You are on page 1of 11

Perspective

https://doi.org/10.1038/s41551-019-0471-7

Opportunities and challenges of translational


3D bioprinting
Sean V. Murphy 1
*, Paolo De Coppi2 and Anthony Atala1

3D-printed orthopaedic devices and surgical tools, printed maxillofacial implants and other printed acellular devices have been
used in patients. By contrast, bioprinted living cellular constructs face considerable translational challenges. In this Perspective,
we first summarize the most recent developments in 3D bioprinting for clinical applications, with a focus on how 3D-printed
cartilage, bone and skin can be designed for individual patients and fabricated using the patient’s own cells. We then discuss
key translational considerations, such as the need to ensure close integration of the living device with the patient’s vascular
network, the development of biocompatible bioinks and the challenges in deriving a physiologically relevant number of cells.
Lastly, we outline untested regulatory pathways, as well as logistical challenges in material sourcing, manufacturing, standard-
ization and transportation.

T
hree-dimensional printing—a type of additive manufactur- still lack essential functional elements, such as vasculature, innerva-
ing—creates 3D objects through the digitally controlled tion, lymphatics, and the number and diversity of functional and
deposition of successive layers of material. The United supporting cell types required for tissues and organs that are large
States Food and Drug Administration (FDA) has cleared several and complex. Owing to these challenges, early successes in the clini-
3D-printed devices for clinical use, including orthopaedic devices, cal translation of 3D-printed living tissues are likely to involve rela-
patient-matched implants, surgical guides and restorative devices tively simple tissues (rather than tissues with complex geometries
such as dental bridges1,2. However, to date most medical applica- at the scales that are clinically relevant). In this Perspective, we pro-
tions of 3D printing have involved static non-living constructs vide an overview of the translational opportunities and challenges
designed to function as structural or space-filling prostheses3. of 3D-printed tissues and organs. We first discuss recent advances
Personalized 3D-printed titanium plates, specifically modelled via in the 3D bioprinting of cartilage, bone and skin, and why these
image reconstruction following magnetic resonance imaging (MRI) bioprinted tissues have progressed to the preclinical stage, and then
and computed tomography (CT), have significantly improved out- outline challenges in the fabrication of more complex 3D-printed
comes in patients undergoing tissue reconstruction after the sur- tissues and organs.
gical removal of bone tumours4. Similarly, 3D-printed genioplasty
template systems can provide greater accuracy in the repositioning Clinically relevant 3D-bioprinted tissues
of the chin than traditional intraoperative measurements5. In 2013, The capabilities of 3D bioprinting are at a stage where multiple
a 3D-printed and personalized bioresorbable external airway splint biomaterials and cell types can be patterned into constructs
was implanted in an infant with tracheobronchomalacia6. In 2015, a approaching clinically relevant sizes and geometries. 3D bioprinted
further three infants received patient-matched 3D-printed splints7. tissues, such as cartilage, bone and skin, exemplify the potential of
These constructs, although non-living, were designed to prevent the technology.
external airway compression over a predetermined period before
they are resorbed by the body to accommodate airway growth. 3D Cartilage. Cartilaginous tissues are avascular and aneural struc-
printing has also been applied in the layer-by-layer manufacturing tures with a relatively low density of chondrocytes. Their simplic-
of oral pills (such as Spritam, the commercial name for the drug ity limits potential hurdles in the fabrication of human cartilage at
levetiracetam) that deliver a high drug dosage while being porous scale. However, because of the heterogeneity (zonal structure) of the
enough to dissolve quickly8. tissue9, it is challenging to reproduce functional articular cartilage.
These recent developments in the design and fabrication of non- The zonal structure of native articular cartilage includes areas with
living 3D-printed implantable constructs illustrate the potential of different cell morphologies and cell arrangements, as well as with
3D-printing technology for the production of advanced patient- different extracellular matrix (ECM) arrangements, constituents
specific devices. By contrast, the bioprinting of living cellular con- and distribution. Such heterogeneity in structure is associated with
structs has faced significant hurdles, in particular: the transition the tissue’s tensile properties, which enable it to resist the sheer, ten-
from synthetic materials—including metals, ceramics and plas- sile and compressive forces imposed by articulation10,11.
tics—to biologically functional materials while maintaining control Chondrocytes can be harvested from different zones of the car-
over the mechanical properties at the microscale and macroscale; tilage12 and, by using 3D printers, deposited in hydrogels (such as
achieving tissue designs with physiological heterogeneity; develop- gelatin and alginate, cartilage-derived ECM, and nanofibrillated
ing methods to derive and expand functional cells from stem cells; cellulose and alginate13–16), with the cells maintaining high viability
and interfacing bioprinted tissues with a physiological vasculature and zone-specific gene-expression profiles17,18. Deposition of human
network. 3D-printed tissues at the preclinical stage of development chondrocytes within a shear-thinning nanofibrillated cellulose can

Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. 2Stem Cells & Regenerative Medicine
1

Section, University College London, Great Ormond Street Institute of Child Health, London, UK. *e-mail: semurphy@wakehealth.edu

370 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng


NaTure BIomeDIcaL EngIneerIng Perspective
a b
Day 0
2 mm

10 mm 5 mm
Red: cells
Green: PCL/TCP 5 months

5 mm

10 mm 10 mm

c d
i ii

iii iv

Fig. 1 | Bioprinted cartilage, bone and skin tissues. a, Anatomically shaped 3D-printed cartilage structures (a human ear, left; and a sheep meniscus,
right). b, A bioprinted human-scale calvarial structure amenable to facial reconstruction. Visualized motion program for 3D-printing of the calvarial bone
construct (green and red indicate the dispensing paths of, respectively, the polymer mixture and cell-laden hydrogel) (top left). Photograph of the printed
construct (left). Implantation in a calvarial bone-defect region in Sprague Dawley rats (top right). Construct 5 months post-implantation (centre right).
Histology image of the construct five months after implantation (bottom). c, In situ skin-bioprinting concept. Wounds are first scanned (i) to obtain
precise information on wound topography (ii), which then guides the print heads to deposit specified materials (iii) and/or cell types (iv) in appropriate
locations. d, Skin bioprinter. Wound scanning with a hand-held ZScanner Z700 scanner (top left). The geometric information obtained by scanning is then
input into software for orienting the scanned images to the standard coordinate system (top right). The scanned data with its coordinate system is used to
generate the fill volume and the path points for the nozzle head (bottom left). Printing of the fill volume (bottom right). Panels reproduced from ref. 16,
ACS (a); ref. 26, Springer Nature Ltd (b); and ref. 49, Springer Nature Ltd (c,d).

also be combined with crosslinkable alginate to print anatomically layered construct of natural and synthetic biomaterials can direct
shaped cartilage structures, such as a human ear and a sheep menis- those cells to differentiate into zone-specific chondrocytes, and to
cus, with high fidelity and stability16 (Fig. 1a). Inkjet 3D-printing create a native-like articular cartilage with mechanical and bio-
has also been used to repair cartilage in preclinical models19, achiev- chemical properties varying with depth23,24. Similarly, hyaline-like
ing a tissue construct with a compressive modulus of the same order cartilaginous tissue expressing collagen type II has been made via
of magnitude as hyaline cartilage20. Another approach involves the the bioprinting of induced pluripotent stem cells (iPSCs) within a
fabrication of tissue constructs using micromass chondrocyte pel- nanocellulose alginate bioink25.
lets to form cartilage strands, with tubular permeable alginate cap- Further advances in 3D bioprinting will enable the creation of
sules working as a reservoir for cell aggregation and tissue-strand patterns of growth factors, mechanical gradients and stem cells
maturation. This approach resulted in strands with a diameter of in each cartilage zonal region, improving the function of biofab-
~500 μm and with significantly increased cell density, as well as ricated cartilage tissue. In fact, it has been shown that 3D-printed
improved post-transplantation maturation and function of the cartilage can have the histological and mechanical characteristics
engineered tissue21. of human auricles after implantation in vivo26. And a tissue-engi-
Combining multiple cell types may also increase the efficiency neered trachea from a 3D-printed biodegradable reticular polycap-
of the bioprinted cartilage. The co-printing of multipotent articular rolactone (PCL) scaffold suspended in culture with chondrocytes
cartilage-resident chondroprogenitor cells, bone marrow mesen- was used in the replacement surgery of the native trachea of rab-
chymal stromal cells (MSCs) and chondrocytes by using a gelatin bits27. Owing to potential limitations in the yield, proliferation and
methacryloyl-based hydrogel, led to generation of constructs with metabolic activity of chondrocytes derived from older patients28,
a layered distribution of collagens and glycosaminoglycans, defin- bone-marrow-derived MSCs can be used as a cell source to
ing a cartilage with superficial and deep regions, each with distinct derive functional chondrocytes. The induction of chondrogenesis
cellular and ECM composition22. The integration of MSCs into a in MSCs can also be readily achieved by the addition of specific

Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng 371


Perspective NaTure BIomeDIcaL EngIneerIng

growth factors29,30, and can be modulated by controlling the biome- functionality. These 3D-printed constructs can mimic the geometry
chanical environment31. and bulk mechanical properties of trabecular-like endochondral
bone with a supporting marrow structure, and undergo endochon-
Bone. Although bone is the most commonly transplanted solid tis- dral ossification following implantation. Scalable mandible and
sue (only blood transfusions are more common32), traditional tech- calvarial structures have been 3D-printed using a similar approach,
niques for bone-tissue biofabrication are associated with significant with sizes and shapes similar to what would be needed for facial
limitations and complications, such as the paucity of available autol- reconstruction (Fig. 1b). Also, 3D-printed mandibular bone con-
ogous materials and donor-site morbidity (especially for segmental structs, comprised of a mixture of PCL and tricalcium phosphate
defects33). Significant efforts have thus focused on the study of the (TCP) with26 or without42 cells and implanted in animal defect mod-
unique structure and mechanical properties of bone. els, can form mature vascularized bone tissue (as shown with con-
Multiple types of biomaterial scaffold afford control over the structs retrieved 3–5 months after implantation).
mechanical properties and degradability of the constructs follow- The combination of multiple biofabrication tools and materi-
ing transplantation34. Two commercially available void-filling bio- als seems to hold promise for the recapitulation of the mechanical
material products—Infuse (Medtronic), an absorbable collagen and biological properties that are essential for bone; but engineer-
sponge that releases bone morphogenetic protein-2 (rhBMP-2); ing bone tissue with histomorphometry and function approaching
and Actifuse (Baxter International), a silicated calcium phosphate– those of native bone tissue remains elusive. The approaches with
hydroxyapatite graft—can accelerate bone-tissue healing in non- best results remain those that successfully direct bone healing to
weight-bearing defects, but they are not printable. fill the injury. Yet the use of bioprinting to guide the innate heal-
Attempts are underway to combine the natural regenerative ing process to fill large bone injuries with an anatomically accurate
potential of bone with the benefits of bioprinting, such as the con- shape (rather than approaches seeking to replicate the developmen-
trol over the shape and precise placement of cells and biomateri- tal stages of bone formation) has clinical potential.
als in bioprinted scaffolds or bioengineered grafts. Several scaffolds
have been developed using synthetic biomimetic nanoceramics, Skin. The skin’s relatively thin (~2.5 mm), layered and structured
particularly calcium phosphate ceramics (such as hydroxyapatite, nature, combined with easy access to cell sources, has facilitated the
biphasic calcium phosphate or tri-calcium phosphate; TCP), and early adoption of 3D bioprinting technology for the manufacturing
fabricated into bone-like architectures through 3D bioprinting35. of skin tissue. There are two main strategies for the 3D bioprint-
For example, a calcium-phosphate–collagen composite bone scaf- ing of skin constructs: in vitro bioprinting and transplantation of
fold has been fabricated using a modified inkjet-based 3D printer36, the printed tissue into the defect site; and direct in situ bioprint-
and the implants were confirmed to be osteoconductive and biode- ing, where the cells and materials are printed directly into the defect
gradable in a segmental murine femoral defect. Also, a case report site. The feasibility of using bioprinting to fabricate skin constructs
described the computer-aided design and fabrication of a medical- in vitro was first shown with multilayered engineered tissue com-
grade PCL–TCP biodegradable scaffold implanted to reconstruct posites of human skin fibroblasts and keratinocytes deposited layer-
a complex cranial defect37. The scaffold appeared to be well-inte- by-layer within a collagen hydrogel, resulting in an inner layer of
grated at the site after six months, with the onset of bone consolida- fibroblasts and an outer layer of keratinocytes42. Since then, owing
tion detected via CT. to laser-based bioprinters43,44 and inkjet-based printers45,46, in vitro
To reproduce the appropriate mechanical properties of cortical skin bioprinting has increased in complexity and accuracy. Areas
bone, many 3D-bioprinting approaches rely mainly on hard scaf- up to 100 cm2 of bilayered skin using primary human fibroblasts
folds; hence, they fail to fully recapitulate the properties of the cel- and keratinocytes, obtained from skin biopsies, can now be printed
lular component. This is a significant limitation. Although many in less than 35 minutes47. Human skin can also be fabricated in
fabrication techniques can produce constructs with suitable inter- amounts and timeframes that should be appropriate for clinical and
connected porosity and mechanical properties, they require the commercial applications.
application of temperatures, solvents or other conditions that can In situ bioprinting of the skin construct directly on the wound
adversely affect living cells. The post-production seeding of cells site relies on the patients’ body serving as the ‘bioreactor’ for the
then results in problems associated with non-uniform cell distribu- functional maturation of the bioprinted tissue. In wound healing,
tion and poor cell attachment. the main advantage of this approach is the rapid coverage of large
By taking advantage soft materials amenable to cell growth and wounds with permanent skin tissue, and their accelerated healing.
tissue formation, bioprinting techniques may meet the mechanical In comparison to the transplantation of in vitro fabricated con-
needs of hard bone tissue while promoting tissue regeneration. One structs, in situ bioprinting avoids the risk of damaging the thin and
approach is the development of hybrid or composite structures with fragile construct during transport and handling, and avoids poten-
a strong, mechanically robust structure, filled with precisely placed tial issues related to the correct placement and orientation of a con-
cells deposited within osteoconductive hydrogel bioinks. These struct with complex 3D topology. In one of the first descriptions
composite scaffolds could enable the fabrication of large and strong of in situ bioprinting, human keratinocytes and fibroblasts were
scaffolds with specific patterns of cells and chemical factors that printed directly into a full-thickness mouse skin-wound model48.
elicit specific tissue development. In one approach, nanohydroxy- Wounds were first scanned to obtain precise information on wound
apatite (nHA) and human osteoprogenitor cells were patterned and topography, which then guided the print heads to deposit speci-
assembled via laser-assisted bioprinting38, with the osteoprogenitors fied materials and cell types in appropriate locations (Fig. 1c). The
maintaining their osteoblastic phenotype and functionality after first layer of a fibrinogen–collagen hydrogel precursor containing
printing (the resulting tissue was, however, not evaluated in vivo). fibroblasts was bioprinted, followed by the simultaneous deposi-
Other methods can produce customized 3D porous structures by tion of thrombin from nozzles to form a fibrin–collagen hydrogel;
building osteochondral tissue, fabricated via the sequential dispens- an additional layer of keratinocytes was then bioprinted on top of
ing of PCL and two alginate solutions containing osteoblasts and the fibroblast layer via a similar deposition approach. The same
chondrocytes39,40, or by using biofabrication to make a mechanically approach was also applied in a porcine model with large full-thick-
reinforced template that supports the development of vascularized ness wounds (Fig. 1d), where in situ bioprinting led to the complete
bone41. In the latter, soft bioinks were supported by a network of rein- re-epithelialization of the large wound after 8 weeks49.
forcing PCL microfibers to enable the fabrication of mechanically Our group has previously used an in-situ skin bioprinter to
reinforced constructs with decoupled biological and mechanical deposit amniotic-fluid-derived stem cells on full-thickness skin

372 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng


NaTure BIomeDIcaL EngIneerIng Perspective
Table 1 | Specific needs in the 3D-printing of tissues, and potential solutions
Specific needs Potential solutions
Materials Accurate reproduction of the functional and Hybrid constructs, consisting of patterned synthetic and natural
biomechanical properties of tissue materials23,24,40
Compatible properties for bioprinter deposition ECM-based hydrogels54,91–93
Robust and controllable post-printing mechanical Chemical or non-chemical modification of ECM and hydrogel
properties materials63,64
Physiological, biochemical and mechanical interactions Synthetic peptides that mimic ECM functional motifs65,66
with the cellular component
Appropriate scaffold-degradation times ‘Smart’ materials whose properties change over time or after the
application of external stimuli122,123
Cells Minimally invasive or non-invasive cell sourcing Small-molecule control over cell function, such as conditional cell
reprogramming70,71
Autologous or non-immunogenic cell sources Mesenchymal cells72,73
Induced pluripotent stem cells80,81
Perinatal/adipose-derived cells74,75
Ex vivo expansion of the cells without loss of phenotype ECM-based 3D hydrogel culture systems91–93
or function
Vascularization Oxygen and nutrient availability across thick tissues Microchannel embedding26,102,103
Fabrication of multiscale hierarchical networks with Patterning of angiogenic growth factors or cells104
complex structures
Direct vascular fabrication or patterning of cells and/or Sacrificial templates for the fabrication of perfusable microchannel
factors that mature either in vivo or in bioreactors networks42
Direct bioprinting of vasculature by using cell patterning or tissue
spheroids109

wounds in mice, using either a fibrin–collagen bioink50 or a hyal- Bioinks. Making tissue constructs with the desired functional and
uronic acid-based gel with tuneable properties tailored for extended biomechanical properties from available biomaterials remains a
cytokine release51. Although the stem cells did not permanently challenge. One approach involves hybrid constructs of patterned
integrate into the regenerated skin, the secretion of trophic fac- synthetic and natural materials (such as ECM-derived hydrogels):
tors accelerated wound-closure rates and promoted angiogenesis. the synthetic materials provide physical integrity and control
Similar strategies would avoid the sourcing of cells from patients over the scaffold’s mechanical, structural and geometrical prop-
with significant loss of healthy skin tissue. erties (such as the elastic modulus, tensile strength, porosity and
No bioprinting approach can yet fully replicate the morphologi- alignment) at the macroscopic level, and the natural materials
cal, biochemical and physiological properties of native skin. The provide an appropriate structural and biochemical environment for
incorporation of additional cell types and the patterning of more cell encapsulation and placement. Hence, the use of hydrogels based
representative ECM components is necessary. For example, achiev- on ECM components in native tissue is desirable for cell encapsu-
ing stratified tri-layered structures containing epidermis, dermis lation because they facilitate the provision of tissue-specific nutri-
and hypodermis would be beneficial, as well as the incorporation of ents and cellular products after printing. For example, a hydrogel
components of the vasculature, nerves, sweat and sebaceous glands, composed of skin-derived decellularized ECM was included in
hair follicles and pigmentation. Moreover, future skin constructs a 3D cell-printing process that enabled the precise generation of
should facilitate the proper development and regulation of hair fol- cell-laden constructs by inducing the simultaneous gelation of the
licles, pigmentation and epidermis formation and maturation. This printed bioinks54.
will need the understanding of the roles of multipotent progeni- However, a major limitation is the need for these materials to be
tor cells present in the upper permanent region of the hair follicle, deposited by bioprinting techniques that often rely on melt-extru-
which contribute to the formation of hair follicles and sebaceous sion, on materials with sheer-thinning properties or on selective
glands52, and of the interaction of these cells with melanocyte stem crosslinking approaches55. A further limitation is that these materi-
cells during skin homeostasis and repair53. als must also establish physiological, biochemical and mechanical
interactions with the cellular component. The approach of combin-
Current challenges and potential solutions ing a synthetic material for mechanical strength, together with a
Most bioprinted tissues and organs are small in scale, contain only softer hydrogel for cell encapsulation and deposition, may lead to
one or two cell types, consist of relatively simple structures, and a construct in which the cells are not exposed to the appropriate
provide limited functionality. Bioprinted tissues generally lack mechanical stimulation necessary for the maturation of tissues such
vascular networks, and thus rely on diffusion for nutrient supply. as articular cartilage and muscle. Furthermore, many tissues have
The bioprinting of more complex and larger tissues with robust, biological and biomechanical heterogeneity, and interface with dif-
tailorable mechanical properties and cell-compatible materials ferent tissue types. In this context, 3D bioprinting enables the for-
requires methods for the derivation and expansion of multiple mation of concentration gradients of materials, cells and biological
types of functional, progenitor and supporting cell types, as well as factors, and hence the reproduction of these heterogeneous tissue
strategies for the integration of a vascular network for oxygen and properties. Examples of solutions to these problems are micro-
nutrient supply (Table 1). fluidic switching nozzles that swap between two different inks on

Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng 373


Perspective NaTure BIomeDIcaL EngIneerIng
a b

Rotating
impelier
Inlet 2

2 cm 2 cm
Inlet 1
Mixing
volume

Nozzle
outlet

4 cm

Printed
structure

Fig. 2 | 3D bioprinting of biomaterials with graded properties. a, A mixing nozzle that can be used to print materials at the microscale, with tuneable
gradients of differing material properties. b, Images of the cross-section of a 3D rectangular lattice with a continuously varying compositional gradient,
showing continuous change in fluorescent pigment concentration under bright light (top left) and UV radiation (top right). 2D lattice structure shows
discretely varying fluorescence gradient at eight different mixing ratios under bright light (middle) and UV radiation (bottom). Dashed white lines mark
the regions of different mixing ratios. Figure reproduced from ref. 57, NAS.

demand56, and mixing nozzles that can be used to print materials at including specific enzymes, antibodies or cells68,69. The ability to
the microscale with tuneable gradients of differing material proper- tightly control the density, patterning, structure and orientation of
ties57 (Fig. 2). Some specific applications may require more complex synthetic peptides within a bioprinted 3D matrix provides opportu-
printing patterns, such as in the reproduction of the zonal mechani- nities for inducing cell responses that would not normally be evoked
cal structure of articular cartilage23,24, the fabrication of vascularized by native matrix molecules. Still, this approach has, to date, been
bone constructs with hierarchical organization58 and the fabrication limited to the coating and functionalization of artificial surfaces and
of muscle–tendon interfaces59. has yet to be incorporated into a 3D bioprinted construct.
Although ECM-derived hydrogels are biocompatible, their weak Another important factor is the requirement for the degrada-
mechanical properties limit their contribution to the physical prop- tion time of the scaffold to be commensurate with the timing of
erties of the tissue. Still, the hydrogels can be chemically modified to host remodelling of the construct. The implanted construct should
enable the material to crosslink and alter, for instance, its mechani- facilitate host ECM production and remodelling, to allow for the
cal strength or degradation time60. For example, synthetic hydrogels replacement of any synthetic or temporary support structures in a
based on poly(ethylene glycol) (PEG) have been modified to cova- time scale that neither delays nor inhibits the formation of native
lently tether ECM-derived biomolecules to the hydrogel network tissue structures, nor poses any substantial risk of premature scaf-
via monovalent, divalent or multivalent reactive groups, such as fold failure. There is thus a need for ECM modifications or ECM-
acrylate, amine, thiol, azide, maleimide and biotin–streptavidin. In mimicking materials that provide stronger mechanical strength and
a similar approach, a photocrosslinkable hyaluronan (HA)–gelatin that maintain a cell-friendly environment, as well as for synthetic
hydrogel was developed for use in the bioprinting of decellularized materials that support cell delivery and post-printing bioactivity
ECM materials61,62. This protocol implemented a methacrylate- and function. Controlling the spatial hierarchies of materials, cells
based photopolymerization system for a two-step photocrosslink- and biological factors, and the dynamics of tissue fabrication offers
ing process, allowing the extrusion of the material through a syringe many possibilities that remain to be exploited.
or printing head and the subsequent increase in elastic modulus63.
The incorporation of additives such as graphene oxide nanosheets Cell sourcing. The production of an adequate number of regen-
can also significantly increase the tensile strength of soft hydro- eration-competent cells that do not elicit an immune response
gels64. Although modifications such as these have expanded the util- following transplantation is an ongoing challenge. 3D bioprinted
ity of hydrogels for 3D printing, the ability to significantly improve tissues have typically contained only a small number of accessible
the mechanical strength of ECM-derived hydrogel scaffolds cell types. One of the simplest methods of obtaining cells involves
remains limited. the harvesting of autologous primary cells (often mature, terminally
Another approach involves the use of synthetic peptides that differentiated cell types) from the patient, followed by their lim-
mimic functional motifs of the ECM. The most common peptide ited expansion in vitro prior to tissue fabrication. For example, for
is Arg-Gly-Asp (RGD), which is found on multiple ECM proteins cartilage tissue engineering, primary chondrocytes are the choice
and is involved in cell adhesion. There are many other self-assem- because they comprise the native tissue population and are isolated
bling peptides with a range of properties and functions65,66 that can easily as homogeneous cell preparations that can be expanded ex
influence cell adhesion, shape, migration and differentiation, and vivo. However, articular chondrocytes cannot be easily harvested in
that are known to be involved in the binding and release of growth significant numbers and, in addition to donor-site morbidity, de-
factors and in the stimulation of tissue repair and regeneration67. differentiate following in vitro expansion.
Also, stimuli-responsive systems can be made responsive to factors Some of the limitations of expanding primary cell types can
such as temperature, pH, and biochemical or biological stimuli, be overcome. One approach is to use conditional reprogramming

374 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng


NaTure BIomeDIcaL EngIneerIng Perspective
to temporarily immortalize primary cells and thus increase their bioprinting and 3D multicellular building blocks comprising cells
expansion potential. Conditional reprograming often includes the and their ECM, and in the form of cell aggregates, strands, fibres
use of small molecules or other factors to promote cell proliferation or more complex organoid or microtissue structures, can be com-
or other cellular functions. For example, culturing primary human bined, as was shown with the mixing of dispensed cell spheroids and
epithelial cells with fibroblast feeder cells combined with the Rho- cell cylinders into a hydrogel bed via the use of extrusion bioprint-
associated kinase (ROCK) inhibitor Y-27632 induces continued ers95–97 (this approach exploited the self-organizing capacity of mul-
cell proliferation while retaining the normal karyotype and remain- ticellular spheroids). Moreover, the development of small molecules
ing non-tumorigenic70,71. Other solutions to limited cell availability of low molecular weight may regulate biological processes such as
include the use of progenitor cells or stem cells. The most com- survival, proliferation and function, and may allow for significantly
monly used progenitor cells are MSCs, which can be isolated from greater expansion of cell numbers without loss of phenotype and
many tissue types, expanded and differentiated either in vitro or function. A combination of approaches is likely to be applied to spe-
in vivo into osteogenic, chondrogenic, tenogenic, myogenic, adipo- cific cell types in the future, with culture conditions designed for
genic and marrow-stromal lineages72–74. Perinatal or amniotic-fluid- each end-stage application.
derived stem cells75, which can also be expanded and differentiated
into multiple lineages, are alternative multipotent stem cell sources. Vascularization. Tissue-engineered constructs in vitro, for instance
Multipotent stem-cell populations will continue to have impor- in perfusion bioreactors98, can be supplied with oxygen and nutri-
tant applications in the biofabrication of mesodermal-derived tis- ents. But after implantation in vivo the supply of oxygen and nutri-
sues such as cartilage, bone and muscle. However, these cell sources ents is often limited by diffusion kinetics. Because in engineered
are associated with complications, in particular their propensity to tissues oxygen diffusion is often slower than its consumption,
undergo senescence following multiple expansion rounds in vitro76. oxygen is the limiting factor in cell survival99; hence, as the thick-
Donor age can also negatively affect cell expansion and differentia- ness of the tissue exceeds the limits for nutrient diffusion, the need
tion77–79. Since the discovery of iPSC in 2007 (refs. 80,81), their easy for vascularization becomes a critical factor. This aspect becomes
accessibility, expandability and ability to give rise to almost any cell increasingly important for tissues with a high volumetric oxygen-
types has motivated the investigation of autologous iPSC-derived cell consumption rate, such as cardiac, pancreas and liver tissues. For
types for regenerative-medicine applications. The first clinical study instance, few cells tolerate distances greater than 200 μm from
involving the evaluation human iPSC-derived cells, specifically the a blood vessel100, with sensitive cells such as islets undergoing
use of human iPSC-derived retinal pigment epithelial cells for the necrosis when the diffusion distance exceeds ∼100 μm. Conversely,
treatment of macular degeneration82,83, initiated in 2014, highlighted cartilage cells are generally more resistant, maintaining viability in
several challenges and risks that will need to be addressed before grafts thicker than 1 mm (ref. 101). Therefore, the fabrication of a
the clinical application of iPSCs can take off. Most importantly, the functional 3D-printed tissue will require the incorporation of mul-
potential for tumorigenicity remains a significant risk84; in fact, an tiscale vascular, lymphatic and/or neural networks. Currently, the
iPSC trial was put on hold following the identification of two genetic majority of tissues fabricated for transplantation using 3D printing
variants in a patient’s iPSCs85,86. Also, although they have the ability lack these components.
to expand indefinitely, the in vitro culture conditions and passage A major roadblock in the fabrication of engineered tissues
number have a significant impact on iPSC phenotype, including the concerns the replication of the complex hierarchical structure of
incidence of mutations87,88. To avoid this problem and to reduce the a native integrated vascular network spanning arteries and veins
cost of preparing autologous iPSCs, there are attempts to establish down to the smallest capillaries. Two main approaches have been
iPSC banks, so that cell donors and recipients can be matched on designed to overcome current limitations: the embedding of micro-
the basis of the major histocompatibility complex class-I and class-II channels to improve the diffusion of nutrients and oxygen in the
human leukocyte antigens (HLAs) involved in the immune recogni- absence of a vasculature26,39,40,102,103 (Fig. 3a), and the patterning
tion of foreign antigens. This may be easier to achieve in countries of angiogenic growth factors or cells within a tissue construct to
with small genetic variability (such as Japan, where it is estimated facilitate vascular development, either in vitro or following trans-
that 10, 75 and 140 cell lines would match about 50%, 80% and 90%, plantation104. However, although both of these approaches allow for
respectively, of the Japanese population89,90). the fabrication of larger constructs, they ultimately rely on endog-
Protocols for the isolation and expansion of new primary cell enous vascularization.
sources are highly sought after. Advances over the past few decades Although current technologies excel at fabricating a specific type
have seen the isolation, characterization and expansion of primary of materials within a relatively small range of scales, the complex
cell types that had been thought to be impossible to culture out- hierarchical 3D architecture of a multiscale vascular network can-
side of the body. However, many cell types cannot yet be expanded not yet be recapitulated. One promising route involves the com-
with retention of their phenotype and function. A greater under- binatorial use of bioprinters with different working principles for
standing of growth factors, culture media and enzymatic subculture the deposition of multiple materials and cell types, and at differ-
techniques significantly contributed to the current ability to expand ent scales. In this regard, direct ink writing has led to remarkable
primary cells. Also, the more recent applications of 3D cell-culture achievements in the high-resolution patterning of matrix materi-
techniques (especially those with mixed cell populations in which als105. One example is omnidirectional printing (ODP)105, whereby
stromal and vascular cell types are combined with other functional fugitive bioinks are printed within a photocurable gel reservoir that
cell types in 3D culture conditions) provide contact-specific and physically supports the patterned features, thereby allowing truly
biochemical supports for the maintenance of cell viability and func- omnidirectional free-form fabrication. The fugitive bioink can
tion in vitro. Additionally, physiologically accurate biomaterial- then be removed to yield a desired microchannel network within
growth environments are replacing the common tissue-culture the matrix (Fig. 3b). Because the nozzle can simultaneously move
plastic. For instance, recent advances in ECM and 3D hydrogel cul- in three dimensions, the major advantage of ODP, compared with
ture systems91–93 have enabled the culture and expansion of clonal multidimensional bioprinting, is that ODP is not limited to stan-
primary adult liver stem cells without changes to cell phenotype94. dard layer-by-layer fabrication. This allows ODP to broaden the
This includes 3D-culture environments that match the physical and network-design space and to deposit materials in highly anisotropic
biochemical composition of the cells’ native environment, includ- structures. Also, in ODP the microchannel diameter can be con-
ing tissue-specific motifs and tissue-derived ECM that provide trolled via dynamic-pressure variation; hence, a single nozzle can
near-physiological interactions with the embedded cell types. 3D pattern microchannels of varying size.

Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng 375


Perspective NaTure BIomeDIcaL EngIneerIng
a b

Fluid filler

PCL Matrix

Ink
3D-printed Nozzle direction
construct
el)
nn
ha
oc
icr
(m
re
Po

Cell A

Cell B
c
t = 0 days t = 2 days

Cell ink 1 Lumen

Vasculature

Cell ink 2 HNDFs


ECM
HUVECs

Fig. 3 | Overcoming diffusion limitations and the need for vascularization. a, A patterning approach for generating a 3D architecture that includes
multiple cell-laden hydrogels, a supporting PCL polymer and microchannel pores for improving the diffusion of oxygen and nutrients. b, Fabrication of
vascular structures via omnidirectional printing. Deposition of a fugitive ink into a physical gel reservoir (top left) allows hierarchical and branching
networks to be patterned. Voids induced by nozzle translation are filled with liquid that migrates from the fluid-capping layer (top right). Subsequent
photopolymerization of the reservoir yields a chemically crosslinked hydrogel matrix (bottom left). The ink is liquefied and removed under modest vacuum
to expose the microvascular channels (bottom right). c, A 3D-bioprinting approach in which vasculature, cells and ECM are co-printed to yield engineered,
vascularized and heterogeneous cell-laden tissue constructs (left, schematic; right, micrographs). Scale bars, 300 μm. Panels reproduced from ref. 26,
Springer Nature Ltd (a); ref. 105, Wiley-VCH (b); and ref. 107, Wiley-VCH (c).

With current technology, it is technically challenging to print and without the need of sacrificial materials. Notably, anastomosis
functional capillaries at the micrometre scale. One alternative is to can occur between the bioprinted endothelial network and the host
let capillaries develop by first fabricating a vascular network that circulation.
then matures in vivo or in bioreactors106. For example, by drawing Despite advances in the direct fabrication of larger conduits, in
on techniques based on sacrificial template materials, one can fab- the incorporation of perfusable microchannel networks and in the
ricate a perfusable microchannel network within the tissue to, for stimulation of vasculogenesis, current fabrication approaches can-
instance, develop a construct consisting of two endothelialized flu- not incorporate a complete multiscale vascular network suitable for
idic channels with a fibrin–endothelial-cell mixture located between the provision of oxygen and nutrients into clinically relevant sized
them. This design results in the formation of adjacent capillary 3D-printed tissues. The patterning of tissues at scales ranging from
networks and, consequently, in the generation of a multiscale vas- micrometres to centimetres will require significant technologi-
cular network that connects millimetre-scale vessels with adjacent cal innovations in material deposition and cell deposition. Recent
microvasculature. Another alternative involves the bioprinting of a advances in microextrusion technology have enabled the pattern-
vasculature network by using patterned cells or tissue spheroids. For ing of multicomponent constructs containing both synthetic and
example, multiple biomaterials encapsulating MSCs and fibroblasts natural materials with resolutions down to 2 μm for biomaterials
within a customized ECM can be co-printed alongside embedded alone and down to 50 μm for encapsulated cells26. Further progress
vasculature subsequently lined with endothelial cells107,108 (Fig. 3c). will require the ability to deposit an even wider range of material
This approach enabled the creation of thick tissues (larger than 1 types concurrently with increased printing resolution and printing
cm) replete with an engineered ECM, embedded vasculature and speed. One promising approach involves parallelization schemes
multiple cell types. Another example of such a bioprinting strategy that use multiple nozzles to deposit materials at the same time: with
involved multiple vascular cell types aggregated as multicellular multinozzle arrays designed with hierarchically branching chan-
vascular tissue spheroids and printed layer-by-layer concomitantly nels, each print-head distributes bioink from a single microchannel
with agarose rods as a template109. The closely placed vascular tis- into repeatedly bifurcated branches, to deposit bioink simultane-
sue spheroids underwent tissue fusion and self-assembly into small ously from multiple nozzles during printing111. This approach can
segments resembling a branched vascular tree. Another strategy print, at high-throughput, planar and multilayered architectures
involves microscale continuous optical bioprinting, which offers composed of single and multicomponent materials. However, it has
speeds, resolutions and flexibility that are superior to those of con- yet to be demonstrated with materials containing cells. Also, bio-
ventional bioprinters110. In this approach, multiple vascular cell types logical advances could lead to improvements in the bioprinting of
are encapsulated directly into hydrogels with precise distribution vasculature: for example, relationships between endothelial cells

376 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng


NaTure BIomeDIcaL EngIneerIng Perspective
and parenchymal constituents, and between interstitial and per- This approach could significantly improve fabrication efficiency and
fusate flows and hydrostatic pressure, are relevant for the optimal reduce fabrication costs, although it may need the determination of
self-assembly of capillary-sized vessels112–115. Although biological appropriate sites for implantation, the promotion of anastomosis,
responses to multivariate stimuli are difficult to model and predict, and the ensuring of long-term integration and function.
such knowledge should enable the engineering of vascular struc-
tures that favour rapid angiogenesis. Automation and personalization. Implementing automated
Another promising general approach is ‘modular tissue design’, designs in personalized manufacturing processes can be used to
where ‘tissue modules’ are scaled to incorporate relevant vascular- understand which control mechanisms can be regulated to ensure
ization strategies that consider the limitations of each approach. For predictable outcomes. Currently, many personalized designs are
example, small tissue modules may incorporate a series of perfus- produced with more artistry than engineering, simply because of the
able microchannels combined with approaches for the stimulation complexity of anatomical designs. Computational processes with
of angiogenic sprouting and growth. These small tissue modules clear design parameters are made possible by modern 3D imaging
could then be assembled with a larger network of directly fabricated and modelling software, which can be adapted and automated to
vessels, and patterned with a branching network of endothelialized generate personalized structures that match the patient’s anatomy
microvasculature connected to millimetre-scale vessels capable of and injury needs117. Such an approach would map predetermined
anastomosis. This will require the development of approaches for hatching architecture, materials and cell types onto appropriate
the high-throughput, high-resolution bioprinting of multiscale vas- regions of the bioprinted construct spatially defined by a 3D model
cular networks within instructive bioinks that promote angiogenic generated via medical imaging, typically via CT or MRI. A detailed
sprouting and neovascularization. understanding of biomaterial self-assembly, of tissue healing and
of the integration of the bioprinted construct, will be needed to
Outlook develop the required design criteria for future bioprinting concepts
To envisage how future advances will influence the next generation to reliably meet patient needs. This understanding is also necessary
of 3D-printed tissues, we can look at current technologies for the for the clinical translation and regulation of personalized bioprinted
3D-printing of cartilage, bone and skin constructs. They can serve constructs. In many ways, bioprinting is a type of 4D printing, as the
as benchmarks for the evaluation of the status of bioprinting tech- cells reorganize and produce tissue, and degradable scaffold materi-
nology. By understanding the factors that have contributed to their als break down over time after implantation. Building the repertoire
success, and by anticipating advances in the bioprinting of materi- of knowledge needed to properly predict the outcomes of such com-
als and cells, we can postulate which of the current limitations may plex 4D prints118 will require research on the relevant in vitro and
be overcome in the short term, and what the impact will be in the in vivo interactions of cells over time.
development of the next generation of 3D-printed tissues. Common
properties of successfully 3D-printed tissues include: (i) the repli- Cost-effectiveness. There are substantial challenges associated
cation of the 3D architecture, size and form of native tissues that with the scaling and commercialization of bioprinted tissues. For
(ii) contain appropriate mechanical properties at the microscale and example, the tissue design, cell sourcing and fabrication logistics
macroscale, (iii) including the cell types necessary for tissue func- developed for each organ in many current approaches are highly
tion and for the maintenance of tissue homeostasis, and (iv) the personalized to the individual needs of each patient. Therefore, the
reproduction of tissue or organ function at a level sufficient for the cost-effectiveness of personalized 3D-printed tissue replacements
replacement, restoration or supplementation of the in vivo tissue. will be a significant challenge. In the short term, companies consid-
In our opinion, the continued development of materials suitable ering investing in this area may consider the development of generic
for the deposition, with a 3D printer, of cells that can also support or universal scaffolds, or of components that would reduce the costs
cell function, tissue structure and biomechanical properties would associated with the need for individualized materials and designs.
have significant impact. Also, the further development of bioprinters The use of acellular scaffolds that are then seeded with the patient’s
could accelerate the fabrication timeline, and provide the resolution own cells may reduce costs while minimizing the risks of alloge-
needed for the fabrication of functional tissues at a clinically relevant neic rejection. When considering a personalized tissue-engineering
scale. Approaches that accelerate manufacturing processes, such as approach such as 3D bioprinting, the cost-effectiveness of a treat-
advances in bioprinter or material technologies, could, in future, ment may require high upfront costs, but these must be balanced
overcome the slow speed of current layer-by-layer printing. Although against the alternative of costly life-long treatments (such as dialysis
not yet tested for the fabrication of medical devices, the continuous- or the management of diabetic complications). For many conditions,
liquid-interface-production approach can in principle print complex a cost-effectiveness analysis may indicate that a single intervention is
solid parts at rates of hundreds of millimetres per hour116. economically preferable than life-long non-curative treatments.

The limits of biomimicry. Whether the current approach of mim- Regulation. The implantation of 3D-bioprinted tissues and organs
icking biological structures will continue to lead to optimal function will face regulatory challenges. Despite the variety of manufacturing
and design efficiencies in larger and more complex tissues is unclear. methods, the FDA currently assesses 3D-printed medical devices
Manufacturing close reproductions of the cellular and extracellular and conventionally made products under the same guidelines. The
components of a tissue or organ has often resulted in approximations 21st Century Cures Act119 describes regenerative-medicine therapies
of tissue function. However, biomimicry may reach a point where that may be eligible for the designation of ‘regenerative medicine
increased complexity no longer improves functional outcomes. advanced therapy’ (RMAT). These include cell therapies, therapeu-
Also, technological or economic limitations may require a departure tic tissue-engineering products, human cell and tissue products,
from biomimicry in the process of enhancing tissue function, tis- combination products that use any such therapies or products, and
sue scale or printing throughput. Therefore, new design approaches gene therapies that lead to durable modifications of cells or tissues.
that prioritize construct function and biofabrication efficiency may To help frame the future of 3D bioprinting for healthcare, the FDA
be needed, possibly with 3D-bioprinted constructs that are efficient has released detailed guidance for 3D-printer manufacturers120.
in replacing or supplementing tissue function but that do not pos- There are a number of regulatory challenges that apply to func-
sess the size, shape or form of the functional tissue type (for exam- tional 3D-bioprinted constructs containing living cells and bio-
ple, one could imagine 3D-printed constructs shaped as a sheet or active materials. First, they are intrinsically different from other
patch and designed to assist liver function or pancreas function). clinical products, owing to the complexity of mechanisms and to

Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng 377


Perspective NaTure BIomeDIcaL EngIneerIng

as-yet-unknown long-term effects in human hosts. Second, the In summary, a clinically relevant bioprinted construct will most
mechanisms of action in tissue constructs depend on their multiple probably need to reach thresholds for functional and supporting
active components, which makes it difficult to meet a regulatory cell types (including stem cells), have biomechanical properties that
definition of potency. Third, the manufacturing and product char- closely mimic those of the native organ at both the microscale and
acterization are likely to be significantly more complex; seemingly macroscale, and feature intact vascular networks spanning arteries,
minor manufacturing changes may have large and unpredicted veins and capillaries. These criteria have been partly met for bio-
effects on the characteristics of the product. Hence, a centralized printed cartilage, bone and skin constructs. Although the scale and
and defined regulatory pathway will be necessary to avoid overbur- functionality of 3D-printed tissue constructs continues to improve,
dened and redundant regulatory pathways. Regulatory frameworks significant challenges remain for the bioprinting of more complex
will need to describe clearly how regulators will apply existing tissues with greater physiological demands. Progress towards the
laws and regulations that govern device manufacturing to non- multiscale and multicomponent fabrication of clinically relevant
traditional manufacturers such as medical facilities and aca- bioprinted tissues and organs will require the incorporation of
demic institutions that create 3D-printed personalized devices. advances in the isolation and expansion of populations of primary
Encouragingly, the current guidelines and the regulatory pro- cells and stem cells, the development of ‘smart’ biomaterials, and the
cesses proposed by the FDA and by other national regulatory bod- integration of complementary bioprinting technologies.
ies suggest that these considerations are known. Also, some of the
regulatory challenges could be addressed via the technological Received: 13 July 2018; Accepted: 30 September 2019;
convergence of production methods, and via the increased stan- Published online: 6 November 2019
dardization and identification of best practices for the design and
production of bioprinted constructs. References
1. Tack, P., Victor, J., Gemmel, P. & Annemans, L. 3D-printing techniques
Standardization. There are currently no standards for 3D bioprint- in a medical setting: a systematic literature review. Biomed. Eng. Online 15,
ing technology, for bioprinting materials (including bioinks and cell 115 (2016).
sources) or for the overall bioprinting process. Despite the existence 2. Di Prima, M., Coburn, J., Hwang, D., Kelly, J., Khairuzzaman, A. &
Ricles, L. Additively manufactured medical products–the FDA perspective.
of standards for additive-manufacturing terminology (ISO/DIS 3D Print. Med. 2, 1 (2016).
17296-1) and the recent first guidance documents for 3D-printer 3. Ventola, C. L. Medical applications for 3D printing: current and projected
manufacturers120, there is an urgent need for increased levels of uses. Pharma. Ther. 39, 704–711 (2014).
standardization and for production guidelines for bioprinting. 4. Ma, L. et al. 3D printed personalized titanium plates improve clinical
A degree of standardization of the bioprinting materials (analogous outcome in microwave ablation of bone tumors around the knee.
Sci. Rep. 7, 7626 (2017).
to standardized cell-expansion basal media or to modular bioink 5. Li, B. et al. Application of a novel three-dimensional printing genioplasty
chemistry) would have a beneficial impact on product-development template system and its clinical validation: a control study. Sci. Rep. 7,
time. The standardization of materials and manufacturing processes 5431 (2017).
would also accelerate the clinical translation of bioprinted con- 6. Zopf, D. A., Hollister, S. J., Nelson, M. E., Ohye, R. G. & Green, G. E.
structs, and enable manufacturers to optimize their processes for a Bioresorbable airway splint created with a three-dimensional printer.
New Eng. J. Med. 368, 2043–2045 (2013).
specific construct or product and to minimize the resources required 7. Morrison, R. J. et al. Mitigation of tracheobronchomalacia with 3D-printed
for the development of base materials and technical processes. personalized medical devices in pediatric patients. Sci. Transl. Med. 7,
Standardized cell-culture media and bioinks, and standardized 285–264 (2015).
quality-control systems for in-line sensing (required to collect qual- 8. Highlights of Prescribing Information—Spritam 2015 (FDA, 2017).
ity-control data throughout the manufacturing process and to ensure 9. Mankin, H. J., Mow, V. C., Buckwalter, J. A., Iannotti, J. P. & Ratcliffe, A.
Articular cartilage structure, composition, and function. Orthopaed. Basic
product safety, efficacy and reproducibility), are also necessary for the Sci. 2, 443–470 (2000).
reduction of the high costs and prolonged development times asso- 10. Buckwalter, J. & Mankin, H. Articular cartilage: tissue design
ciated with the manufacturing of regenerative-medicine products121. and chondrocyte-matrix interactions. Instr. Course Lect. 47,
477–486 (1998).
11. Hunziker, E., Quinn, T. & Häuselmann, H.-J. Quantitative structural
Logistics. Because products that contain living cells and tissues are
organization of normal adult human articular cartilage. Osteoarth. Cart. 10,
environmentally and time-sensitive, logistical demands for these 564–572 (2002).
products are especially challenging. The logistical chain can be par- 12. Sharma, B. et al. Designing zonal organization into tissue-engineered
ticularly complex when considering the manufacturing of patient- cartilage. Tissue Eng. 13, 405–414 (2007).
specific tissues or organs, as it is unlikely that patient-care facilities 13. Kesti, M. et al. Bioprinting complex cartilaginous structures with clinically
compliant biomaterials. Adv. Funct. Mat. 25, 7406–7417 (2015).
will possess 3D-printing capabilities or be capable of the clinical- 14. Schuurman, W. et al. Gelatin‐methacrylamide hydrogels as potential
grade production of the necessary cells and materials. Current cold- biomaterials for fabrication of tissue‐engineered cartilage constructs.
storage shipping procedures, originally developed decades ago, do Macromol. Biosci. 13, 551–561 (2013).
not meet the precision or performance required for living biological 15. Ávila, H. M., Schwarz, S., Rotter, N. & Gatenholm, P. 3D bioprinting of
products. A new shipping system that ensures the required stabil- human chondrocyte-laden nanocellulose hydrogels for patient-specific
auricular cartilage regeneration. Bioprinting 1, 22–35 (2016).
ity, communication, data collection and documented compliance 16. Markstedt, K. et al. 3D bioprinting human chondrocytes with
will thus need to be designed. This includes the transfer of medical nanocellulose–alginate bioink for cartilage tissue engineering applications.
data for the design of biofabricated tissues or organs. Furthermore, Biomacromolecules 16, 1489–1496 (2015).
because few facilities possess the expertise, technology and resources 17. Hwang, N. S. et al. Response of zonal chondrocytes to extracellular
to manufacture tissues and organs for transplantation, a centralized matrix‐hydrogels. FEBS Lett. 581, 4172–4178 (2007).
18. Coates, E. & Fisher, J. P. Gene expression of alginate‐embedded
logistical model may need to be used, at least initially; several major chondrocyte subpopulations and their response to exogenous IGF‐1
facilities could then be strategically located. Hence, patient-derived delivery. J. Tissue Eng. Regen. Med. 6, 179–192 (2012).
cells and materials would need to be transported to a centralized 19. Cui, X., Breitenkamp, K., Finn, M. G., Lotz, M. & D’Lima, D. D. Direct
biofabrication facility, and the biofabricated organs would then be human cartilage repair using three-dimensional bioprinting technology.
shipped back to the patient. The shipping logistics could take a page Tissue Eng. 18, 1304–1312 (2012).
20. Gao, G. et al. Improved properties of bone and cartilage tissue from 3D
from traditional organ-procurement and transplantation programs, inkjet-bioprinted human mesenchymal stem cells by simultaneous
which focus on best systems and practices for organ procurement, deposition and photocrosslinking in PEG-GelMA. Biotechnol. Lett. 37,
multi-site coordination, preservation and transportation. 2349 (2015).

378 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng


NaTure BIomeDIcaL EngIneerIng Perspective
21. Yu, Y. et al. Three-dimensional bioprinting using self-assembling scalable 50. Skardal, A. et al. Bioprinted amniotic fluid‐derived stem cells
scaffold-free “tissue strands” as a new bioink. Sci. Rep. 6, 28714 (2016). accelerate healing of large skin wounds. Stem Cell. Transl. Med. 1,
22. Levato, R. et al. The bio in the ink: cartilage regeneration with bioprintable 792–802 (2012).
hydrogels and articular cartilage-derived progenitor cells. Acta Biomater. 61, 51. Skardal, A. et al. A tunable hydrogel system for long‐term release of
41–53 (2017). cell‐secreted cytokines and bioprinted in situ wound cell delivery. J. Biomed.
23. Nguyen, L. H., Kudva, A. K., Saxena, N. S. & Roy, K. Engineering articular Mat. Res. Appl. Biomat. 105, 1986–2000 (2017).
cartilage with spatially-varying matrix composition and mechanical 52. Ito, M. et al. Stem cells in the hair follicle bulge contribute to wound repair
properties from a single stem cell population using a multi-layered but not to homeostasis of the epidermis. Nat. Med. 11, 1351 (2005).
hydrogel. Biomaterials 32, 6946–6952 (2011). 53. Hsu, Y.-C., Li, L. & Fuchs, E. Emerging interactions between skin stem cells
24. Nguyen, L. H., Kudva, A. K., Guckert, N. L., Linse, K. D. & Roy, K. and their niches. Nat. Med. 20, 847–856 (2014).
Unique biomaterial compositions direct bone marrow stem cells into 54. Ahn, G. et al. Precise stacking of decellularized extracellular matrix based
specific chondrocytic phenotypes corresponding to the various zones of 3D cell-laden constructs by a 3D cell printing system equipped with heating
articular cartilage. Biomaterials 32, 1327–1338 (2011). modules. Sci. Rep. 7, 8624 (2017).
25. Nguyen, D. et al. Cartilage tissue engineering by the 3D bioprinting of iPS 55. Murphy, S. V., Skardal, A. & Atala, A. Evaluation of hydrogels for
cells in a nanocellulose/alginate bioink. Sci. Rep. 7, 658 (2017). bio‐printing applications. J. Biomed. Mat. Res. 101, 272–284 (2013).
26. Kang, H.-W. et al. A 3D bioprinting system to produce human-scale tissue 56. Hardin, J. O., Ober, T. J., Valentine, A. D. & Lewis, J. A. Microfluidic
constructs with structural integrity. Nat. Biotechnol. 34, 312–319 (2016). printheads for multimaterial 3D printing of viscoelastic inks. Adv. Mater.
27. Gao, M. et al. Tissue-engineered trachea from a 3D-printed scaffold 27, 3279–3284 (2015).
enhances whole-segment tracheal repair. Sci. Rep. 7, 5246 (2017). 57. Ober, T. J., Foresti, D. & Lewis, J. A. Active mixing of complex fluids at the
28. Barbero, A. et al. Age related changes in human articular chondrocyte microscale. Proc. Natl. Acad. Sci. USA 112, 12293–12298 (2015).
yield, proliferation and post-expansion chondrogenic capacity. Osteoarth. 58. Cui, H. et al. Hierarchical fabrication of engineered vascularized bone
Cartil. 12, 476–484 (2004). biphasic constructs via dual 3D bioprinting: integrating regional bioactive
29. Huang, A. H., Stein, A., Tuan, R. S. & Mauck, R. L. Transient exposure to factors into architectural design. Adv. Health Mater. 5, 2174–2181 (2016).
transforming growth factor beta 3 improves the mechanical properties of 59. Merceron, T. K. et al. A 3D bioprinted complex structure for engineering
mesenchymal stem cell–laden cartilage constructs in a density-dependent the muscle–tendon unit. Biofabrication 7, 035003 (2015).
manner. Tissue Eng. 15, 3461–3472 (2009). 60. Huang, G. et al. Engineering three-dimensional cell mechanical
30. Majumdar, M. K., Banks, V., Peluso, D. P. & Morris, E. A. Isolation, microenvironment with hydrogels. Biofabrication 4, 042001 (2012).
characterization, and chondrogenic potential of human bone marrow‐ 61. Skardal, A. et al. A hydrogel bioink toolkit for mimicking native tissue
derived multipotential stromal cells. J. Cell. Phys. 185, 98–106 (2000). biochemical and mechanical properties in bioprinted tissue constructs.
31. Huang, A. H., Farrell, M. J., Kim, M. & Mauck, R. L. Long-term dynamic Acta Biomater. 25, 24–34 (2015).
loading improves the mechanical properties of chondrogenic mesenchymal 62. Zarembinski, T. I. & Skardal, A. in Hydrogels-Smart Materials for Biomedical
stem cell-laden hydrogels. Eur. Cell. Mat. 19, 72 (2010). Applications (ed. Popa, L.) Ch. 5 (IntechOpen, 2018).
32. Shegarfi, H. & Reikeras, O. Bone transplantation and immune response. 63. Skardal, A. et al. Photocrosslinkable hyaluronan-gelatin hydrogels for
J. Orthopaed. Surg. 17, 206–211 (2009). two-step bioprinting. Tissue Eng. 16, 2675 (2010).
33. Henkel, J. et al. Bone regeneration based on tissue engineering 64. Fan, H. et al. Fabrication, mechanical properties, and biocompatibility
conceptions—a 21st century perspective. Bone Res. 1, 216 (2013). of graphene-reinforced chitosan composites. Biomacromolecules 11,
34. Olszta, M. J. et al. Bone structure and formation: a new perspective. 2345–2351 (2010).
Mat. Sci. Eng. Rep. 58, 77–116 (2007). 65. Zhang, S. Emerging biological materials through molecular self-assembly.
35. Bose, S., Vahabzadeh, S. & Bandyopadhyay, A. Bone tissue engineering Biotechnol. Adv. 20, 321–339 (2002).
using 3D printing. Mat. Today 16, 496–504 (2013). 66. Zhang, S. Fabrication of novel biomaterials through molecular self-
36. Inzana, J. A. et al. 3D printing of composite calcium phosphate assembly. Nat. Biotechnol. 21, 1171–1178 (2003).
and collagen scaffolds for bone regeneration. Biomaterials 35, 67. Gelain, F., Horii, A. & Zhang, S. Designer self‐assembling peptide scaffolds
4026–4034 (2014). for 3‐D tissue cell cultures and regenerative medicine. Macromol. Biosci. 7,
37. Probst, F., Hutmacher, D., Müller, D., Machens, H. & Schantz, J. 544–551 (2007).
Calvarial reconstruction by customized bioactive implant. (German trans.) 68. Mano, J. F. Stimuli‐responsive polymeric systems for biomedical
Handchir. Mikrochir. Plast. Chir. 42, 369–373 (2010). applications. Adv. Eng. Mat. 10, 515–527 (2008).
38. Catros, S. et al. Laser-assisted bioprinting for creating on-demand patterns 69. Karbarz, M., Mackiewicz, M., Kaniewska, K., Marcisz, K. & Stojek, Z.
of human osteoprogenitor cells and nano-hydroxyapatite. Biofabrication 3, Recent developments in design and functionalization of micro-and
025001 (2011). nanostructural environmentally-sensitive hydrogels based on
39. Shim, J.-H., Lee, J.-S., Kim, J. Y. & Cho, D.-W. Bioprinting of a mechanically N-isopropylacrylamide. Appl. Mat. Today 9, 516–532 (2017).
enhanced three-dimensional dual cell-laden construct for osteochondral 70. Liu, X. et al. ROCK inhibitor and feeder cells induce the conditional
tissue engineering using a multi-head tissue/organ building system. reprogramming of epithelial cells. Am. J. Path. 180, 599–607 (2012).
J. Micromech. Microeng. 22, 085014 (2012). 71. Chapman, S., Liu, X., Meyers, C., Schlegel, R. & McBride, A. A. Human
40. Shim, J.-H., Kim, J. Y., Park, M., Park, J. & Cho, D.-W. Development of a keratinocytes are efficiently immortalized by a Rho kinase inhibitor.
hybrid scaffold with synthetic biomaterials and hydrogel using solid J. Clin. Investig. 120, 2619 (2010).
freeform fabrication technology. Biofabrication 3, 034102 (2011). 72. Dominici, M. et al. Minimal criteria for defining multipotent mesenchymal
41. Daly, A. C. et al. 3D bioprinting of developmentally inspired templates for stromal cells. The International Society for Cellular Therapy position
whole bone organ engineering. Adv. Health Mater. 5, 2353–2362 (2016). statement. Cytotherapy 8, 315–317 (2006).
42. Lee, W. et al. Multi-layered culture of human skin fibroblasts and 73. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal
keratinocytes through three-dimensional freeform fabrication. Biomaterials stem cells. Science 284, 143–147 (1999).
30, 1587–1595 (2009). 74. Zuk, P. A. et al. Human adipose tissue is a source of multipotent stem cells.
43. Koch, L. et al. Skin tissue generation by laser cell printing. Biotechnol. Mol. Biol. Cell. 13, 4279–4295 (2002).
Bioeng. 109, 1855–1863 (2012). 75. De Coppi, P. et al. Isolation of amniotic stem cell lines with potential for
44. Michael, S. et al. Tissue engineered skin substitutes created by laser-assisted therapy. Nat. Biotechnol. 25, 100–106 (2007).
bioprinting form skin-like structures in the dorsal skin fold chamber in 76. Wagner, W. et al. Replicative senescence of mesenchymal stem cells: a
mice. PLoS ONE 8, e57741 (2013). continuous and organized process. PLoS ONE 3, e2213 (2008).
45. Lee, W. et al. On‐demand three‐dimensional freeform fabrication of 77. Zhou, S. et al. Age‐related intrinsic changes in human bone‐marrow‐
multi‐layered hydrogel scaffold with fluidic channels. Biotechnol. Bioeng. derived mesenchymal stem cells and their differentiation to osteoblasts.
105, 1178–1186 (2010). Aging Cell 7, 335–343 (2008).
46. Lee, V. et al. Design and fabrication of human skin by three-dimensional 78. Choudhery, M. S., Badowski, M., Muise, A., Pierce, J. & Harris, D. T.
bioprinting. Tissue Eng. Meth. 20, 473–484 (2013). Donor age negatively impacts adipose tissue-derived mesenchymal stem cell
47. Cubo, N., Garcia, M., del Cañizo, J. F., Velasco, D. & Jorcano, J. L. 3D expansion and differentiation. J. Transl. Med. 12, 8 (2014).
bioprinting of functional human skin: production and in vivo analysis. 79. Stolzing, A., Jones, E., McGonagle, D. & Scutt, A. Age-related changes in
Biofabrication 9, 015006 (2016). human bone marrow-derived mesenchymal stem cells: consequences for
48. Binder, K. W. In situ bioprinting of the skin. PhD thesis, Wake Forest cell therapies. Mech. Ageing Dev. 129, 163–173 (2008).
University (2011). 80. Takahashi, K. et al. Induction of pluripotent stem cells from adult human
49. Albanna, M. et al. In situ bioprinting of autologous skin cells accelerates fibroblasts by defined factors. Cell 131, 861–872 (2007).
wound healing of extensive excisional full-thickness wounds. Sci. Rep. 9, 81. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic
1856 (2019). cells. Science 318, 1917–1920 (2007).

Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng 379


Perspective NaTure BIomeDIcaL EngIneerIng
82. Kimbrel, E. A. & Lanza, R. Current status of pluripotent stem cells: 108. Kolesky, D. B., Homan, K. A., Skylar-Scott, M. A. & Lewis, J. A. Three-
moving the first therapies to the clinic. Nat. Rev. Drug Discov. 14, dimensional bioprinting of thick vascularized tissues. Proc. Natl Acad. Sci.
681–692 (2015). USA 113, 3179–3184 (2016).
83. Scudellari, M. How iPS cells changed the world. Nature 534, 109. Norotte, C., Marga, F. S., Niklason, L. E. & Forgacs, G. Scaffold-free
310–312 (2016). vascular tissue engineering using bioprinting. Biomaterials 30,
84. Lee, A. S., Tang, C., Rao, M. S., Weissman, I. L. & Wu, J. C. Tumorigenicity 5910–5917 (2009).
as a clinical hurdle for pluripotent stem cell therapies. Nat. Med. 19, 110. Zhu, W. et al. Direct 3D bioprinting of prevascularized tissue
998–1004 (2013). constructs with complex microarchitecture. Biomaterials 124,
85. Trounson, A. & DeWitt, N. D. Pluripotent stem cells progressing to the 106–115 (2017).
clinic. Nat. Rev. Mol. Cell Biol. 17, 194–200 (2016). 111. Hansen, C. J. et al. High‐throughput printing via microvascular multinozzle
86. Mandai, M. et al. Autologous induced stem-cell-derived retinal cells for arrays. Adv. Mater. 25, 96–102 (2013).
macular degeneration. New Engl. J. Med. 376, 1038–1046 (2017). 112. Traore, M. A. & George, S. C. Tissue Engineering the Vascular Tree.
87. Simonson, O. E., Domogatskaya, A., Volchkov, P. & Rodin, S. The Tissue Eng. Rev. 23, 505–514 (2017).
safety of human pluripotent stem cells in clinical treatment. Ann. Med. 47, 113. Ehsan, S. M. & George, S. C. Nonsteady state oxygen transport in
370–380 (2015). engineered tissue: implications for design. Tissue Eng. 19, 1433–1442 (2013).
88. Ma, H. et al. Abnormalities in human pluripotent cells due to 114. Ehsan, S. M. & George, S. C. Vessel network formation in response
reprogramming mechanisms. Nature 511, 177–183 (2014). to intermittent hypoxia is frequency dependent. J. Biosci. Bioeng. 120,
89. Okita, K. et al. A more efficient method to generate integration-free human 347–350 (2015).
iPS cells. Nat. Methods 8, 409–412 (2011). 115. Hsu, Y. H., Moya, M. L., Hughes, C. C., George, S. C. & Lee, A. P.
90. Umekage, M., Sato, Y. & Takasu, N. Overview: an iPS cell stock at CiRA. A microfluidic platform for generating large-scale nearly identical
Inflamm. Regen. 39, 17 (2019). human microphysiological vascularized tissue arrays. Lab Chip 13,
91. Deegan, D. B., Zimmerman, C., Skardal, A., Atala, A. & Shupe, T. D. 2990–2998 (2013).
Stiffness of hyaluronic acid gels containing liver extracellular matrix 116. Tumbleston, J. R. et al. Additive manufacturing: continuous liquid interface
supports human hepatocyte function and alters cell morphology. production of 3D objects. Science 347, 1349–1352 (2015).
J. Mech. Behav. Biomed. Mater. 55, 87–103 (2016). 117. Kengla, C. et al. Clinically relevant bioprinting workflow and imaging
92. Loneker, A. E., Faulk, D. M., Hussey, G. S., D’Amore, A. & Badylak, S. F. process for tssue construct design and validation. 3D Print. Addit. Manuf. 4,
Solubilized liver extracellular matrix maintains primary rat hepatocyte 239–247 (2017).
phenotype in‐vitro. J. Biomed. Mat. Res. 104, 957–965 (2016). 118. Tibbits, S. Design to self-Assembly. Arch. Des. 82, 68–73 (2012).
93. Kobayashi, J., Akiyama, Y., Yamato, M. & Okano, T. ECM-mimicking 119. Bonamici, S. H. R.34 - 21st Century Cures Act Senate Report 114–146
thermoresponsive surface for manipulating hepatocyte sheets with (United States Congress, 2016).
maintenance of hepatic functions. in 2016 International Symposium on 120. Technical Considerations for Additive Manufactured Medical Devices
Micro-NanoMechatronics and Human Science (MHS) (IEEE, 2016) - Guidance for Industry and Food and Drug Administration Staff
94. Huch, M. et al. Long-term culture of genome-stable bipotent stem cells (FDA, 2017).
from adult human liver. Cell 160, 299–312 (2015). 121. Hunsberger, J. G., Goel, S., Allickson, J. & Atala, A. Five critical areas that
95. Moroni, L. et al. Biofabrication: a guide to technology and terminology. combat high costs and prolonged development times for regenerative
Trends Biotechnol. 36, 348–402 (2017). medicine manufacturing. Curr. Stem Cell Rep. 3, 77–82 (2017).
96. Mironov, V. et al. Organ printing: tissue spheroids as building blocks. 122. Gladman, A. S., Matsumoto, E. A., Nuzzo, R. G., Mahadevan, L. &
Biomaterials 30, 2164–2174 (2009). Lewis, J. A. Biomimetic 4D printing. Nat. Mater. 15, 413–418 (2016).
97. Jakab, K. et al. Tissue engineering by self-assembly of cells printed into 123. Kuribayashi-Shigetomi, K., Onoe, H. & Takeuchi, S. Cell origami:
topologically defined structures. Tissue Eng. 14, 413–421 (2008). self-folding of three-dimensional cell-laden microstructures driven by cell
98. Janssen, F. W., Oostra, J., van Oorschot, A. & van Blitterswijk, C. A. traction force. PLoS ONE 7, e51085 (2012).
A perfusion bioreactor system capable of producing clinically relevant
volumes of tissue-engineered bone: in vivo bone formation showing proof
of concept. Biomaterials 27, 315–323 (2006). Acknowledgements
99. Brown, D. A. et al. Analysis of oxygen transport in a diffusion‐limited P.D.C. is supported by the National Institute for Health Research
model of engineered heart tissue. Biotechnol. Bioeng. 97, 962–975 (2007). (NIHR-RP-2014-04-046) and by the NIHR Great Ormond Street Hospital Biomedical
100. Jain, R. K., Au, P., Tam, J., Duda, D. G. & Fukumura, D. Engineering Research Centre.
vascularized tissue. Nat. Biotechnol. 23, 821–823 (2005).
101. Muschler, G. F., Nakamoto, C. & Griffith, L. G. Engineering principles of
clinical cell-based tissue engineering. JBJS 86, 1541–1558 (2004).
Author contributions
S.V.M, P.D.C and A.A. discussed the content, researched the literature and wrote
102. Bertassoni, L. E. et al. Direct-write bioprinting of cell-laden methacrylated
the manuscript.
gelatin hydrogels. Biofabrication 6, 024105 (2014).
103. Bertassoni, L. E. et al. Hydrogel bioprinted microchannel networks
for vascularization of tissue engineering constructs. Lab Chip 14, Competing interests
2202–2211 (2014). The authors declare no competing interests.
104. Poldervaart, M. T. et al. Prolonged presence of VEGF promotes
vascularization in 3D bioprinted scaffolds with defined architecture.
J. Contr. Release 184, 58–66 (2014). Additional information
105. Wu, W., DeConinck, A. & Lewis, J. A. Omnidirectional printing of 3D Correspondence should be addressed to S.V.M.
microvascular networks. Adv. Mater. 23, 178–183 (2011). Reprints and permissions information is available at www.nature.com/reprints.
106. Ozbolat, I. T. Bioprinting scale-up tissue and organ constructs for
transplantation. Trends Biotechnol. 33, 395–400 (2015). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
107. Kolesky, D. B. et al. 3D bioprinting of vascularized, heterogeneous published maps and institutional affiliations.
cell‐laden tissue constructs. Adv. Mater. 26, 3124–3130 (2014). © Springer Nature Limited 2019

380 Nature Biomedical Engineering | VOL 4 | April 2020 | 370–380 | www.nature.com/natbiomedeng

You might also like