
Upon printing, the gelatin was sacrificed and the printed tis- tissue constructs was developed by the Chen group,[147] where
sues could be retrieved. A similar report was also published the authors used an inexpensive RepRap extrusion 3D printer
around the same time, where a different group used a similar to dispense a glass carbohydrate type of material that offered
strategy to bioprint miniaturized beating hearts,[143] but instead sufficient rigidity upon printing. After coating with a protective
of using the FRESH approach, this team developed a simpler PLGA layer, the carbohydrate glass could be fully covered with
strategy to dispense an omentum-derived hydrogel on a sup- a cell-laden hydrogel pre-polymer, which was then polymerized
porting medium containing sodium alginate, xanthan gum, before sacrificing the 3D-printed mold via dissolution. This
and calcium carbonate. It is worth noting here that the biolog- was a remarkable development in the field of tissue printing
ical complexity of orchestrating the multi-typic cell–cell inter- at the time, since it enabled the fabrication of much larger vas-
actions that ensure the function of a continuously (long-term) cularized tissues by ensuring that cells across the thickness of
beating heart in these reports was not yet possible;[134,143] which larger constructs could receive sufficient oxygen and nutrients
highlights the immense challenges associated with the biology to survive, irrespective of the tissue type and dimensions. This
of printing long-term functional tissues and organs, which are work also builds significantly on previous projects that used a
further discussed in Section 4. similar methodology to microfabricate planar microchannels
In summary, while recent examples of bioprinting of cell- in cell-laden tissue constructs. These constructs then needed
laden hydrogels supported by rigid inks and sacrificial bioma- to be stacked to form more complex tissue structures,[119,148–153]
terials have provided, perhaps, the most advanced examples of which highlights the importance of adding a third dimension
replicating structure and function in human-sized bioprinted to engineered tissues via bioprinting. A few years later, a sim-
constructs, it appears as though the bioprinting of materials in ilar strategy was published using a pluronic gel as a sacrificial
granular media is where the state-of-the-art methods of repli- template for the same goals of creating a circulatory system
cating organ complexity will derive from. Advances in light inside of a cell-laden hydrogel,[154,155] and indeed such methods
photopolymerization printing also have strong chances of pro- enabled the creating of pericyte-supported cell-dense vascu-
viding substantial developments in the field, and these are dis- larized tissues that responded biologically as expected from a
cussed more in depth in the following section. fully vascularized human tissue.[154] Contemporary work from
our team utilized a similar methodology to 3D print with an
agarose ink, which did not require coating or dissolution of
3. The Complexity of Tissue Connectivity— the template mold. Instead, the 3D-printed gels could be cov-
ered with cell-laden biomaterials and subsequently aspirated or
Bioprinting the Human Vasculature
pulled out of the construct to avoid any interaction between the
While bioprinting of specific tissues and organs of high cellular template materials with the surrounding cells. We were then
heterogeneity and complex structural organization still poses a able to demonstrate that the 3D-printed vasculature enhanced

Adv. Mater. 2022, 34, 2101321 2101321  (7 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 3.  A) 3D printing of alveolar model topologies with entangled vascular networks. i,ii) Images showing fabrication of entangled vessel topologies
(interpenetrating Hilbert curves and bicontinuous cubic lattice) within the PEGDA hydrogels. iii) Architectural design of an alveolar model topology
based on a Weaire–Phelan 3D tessellation with vasculature and shared airway atrium. iv) Photograph of a printed hydrogel shows concave regions of
the airway (dashed black circles) squeezing adjacent blood vessels and causing RBC clearance. v) A computational model of airway inflation demon-
strates increased displacement at concave regions (dashed yellow circles). A) Reproduced with permission.[164] Copyright 2019, The Authors, published
by AAAS. B) Generation of model tissues with dendritic vascular networks via sacrificial laser-sintered (SLS) carbohydrate templates. i) Schematic
showing the workflow for the additive fabrication of architectural motifs of the smooth vasculature, hierarchical branching, and unsupported overhangs
via SLS. ii) Volumetric reconstruction demonstrating that the metabolically active zone of cells closely follows the perfusable dendritic vascular network.
B) Reproduced with permission.[159] Copyright 2020, The Authors, published by Springer Nature.

the differentiation and viability of osteoprogenitor cells, and Stevens’ groups utilized a laser sintering 3D printing approach to
that the formed vessels remained patent and stable in solution, fabricate complex patterns of laser sintered isomalt that, like their
even after being dissected out of the surrounding gels.[156] This earlier work on extruded carbohydrate glass, could be sacrificed
demonstrated that the printed vessels had the cell–cell commu- after covering with a cell-laden hydrogel (Figure  3b).[159] Several
nication that is typical of native vessels, enabling their potential other methods that rely on extrusion printing of one form or
manipulation for grafting and suturing, which remains to be another or sacrificial bioink have been reported, and the reader is
tested. Moreover, this same method was used for a dual-ink 3D encouraged to refer to recent reviews on the topic for a closer look
printing approach where strands of vascular capillaries were at the specific details of each one of these methods.[160–163]
printed adjacent to strands of a rigid beta tricalcium phosphate Another method that has changed the landscape on the
bone scaffold material, paving the way for fabrication of larger fabrication of complex vascularized tissues uses a completely
pre-vascularized bone grafts.[157] This same method was then different approach to that of sacrificial 3D printing. This strategy
used to manipulate fluid flow as a reconfigurable (movable) utilizes the photopolymerization chemistry of light-activated
microfluidic valve in 3D-printed hydrogel channels, showing hydrogel pre-polymers, together with the long-standing tech-
some additional versatility to this approach.[158] nology of Digital Light Processing (DLP) 3D printing, to pattern
Many other strategies have emerged to address the issue of layered structures that will result in the formation of complex
connectivity between cells in a tissue. One noteworthy develop- channels. Attempts to DLP 3D print hydrogel microchannels
ment in recent years integrates two of the recent developments are not new,[165–167] and, one of the greatest challenges has been
cited above to create a technique called “sacrificial writing into to control the specificity of light interaction with the existing
functional tissues” (SWIFT).[141] In this method cell aggregates monomers, such that light does not “bleed” through the regions
and organoids are fabricated in high-throughput and then com- of interest ,and prevent the formation of an effective hollow
pacted via centrifugation to result in a granular supporting bath channel. Grigoryan et  al. addressed this challenge by using
that is comparable to those utilized in the FRESH method, with molecules that absorb light at specific wave lengths to fine-
the caveat that the SWIFT approach uses living cells as the non- tune the control of photopolymerization depth, and with that
Newtonian supporting bath, as opposed to granular microgels. enabled the printing of very complex microchannel geometries
At low temperatures of 0–4 °C, the high-density cell slurry is soft in biocompatible, and even cell-laden, hydrogels.[164] Using this
enough for omnidirectional movement of the print head, while method, the team was able to replicate the complex architecture
it dispenses any material without damaging cells, but viscous of the alveoli in the lung, inclusive of the ability to oxygenate
enough to hold its shape as this occurs. This allows for a straight- incoming blood via induction of breathing-like movements,
forward dispensing of gelatin inks, which harden at low tempera- that were activated mechanically on the 3D-printed hydrogels
ture and can be easily sacrificed afterward.[141] Using this method, (Figure  3a). In summary, 3D biofabrication and printing of
Skylar-Scott et  al. 3D-printed complex vascular capillaries within complex vascularized constructs has evolved at a rapid pace.
constructs made of high-density cell aggregates, which enables Many challenges remain, and again, these seem to pertain the
the adequate interconnection between cells by the presence of a specific biological complexity of replicating the biology of a
circulatory system fabricated via printing. Another vasculature 3D functioning blood vessel,[146] more so then simply creating con-
printing strategy that has relied on a sacrificial approach, builds on duits of fluids, which appears to be more attainable to date.
the concept of sacrificing sugar glass (isomalt) to generate com- For instance, while the fabrication of endothelialized
plex vascular capillary structures. A recent paper by the Miller and 3D-printed channels has been accomplished and characterized

Adv. Mater. 2022, 34, 2101321 2101321  (8 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

with extensive success, only a few papers have included the pres- regarding the need for close communication and evolution of
ence of perivascular cells, which are known to regulate impor- regulatory guidelines that need to be modified in order to allow
tant physiologic process of endothelial barrier function, among for adequate translation of bioprinted tissues into the clinic. One
others.[154,157] Extensive work by our group has been conducted consequence of this, is that clinical trials with bioprinted pre-
to characterize the specific microenvironmental and tissue fab- vascularized grafts has remained little active in the recent past.
rication conditions that enable the consistent and reproduc-
ible fabrication of vascular capillaries in vitro,[168–172] and many
other groups have generated vascularized organoids as well.[173] 4. Engineering Challenges to Replicate Microscale
It has been demonstrated that biological requirements, such
Organ Heterogeneity
as stem cell source,[169] mechanical properties,[172] hydrogel and
medium composition,[157] cell ratios,[157] and a variety of different There are a growing number of bioprinting methods that
conditions, all play a significant role in ensuring adequate have emerged since the implementation of the field, and it is
endothelial-perivascular cell communication. These are aspects increasingly hard to categorize the specific strategies that are
that are likely to be needed if one intends to reliably print fully available, owning to their growing intersections and commu-
vascularized and functional solid organs. Moreover, the presence nalities. But generally speaking, the methods have been divided
of endothelialized channels in bioprinted tissues and organs, in the form of extrusion, ink-jet, light, and laser bioprinting, as
does not translate into a functional circulatory system. Vessels described above.[76] There have been several dozens of review
have complex blood carrying functions, barrier properties, parac- papers on the details, advantages, and disadvantages of each spe-
rine signaling, and a myriad of vascular biology specific require- cific method,[3,6–8,57,58,174–178] including some of our own.[14,51,146]
ments that tend to be less studied when teams have attempted Therefore, we encourage the reader to refer to these earlier publi-
to bioprint vessels and capillaries.[146] Understandably, the chal- cations. Here, we will focus on the specific challenges of each bio-
lenges associated with printing blood vessels is substantial, but printing method in addressing the biological needs of building
until these rudimentary vessels demonstrate complex regulatory complex tissues and organs, with a special emphasis on recent
functions and long-term stability, they are unlikely to be used as developments that have received lesser attention in previous
reliable substitutes for native vessels. Moreover, it is important to reviews.
acknowledge that complex organs are vascularized by complex In keeping in line with the challenges of replicating the
vascular trees, where larger vessels feed very small capillaries structural and organizational complexity of human tissues and
(<20  µm diameter), which we discuss in a recent review.[146] To organs, one of the main questions that remain is what are the
date, although many examples of 3D-printed bifurcating vas- engineering challenges that need to be addressed in order to
cular trees exist, the far majority of bioprinting methods have enable meaningful mimicry of such complexity? Previous sec-
concentrated on larger diameter channels, and even when nar- tions hinted to some of these needs from a biological stand-
rower channels are printed, they typically to not fully replicate point, but here, some of the recent engineering challenges are
the architectural complexity of the native vascular plexus in solid discussed more specifically, without getting so much into the
organs. On that note, extrusion methods of sacrificial vascular hardware aspects, but rather the engineering challenges that
printing have generally enabled relatively simpler vascular struc- enable biology. First and foremost, arguably the primary engi-
tures, whereas more recent examples of DLP biofabrication have neering challenge toward fabrication of meaningful organs, is
addressed some of the complexity of smaller and more intri- the ability to recreate tissue heterogeneity with precision. To a
cate vascular capillaries with higher resolution.[164] It appears as great extent, despite the development of many multi-ink bio-
though the existing methods of vasculature printing will facili- printers in recent years,[46,122,179] methods that enable a system
tate the fabrication of perfusable channels that are sufficient to to replicate the accuracy and precision at which tissue and cell
let endothelial and perivascular cells communicate to generate heterogeneity is displayed in the body remain limited. Extru-
the very small vascular capillaries via endothelial morphogen- sion methods have been particularly good at attempting to
esis. Importantly, without the 3D-printed channels, these cells recreate tissue heterogeneity (Figure 4a),[180] owning to the rela-
themselves cannot get the nutrients required to remodel into tive ease at which syringe print heads can be positioned on a
capillaries in the core of larger cell-laden biomaterials or cell- printing axis and used interchangeably. A critical limitation to
aggregated organoids/assembloids. Thus, a likely approach is these developments, however, is that the true heterogeneity of
one where larger vessels are printed to support self-assembled complex tissues typically happens at a scale that can be orders
capillaries of small diameter, and together, these can oxygenate of magnitude lower than what current extrusion printers can
an entire 3D solid organ construct. achieve. For instance, even though extrusion bioprinting inks
It must also be highlighted that printing and biofabrication have been developed to be dispensed with the theoretical width
of tissues with terminally differentiated cells, such as endothe- of a single cell,[57] these bioinks are generally dispensed as
lial cells, is likely to pose a challenge for translation. The cur- fibers, which makes it extremely difficult to micropattern tissue
rent FDA regulation of cell therapies is restricted to a narrow regions that are discrete with areas that as small as clusters of
window of isolated stem cells or primary differentiated cells that 2 or 3 cells, surrounded by areas of a different cell type, inter-
are not subjected to further processing, except for rare circum- connected by 15–30 µm blood capillaries and innervation. Most
stances. This means that the conventional process of cell expan- systems are just not on par with this level of complexity. Laser
sion that is currently needed to obtain sufficient endothelial cell and inkjet printers, can approximate this precision and resolu-
numbers for a graft is not yet regulated, neither is the process tion to a far greater extent, however, despite growing improve-
of iPSC cell implanation. This brings another important point ments in printing with multiple inks (cell types) using inkjet

Adv. Mater. 2022, 34, 2101321 2101321  (9 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 4.  A) Design of the digitally tunable continuous multi-material extrusion bioprinter. i) Schematics and photographs showing the seven-
channel printhead design connected to reservoirs that are individually actuated by programmable pneumatic valves to print microfibers of seven
bioinks. ii) Images showing a printed multi-component cell-laden heart-like structure. A) Reproduced with permission.[180] Copyright 2016, Wiley-VCH.
B) Microfluidics-enabled multi-material maskless stereolithographic bioprinting. iii) The defined CFD model and the velocity profile of PEGDA in the
closed microfluidic chamber under sinusoidal fluid flow. iv) The operation of the microfluidic device for consecutive injection of different bioinks.
v) Schematics showing the skeletal muscle tissue and tendon-to-bone insertion models, the mask for printing, and the fluorescence images of bio-
printed GelMA structures containing cells of various types. B) Reproduced with permission.[184] Copyright 2018, Wiley-VCH.

and laser printers, it remains remarkably difficult to build cleaning, vat tilting, build platform movement, etc.). Currently,
tissue layers at a reasonably high speeds in 3D when using the far majority of light polymerization based bioprinters have
these printers.[181–183] Even more important than limitations the capacity to comfortably pattern regions as little as ≈40 µm in
associated with printing speed, is the fact that laser and inkjet XY. In theory, this would enable micropatterning of very com-
systems have a far narrower window of printability, and thus plex regions that are as small as a single cell, potentially ena-
have far more limited number of compatible bioinks. Moreover, bling very complex tissues to be fabricated. However, this is
the structural weakness of bioprinted constructs, which is a typically listed as the nominal resolution of the machine, and
direct result of the limited ink viscosity, restricts the size of the the reality ends up being far removed from that once the mate-
bioprinted structures to very small dimensions, despite the fact rial is encapsulated with cells and other biological molecules.
that these methods can indeed provide very high accuracy of Not only the nominal resolution of the printer does not account
tissue positioning and resolution. for a number of light refraction and blocking issues that arise
On that note, the speed and precision of light polymeriza- from the presence of light-scattering components (such as cells);
tion based printers has been a focus of great attention.[53,185–190] but also, they typically underestimate the complexity of printing
A great deal of effort has been expended toward increasing with heavily hydrated monomers that are composed of a very
the speed of light polymerization printers (DLPs, 2PP, ste- low concentration of photosensitive moieties, which decreases
reolithogrpahy, etc.).[53,185–189] This has been done mostly by the precision of the monomer–light interactions substantially.
developing improved chemistries that that are compatible with In other words, a machine that can print very precise cell-free
speed,[190] precision and scalability.[191–195] Another strategy pure monomers will likely have only a fraction of its accuracy
has been to adapt the hardware to improve functionalities and when printing with cell-laden hydrogels at their typical low con-
overcome limitations that are built into the process, and are centration monomers. Extensive improvements on hydrogel and
known to slow down the fabrication (i.e., calibration, priming, tip process conditions have been achieved recently on exactly these

Adv. Mater. 2022, 34, 2101321 2101321  (10 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 5.  A) DLP bioprinted microarray of cell-laden microgels with controllable geometry and mechanics. Reproduced with permission.[196] Copyright
2021, IOP Publishing. B) Each microgel can be: i) loaded with a co-culture of HUVECs and hMSCs that ii) self-assemble into C) a pericyte-supported
microvascular capillaries in less than 72 h. D) The pre-vascularized microgels can be injected with a built-in vasculature and show ability to anastomose
with adjacent capillaries in vitro. E) These microgels can also be injected into selective regions within 3D-printed stackable microcages, to result in
complex 3D constructs with predefined tissue regions. E-i,iii,iv) Reproduced with permission.[202] Copyright 2020, Wiley-VCH.

fronts, with modification of the biomaterial using light blockers to a microfluidic syringe pump, and flow it to a microdevice
and other strategies,[164,191–196] but this remains a topic of further in a coordinated fashion with the exposure of light, such that
development. different shapes and structures could be built to result in het-
Despite the important advances mentioned above, per- erotypic constructs (Figure  4b).[184] This strategy may point to
haps the major current limitation with light based bioprinters smart alternatives in the future, where tissues can also be built
remains that of building heterotypic complexity in tissue con- in Z, for increased volumes, as opposed to remaining limited
structs. The far majority of light activated bioprinters, both DLP, to flat and thin footprints. On that note, another avenue that
two-photon polymerization and light/laser rastering lithography, may be worth pursuing is that of bioprinted microgels that can
function by using a vat that is filled with a photocrosslinkable be fabricated with microscale heterogeneity and then assembled
biomaterial, and for each added layer, a fresh coat of material in the form of supramolecular granular hydrogels.[197,198] Several
needs to be layered onto the printed part. A variety of strategies microgel assembly methods have been developed over the years
are used for this. Some rely on the tilting movement of the vat and this may point to a simpler strategy of tissue biofabrication
itself, some rely on the coordinated motion of the robotic arm and assembly.[199–201] For instance, we have recently developed
holding the build platform in X–Y, or even up and down. The methods for high-throughput bioprinting of injectable pre-
issue with that strategy is that coverage of the printed biomate- vascularized microgels[196] (Figure 5a,b) which can be injected
rial with a fresh layer of ink is the most straightforward oppor- (Figure 5c) into stackable 3D-printed microcages with pre-deter-
tunity to provide different composition of the tissue, either via mined functional regions of tissue composition or spatiotem-
a different biomaterial ink, or a different cell type. The obvious poral release of growth factors (Figure  5e).[202] Interestingly,
challenge with that method is that the body is not built in flat when these pre-vascularized microgels are implanted and com-
and even layers, and hence true tissue heterogeneity is difficult pared to non-printed cell-laden hydrogels of the same polymer
to achieve. One method that has been developed to address and cellular composition, the formation of vascularized tissue
this goal was to connect hydrogels of different compositions is significantly improved with the pre-vascularized matrix (data

Adv. Mater. 2022, 34, 2101321 2101321  (11 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

not shown). That is because vessels in the microgels can anas- cells in their designated reservoirs, with their preferred
tomose with one another (Figure 5d) and with the host from the medium, and rapidly switching the dispense of many cell and
onset of tissue remodeling, as opposed to being dependent on material types in the middle of a print run (Figure  6a,b). One
the assembly of vascular capillaries in the core of conventional commercially available system that has taken significant advan-
constructs, which have poor access to oxygenation. tage of this concept is the Biopixlar, which uses a multi-channel
On the specific advantages of extrusion, versus light and microfluidic device integrated to a four-chamber microdevice,
inkjet/laser bioprinting, specifically for the fabrication of complex and a controller enables the precise opening and closing of
heterotypic organ constructs, it appears that extrusion methods microfluidic valves that coordinate the flow of each chamber
have an important edge, which is the ease with which newer component down onto an extrusion port (Figure 6b–d). Remark-
methods have to dispense multiple cell/material types simultane- ably, the flow regimes that are created at the end of the print
ously. The major disadvantage, remains the relative low resolu- head, result in a dispense–aspirate system where the outflow
tion of conventionally extruded cell-laden inks, which still revolve of cells, even at the single cell level, is controllable to an area of
around the >100 µm mark, for the most part. On the other hand, a few micrometers (Figure  6b,d).[206] Once these flow regimes
the precision and speed of light polymerization bioprinters has are coordinated with the speed of the dispensing robot, one can
shown amazing success in recent publications, with a remarkable deposit even single cells with extensive precision onto a sub-
ability to build up complex tissue structures in 3D. Nevertheless, strate. This is a great example of how microfluidics engineering
the ability to bioprint with multiple cell/material types in 3D has has been utilized to favor the biology of bioprinting. One key
remained limited, and this is a major disadvantage for complex advantage of a system like this, is that it enables rapid switching
heterotypic organ printing. Laser and inkjet printers, as discussed between various cell types, in the middle of the printing run,
in other sections of this paper, have lagged behind mostly due to with relative ease, and as many times as needed. Consequently,
their limited range of viable inks, and undesirable ink properties the idea of building tissue complexity and cell heterogeneity in
for manufacturing of large-scale constructs, despite their ability a construct becomes a lot more attainable (Figure 6c). The main
of patterning tissues with single cell resolution. disadvantage of this system, which is not small considering the
Another recent topic that has received tremendous atten- tissues in the body are 3D and printing leverages precisely that
tion in the 3D printing community, and is slowly finding its aspect, is that cells can only be deposited in 2D, and building
way onto the tissue bioprinting in the literature, is that of volu- 3D architectures requires stacking of many tissue layers in a
metric/holographic printing, where the idea of additive manu- labor-intensive sequence of cell deposition, attachment, cov-
facturing derived from the concept of building layers one on erage with a cell-adhesive solution, and follow-on deposition of
top of the other, is challenged in favor of printing entire 3D cells, usually after several hours (Figure 6d). Nevertheless, there
constructs from the outside in, so to speak.[203] These technolo- is a list of possibilities to be explored on the interface of micro-
gies are still nascent and pose a set of challenges for biologists fluidics, printing and biology, which appear to be only begin-
and tissue engineers that are only beginning to be explored. ning to be addressed.
But are likely to provide some new impetus in the field. An additional aspect that has received little attention in com-
Similarly, one other aspect that is noteworthy on the engi- parison to bioink and hardware development for bioprinting is
neering side, as we have hinted to in previous sections, is that the progress on methods that enable precise and quantitative
pertaining to the interface of microfluidics and bioprinting. characterization of bioprinting precision. For instance, if the goal
Microfluidics has a long history of assisting the development is to position cells with as high precision as possible to match that
of novel printing methods. Very early approaches developed of a native organ, then there should be methods that allow one to
controllable microfluidic-based systems to dispense highly quantify these outcomes in 3D relative to larger organ structures.
intricate fiber morphologies with topographical features with While confocal microscopy enables imaging and quantification of
extensive level of complexity in their composition.[196] These cellular processes in 3D, quantification of cell positioning in engi-
earlier methods were not attached to an X–Y–Z robot capable neering tissues with single cell precision has remained poorly
of reading G-code, but they paved the way for a long range explored. Combinations of clarity microscopy[208] and computa-
of papers utilizing microfluidics control of ink dispensing tional analyses of high-content cell imaging are possible solutions
capacity to generate tissue complexity.[180] An interesting out- toward that end,[209] and should be further explored.
come from these initial methods benefits a lot from materials
that have rapid crosslinking in the presence of specific fluids,
such as alginate, which has been very extensively character- 5. Materials Challenges to Replicate the
ized for tissue engineering. For instance, using these alginate
Extracellular Matrix and Organ Complexity
based systems, Colosi et  al. reported on the development of
what has been known as universal bioinks,[184,204,205] where One last relevant aspect pertaining to replicating true tissue
the alginate functions as a carrier material for secondary com- and organ complexity relates to the nature of the materials
position of hydrogel, and once the printing is complete, the that make up the native extracellular microenvironment in
alginate is either dissolved, degraded, or maintained in place, the body. It has become increasingly clear that, while cells are
enabling straightforward dispensing of multiple types of inks, embedded in a protein-rich 3D matrix of relatively consistent
ranging from dental proteins,[120] to methacrylated hydrogels composition (mostly collagenous with non-collagenous macro-
and others.[184,204,205] Another remarkable feature of integrating molecules), there is a significant degree of organization to the
microfluidics with bioprinting, which has led to the development interaction and composition of these proteins and other bio-
of some companies altogether, is the possibility of maintaining logical molecules. This manifests itself in the form of gradients

Adv. Mater. 2022, 34, 2101321 2101321  (12 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Figure 6.  A) Schematic diagram showing: i) a PDMS microfluidic pipette tip, ii) the holder, iii) the pneumatic interface between the holder and the
tip, wells, and iii–iv) the microfluidic switching junction near the tip. A) Reproduced with permission.[206] Copyright 2012, Royal Society of Chemistry.
B) i) The microfluidic printhead is connected a controller hardware and software interface, which regulates the deposition of various cell types at a time,
by allowing for the formation of ii–v) a recirculating fluid zone at the end of the printhead within a liquid bath. vi–viii) The translational movement of
the printhead and X and Y, coordinated with the adhesion of the cells to treated substrate, allows for ix,x) the patterning of cells with multi- (ix) or single
(x) cell resolution. C) The bioprinter enables fabrication of multi-cellular 2D (planar) tissues, in this case skin cancer cells (A431, red) surrounded by
epithelial cells (HaCaT, green). Scale bars: 200 µm. D) 3D tissue constructs where the base cell layer was composed of A431 cells (green), the middle
layer being HaCaT cells (red), and the top layer being A431 cells (blue) (v,vii); in (vi,viii), a patch of liver cancer cells (HepG2, in red) is surrounded by
fibroblasts (3T3-J2, in blue). Scale bars: 300 µm. B,C) Reproduced under the terms of the CC-BY Creative Commons Attribution 4.0 International license
(https://creativecommons.org/licenses/by/4.0).[207] Copyright 2020, The Authors, published by Springer Nature.

of concentration of soluble molecules, alignment of fibrillar on strategies that are based on hydrogel patterning versus
structures, hierarchical organization of assembled proteins, and direct cell patterning. For instance, despite the extensive work
many more. For the most part, the field has devoted significant of Forgacs, Mirinov et  al.[23,25,26,103,105,106] in implementing bio-
efforts to determine the materials characteristics that are printing through the dispensing and pattering of high den-
required to enable reliable printing via extrusion, light poly­ sity cell aggregates, the field has concentrated on cell-laden
merization, and several other setting reaction methods. A few hydrogels as bioinks for many years. However, it should be
reviews have been published on this topic to provide guidance acknowledged that developmental biology stems from the close
on the material properties leading to improved printability. Nev- interaction of cells in a low concentration of matrix, where cell-
ertheless, the perspective of how these bioprintable materials laden gels propose the exact opposite, high concentrations of
facilitate replicating the heterotypic function and complexity matrix with a low percentage of cells. Recent successful exam-
of tissues and organs has been superficially addressed. For ples of complex organ models, such as the heart[134] and the
instance, while many studies have pointed toward the desir- vasculature,[141] have gone back to mimicking this cell-dense
able combination of biomaterials presenting shear-thinning approach used by nature, where the material was only utilized
properties and cell-adhesive motifs, these materials of single as temporary supporting medium for fabrication, or at very low
composition fail to acknowledge that the matrix composition concentration. This brings up the question of where the field
can also vary according to the location in a given organ. These should focus more, if the intention is to replicate organ func-
questions point to an urgent need to concentrate efforts on the tion. One perspective that may be relevant to answer this ques-
development of printing methods that are amendable to rapid tion is, again, the original characteristics of the tissue or organ
exchange of inks, as to allow for the fabrication of more com- that is being mimicked. Connective tissues (i.e., cartilage, bone)
plex heterotypic constructs. Another debate on this question are matrix-dense by design. Solid vital organs have very little
is regarding the emphasis of the biomaterial ink development matrix in comparison, and are far more cell-dense. (i.e., liver,

Adv. Mater. 2022, 34, 2101321 2101321  (13 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

heart). This original organization should provide a starting matrix of the native calcified tissues,[120] or the bone matrix was
point in the design of bioinks that will facilitate an approxima- functionalized with methacryloil to give rise to a bone-derived
tion of the desired organ function. phtocrosslinkable bioink for DLP bioprinting (BoneMA).[196] Sev-
While the liver is an interesting example to describe the chal- eral other examples are available for tissue-derived bioinks for
lenges of replicating cellular heterogeneity and architectural a many different organs.[227,228] Despite these advances, from
complexity for bioprinting, as it was discussed in earlier sec- a matrix biology standpoint, simplified porous hydrogels fail
tions, bone tissue can be used as an interesting example of a to mimic the nanostructural organization or protein composi-
seemingly simpler tissue, from a cellular perspective, but much tion of the native extracellular matrix in the body. For instance,
more complex from a matrix biology perspective. For instance, while many natural and synthetic hydrogels have been chemi-
at the microstructural level, compact bone is organized pri- cally modified with RGD and other cell-adhesive sites[196,229] to
marily by the lateral positioning of osteon units.[210–212] Each promote simple integrin-binding and cellular traction, these
osteon is structured in the form of concentric rings of osteo- interactions fail to recapitulate the hierarchical interactions
cytes, which result in a unit measuring about 200 µm in diam- that occur in the extracellular microenvironment in the native
eter. The osteon has a precise organization that is defined by the tissues. For instance, the structure and mechanics of native
alignment of groups of collagen fibers, which essentially show fibrillar collagen, which dictate cell response in the ECM in the
a pattern of counter-posed inclination, where groups of fibers body, is largely determined by variable modifications of intrafi-
in one layer are inclined in the opposite direction of the next. brillar and extrafibrillar crosslinking,[230] the presence and inter-
This organization is responsible for a remarkable degree of action of interfibrillar non-collagenous proteins, the slipping and
self-toughening, which makes bone stiff and tough at the same sliding ability of interconnecting proteoglycans and glycosami-
time.[213–216] One question then is how well can existing bio- noglycans,[231] as well as the ability of other soluble molecules
printers replicate this level of detail in the extracellular matrix, to interact and bind to these structural proteins.[232] Virtually all
for an example? Certainly this question depends on the inten- of these nuanced aspects of matrix biology are lost in the cur-
tions for the printed tissue. For instance, if the end goal is to rent reductionist approach to bioink development. Certainly, this
simply bioprinting of “scaffolds” loaded with cells, then we may reductionist approach to bioink development is not a subject of
not require the same level of precision that is intended for a concern when engineering regenerative materials,[233,234] given
true permanent body replacement part, since the biodegradable the temporary nature of the scaffolds. However, these questions
scaffold should only stimulate accelerated healing.[12,17] On the may become more significant when organs are to be bioprinted
other hand, if the intention is to achieve a material system that with the desired level of biological precision that will be required
replicates the same level of “tissue quality” as the native tissue, to manufacture functional body parts in the lab from the get go.
then that would be desirable; especially if it is understood that Again, using bone as an example of such complexity, and
this is an important component to achieve the desired function, highlighting the broad gap between native tissues and engi-
as it is the case with bone. In other words, from a clinical stand- neered inks, it is well known that the bone matrix is composed
point, if a patient loses a vital organ, the said patient cannot of a hierarchically organized matrix material, mostly consti-
wait until a scaffold matures into a vital tissue over the course tuted of collagen and hydroxyapatite. The majority of bone
of several months, as it is currently the case. That is the exact bioinks are composed of either simple osteoinductive hydro-
purpose of organ transplantation; to provide an immediate gels,[235] or basic calcified ceramics and extrudable cements,[51]
remedy for a failed organ structure and immediately recover rather than a matrix that mimics the complexity of bone tissue.
organ function. Ultimately, if bioprinting is to deliver on the At some point, there should be a shift to where designer inks
promise of circumventing the problems of organ donation and can accommodate the matrix complexity of the native tissues,
transplant, the field should strive for these aspirational goals, and enable patterning of cells and biological materials that will
which poses a significant challenge. result in the manufacturing of fully (or at least more highly)
Still using bone as an example, another aspect that deserves biomimetic tissue constructs right out of the printer, rather
recognition with respect to the development of biomaterial inks, than requiring a long process of cell differentiation, remode-
is the relative nano and microscale simplicity of existing inks in ling of a scaffold system, and eventual maturation into a func-
comparison to the native matrix in the body. For instance, there tional organ, which is currently the case.[77] Examples of how
is an incredibly high number of reports developing bioinks for bioprinting can accommodate for these nuanced questions of
bone regeneration.[87,217–226] The far majority of these existing materials properties and micro- to nanoscale matrix organiza-
methods use a reductionist process of loading cells in a single tion already exist. For instance, Martin et al. developed a mag-
component hydrogel that stimulates differentiation of osteopro- netic printing protocol that allow one to replicate not only the
genitor cells with some form of osteoinductive ingredient. These structure of microscale osteon-like geometries, but also the
hydrogel biomaterials, in their large majority, are composed specific alignment of mineral particles relative to the Haversian
of either broken down biological molecules that are modi- canal in the native bone using magnetic control of the ink in
fied to endow the material with controllable properties, such question.[236] The mechanical consequence of a biomimetic
as photo-crosslinking control, desirable chemistry, or tunable “organized” particle distribution in comparison to a random
mechanics.[87,217–226] A number of relevant studies developing orientation of loaded particles, as expected, was significant;
tissue specific ECM-derived bioinks have followed this trajec- pointing to the perspective stated above, that perhaps nanoscale
tory, including some of our own. We have reported dental and engineering of biological inks may be more important for cer-
bone-derived bioinks, where a dentinal tissue was processed tain tissues than originally predicted. Of note, the ability of
to result in an extrusion bioink composed of the extracellular 3D printing the microscale morphology of osteons in bone

Adv. Mater. 2022, 34, 2101321 2101321  (14 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

via DLP printing, while enabling control of the directionality Perhaps a more balanced approach would be to develop ena-
of organic and inorganic components that make up the bone bling engineering tools that are rooted on biological problems,
tissue can be highly relevant to mimic the hierarchical (nano to rather than heavily focused on speed, precision, and cost. For
micro) complexity of bone tissue, since not only osteons have instance, it may be better to identify the critical barriers to bio-
a complex architectural organization of interconnected Haver- printing heterogeneous, complex, vascularized, and innervated
sian canals, but also precisely angled collagen fibrils that are tissues that function, rather than simply being able to dis-
reinforced with hydroxyapatite.[236] Our recent work on engi- pense biomaterials with high resolution but limited biological
neered bone tissue replicating its nanoscale requirements, function. Ultimately, it is a well-balanced synergistic effort of
strongly advocates for this perspective, since simply mimicking engineering and biology that will show us the viable path to
the nanoscale feature of the native tissue resulted in even fabrication of ideal organs on the lab bench for implantation in
better stem cell differentiation and bone-like response than humans, which is the overarching goal of of bioprinting.
cells treated with gold-standard osteoinductive supplements.[171] Another aspect that will shape the future outlook of the
Perhaps a combination of a bone-derived photo-crosslinkable field is how these technologies are regulated. While we may be
bioink (i.e., BoneMA[196]), with the nanoscale method of engi- years away from discussing full cellularized bioprinted organs
neering the bone matrix,[171] bioprinted with cells, using the 3D as an alternative to transplantation as a mainstream treatment
magnetic printing method could closely approximate the com- method, the validation of such engineered systems should be
plexity of human bone. This could form the basis for future the starting point driving the use of these bioprinted tissues
studies. for medical use. This brings up an extremely important point
Another aspect that requires further consideration is how that will advance the field, which is the use of bioprinted tis-
materials can be developed to ensure that cell growth is guided sues as reliable and reproducible subjects of disease models,
in a way that is conducive to the desirable structure as the cells drug testing, and in vitro models of healthy tissues. While it
expand and process the matrix. Preliminary data in our own lab may take some time until these bioprinted organs are validated
has shown that hydrogels of low stiffness lose their bioprinted clinically, there is an urgent need for better models of disease
shape far more quickly than hydrogels of higher stiffness, that can replicate the complexity of drug interactions with mul-
which is expected. However, this loss of shape fidelity over time tiple cell types, or cell microenvironment, and the progressive
also has biological implications, since tissue shape and mor- nature of disease development. All of these aspects are diffi-
phology are well known regulators of development.[237] Further cult to study with animal models, which is hard to control and
research is also required on this aspect. visualize in real time, or with conventional 2D models of cell
culture. Yet, the overwhelming majority of in vitro and pre-
clinical knowledge available about disease conditions and drug
6. Conclusion and Future Outlook response, comes from these models. Engineering miniaturized
and reproducible bioprinted organ constructs that replicate
The progress in the field of bioprinting in the past 10–15 years is the complexity of human biology on a dish, or on-a-chip,[238]
remarkable. The field has progressed from a few cells dispensed is a mutually beneficial step. Not only will this generate valu-
on a plate, to 3D-printed structural alveolar models,[164] beating able knowledge to guide future medical treatments and under-
heart-like tissue constructs,[134] contracting muscles,[77] bendable standing, but also it can validate much of the function that is
cartilage,[77] and endothelialized vascular channels.[147,154,156] All expected from bioprinted constructs before they need to be
addressing both engineering developments, as well biological implanted as organ substitutes. In other words, bioprinting
challenges. One may argue that the challenges before us are of diseased tissues and organs should, perhaps, be given
nothing short of extraordinary. Piecing together an entire organ, as much importance as bioprinting of healthy implantable
cell-by-cell, protein-by-protein, using a machine, is not a small constructs.
feat, and if successful, the outcomes are truly transformative In summary, there are many challenges ahead, but the
for medicine and engineering. Therefore, all of the challenges future of bioprinting is warranted, and the speed of recent
stated above need to be read with a clear sense of the true dif- developments point to a bright outlook where many of these
ficulties behind tackling them. In other words, it is not easy to seemingly distant challenges may already be in the works in a
build organs and body parts from scratch, but it has been more lab somewhere.
than proved by now that this is an achievable goal. Therefore,
future work should focus initially on delineating the critical
challenges that will truly progress the field forward, rather than
Acknowledgements
laterally. For instance, there has been an explosion of bioprinting
hardware development and commercialization in recent years. The author acknowledges funding from the NIH/National Institute of
The cost of bioprinters ranging anywhere from a few thousand Dental and Craniofacial Research (R01DE026170 and R01DE029553 to
L.E.B.) and The OHSU Silver Family Innovation Award. The author also
to several hundred thousand. Biologists may argue that many of
thanks Dr. Ramesh Subbiah and Anthony Tahayeri for assistance with
the expensive systems in the market do not necessarily address preparation of figures of this manuscript.
the most critical problems that are required to actually rebuild
the biology of functional tissues. Engineers, on the other hand,
may argue that the hardware is highly capable of dispensing
biological materials with precision, and it is up to biologists to Conflict of Interest
understand the critical aspects required for ultimate function. The author declares no conflict of interest.

Adv. Mater. 2022, 34, 2101321 2101321  (15 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Keywords [28] Y.  Petrenko, E.  Syková, Š.  Kubinová, Stem Cell Res. Ther. 2017,
8, 94.
3D printing, bioprinting, organ engineering, organoids, vasculature [29] M.  Kijanska, J.  Kelm, Assay Guidance Manual [Internet], (Eds:
S. Markossian, G. S. Sittampalam, A. Grossman), Eli Lilly & Com-
Received: February 16, 2021 pany and the National Center for Advancing Translational Sci-
Revised: April 20, 2021 ences, Bethesda, MD, USA 2004.
Published online: November 5, 2021 [30] B. Desoize, D. Gimonet, J. C. Jardiller, Anticancer Res. 1998, 18, 4147.
[31] G. Pettinato, X. Wen, N. Zhang, Stem Cells Dev. 2015, 24, 1595.
[32] G. Q. Daley, Ann. N. Y. Acad. Sci. 2003, 996, 122.
[33] I. Desbaillets, U. Ziegler, P. Groscurth, M. Gassmann, Exp. Physiol.
[1] L.  Moroni, J. A.  Burdick, C.  Highley, S. J.  Lee, Y.  Morimoto, 2000, 85, 645.
S. Takeuchi, J. J. Yoo, Nat. Rev. Mater. 2018, 3, 21. [34] K. E. Foley, Nat. Methods 2017, 14, 559.
[2] M.  Castilho, M.  De Ruijter, S.  Beirne, C. C.  Villette, K.  Ito, [35] N. Vogt, Nat. Methods 2021, 18, 27.
G. G. Wallace, J. Malda, Trends Biotechnol. 2020, 38, 1316. [36] J.  Andersen, O.  Revah, Y.  Miura, N.  Thom, N. D.  Amin,
[3] J.  Groll, T.  Boland, T.  Blunk, J. A.  Burdick, D.-W.  Cho, K. W.  Kelley, M.  Singh, X.  Chen, M. V.  Thete, E. M.  Walczak,
P. D. Dalton, B. Derby, G. Forgacs, Q. Li, V. A. Mironov, L. Moroni, H. Vogel, H. C. Fan, S. P. Paşca, Cell 2020, 183, 1913.
M.  Nakamura, W.  Shu, S.  Takeuchi, G.  Vozzi, T. B. F.  Woodfield, [37] E.  Kim, S.  Choi, B.  Kang, J.  Kong, Y.  Kim, W. H.  Yoon, H.-R.  Lee,
T. Xu, J. J. Yoo, J. Malda, Biofabrication 2016, 8, 013001. S. Kim, H.-M. Kim, H. Lee, C. Yang, Y. J. Lee, M. Kang, T.-Y. Roh,
[4] J.  Malda, J.  Visser, F. P.  Melchels, T.  Jüngst, W. E.  Hennink, S. Jung, S. Kim, J. H. Ku, K. Shin, Nature 2020, 588, 664.
W. J. A. Dhert, J. Groll, D. W. Hutmacher, Adv. Mater. 2013, 25, 5011. [38] Y.  Miura, M.-Y.  Li, F.  Birey, K.  Ikeda, O.  Revah, M. V.  Thete,
[5] V. Mironov, T. Trusk, V. Kasyanov, S. Little, R. Swaja, R. Markwald, J.-Y. Park, A. Puno, S. H. Lee, M. H. Porteus, S. P. Pașca, Nat. Bio-
Biofabrication 2009, 1, 022001. technol. 2020, 38, 1421.
[6] W.  Sun, B.  Starly, A. C.  Daly, J. A.  Burdick, J.  Groll, G.  Skeldon, [39] L. E. Bertassoni, J. Calif. Dent. Assoc. 2020, 47, 645.
W. Shu, Y. Sakai, M. Shinohara, M. Nishikawa, J. Jang, D.-W. Cho, [40] A. Jain, K. K. Bansal, A. Tiwari, A. Rosling, J. M. Rosenholm, Curr.
M.  Nie, S.  Takeuchi, S.  Ostrovidov, A.  Khademhosseini, Pharm. Des. 2018, 24, 4979.
R. D.  Kamm, V.  Mironov, L.  Moroni, I. T.  Ozbolat, Biofabrication [41] J.  Ni, H.  Ling, S.  Zhang, Z.  Wang, Z.  Peng, C.  Benyshek,
2020, 12, 022002. R.  Zan, A. K.  Miri, Z.  Li, X.  Zhang, J.  Lee, K.-J.  Lee, H.-J.  Kim,
[7] A. C. Daly, M. E. Prendergast, A. J. Hughes, J. A. Burdick, Cell 2021, P.  Tebon, T.  Hoffman, M. R.  Dokmeci, N.  Ashammakhi, X.  Li,
184, 18. A. Khademhosseini, Mater. Today Bio 2019, 3, 100024.
[8] R.  Levato, T.  Jungst, R. G.  Scheuring, T.  Blunk, J.  Groll, J.  Malda, [42] Z.  Jiang, B.  Diggle, M. L.  Tan, J.  Viktorova, C. W.  Bennett,
Adv. Mater. 2020, 32, 1906423. L. A. Connal, Adv. Sci. 2020, 7, 2001379.
[9] S. V. Murphy, P. De Coppi, A. Atala, Nat. Biomed. Eng. 2020, 4, 370. [43] J. K. Placone, A. J. Engler, Adv. Healthcare Mater. 2018, 7, 1701161.
[10] M.  Lenda, P.  Skórka, B.  Mazur, A.  Ward, K.  Wilson, Science 2018, [44] L. Ning, X. Chen, Biotechnol. J. 2017, 12, 1600671.
360, 277. [45] Y. J. Choi, H. Park, D.-H. Ha, H.-S. Yun, H.-G. Yi, H. Lee, Polymers
[11] R. Klebe, Exp. Cell Res. 1988, 179, 362. (Basel) 2021, 13, 366.
[12] A. Khademhosseini, R. Langer, Nat. Protoc. 2016, 11, 1775. [46] X.  Cui, J.  Li, Y.  Hartanto, M.  Durham, J.  Tang, H.  Zhang,
[13] V. Mironov, N. Reis, B. Derby, Tissue Eng. 2006, 12, 631. G.  Hooper, K.  Lim, T.  Woodfield, Adv. Healthcare Mater. 2020, 9,
[14] N. Annabi, A. Tamayol, J. A. Uquillas, M. Akbari, L. E. Bertassoni, 1901648.
C.  Cha, G.  Camci-Unal, M. R.  Dokmeci, N. A.  Peppas, [47] P. Rastogi, B. Kandasubramanian, Biofabrication 2019, 11, 042001.
A. Khademhosseini, Adv. Mater. 2014, 26, 85. [48] Y. Luo, X. Wei, P. Huang, J. Biomed. Mater. Res., Part B 2019, 107,
[15] J.  Thiele, Y.  Ma, S. M. C.  Bruekers, S.  Ma, W. T. S.  Huck, Adv. 1695.
Mater. 2014, 26, 125. [49] A. N.  Leberfinger, S.  Dinda, Y.  Wu, S. V.  Koduru, V.  Ozbolat,
[16] H. Lipson, M. Kurman, in Fabricated: The New World of 3D Printing, D. J. Ravnic, I. T. Ozbolat, Acta Biomater. 2019, 95, 32.
(Eds: H. Lipson, M. Kurman), John Wiley & Sons, Hoboken, NJ, [50] P.  Datta, A.  Barui, Y.  Wu, V.  Ozbolat, K. K.  Moncal, I. T.  Ozbolat,
USA 2013. Biotechnol. Adv. 2018, 36, 1481.
[17] R. Langer, J. P. Vacanti, Science 1993, 260, 920. [51] F.  Obregon, C.  Vaquette, S.  Ivanovski, D. W.  Hutmacher,
[18] Y. S. Zhang, A. Khademhosseini, Science 2017, 356, eaaf3627. L. E. Bertassoni, J. Dent. Res. 2015, 94, 143S.
[19] X.  Guan, M.  Avci-Adali, E.  Alarçin, H.  Cheng, S. S.  Kashaf, Y.  Li, [52] A. Panwar, L. Tan, Molecules 2016, 21, 685.
A. Chawla, H. L. Jang, A. Khademhosseini, Biotechnol. J. 2017, 12, [53] D. Nieto, J. A. Marchal Corrales, A. Jorge De Mora, L. Moroni, APL
1600394. Bioeng. 2020, 4, 041502.
[20] R.  Gauvin, R.  Parenteau-Bareil, M. R.  Dokmeci, W. D.  Merryman, [54] S. H.  Kim, D. Y.  Kim, T. H.  Lim, C. H.  Park, Adv. Exp. Med. Biol.
A.  Khademhosseini, Wiley Interdiscip. Rev.: Nanomed. Nanobio- 2020, 1249, 53.
technol. 2012, 4, 235. [55] I. Angelopoulos, M. C. Allenby, M. Lim, M. Zamorano, Biotechnol.
[21] A. Khademhosseini, R. Langer, Biomaterials 2007, 28, 5087. Bioeng. 2020, 117, 272.
[22] S. R. Caliari, J. A. Burdick, Nat. Methods 2016, 13, 405. [56] R.  Devillard, E.  Pagès, M. M.  Correa, V.  Kériquel, M.  Rémy,
[23] F. Marga, A. Neagu, I. Kosztin, G. Forgacs, Birth Defects Res., Part J. Kalisky, M. Ali, B. Guillotin, F. Guillemot, Methods Cell Biol. 2014,
C 2007, 81, 320. 119, 159.
[24] S. A. Newman, G. Forgacs, G. B. Muller, Int. J. Dev. Biol. 2006, 50, [57] J.  Groll, J. A.  Burdick, D.-W.  Cho, B.  Derby, M.  Gelinsky,
289. S. C.  Heilshorn, T.  Jüngst, J.  Malda, V. A.  Mironov, K.  Nakayama,
[25] K.  Jakab, C.  Norotte, F.  Marga, K.  Murphy, G.  Vunjak-Novakovic, A.  Ovsianikov, W.  Sun, S.  Takeuchi, J. J.  Yoo, T. B. F.  Woodfield,
G. Forgacs, Biofabrication 2010, 2, 022001. Biofabrication 2018, 11, 013001.
[26] V.  Mironov, R. P.  Visconti, V.  Kasyanov, G.  Forgacs, C. J.  Drake, [58] L.  Moroni, T.  Boland, J. A.  Burdick, C.  De Maria, B.  Derby,
R. R. Markwald, Biomaterials 2009, 30, 2164. G.  Forgacs, J.  Groll, Q.  Li, J.  Malda, V. A.  Mironov, C.  Mota,
[27] K. Białkowska, P. Komorowski, M. Bryszewska, K. Miłowska, Int. J. M.  Nakamura, W.  Shu, S.  Takeuchi, T. B. F.  Woodfield, T.  Xu,
Mol. Sci. 2020, 21, 6225. J. J. Yoo, G. Vozzi, Trends Biotechnol. 2018, 36, 384.

Adv. Mater. 2022, 34, 2101321 2101321  (16 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

[59] L. E.  Bertassoni, J. C.  Cardoso, V.  Manoharan, A. L.  Cristino, [92] K. Yuki, N. Cheng, M. Nakano, C. J. Kuo, Trends Immunol. 2020, 41,
N. S.  Bhise, W. A.  Araujo, P.  Zorlutuna, N. E.  Vrana, 652.
A. M.  Ghaemmaghami, M. R.  Dokmeci, A.  Khademhosseini, [93] K. Homicsko, Curr. Opin. Biotechnol. 2020, 65, 242.
Biofabrication 2014, 6, 024105. [94] H. C. H.  Lau, O.  Kranenburg, H.  Xiao, J.  Yu, Nat. Rev. Gastroen-
[60] A.  Gough, A.  Soto-Gutierrez, L.  Vernetti, M. R.  Ebrahimkhani, terol. Hepatol. 2020, 17, 203.
A. M. Stern, D. L. Taylor, Nat. Rev. Gastroenterol. Hepatol. 2020, 18, 252. [95] H. Fan, U. Demirci, P. Chen, J. Hematol. Oncol. 2019, 12, 142.
[61] L.  Ma, Y.  Wu, Y.  Li, A.  Aazmi, H.  Zhou, B.  Zhang, H.  Yang, Adv. [96] S.  Montes-Olivas, L.  Marucci, M.  Homer, Front. Genet. 2019, 10,
Healthcare Mater. 2020, 9, 2001517. 873.
[62] A.  Da Silva Morais, S.  Vieira, X.  Zhao, Z.  Mao, C.  Gao, [97] E. M.  Holloway, M. M.  Capeling, J. R.  Spence, Development 2019,
J. M. Oliveira, R. L. Reis, Adv. Healthcare Mater. 2020, 9, 1901435. 146, dev166173.
[63] L. A. Van Grunsven, Adv. Drug Delivery Rev. 2017, 121, 133. [98] S. Grebenyuk, A. Ranga, Front. Bioeng. Biotechnol. 2019, 7, 39.
[64] D. G. Nguyen, S. L. Pentoney, Drug Discovery Today: Technol. 2017, [99] D. Blondel, M. P. Lutolf, Chimia (Aarau) 2019, 73, 81.
23, 37. [100] G. Rossi, A. Manfrin, M. P. Lutolf, Nat. Rev. Genet. 2018, 19, 671.
[65] D. G.  Nguyen, J.  Funk, J. B.  Robbins, C.  Crogan-Grundy, [101] R. M. Marton, S. P. Pașca, Trends Cell Biol. 2020, 30, 133.
S. C. Presnell, T. Singer, A. B. Roth, PLoS One 2016, 11, e0158674. [102] A. A.  Panoutsopoulos, Neuroscientist, https://doi.
[66] S. Ben-Moshe, S. Itzkovitz, Nat. Rev. Gastroenterol. Hepatol. 2019, org/10.1177/1073858420960112.
16, 395. [103] A. Atala, G. Forgacs, Stem Cells Transl. Med. 2019, 8, 744.
[67] M.  Hundt, H.  Basit, S.  John, Physiology, Bile Secretion, StatPearls [104] M. Mccune, A. Shafiee, G. Forgacs, I. Kosztin, Soft Matter 2014, 10,
Publishing, Treasure Island, FL, 2020. 1790.
[68] Y. Nakanishi, R. Saxena, Arch. Pathol. Lab. Med. 2015, 139, 858. [105] C.  Norotte, F. S.  Marga, L. E.  Niklason, G.  Forgacs, Biomaterials
[69] D. Häussinger, C. Kordes, Biol. Chem. 2019, 401, 81. 2009, 30, 5910.
[70] A. M. Jorgensen, J. J. Yoo, A. Atala, Chem. Rev. 2020, 120, 11093. [106] K.  Jakab, B.  Damon, A.  Neagu, A.  Kachurin, G.  Forgacs, Biorhe-
[71] E. E. Hui, S. N. Bhatia, Proc. Natl. Acad. Sci. USA 2007, 104, 5722. ology 2006, 43, 509.
[72] B. D. Humphreys, Nat. Mater. 2021, 20, 128. [107] B.  Ayan, N.  Celik, Z.  Zhang, K.  Zhou, M. H.  Kim, D.  Banerjee,
[73] K. T.  Lawlor, J. M.  Vanslambrouck, J. W.  Higgins, A.  Chambon, Y. Wu, F. Costanzo, I. T. Ozbolat, Commun. Phys. 2020, 3, 183.
K.  Bishard, D.  Arndt, P. X.  Er, S. B.  Wilson, S. E.  Howden, [108] B. Ayan, D. N. Heo, Z. Zhang, M. Dey, A. Povilianskas, C. Drapaca,
K. S. Tan, F. Li, L. J. Hale, B. Shepherd, S. Pentoney, S. C. Presnell, I. T. Ozbolat, Sci. Adv. 2020, 6, eaaw5111.
A. E. Chen, M. H. Little, Nat. Mater. 2021, 20, 260. [109] A. C. Daly, M. D. Davidson, J. A. Burdick, Nat. Commun. 2021, 12,
[74] L.  Serex, K.  Sharma, V.  Rizov, A.  Bertsch, J. D.  McKinney, 753.
P. Renaud, Biofabrication 2020, 13, 025006. [110] J.  Duisit, H.  Amiel, T.  Wüthrich, A.  Taddeo, A.  Dedriche,
[75] J. A.  Brassard, M.  Nikolaev, T.  Hübscher, M.  Hofer, M. P.  Lutolf, V. Destoop, T. Pardoen, C. Bouzin, V. Joris, D. Magee, E. Vögelin,
Nat. Mater. 2021, 20, 22. D.  Harriman, C.  Dessy, G.  Orlando, C.  Behets, R.  Rieben,
[76] S. V. Murphy, A. Atala, Nat. Biotechnol. 2014, 32, 773. P. Gianello, B. Lengelé, Acta Biomater. 2018, 73, 339.
[77] H.-W.  Kang, S. J.  Lee, I. K.  Ko, C.  Kengla, J. J.  Yoo, A.  Atala, Nat. [111] A. J. Mellott, H. E. Shinogle, J. G. Nelson-Brantley, M. S. Detamore,
Biotechnol. 2016, 34, 312. H. Staecker, Stem Cell Res. Ther. 2017, 8, 41.
[78] N. S.  Bhise, V.  Manoharan, S.  Massa, A.  Tamayol, M.  Ghaderi, [112] J. Xue, B. Feng, R. Zheng, Y. Lu, G. Zhou, W. Liu, Y. Cao, Y. Zhang,
M.  Miscuglio, Q.  Lang, Y.  Shrike Zhang, S. R.  Shin, G.  Calzone, W. J. Zhang, Biomaterials 2013, 34, 2624.
N.  Annabi, T. D.  Shupe, C. E.  Bishop, A.  Atala, M. R.  Dokmeci, [113] L.  Zhou, I.  Pomerantseva, E. K.  Bassett, C. M.  Bowley, X.  Zhao,
A. Khademhosseini, Biofabrication 2016, 8, 014101. D. A.  Bichara, K. M.  Kulig, J. P.  Vacanti, M. A.  Randolph,
[79] X. Ma, X. Qu, W. Zhu, Y.-S. Li, S. Yuan, H. Zhang, J. Liu, P. Wang, C. A. Sundback, Tissue Eng., Part A 2011, 17, 1573.
C. S. E.  Lai, F.  Zanella, G.-S.  Feng, F.  Sheikh, S.  Chien, S.  Chen, [114] A. Haisch, Adv. Oto-Rhino-Laryngol. 2010, 68, 108.
Proc. Natl. Acad. Sci. USA 2016, 113, 2206. [115] Y.  Liu, L.  Zhang, G.  Zhou, Q.  Li, W.  Liu, Z.  Yu, X.  Luo, T.  Jiang,
[80] D.  Kang, G.  Hong, S.  An, I.  Jang, W.-S.  Yun, J.-H.  Shim, S.  Jin, W. Zhang, Y. Cao, Biomaterials 2010, 31, 2176.
Small 2020, 16, 1905505. [116] T. H.  Jovic, E. J.  Combellack, Z. M.  Jessop, I. S.  Whitaker, Front.
[81] D.  Kang, G.  Ahn, D.  Kim, H.-W.  Kang, S.  Yun, W.-S.  Yun, Surg. 2020, 7, 44.
J.-H. Shim, S. Jin, Biofabrication 2018, 10, 035008. [117] E.  Mussi, R.  Furferi, Y.  Volpe, F.  Facchini, K. S.  McGreevy,
[82] L. M.  Norona, D. G.  Nguyen, D. A.  Gerber, S. C.  Presnell, F. Uccheddu, Bioengineering (Basel) 2019, 6, 14.
M. Mosedale, P. B. Watkins, PLoS One 2019, 14, e0208958. [118] J.-S. Lee, J. M. Hong, J. W. Jung, J.-H. Shim, J.-H. Oh, D.-W. Cho,
[83] K. G. Battiston, J. W. C. Cheung, D. Jain, J. P. Santerre, Biomaterials Biofabrication 2014, 6, 024103.
2014, 35, 4465. [119] Y.  Ling, J.  Rubin, Y.  Deng, C.  Huang, U.  Demirci, J. M.  Karp,
[84] N. K.  Paschos, W. E.  Brown, R.  Eswaramoorthy, J. C.  Hu, A. Khademhosseini, Lab Chip 2007, 7, 756.
K. A. Athanasiou, J. Tissue Eng. Regener. Med. 2015, 9, 488. [120] A.  Athirasala, A.  Tahayeri, G.  Thrivikraman, C. M.  França,
[85] Y.  Miki, K.  Ono, S.  Hata, T.  Suzuki, H.  Kumamoto, H.  Sasano, N.  Monteiro, V.  Tran, J.  Ferracane, L. E.  Bertassoni, Biofabrication
J. Steroid Biochem. Mol. Biol. 2012, 131, 68. 2018, 10, 024101.
[86] B. Van Der Sanden, M. Dhobb, F. Berger, D. Wion, J. Cell. Biochem. [121] M. B.  Łabowska, K.  Cierluk, A. M.  Jankowska, J.  Kulbacka,
2010, 111, 801. J. Detyna, I. Michalak, Materials (Basel) 2021, 14, 858.
[87] P. Abdollahiyan, F. Oroojalian, A. Mokhtarzadeh, M. Guardia, Bio- [122] C. C. Piras, D. K. Smith, J. Mater. Chem. B 2020, 8, 8171.
technol. J. 2020, 15, 2000095. [123] E. Axpe, M. Oyen, Int. J. Mol. Sci. 2016, 17, 1976.
[88] F. You, B. F. Eames, X. Chen, Int. J. Mol. Sci. 2017, 18, 1597. [124] S. H. Ahn, H. J. Lee, J.-S. Lee, H. Yoon, W. Chun, G. H. Kim, Sci.
[89] E.  Garreta, R. D.  Kamm, S. M.  Chuva De Sousa Lopes, Rep. 2015, 5, 13427.
M. A. Lancaster, R. Weiss, X. Trepat, I. Hyun, N. Montserrat, Nat. [125] N.  Faramarzi, I. K.  Yazdi, M.  Nabavinia, A.  Gemma, A.  Fanelli,
Mater. 2021, 20, 145. A.  Caizzone, L. M.  Ptaszek, I.  Sinha, A.  Khademhosseini,
[90] S. M. Kang, D. Kim, J.-H. Lee, S. Takayama, J. Y. Park, Adv. Health- J. N. Ruskin, A. Tamayol, Adv. Healthcare Mater. 2018, 7, 1701347.
care Mater. 2021, 10, 2001284. [126] D.  Choudhury, S.  Anand, M. W.  Naing, Int. J. Bioprint. 2018, 4,
[91] Y. Li, P. Tang, S. Cai, J. Peng, G. Hua, Cell Regen. 2020, 9, 21. 139.

Adv. Mater. 2022, 34, 2101321 2101321  (17 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

[127] A. Urciuolo, I. Poli, L. Brandolino, P. Raffa, V. Scattolini, C. Laterza, [155] D. B.  Kolesky, R. L.  Truby, A. S.  Gladman, T. A.  Busbee,
G. G.  Giobbe, E.  Zambaiti, G.  Selmin, M.  Magnussen, L.  Brigo, K. A. Homan, J. A. Lewis, Adv. Mater. 2014, 26, 3124.
P.  De Coppi, S.  Salmaso, M.  Giomo, N.  Elvassore, Nat. Biomed. [156] L. E.  Bertassoni, M.  Cecconi, V.  Manoharan, M.  Nikkhah,
Eng. 2020, 4, 901. J.  Hjortnaes, A. L.  Cristino, G.  Barabaschi, D.  Demarchi,
[128] I. T. Ozbolat, Trends Biotechnol. 2015, 33, 395. M. R.  Dokmeci, Y.  Yang, A.  Khademhosseini, Lab Chip 2014, 14,
[129] S. Singh, D. Choudhury, F. Yu, V. Mironov, M. W. Naing, Acta Bio- 2202.
mater. 2020, 101, 14. [157] C. Twohig, M. Helsinga, A. Mansoorifar, A. Athirasala, A. Tahayeri,
[130] N. Hong, G.-H. Yang, J. Lee, G. Kim, J. Biomed. Mater. Res., Part B C. M.  França, S. A.  Pajares, R.  Abdelmoniem, S.  Scherrer,
2018, 106, 444. S.  Durual, J.  Ferracane, L. E.  Bertassoni, Mater. Sci. Eng., C 2021,
[131] R.-Z.  Lin, Y.-C.  Chen, R.  Moreno-Luna, A.  Khademhosseini, 123, 111976.
J. M. Melero-Martin, Biomaterials 2013, 34, 6785. [158] A. Mansoorifar, A. Tahayeri, L. E. Bertassoni, Lab Chip 2020, 20, 1713.
[132] J.  Elisseeff, K.  Anseth, D.  Sims, W.  Mcintosh, M.  Randolph, [159] I. S.  Kinstlinger, S. H.  Saxton, G. A.  Calderon, K. V.  Ruiz,
M. Yaremchuk, R. Langer, Plast. Reconstr. Surg. 1999, 104, 1014. D. R. Yalacki, P. R. Deme, J. E. Rosenkrantz, J. D. Louis-Rosenberg,
[133] J.  Elisseeff, K.  Anseth, D.  Sims, W.  Mcintosh, M.  Randolph, F.  Johansson, K. D.  Janson, D. W.  Sazer, S. S.  Panchavati,
R. Langer, Proc. Natl. Acad. Sci. USA 1999, 96, 3104. K.-D. Bissig, K. R. Stevens, J. S. Miller, Nat. Biomed. Eng. 2020, 4,
[134] A. Lee, A. R. Hudson, D. J. Shiwarski, J. W. Tashman, T. J. Hinton, 916.
S.  Yerneni, J. M.  Bliley, P. G.  Campbell, A. W.  Feinberg, Science [160] N. Puluca, S. Lee, S. Doppler, A. Münsterer, M. Dreßen, M. Krane,
2019, 365, 482. S. M. Wu, Curr. Cardiol. Rep. 2019, 21, 90.
[135] C. B. Highley, K. H. Song, A. C. Daly, J. A. Burdick, Adv. Sci. 2019, [161] S. Abdollahi, J. Boktor, N. Hibino, Transl. Res. 2019, 211, 123.
6, 1801076. [162] A.  Morss Clyne, S.  Swaminathan, A.  Díaz Lantada, Biofabrication
[136] C. S. O’bryan, T. Bhattacharjee, S. Hart, C. P. Kabb, K. D. Schulze, 2019, 11, 032001.
I. Chilakala, B. S. Sumerlin, W. G. Sawyer, T. E. Angelini, Sci. Adv. [163] M. D. Sarker, S. Naghieh, N. K. Sharma, X. Chen, J. Pharm. Anal.
2017, 3, e1602800. 2018, 8, 277.
[137] K. J. Leblanc, S. R. Niemi, A. I. Bennett, K. L. Harris, K. D. Schulze, [164] B. Grigoryan, S. J. Paulsen, D. C. Corbett, D. W. Sazer, C. L. Fortin,
W. G. Sawyer, C. Taylor, T. E. Angelini, ACS Biomater. Sci. Eng. 2016, A. J.  Zaita, P. T.  Greenfield, N. J.  Calafat, J. P.  Gounley, A. H.  Ta,
2, 1796. F. Johansson, A. Randles, J. E. Rosenkrantz, J. D. Louis-Rosenberg,
[138] T.  Bhattacharjee, C. J.  Gil, S. L.  Marshall, J. M.  Urueña, P. A. Galie, K. R. Stevens, J. S. Miller, Science 2019, 364, 458.
C. S.  O’bryan, M.  Carstens, B.  Keselowsky, G. D.  Palmer, [165] A.  Naderi, N.  Bhattacharjee, A.  Folch, Annu. Rev. Biomed. Eng.
S.  Ghivizzani, C. P.  Gibbs, W. G.  Sawyer, T. E.  Angelini, ACS Bio- 2019, 21, 325.
mater. Sci. Eng. 2016, 2, 1787. [166] N.  Bhattacharjee, A.  Urrios, S.  Kang, A.  Folch, Lab Chip 2016, 16,
[139] T. Bhattacharjee, S. M. Zehnder, K. G. Rowe, S. Jain, R. M. Nixon, 1720.
W. G. Sawyer, T. E. Angelini, Sci. Adv. 2015, 1, e1500655. [167] A. K.  Au, W.  Huynh, L. F.  Horowitz, A.  Folch, Angew. Chem., Int.
[140] E.  Mirdamadi, J. W.  Tashman, D. J.  Shiwarski, R. N.  Palchesko, Ed. Engl. 2016, 55, 3862.
A. W. Feinberg, ACS Biomater. Sci. Eng. 2020, 6, 6453. [168] R.  Subbiah, G.  Thrivikraman, S. P.  Parthiban, J. M.  Jones,
[141] M. A.  Skylar-Scott, S. G. M.  Uzel, L. L.  Nam, J. H.  Ahrens, A. Athirasala, H. Xie, L. E. Bertassoni, J. Tissue Eng. Regener. Med.
R. L. Truby, S. Damaraju, J. A. Lewis, Sci. Adv. 2019, 5, eaaw2459. 2021, 15, 219.
[142] T. J.  Hinton, Q.  Jallerat, R. N.  Palchesko, J. H.  Park, [169] S. P.  Parthiban, W.  He, N.  Monteiro, A.  Athirasala, C. M.  França,
M. S.  Grodzicki, H.-J.  Shue, M. H.  Ramadan, A. R.  Hudson, L. E. Bertassoni, Sci. Rep. 2020, 10, 21579.
A. W. Feinberg, Sci. Adv. 2015, 1, e1500758. [170] L. E. Bertassoni, J. Endod. 2020, 46, S90.
[143] N. Noor, A. Shapira, R. Edri, I. Gal, L. Wertheim, T. Dvir, Adv. Sci. [171] G.  Thrivikraman, A.  Athirasala, R.  Gordon, L.  Zhang, R.  Bergan,
2019, 6, 1900344. D. R.  Keene, J. M.  Jones, H.  Xie, Z.  Chen, J.  Tao, B.  Wingender,
[144] H.  Bae, A. S.  Puranik, R.  Gauvin, F.  Edalat, B.  Carrillo-Conde, L. Gower, J. L. Ferracane, L. E. Bertassoni, Nat. Commun. 2019, 10,
N. A. Peppas, A. Khademhosseini, Sci. Transl. Med. 2012, 4, 160ps23. 3520.
[145] I. S. Kinstlinger, J. S. Miller, Lab Chip 2016, 16, 2025. [172] N. Monteiro, W. He, C. M. Franca, A. Athirasala, L. E. Bertassoni,
[146] S. P. Parthiban, P. Jurney, A. Athirasala, C. M. França, A. Tahayeri, ACS Biomater. Sci. Eng. 2018, 4, 2563.
P.  Menezes, L. E.  Bertassoni, in Emerging Technologies for Bio- [173] S. Zhang, Z. Wan, R. D. Kamm, Lab Chip 2021, 21, 473.
fabrication and Biomanufacturing, (Eds: U. Demirci, R. El Assal, [174] A. Zimmerling, X. Chen, Bioprinting 2020, 20, e00104.
P. Cheng), World Scientific Publishing, Singapore 2020. [175] A. Arslan-Yildiz, R. El Assal, P. Chen, S. Guven, F. Inci, U. Demirci,
[147] J. S. Miller, K. R. Stevens, M. T. Yang, B. M. Baker, D.-H. T. Nguyen, Biofabrication 2016, 8, 014103.
D. M. Cohen, E. Toro, A. A. Chen, P. A. Galie, X. Yu, R. Chaturvedi, [176] S. Tasoglu, U. Demirci, Trends Biotechnol. 2013, 31, 10.
S. N. Bhatia, C. S. Chen, Nat. Mater. 2012, 11, 768. [177] Y. S.  Zhang, K.  Yue, J.  Aleman, K.  Mollazadeh-Moghaddam,
[148] K. H. K. Wong, J. M. Chan, R. D. Kamm, J. Tien, Annu. Rev. Biomed. S. M.  Bakht, J.  Yang, W.  Jia, V.  Dell’erba, P.  Assawes, S. R.  Shin,
Eng. 2012, 14, 205. M. R.  Dokmeci, R.  Oklu, A.  Khademhosseini, Ann. Biomed. Eng.
[149] A. P. Golden, J. Tien, Lab Chip 2007, 7, 720. 2017, 45, 148.
[150] J. Tien, C. M. Nelson, C. S. Chen, Proc. Natl. Acad. Sci. USA 2002, [178] P. D. Dalton, T. B. F. Woodfield, V. Mironov, J. Groll, Adv. Sci. 2020,
99, 1758. 7, 1902953.
[151] Y. S.  Song, R. L.  Lin, G.  Montesano, N. G.  Durmus, G.  Lee, [179] C. Twohig, M. Helsinga, A. Mansoorifar, A. Athirasala, A. Tahayeri,
S.-S.  Yoo, E.  Kayaalp, E.  Hæggström, A.  Khademhosseini, C. M.  França, S. A.  Pajares, R.  Abdelmoniem, S.  Scherrer,
U. Demirci, Anal. Bioanal. Chem. 2009, 395, 185. S.  Durual, J.  Ferracane, L. E.  Bertassoni, Mater. Sci. Eng., C 2021,
[152] A. Khademhosseini, J. Yeh, S. Jon, G. Eng, K. Y. Suh, J. A. Burdick, 123, 111976.
R. Langer, Lab Chip 2004, 4, 425. [180] W.  Liu, Y. S.  Zhang, M. A.  Heinrich, F.  De Ferrari, H. L.  Jang,
[153] A.  Khademhosseini, K. Y.  Suh, S.  Jon, G.  Eng, J.  Yeh, G.-J.  Chen, S. M.  Bakht, M. M.  Alvarez, J.  Yang, Y.-C.  Li, G.  Trujillo-de
R. Langer, Anal. Chem. 2004, 76, 3675. Santiago, A. K.  Miri, K.  Zhu, P.  Khoshakhlagh, G.  Prakash,
[154] D. B.  Kolesky, K. A.  Homan, M. A.  Skylar-Scott, J. A.  Lewis, Proc. H. Cheng, X. Guan, Z. Zhong, J. Ju, G. H. Zhu, X. Jin, S. R. Shin,
Natl. Acad. Sci. USA 2016, 113, 3179. M. R. Dokmeci, A. Khademhosseini, Adv. Mater. 2017, 29, 1604630.

Adv. Mater. 2022, 34, 2101321 2101321  (18 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

[181] L.  Koch, M.  Gruene, C.  Unger, B.  Chichkov, Curr. Pharm. Bio- [206] A. Ainla, G. D. M. Jeffries, R. Brune, O. Orwar, A. Jesorka, Lab Chip
technol. 2013, 14, 91. 2012, 12, 1255.
[182] X.  Cui, T.  Boland, D. D.  D’lima, M. K.  Lotz, Recent Pat. Drug [207] G. D. M.  Jeffries, S.  Xu, T.  Lobovkina, V.  Kirejev, F.  Tusseau,
Delivery Formulation 2012, 6, 149. C.  Gyllensten, A. K.  Singh, P.  Karila, L.  Moll, O.  Orwar, Sci. Rep.
[183] P. G. Campbell, L. E. Weiss, Expert Opin. Biol. Ther. 2007, 7, 1123. 2010, 10, 19529.
[184] A. K.  Miri, D.  Nieto, L.  Iglesias, H.  Goodarzi Hosseinabadi, [208] K.  Chung, J.  Wallace, S.-Y.  Kim, S.  Kalyanasundaram,
S.  Maharjan, G. U.  Ruiz-Esparza, P.  Khoshakhlagh, A.  Manbachi, A. S.  Andalman, T. J.  Davidson, J. J.  Mirzabekov, K. A.  Zalocusky,
M. R.  Dokmeci, S.  Chen, S. R.  Shin, Y. S.  Zhang, J.  Mattis, A. K.  Denisin, S.  Pak, H.  Bernstein, C.  Ramakrishnan,
A. Khademhosseini, Adv. Mater. 2018, 30, 1800242. L. Grosenick, V. Gradinaru, K. Deisseroth, Nature 2013, 497, 332.
[185] W.  Li, L. S.  Mille, J. A.  Robledo, T.  Uribe, V.  Huerta, Y. S.  Zhang, [209] M. K. Driscoll, A. Zaritsky, J. Cell Sci. 2021, 134, jcs254292.
Adv. Healthcare Mater. 2020, 9, 2000156. [210] R. Weinkamer, P. Kollmannsberger, P. Fratzl, Curr. Osteoporosis Rep.
[186] W. L. Ng, J. M. Lee, M. Zhou, Y.-W. Chen, K.-.X. A. Lee, W. Y. Yeong, 2019, 17, 186.
Y.-F. Shen, Biofabrication 2020, 12, 022001. [211] P. Roschger, E. P. Paschalis, P. Fratzl, K. Klaushofer, Bone 2008, 42,
[187] J.  Van Hoorick, L.  Tytgat, A.  Dobos, H.  Ottevaere, J.  Van Erps, 456.
H. Thienpont, A. Ovsianikov, P. Dubruel, S. Van Vlierberghe, Acta [212] O.  Paris, I.  Zizak, H.  Lichtenegger, P.  Roschger, K.  Klaushofer,
Biomater. 2019, 97, 46. P. Fratzl, Cell. Mol. Biol. 2000, 46, 993.
[188] A. J.  Guerra, H.  Lara-Padilla, M. L.  Becker, C. A.  Rodriguez, [213] M.-G. Ascenzi, Front. Biosci., Landmark Ed. 2012, 17, 1551.
D. Dean, Curr. Drug Targets 2019, 20, 823. [214] A. S.  Avrunin, R. M.  Tikhilov, I. I.  Shubniakov, L. A.  Parshin,
[189] K. C.  Hribar, P.  Soman, J.  Warner, P.  Chung, S.  Chen, Lab Chip B. E. Mel’nikov, D. G. Pliev, Morfologiia 2010, 137, 69.
2014, 14, 268. [215] N. D. Sahar, S.-I. Hong, D. H. Kohn, Micron 2005, 36, 617.
[190] J. R.  Tumbleston, D.  Shirvanyants, N.  Ermoshkin, R.  Janusziewicz, [216] J. A. Buckwalter, R. R. Cooper, Instr. Course Lect. 1987, 36, 27.
A. R.  Johnson, D.  Kelly, K.  Chen, R.  Pinschmidt, J. P.  Rolland, [217] Y.  Zhang, D.  Wu, X.  Zhao, M.  Pakvasa, A. B.  Tucker, H.  Luo,
A. Ermoshkin, E. T. Samulski, J. M. Desimone, Science 2015, 347, 1349. K. H.  Qin, D. A.  Hu, E. J.  Wang, A. J.  Li, M.  Zhang, Y.  Mao,
[191] K. S. Lim, F. Abinzano, P. N. Bernal, A. Albillos Sanchez, P. Atienza- M.  Sabharwal, F.  He, C.  Niu, H.  Wang, L.  Huang, D.  Shi, Q.  Liu,
Roca, I. A.  Otto, Q. C.  Peiffer, M.  Matsusaki, T. B. F.  Woodfield, N.  Ni, K.  Fu, C.  Chen, W.  Wagstaff, R. R.  Reid, A.  Athiviraham,
J. Malda, R. Levato, Adv. Healthcare Mater. 2020, 9, 1901792. S.  Ho, M. J.  Lee, K.  Hynes, J.  Strelzow, T.-C.  He, M.  El Dafrawy,
[192] B. G.  Soliman, G. C. J.  Lindberg, T.  Jungst, G. J.  Hooper, J.  Groll, Front. Bioeng. Biotechnol. 2020, 8, 598607.
T. B. F. Woodfield, K. S. Lim, Adv. Healthcare Mater. 2020, 9, 1901544. [218] M. Bahraminasab, Biomed. Eng. Online 2020, 19, 69.
[193] K. S. Lim, J. H. Galarraga, X. Cui, G. C. J. Lindberg, J. A. Burdick, [219] M.  Salah, L.  Tayebi, K.  Moharamzadeh, F. B.  Naini, Maxillofac.
T. B. F. Woodfield, Chem. Rev. 2020, 120, 10662. Plast. Reconstr. Surg. 2020, 42, 18.
[194] S.  Bertlein, G.  Brown, K. S.  Lim, T.  Jungst, T.  Boeck, T.  Blunk, [220] G. S.  Kronemberger, R. A. M.  Matsui, G. A. S. C.  Miranda,
J.  Tessmar, G. J.  Hooper, T. B. F.  Woodfield, J.  Groll, Adv. Mater. J. M. Granjeiro, L. S. Baptista, World J. Stem Cells 2020, 12, 110.
2017, 29, 1703404. [221] E. J. Sheehy, D. J. Kelly, F. J. O’Brien, Mater. Today Bio 2019, 3, 100009.
[195] B. Grigoryan, D. W. Sazer, A. Avila, J. L. Albritton, A. Padhye, A. H. Ta, [222] O.  Tao, J. K.  -Mascort, Y.  Lin, H. M.  Pham, A. M.  Charbonneau,
P. T. Greenfield, D. L. Gibbons, J. S. Miller, Sci. Rep. 2021, 11, 3171. O. A.  ElKashty, J. M.  Kinsella, S. D.  Tran, Micromachines (Basel)
[196] S. P.  Parthiban, A.  Athirasala, A.  Tahayeri, R.  Abdelmoniem, 2019, 10, 480.
A. George, L. E. Bertassoni, Biofabrication 2020, 13, 035031. [223] L. Zhang, G. Yang, B. N. Johnson, X. Jia, Acta Biomater. 2019, 84, 16.
[197] M. H. Chen, J. J. Chung, J. E. Mealy, S. Zaman, E. C. Li, M. F. Arisi, [224] A.  Dhawan, P. M.  Kennedy, E. B.  Rizk, I. T.  Ozbolat, J Am. Acad.
P. Atluri, J. A. Burdick, Macromol. Biosci. 2019, 19, 1800248. Orthop. Surg. 2019, 27, e215.
[198] C. B.  Rodell, C. B.  Highley, M. H.  Chen, N. N.  Dusaj, C.  Wang, [225] S. Zhang, H. Wang, SLAS Technol. 2019, 24, 70.
L. Han, J. A. Burdick, Soft Matter 2016, 12, 7839. [226] L.  Roseti, V.  Parisi, M.  Petretta, C.  Cavallo, G.  Desando,
[199] Y.  Dong, J.  Wang, X.  Guo, S.  Yang, M. O.  Ozen, P.  Chen, X.  Liu, I. Bartolotti, B. Grigolo, Mater. Sci. Eng., C 2017, 78, 1246.
W. Du, F. Xiao, U. Demirci, B.-F. Liu, Nat. Commun. 2019, 10, 4087. [227] F. Kabirian, M. Mozafari, Methods 2020, 171, 108.
[200] S. Tasoglu, E. Diller, S. Guven, M. Sitti, U. Demirci, Nat. Commun. [228] D. Choudhury, H. W. Tun, T. Wang, M. W. Naing, Trends Biotechnol.
2014, 5, 3124. 2018, 36, 787.
[201] H.  Qi, M.  Ghodousi, Y.  Du, C.  Grun, H.  Bae, P.  Yin, [229] G. M.  Cunniffe, T.  Gonzalez-Fernandez, A.  Daly, B. N.  Sathy,
A. Khademhosseini, Nat. Commun. 2013, 4, 2275. O.  Jeon, E.  Alsberg, D. J.  Kelly, Tissue Eng., Part A 2017, 23,
[202] R.  Subbiah, C.  Hipfinger, A.  Tahayeri, A.  Athirasala, 891.
S.  Horsophonphong, G.  Thrivikraman, C. M.  França, D. A.  Cunha, [230] M. Saito, K. Marumo, Calcif. Tissue Int. 2015, 97, 242.
A.  Mansoorifar, A.  Zahariev, J. M.  Jones, P. G.  Coelho, L.  Witek, [231] L. E. Bertassoni, M. V. Swain, J. Mech. Behav. Biomed. Mater. 2014,
H. Xie, R. E. Guldberg, L. E. Bertassoni, Adv. Mater. 2020, 32, 2001736. 38, 91.
[203] M.  Regehly, Y.  Garmshausen, M.  Reuter, N. F.  König, E.  Israel, [232] E. Jabbari, Curr. Pharm. Des. 2013, 19, 3391.
D. P.  Kelly, C.-Y.  Chou, K.  Koch, B.  Asfari, S.  Hecht, Nature 2020, [233] O.  Messaoudi, C.  Henrionnet, K.  Bourge, D.  Loeuille, P.  Gillet,
588, 620. A. Pinzano, Cells 2020, 10, 2.
[204] E.  Davoodi, E.  Sarikhani, H.  Montazerian, S.  Ahadian, [234] G. Cidonio, M. Glinka, J. I. Dawson, R. O. C. Oreffo, Biomaterials
M.  Costantini, W.  Swieszkowski, S. M.  Willerth, K.  Walus, 2019, 209, 10.
M.  Mofidfar, E.  Toyserkani, A.  Khademhosseini, N.  Ashammakhi, [235] M.  Askari, M.  Afzali Naniz, M.  Kouhi, A.  Saberi, A.  Zolfagharian,
Adv. Mater. Technol. 2020, 5, 1901044. M. Bodaghi, Biomater. Sci. 2021, 9, 535.
[205] C.  Colosi, S. R.  Shin, V.  Manoharan, S.  Massa, M.  Costantini, [236] J. J. Martin, B. E. Fiore, R. M. Erb, Nat. Commun. 2015, 6, 8641.
A.  Barbetta, M. R.  Dokmeci, M.  Dentini, A.  Khademhosseini, [237] K. B. Blagoev, Proc. Natl. Acad. Sci. USA 2011, 108, 19216.
Adv. Mater. 2016, 28, 677. [238] S. N. Bhatia, D. E. Ingber, Nat. Biotechnol. 2014, 32, 760.

Adv. Mater. 2022, 34, 2101321 2101321  (19 of 20) © 2021 Wiley-VCH GmbH
www.advancedsciencenews.com www.advmat.de

Luiz E. Bertassoni is an associate professor at Oregon Health & Science University, where he
holds appointments in the School of Dentistry, the Department of Biomedical Engineering, and
the Cancer Early Detection Advanced Research Center (CEDAR) at the Knight Cancer Institute.
He is a clinician/scientist with a dental degree, a Ph.D. in biomaterials from the University
of Sydney, and two postdoctoral fellowships, at University of California San Francisco and
the Harvard–MIT Division of Health Science and Technology. His work encompasses various
aspects of bioprinting and microscale technologies for tissue regeneration, and different aspects
of mineralized tissue engineering, organs-on-a-chip and cancer.

Adv. Mater. 2022, 34, 2101321 2101321  (20 of 20) © 2021 Wiley-VCH GmbH

You might also like