You are on page 1of 18

876286

review-article2019
JAO0010.1177/0391398819876286The International Journal of Artificial OrgansPerić Kačarević et al.

IJAO The International


Journal of Artificial
Organs
Review

The International Journal of Artificial

An introduction to bone tissue engineering


Organs
1­–18
© The Author(s) 2019
Article reuse guidelines:
sagepub.com/journals-permissions
https://doi.org/10.1177/0391398819876286
DOI: 10.1177/0391398819876286
journals.sagepub.com/home/jao
Željka Perić Kačarević1, Patrick Rider2 , Said Alkildani3,
Sujith Retnasingh4, Marija Pejakić5, Reinhard Schnettler6,7,
Martin Gosau6,7, Ralf Smeets6,7, Ole Jung7,*
and Mike Barbeck2,6,8,*

Abstract
Bone tissue has the capability to regenerate itself; however, defects of a critical size prevent the bone from regenerating
and require additional support. To aid regeneration, bone scaffolds created out of autologous or allograft bone can be
used, yet these produce problems such as fast degradation rates, reduced bioactivity, donor site morbidity or the risk of
pathogen transmission. The development of bone tissue engineering has been used to create functional alternatives to
regenerate bone. This can be achieved by producing bone tissue scaffolds that induce osteoconduction and integration,
provide mechanical stability, and either integrate into the bone structure or degrade and are excreted by the body. A
range of different biomaterials have been used to this end, each with their own advantages and disadvantages. This review
will introduce the requirements of bone tissue engineering, beginning with the regeneration process of bone before
exploring the requirements of bone tissue scaffolds. Aspects covered include the manufacturing process as well as the
different materials used and the incorporation of bioactive molecules, growth factors and cells.

Keywords
Bone tissue engineering, osteoinductive, osteoconductive, bone remodelling, macrophages

Date received: 3 April 2019; accepted: 26 July 2019

Introduction 100–230 MPa, while trabecular or cancellous bone has a


higher porosity between 30% and 90% and a lower com-
Bone tissue pressive strength of 2–12 MPa.2
Bone is a composite of organic and inorganic phases.1
The organic components are mainly collagen type I 1Department of Anatomy Histology, Embryology, Pathology Anatomy
(COL-I) (~30%) and inorganic components are mostly and Pathology Histology, Faculty of Dental Medicine and Health, Josip
calcium phosphates (~70%) in the form of hydroxyapa- Juraj Strossmayer University of Osijek, Osijek, Croatia
2Research and Development, botiss biomaterials GmbH, Berlin, Germany
tite (HA). The nanoscale organization of the organic and
3Department of Biomedical Engineering, School of Applied Medical
inorganic phases is responsible for producing the
Sciences, German Jordanian University, Amman, Jordan
mechanical strength and durability that characterizes 4Institute for Environmental Toxicology, Martin-Luther-Universität

bone. Microfibrils of collagen, synthesized by osteo- Halle-Wittenberg, Halle, Germany


blasts, aggregate laterally and longitudinally; forming 5Department of Dental Medicine, Faculty of Dental Medicine and

fibres that are then called an osteoid. The osteoblasts Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
6Department of Oral and Maxillofacial Surgery, University Hospital
deposit HA crystals into the voids between the collagen
Hamburg-Eppendorf, Hamburg, Germany
fibrils during a process called biomineralization. The 7Department of Oral and Maxillofacial Surgery, Division of

degree of biomineralization determines the mechanical Regenerative Orofacial Medicine, University Medical Center Hamburg-
properties of the bone tissue; a higher degree of mineral- Eppendorf, Hamburg, Germany
8BerlinAnalytix GmbH, Berlin, Germany
ized bone matrix provides a stiffer structure, however,
one that is more susceptible to fracture. *The authors contributed equally.
The two main types of bone are trabecular and cortical.
Corresponding author:
The bone types differ from each other by their structure. Patrick Rider, Research and Development, botiss biomaterials GmbH,
Cortical, also known as compact bone, has a porosity Hauptstaße 28, 15806 Zossen, Germany.
between 5% and 30% and a compressive strength of Email: patrick.rider@botiss.com
2 The International Journal of Artificial Organs 00(0)

Figure 1.  Demonstration of the tissue reaction to the biphasic bone substitute granules 2 months postimplantation and the bony
integration of the biphasic granules into new-built bone tissue within the peripheral regions of the implant beds neighboured to the
defect borders. The detection of (a) M1 and (b) M2 positive macrophages into the implantation beds. Representative biomaterial;
synthetic bone graft substitute (maxresorb, botiss biomaterials GmbH, Germany). Immunohistochemical detection of macrophages
and their M1 and M2 subforms by means of antibodies against the pro-inflammatory and anti-inflammatory molecules, that is,
haemoglobin scavenger receptor (CD163) and mannose receptor (MR; also known as CD206): (a) 400× magnification and (b)
100× magnification. *: Synthetic bone graft substitute; →: macrophages.

Bone regeneration substitute material.4 Once a material is implanted, the


blood/biomaterial interaction begins immediately from the
Before designing a scaffold for bone tissue engineering moment the implant is inserted in to the body.5 During
(BTE), it is important to understand the fundamentals of implantation, the body triggers a tissue response involving
bone regeneration, which occurs after injury or trauma inflammatory and wound healing responses. Immediately,
such as after the implantation of a BTE scaffold. after the implantation in the injury site, changes in blood
The fundamental process of bone formation, otherwise flow results in the discharging of fluids, cells and the pro-
known as osteogenesis, can happen via two routes: endo- teins from the surrounding tissues. These adsorb onto the
chondral ossification or intramembranous ossification.3 surface of the implanted materials and induce thrombus
Both processes begin with the condensation of specialized formation. The thrombus is composed of platelets, fibrin
mesenchymal stem cells (MSCs) (discussed in more detail fibres, as well as red and white blood cells. The combina-
below) and are dependent on the differentiation of this tion of these components enables the thrombus to form a
condensate. In endochondral ossification, the MSCs dif- provisional blood-based matrix that is rich with growth fac-
ferentiate into chondrocytes that deposit cartilage tissue tors, cytokines and chemo-attractants that recruit and medi-
which is then mineralized by osteoblasts. The chondro- ate the innate immune cells residing in the bone marrow.
cytes and cartilage matrix reach a terminal and hyper- When stimulated, the innate immune cells enter the
trophic state, and are then engulfed and resorbed by blood circulation and travel towards the injured site. The
preosteoblasts, osteoblasts and blood vessels. This struc- arrival of these cells orchestrates the healing process. First
ture is eventually replaced by bone marrow. Therefore, the to arrive are neutrophils that initiate an acute inflammatory
endochondral ossification process is limited to the forma- reaction. Neutrophils begin to debride the wound, kill path-
tion of marrow-containing bone types in the body. ogens and release more soluble factors for the further
Intramembranous ossification involves direct osteogenic recruitment of immune cells. Once their function is com-
differentiation of the MSCs condensate; hence, does not plete, the neutrophils undergo apoptosis. Along with the
involve and intermediate chondrogenic phase. This pro- neutrophils, monocytes are also recruited to the site of injury
cess is used in the formation of facial and cranial bones. via chemotaxis. These enter the blood circulation and dif-
These osteogenic processes are effective at repairing and ferentiate en route into macrophages. The macrophages
restoring bone after trauma and injury. However, should remove apoptosed neutrophils via phagocytosis and secrete
the size of the defect reach a critical size, the regeneration cytokines and growth factors. Macrophages change their
process requires external support. phenotype and the cytokines they secrete depend on molec-
Current evidence around the definition of a critical sized ular signalling and their cellular environment that are influ-
bone defect ranges from 2.5–3 cm or greater. The regenera- enced via their interaction with the biomaterial.
tive process is unable to repair the bone without external Macrophages have several different phenotypes, two of
intervention, often in the form of an implanted bone them being: M1 and M2. Each phenotype is not terminally
Perić Kačarević et al. 3

differentiated, but are instead reversible, responding to


external cues. Pro-inflammatory mediators from a cell-
mediated immune response will activate macrophages into
an M1 phenotype, commonly referred to as ‘classically
activated macrophages’. These cells take on a defensive
role when interacting with a biomaterial, performing
phagocytotic functions to remove pathogens and debris
from the injury site, as well as performing pro-inflamma-
tory functions by secreting tumour necrosis factor-alpha
(TNF-α) and interleukin-6 (IL-6) cytokines.6 M2 mac-
rophages, commonly referred to as ‘alternatively activated
macrophages’, act to instigate repair functions. Figure 1
contains two histological images of chemo-stained M1 and
M2 macrophages in implanted bone grafts.
M2 macrophages are the phenotype of resident tissue
macrophages, and are activated by IL-4 and IL-13 as well
as the phagocytosis of apoptosed neutrophils.6 These cells
perform anti-inflammatory functions by secreting
cytokines such as IL-10,6 as well as increase the produc-
tion of extracellular matrix (ECM). The M2 phenotype can
be further split into four types: M2a, M2b, M2c and M2d;
however, the precise function of each group has yet to be
precisely defined. Macrophages residing on the implant
bed have been demonstrated to promote vascularization,
bone cell differentiation and overall bone growth via the
expression of vascular endothelial growth factor (VEGF).7 Figure 2.  A depiction of the cyclic process of bone
remodelling in healthy bone, along with the influence of
Some studies have shown a correlation of increased vascu-
the different phenotypes of macrophages during healing.
larization with higher numbers of macrophages.8 In soft Osteoblasts, ascending of mesenchymal stem cells (MSCs)
tissue it has been shown that without the presence of M2 lineage, deposit and organize both organic and inorganic
macrophages during mid-stage healing, there was a lack a phases during regeneration. Osteoblasts activity is increased by
vascularization of the wound bed.6 Figure 2 depicts the anti-inflammatory cytokines secreted from M2 macrophages.
remodelling process of bone and the role of macrophages Osteoclasts are multinucleated cells with monocytic origin that
in healing. breakdown bone tissue during resorption. Osteoclast activity
is increased by pro-inflammatory cytokines secreted from M1
Macrophages can remain mononucleated individual macrophages. Regeneration and resorption occur in tandem in
cells, or fuse into multinucleated giant cells (MGCs) healthy bone as a lifelong ongoing process of bone remodelling.
depending upon their interaction with the biomaterial. If MSCs: mesenchymal stem cells; HSCs: haematopoietic stem cells.
the biomaterial is too large to phagocytose, the MGCs can *Secretion of pro-inflammatory cytokines.
**Secretion of anti-inflammatory cytokines.
fuse together, enabling them to surround the biomaterial.
Yet as they increase in size, their ability to perform phago-
cytosis decreases. Instead, as the multinucleated cell size surrounding tissue matrix. Due to their perceived similarity
increases, its capability for secreting extracellular degra- to osteoclasts, the MGCs were described as foreign body
dative enzymes improves. If the particle remains too large giant cells (FBGCs). However, studies have found MGCs
to be phagocytosed, the MGCs remain at the interface residing next to bone substitute materials years after
between the biomaterial and tissue for the duration of time implantation, surrounded by a flourishing bony matrix,
the implanted device remains in situ. thus contradicting this theory. This indicates that some
For biomaterials with a slow rate of degradation, MGCs MGCs have an inability to resorb biomaterial and there-
are observed to persist on the biomaterial surface and dem- fore, there may be differences between MGCs and the func-
onstrate similar histological features to that of osteoclasts. tions they perform.10 The presence of MGCs around bone
However, unlike osteoclasts, the MGCs do not show evi- grafts over prolonged period of time has shown evidence of
dence of particle resorption over prolonged periods.9 inducing vascularization and bone formation.8,9,11
Osteoclasts are bone-absorbing MGCs that work in tandem
with osteoblasts in natural process called bone remodula-
Growth factors in BTE
tion. It had previously been assumed that these MGCs
residing on the surface of biomaterials were similar to oste- Bone is a dynamic tissue that undergoes remodelling
oclasts and would breakdown the foreign body and throughout life. Growth factors are important for regulating
4 The International Journal of Artificial Organs 00(0)

bone formation because their regulatory functions include


cell adhesion, proliferation and differentiation. Bone
remodelling is executed by a functional and anatomic struc-
ture known as basic multicellular units (BMUs), which
includes four major bone cells: bone-lining cells, osteo-
cytes, osteoclasts and osteoblasts. The surface of the bone
is covered by a monolayer of bone-lining cells. Osteocytes
are the most abundant bone cells and play a vital role in the
initiation of bone remodelling, which is important for
maintaining the strength of the bone matrix. Osteoclasts are
MGCs that have differentiated from mononuclear cells by
two different growth factors: the monocyte/macrophage
colony stimulating factor (M-CSF) and the receptor activa-
tor of nuclear factor kappa-Β ligand (RANKL).12 Various
concentration levels of RANKL throughout several organs
reconfirm the importance of RANKL in tissue growth (par-
ticularly bone growth) and immune functions within the Figure 3.  Tissue reaction to biphasic bone substitute granules
body. Osteoblasts produce bone and are derived from 6 months postimplantation. Bony integration of the biphasic
MSCs. Osteoblasts are responsible for the deposition of granules into new-built bone tissue within the peripheral
collagen fibres and HA and the activation of osteoclast by regions of the implant beds neighboured to the defect borders.
expressing RANKL and M-CSF growth factors. Osteoclasts The granules were optimally integrated into bone tissue up
to their pores without any histological signs of exaggerated
secrete different growth factors including bone morpho- inflammatory tissue reactions. Representative biomaterial: *
genic proteins (BMPs) that influence the formation of new synthetic bone graft substitute (botiss biomaterials GmbH,
bone.13 However, many skeletal defects or extensive bone Germany), + new bone, and  soft tissue (haematoxylin and
loss through deterioration, may result in failure of bone eosin (H&E)-staining, 100× magnification).
healing.
The essential growth factors for BTE include VEGF,
been shown to be successful in bone regeneration in vitro,
fibroblast growth factors (FGFs) and BMPs.12,14,15 VEGF is
larger pore sizes, such as those similar to cancellous bone
an angiogenic protein which regulates the endothelial cell
(between 200 and 500 µm), are found to provide optimal
proliferation. BMPs induce osteogenesis through MSC dif-
tissue penetration and enable vascularization to occur in
ferentiation to osteoblasts. The combined delivery of VEGF
vivo.3,12,13
and BMPs could provide a consorted effect to induce angi-
The material used to fabricate the scaffold will also
ogenesis and osteogenesis, as the VEGF has the potential to
impact the success of the implant. Non-resorbable implants
enhance BMP-induced bone formation.12 FGFs are essen-
are useful for immediate load-bearing applications and are
tial for angiogenesis as FGFs induce endothelial and osteo-
also important for patients with a low bone metabolism,
blast cell proliferation, crucial for normal vascular
such as patients suffering from cancer or osteoporosis.10 In
developmental stage. In humans, 22 different FGFs (FGF1–
healthy bone, a biodegradable implant is ideal as it is
FGF23, although there is no human FGF15) have been
identified. FGF2, FGF9 and FGF18 are based on geneti- replaced by the patient’s native bone. Figure 3 is a histo-
cally modified temporal expression and have been identi- logical image of new bone regeneration during bone graft
fied as the best candidates for bone formation.14 resorption.
The rate of biodegradation should relate to the rate of
new bone formation, so that the biomaterial is replaced by
Scaffold properties new bone in a process called creeping substitution.10 A
For successful integration and support of the regeneration BTE scaffold must endure mechanical stresses that are
of bone, scaffolds for BTE should provide characteristics physiologically relevant to the surrounding bone.16
similar to the native bone structure. Important characteris- Overall, the mechanical properties need to be taken into
tics to be considered for bone scaffolds include surface consideration when designing scaffold architecture, as the
roughness, porosity and pore size, interconnectivity, deg- mechanical strength can be compromised by increased
radation, mechanical properties and biocompatibility.16 porosity and degradation rates.
The microarchitecture of the scaffold, which includes The inflammatory reaction caused by a biocompatible
porosity, pore size and interconnectivity between pores, is biomaterial should resolve within 2 weeks. Chronic
essential for proper mass transfer of nutrients and waste inflammation could be an indication of an infection, or a
products, as well as vascularization and tissue infiltration. response to the degradative by-products of the biomateri-
Although smaller pore sizes between 75 and 100 µm have als used. Biomaterials in BTE must have an osteoinductive
Perić Kačarević et al. 5

capacity, whereby they should instigate MSCs to differen- osteoconduction. Osteoinduction is a regular phenomenon
tiate into bone-forming cells called osteoblasts. However, seen in any type of bone healing processes and is the stim-
many biomaterials do not possess this property and are ulation of immature cells (MSCs) to develop into preosteo-
therefore incorporated with bioactive inclusions; for exam- blasts. Bone growth on an implant surface depends on the
ple, demineralized bone matrix, bioceramics or BMPs.17 differentiation of bone cells, recruited from pre-existing
preosteoblasts/osteoblasts, that are activated by primitive
mesenchymal cells via osteoinduction. Grafts should also
Biomaterials
be osteoconductive, whereby they easily integrate with the
Due to the highly regenerative properties of bone, small adjacent bone. In the case of implants, osteoconduction is
defects are able to spontaneously begin the healing process dependent on the biomaterials used and its morphology.24
without intervention. Large-scale defects caused by trauma Osteointegration is the direct structural and functional
or genetic disorders require surgical intervention to implant integration between the host bone cells and the implants.
materials that guide and accelerate the healing process.
Biomaterials provide an essential component for bone
Natural polymers
regeneration. Their overall characteristics such as their
surface and bulk properties will ultimately affect the suc- Natural polymers benefit from a low immunological
cess of the scaffold. An idealized biomaterial must fulfil a potential and possible bioactive behaviour.25 Natural poly-
specific criteria: be biocompatible, be replaced by the host mers that have been investigated for BTE include poly-
bone at a rate matching new bone formation, degrade into mers naturally occurring in humans; collagen, fibrin and
non-toxic components and not to elicit a chronic immune elastin, as well as other natural polymers such as silk, chi-
response. Bio-incompatibility results in an immune system tosan and alginate.
activation, tumour formation and inflammation. Collagen is the main component of the ECM, a network
Biodegradability of the material refers to the chemical or of macromolecules that provides structural and mechani-
enzymatic breakdown of the material over time when cal support to cells within tissues. Collagen can be
introduced into living organisms. The material should extracted from animal sources for use in BTE. It can be
degrade into non-toxic components that are either recycled removed from animal tissue either by decellularization or
or excreted by the body.18 demineralization of an organ, or via the process of extrac-
Bone grafting materials can be divided into autolo- tion, purification and polymerization.26 The extracted col-
gous, allogeneic, xenogeneic and alloplastic. The gold lagen is typically ground down to be further processed into
standard material for repairing bone tissue is to use autol- hydrogels, foams and fibres that are used to mimic the
ogous bone. Autologous bone is bone harvested from the native ECM. As one of the most commonly used scaffold
same individual receiving the treatment. Although this materials, collagen and its denatured form gelatine are
can produce excellent healing results, the necessity of a favoured due to their fast biodegradability, weak anti-
secondary surgical site increases patient trauma and the genicity, as well as excellent biocompatibility and cell
risk of infection.19 Allografts are bone tissue harvested adhesion.27 Collagen scaffolds have also reported the
from one individual and transplanted between genetically potential to induce fast vascularization.28 However, colla-
non-identical individuals of the same species.20 Allografts gen has a poor elasticity and processed collagen can suffer
provide an alternative to autografts, however, can suffer from poor mechanical strength and requires methods such
from poor quality as the material is usually sourced from as crosslinking or blending with other polymers for
cadavers or from bone removed during surgical opera- improving the mechanical properties.29,30 Another alterna-
tions. However, when the quality of the sourced material tive is to produce a composite material by mixing the col-
is high, allografts can provide results similar to that of lagen with a ceramic such as beta-tricalcium phosphate
autologous grafts.21 Xenografts are tissue transferred from (β-TCP). The combined effects of increased mechanical
one species to another. Xenografts are readily available strength and the osteogenic characteristics of β-TCP were
and also provide excellent bone tissue regeneration in shown to improve bone regeneration in rabbit femoral
comparison to autologous bone,11 but risk disease trans- defects, when compared to a pure collagen scaffold.31
mission and can be unsuitable for patients from certain Silk has had a long history of use as a biomaterial.32 It
religious groups.22 Alloplastic grafts are of a synthetic ori- is highly utilized in the field of biomaterials as it has the
gin; this provides the benefit of preventing disease trans- ability to absorb and release water easily, demonstrates
mission and improving availability. One of the most excellent mechanical strength, and has a resistance to
commonly used types of alloplastic materials in BTE are ultraviolet (UV) and oxidation, thereby providing a versa-
calcium phosphates, the properties of which can be tai- tility for its sterilization.33 Silk is also highly processable,
lored by their composition.23 and has been used to fabricate sponges, films, hydrogels
Ultimately the bone grafts should provide specific and mats.34,35 Many of its characteristics can be adapted,
characteristics: osteointegration, osteoinduction and such as its mechanical strength and degradation rate,
6 The International Journal of Artificial Organs 00(0)

controlled via its crystallinity.36–38 As part of the silkworm Synthetic polymers


cocoon, silk has a tensile strength of 27.6 MPa;39 however,
in its regenerated form, this can drop down to 3.45 MPa.40 Synthetic polymers are more predictable and reproducible
The ability to domesticate silkworms means that silk is in their mechanical and physical properties as they are syn-
widely available. The silkworms must be treated under a thesized under controlled conditions. As they are synthetic,
controlled environment as the properties of silk can be they do not pose any immunological risks. Aliphatic poly-
drastically influenced by the silkworms’ diet. The structure esters are the most commonly used synthetic polymers for
of silk fibres spun by the silkworm includes an outer coat BTE and include poly(ε-caprolactone) (PCL), polylactide
of a protein called sericin, which acts as a glue to hold the (PLA), polyglycolic acid (PGA) and their copolymers.49
silk fibres together when spinning the silkworm cocoon. Polyesters degrade through the hydrolysis of ester groups
Sericin is generally considered to be a negative component along their molecular backbone. By controlling the poly-
of silk fibres as it triggers immunological responses mer molecular weight, crystallinity, molecular side groups
(although with some conflicting reports)41,42 and therefore as well as by their blending or co-polymerization, the
it must be removed in a process called ‘degumming’, a required mechanical and degradative rates are adapted.
process well established in the textile industry.34,35 The degradative products of polyesters cause a reduction
The polysaccharide chitosan is a good candidate in the in pH, which if left to accumulate in the surrounding envi-
field of biomaterials due to its highly antibacterial/anti- ronment can have cytotoxic effects, increase the inflam-
fungal, biocompatible and biodegradable properties. matory reaction, as well as increase the degradation rate.25
While it is not naturally formed, it is extracted from the PCL is a semi-crystalline, biodegradable and non-toxic
natural polymer chitin, commonly found in invertebrates, polyesters that can be used for the fabrication of scaffolds
and it is therefore abundantly available and easy to source. for BTE. It is highly processable, as it has a low melting
Mushrooms can also be a source of chitin and offer a con- point (55°C–60°C) and glass transition temperature
sistent source all year round, avoiding inconsistent physi- (60°C), is soluble in a wide range of organic solvents.50
ochemical characteristics. Biodegradation rate of chitosan PCL is a ductile polymer with a high elongation to break
correlates to the deacetylation degree (DD)43 which is (>700%).50 Its main disadvantages are its hydrophobicity
related to its crystallinity. DD can be controlled by alka- and its slow degradation, as PCL can remain in situ for up
line/acid conditions as well as by chitin deacetylases. to 24 months.51 These problems can be addressed by blend-
Crystallinity reaches a maximum when DD is 0% or 100% ing with different polymers or producing a composite
and decreases with intermediate DD. As polymer crystal- material. It was reported that PCL/calcium silicate com-
linity is inversely related to degradation kinetics, when posites, coated with decellularized ECM reduced the
the DD of chitosan decreases to an intermediate point, hydrophobic nature of PCL and provided excellent bio-
there is an in increase in the rate of biodegradation. compatibility. The incorporation of calcium silicate and
Chitosan and chitin are mechanically weak and are there- ECM effectively enhanced cellular adhesion, proliferation
fore usually blended with other more mechanically stable and differentiation in both in vitro and in vivo.52
polymers.44,45 PLA is highly biocompatible and can be made from
Alginate is a natural polysaccharide sourced from renewable sources such as through the fermentation of
brown seaweed. It has a good biocompatibility and low sugars.53 PLAs are composed of a chiral molecule that
toxicity when processed to a high purity. When high- exists as two enantiomers: l- and d-lactic acid, and has
purity alginate is implanted, it causes only a minimal several stereoisomers such as poly(l-lactide) (PLLA) and
foreign body reaction.46 Alginate can be gelated into a poly(d-lactide) (PDLA). It generally has a slow degrada-
three-dimensional (3D) structure, most commonly via tion rate (3–5 years).53 Porous PLA scaffold showed excel-
ionic crosslinking using divalent cations.47 Although lent biocompatibility with homogeneous distribution of
alginate is non-degradable in mammals as they do not cells in vitro.54 PLA scaffolds with a pore size of 750 µm
possess the correct enzyme (alginase), ionically has been demonstrated to enhance osteoblast proliferation
crosslinked gels can break down by the release of the and metabolic activity.55 PLA can be combined with differ-
divalent cation. Increasing the molecular weight of algi- ent calcium phosphates to improve the hydrophilic nature
nate will improve its mechanical properties; however, and osteoblast activity.56 Combining PLA with HA has
this will increase its gelatinous viscosity, which can also been shown to improve its mechanical properties,
cause problems with its processing. An alternative such as its flexural strength.57
method to controlling alginate mechanical properties is
via its level of methacrylation in photocrosslinkable alg-
inate. By increasing the level of methacrylation from
Bioceramics
~7% to ~25%, the elastic modulus can be increased from Bioceramics have long been applied as bone grafting
34.29 ± 9.22 kPa to 174.77 ± 14.88 kPa, as well as materials due to their promising biocompatibility, bioac-
reduce its degradation rate.48 tivity, osteoconductivity and mechanical strength.
Perić Kačarević et al. 7

Bioceramics enhance the osteo-potential of scaffolds and Metallic scaffolds


promote new bone formation.58 The most commonly used
bioceramics include calcium phosphates, calcium sul- Although polymer and bioceramics scaffolds have
phates and bioactive glasses (BGs). The most extensively received the most attention as bone tissue regeneration
used bioceramics are calcium phosphates, due to their materials, they suffer from poor mechanical properties,
similarity to the mineral phase of bone, HA. The majority either in strength or ductility. Certain metals have demon-
of calcium phosphates used are HA, tricalcium phosphates strated excellent biocompatibility, also known as biomet-
(β-TCP/α-TCP) and biphasic calcium phosphates (BCPs). als and are well established as a bone replacement
HA is the main mineral component of natural bone material. Metals have a long history of use as for the
repair of bones, such as stainless steel, titanium-alloy,
which represents approximately 60% of the weight of the
cobalt–chromium-based, aluminium, lead and silver.
bone tissue.59 Although HA is already found within the
These materials were used because of their excellent
bone structure, it has very slow resorption and remodelling
mechanical strength and fatigue resistance and ability to
rates, meaning that its properties might not be ideal for all
be rehabilitate load-bearing bone defects. However, these
BTE applications. Due to its long remodelling and resorp-
metals are non-degradable, and either require a secondary
tion times, nano-hydroxyapatite (nHA) has also shown to
surgery for their removal, or remain in situ for the lifespan
offer some of the best properties for BTE, as nHA more
of the metallic implant.
closely resembles the mineral size and structure found in
There are several biometals that are also biodegradable,
natural bone and is therefore more easily incorporated into
such as magnesium (Mg) and iron (Fe) that are gaining
a regenerating bone structure.60 As HA lacks osteoinduc-
increasing interest for bone tissue scaffolds due to their
tivity, using an adsorption method, Zhou et al. incorpo- similar mechanical properties to bone.68 The release of
rated BMP-2 onto the surface of the nHA. Their method metallic ions could also be beneficial for the regeneration
provided a sustained release of growth factor over a 35-day of bone as many metallic ions are important for the proper
period, enhanced the expression of osteogenic markers and function of the body.69
showed new ectopic bone formation in mice.61 Fe has excellent mechanical properties (similar to alu-
β-TCP/α-TCP offers faster resorption times than HA. minium), however, has a very slow rate of degradation in
α-TCP has a much higher biocompatibility than β-TCP, vivo that can cause it to induce a similar tissue reaction to
however, is also more soluble, hence degrades faster in that of a permanent scaffold.70 In contrast, Mg has more
vivo.62 β-TCP, although demonstrating a lower biocom- ductile mechanical properties, similar to bone, but has a
patibility to α-TCP, has a more stable degradation rate and fast degradation rate.70 Alloying each metal with ions such
can be used as an alternative to HA.63 TCP can be com- as strontium or calcium can be useful for adapting the
bined with HA to form BCP to provide the ideal resorption mechanical and corrosive properties, as well as be benefi-
rates and biocompatibility. Many studies have shown that cial to bone regeneration.69,71 Open porous degradable
a ratio of 60:40 HA to TCP will provide the best ratio for magnesium scaffolds fabricated with pore sizes of 250 and
use in BTE.23 400 µm have demonstrated mechanical properties within
BG is a silicate-based glass that elicits biological the range of human cancellous bone. In vivo experiments
response upon contact with bodily fluids, forming bonds demonstrated a high degree of vascularization and bone
with both hard and soft tissues via the formation of amor- formation.72
phous calcium phosphates or crystalline HA on the surface
of the glass.64 The first and most famous BG composition
belongs to Hench,65 registered as 45S5 Bioglass®. 45S5 BG Fabrication techniques
is composed of 45% SiO2 and 55% of sodium, calcium and Fabricating materials with satisfactory mechanical and
phosphorous oxides. The inclusion of the phosphorous bioactive properties are one of the great challenges in
oxides on the surface of the glass is highly reactive and attempting to design bone or cartilage scaffolds, as the
results in strong bonding with host tissue. A main advantage implanted structure should be equivalent to the anatomical
of BG is its controllable degradation rate that can be matched site. Many materials with good mechanical properties have
with the regular rate of remodelling of native bone.66 During shown great potential in vitro, but failed when implanted
degradation, released Na, Ca, Si and P ions increase the in vivo due to an imbalance between the mechanical prop-
induction of osteogenesis and angiogenesis. Some of the erties and porosity of the structure, as well as by its vascu-
additional elements like fluorine, magnesium, strontium and larization,22 all properties influenced by the fabrication
zinc can reinforce the function of BG.67 However, the major process.
downfall of BG is the low mechanical competence in BTE
due to its brittleness. An extensive review of mechanical
properties on the different compositions of BG was done by
Conventional methods
Fu et al.,66 with some compositions reaching the compres- Solvent casting and particulate leaching.  In solvent casting
sive strength range of cortical bone.59 and particulate leaching (SCPL), a solvent is used to
8 The International Journal of Artificial Organs 00(0)

Figure 5.  Depiction of freeze-drying. An emulsion of polymer


solution and water is prepared, frozen and vacuum dried to
create porous structures.

compromise the porosity. Moreover, it is limited to a thick-


ness between 3 and 4 mm to ensure efficient leaching, hin-
dering its use in defects with thicker bone.
Figure 4.  Depiction of solvent casting and particulate leaching.
Particulates are dispersed in the polymer solution, solvent
evaporates, and the particulates leach out, eroding the polymer Freeze-drying.  In freeze-drying, or lyophilization, an emul-
and creating porous structures. sion of polymer solution and water is cooled down below
its freezing temperature and subjected to vacuum drying
that evaporates the water and solvent via sublimation. As
dissolve a polymer with salt/polymer particles. After the the water leaves the frozen polymer it creates intercon-
solvent evaporates, the matrix of polymer and salt/polymer nected porosities.3 The freeze-drying process is depicted in
particles is submerged in water, causing the particulates to Figure 5. Freeze-drying is a type of thermally induced
leach out; creating a porous structure.73 This process is phase separation (TIPS) process, and is discussed further
depicted in Figure 4. SCPL is a simple cost-effective pro- in section ‘TIPS’.
cedure, which has controllability over porosity and pore This technique does not require high temperatures,
size. Achievable porosity and pore sizes are 20%–50% and which allows for the incorporation of natural polymers and
30–300 µm, respectively.74 Disadvantages of this tech- biomolecules. Problems with the technique include the
nique are cytotoxic residual of the solvent and/or particu- potential for solvent residuals to remain within the struc-
late and low controllability over spatial geometry, as well ture as well as an irregular pore size. Achievable porosity
as low-pore interconnectivity. and pore size are reported to be >90% and 20–400 µm.78
Cao and Kuboyama75 fabricated composite scaffolds Li et al.79 freeze-dried blends of poly(lactide-co-gly-
made of PGA/β-TCP with different weight ratios (1:1 and colide) (PLGA), PCL and HA. The scaffolds had a poros-
1:3 PGA:β-TCP) via SCPL. The solution was mixed with ity of 75.74% ± 1.21% and a pore size of 179.07 ± 0.75
NaCl particles, with a particle size of 425–500 µm. µm. Compressive strength was 47 ± 2 MPa. The scaffolds
Porosities of the 1:1 and 1:3 scaffolds were 93.6% ± promoted human MSCs to differentiate into osteoblasts
2.04% and 88.4% ± 0.7%, respectively. The scaffolds after 21 days in culture. For in vivo assessment, the scaf-
were implanted and assessed in femoral defects in rats folds were implanted into rabbit calvarial defects. After 12
over 3 months. New bone formation was the highest in 1:3 weeks, the PLGA/PCL/HA scaffolds exhibited higher vol-
composite scaffolds, compared to 1:1 scaffold. After 90 umes of new bone ingrowth compared to PLGA/HA, PCL/
days, the wound site had healed by >96% and all the HA and a negative control. The PLGA/PCL/HA scaffolds
PGA/β-TCP scaffolds had degraded. A higher content of were sufficiently degradable, biocompatible and induced
β-TCP improved the osteoconductivity of the scaffolds. bone tissue regeneration.
Instead of mixing the porogen with the polymer in a Due to the fact that this technique does not require high
solution composed of a polymer and an organic solvent, temperatures during fabrication, natural polymers can be
the polymers can be melted and mixed with the porogen. used80 as well as biomolecules.81 Research scholars have
PLA was combined with demineralized bone particles successfully combined the mechanical properties and pro-
(DBPs) in SCPL.76 The scaffolds proved to be osteoinduc- cessability of silk with the osteogenic capacity of HA to
tive; however, an increase in the DBP decreased porosity fabricate scaffolds via freeze-drying. By combining silk
of the resultant scaffolds. In SCPL, bioglass powders have and HA resulted in scaffolds that have mechanical proper-
been mixed with the base polymers.77 The addition of bio- ties with similar load-bearing properties to bone, while also
glass enhanced hydrophilicity, degradability and bioactiv- mimicking its native ECM. Sangkert et al.80 fabricated
ity; however, increased content of bioglass decreased scaffolds with silk fibroin via freeze-drying, however
porosity. incorporated different biological components to improve
SCPL has proved to be a successful technique in fabri- its biological response. The scaffolds were modified with
cating BTE scaffolds with many different bone-inductive collagen, decellularized pulp and fibronectin coating. The
additives;73 however, addition of these particles could silk provided a mechanical support structure, while the
Perić Kačarević et al. 9

collagen improved cellular adhesion and proliferation. The


human decellularized tooth pulp was used to stimulate a
higher biofunctionality of the scaffolds, while the fibronec-
tin induced mineralization. The coating provided the sam-
ples with an osteoinductive bioactivity; exhibiting a higher
alkaline phosphatase (ALP) expression after 21 days in cul-
ture with MG-63 osteoblasts when compared to non-modi- Figure 6.  Depiction of gas foaming. Polymer discs are put
fied samples. under a high-pressure inert gas, creating a foam. A drop of
Using the freeze-drying method, it is possible to create pressure will create a phase separation, resulting in a porous
scaffolds that gradually release bioactive compounds. structure.
Menon et al.81 fabricated bone tissue scaffolds from cellu-
lose, chitosan, nHA and chrysin via freeze-drying. Chrysin on the scaffolds for in vitro assessment. After 14 days, the
is a flavonoid that promotes osteogenic growth.82 The scaf- modified scaffolds exhibited a higher proliferation, cell
folds were seeded with mouse MSCs and incubated for 7 number and osteogenic gene expressions than pure PLLA
days. The sustained release of chrysin upregulated the scaffolds. The scaffolds were implanted in mouse calvarial
expression of osteogenic differentiation marker genes. critical-size defects. Groups with modified PLLA scaf-
Freeze-drying has great potential in BTE with the pos- folds exhibited 90% regeneration of the defects, after 12
sibility of using natural biomaterials and biomolecules. days.
Despite its promising outcomes in tissue engineering and TIPS appear to be a suitable technique for the fabrica-
drug delivery, this technique is time-consuming and has tion of BTE, a wide span of pore sizes and morphologies
high energy-consumption, as well as providing only a lim- are achievable. Due to the relatively higher temperatures,
ited control of scaffold architecture. however, natural biomaterials and biomolecules cannot be
incorporated during the fabrication process but must be
TIPS.  In TIPS, a homogeneous polymer solution is sepa- added in a post-fabrication modification. It has also been
rated into polymer and solvent phases by the change of observed that the incorporation of bone substitutes during
thermal energies.83 This liquid–liquid phase separation pro- the fabrication process decreased porosities and resulted in
cess results in polymer-rich and solvent-rich phases. After smaller irregular pore sizes.
using thermal energy to separate the solution, freeze-drying
is performed to evaporate the solvent, resulting in a Gas foaming.  Polymer discs are subjected to high-pressure
microporous structure. Although this technique can pro- inert gases like CO2, N2 or He, to saturate and create foams
duce large volumes of pores with controllable size and of the polymer and gas.86 A drop of pressure to ambient
morphology, it requires the use of polymers with low melt- levels will cause the gas phase to separate, instigating the
ing temperatures and organic solvents. Achievable porosity nucleation of bubbles. The gas then diffuses through the
and pore sizes are <97% and <200 µm, respectively.74 polymer, resulting in the growth of pores. Figure 6 demon-
Carfi Pavia et al.84 fabricated PLLA/HA scaffolds with strates the gas foaming technique. Alternative chemical-
ratios of 95:5, 90:10, 70:30, 50:50 and 34:66. HA was sus- based gas foaming uses foaming agents, polymer and
pended in PLLA solution and the scaffolds were fabricated binding agents. The semi-solidified mixture is then sub-
via TIPS and freeze-dried. Porosity decreased with an merged in chemicals that react with the foaming agents,
increase in the HA content, although the results were statis- creating gas bubbles that diffuse through the polymer and
tically insignificant, and all ratios produced porosities have an erosion effect that creates a porous structure. This
>90%. Compressive moduli increased with the increase in technique is fast and simple; however, the resulting porous
the HA content until a concentration of 30% HA was structure contains closed pores that lack interconnectivity.
reached. It is thought that higher concentrations caused a The interconnectivity can be improved through combining
less homogeneous distribution of HA, resulting in a decrease the process with salt/polymer leaching or freeze-drying.3
of the modulus. MC3T3-E1 preosteoblasts were seeded on Complex structures require negative scaffolds as moulds,
PLLA/HA (95:5) and neat PLLA scaffolds. After 21 days, which is not cost-effective. Achievable porosity and pore
the PLLA/HA scaffolds exhibited higher ALP activity. sizes of >93% and 40–800 µm have been reported with
Chen et al.85 fabricated PLLA scaffolds via TIPS. The this technique.3,87
scaffolds had a porosity of >94%, pore size of 300 µm and Duarte et al.88 fabricated PCL scaffolds loaded with β-
compressive strength of 1 MPa. To improve the cellular TCP and dexamethasone. A concentration of 10 wt% β-
attachment and degradation kinetics of PLLA, the scaf- TCP was and 5 or 10 wt% dexamethasone was used. The
folds were modified with chitosan coating to increase their fabrication was carried out with subcritical CO2 that
hydrophilicity. The TIPS-fabricated scaffolds were sub- resulted in milder processing conditions. Resultant poros-
merged in high concentration of chitosan and freeze-dried. ity ranged between 73% and 99% and pore sizes between
Mouse bone marrow stromal cells (mBMSCs) were seeded 164 and 882 µm. Young’s modulus ranged between 1.76
10 The International Journal of Artificial Organs 00(0)

and capillary tube, temperature and humidity. Due to the


latter two, the process should happen under a well-con-
trolled atmosphere. Electrospinning produces structures
with a high surface area that mimic natural tissue ECM
and has the potential of incorporating natural biomaterials/
biomolecules as it is a low-temperature process. Although
this technique is versatile, simple and inexpensive, the
resulted structures lack mechanical competence.90
Sharifi et al.91 fabricated nanofibrous scaffolds via elec-
trospinning, with fibre diameters of 356 nm. The scaffolds
were based on PCL and carboxylmethyl chitosan.
Carboxylmethylation of the amine and hydroxyl groups of
chitosan enhances its solubility in water and surface hydro-
philicity. Scaffolds were seeded with MG-63 osteoblasts
and after 3 days, the PCL/carboxylmethyl chitosan exhib-
ited higher proliferation rate compared to the PCL and
Figure 7.  Depiction of electrospinning. Polymer is fed to the
PCL/chitosan scaffolds.
capillary tube and a high voltage is used to introduce a polarity
that accelerates filaments of the polymer from the syringe tip Aragón et al. incorporated BMP-2-loaded PLGA parti-
towards the collector. cles with electrospun PCL/HA fibres via electrospraying.
The fibres had PCL shells and PCL/HA cores, with an
overall fibre diameter of 380 ± 108 nm. Core-shell nanofi-
and 2.92 MPa, decreasing with increasing β-TCP content. bres can be fabricated using coaxial electrospinning, a
The addition of the osteoinductive and osteoconductive developed method to created coated fibres.92 BMP-2 con-
additives did not affect internal morphology of the scaf- tent ranged between 1.2 and 1.7 µg/g and HA nanoparti-
folds and the dexamethasone provided a sustained release cles between 8.8 and 12.6 wt%. The scaffolds loaded with
over 35 days. PLGA/BMP-2 particles exhibited higher ALP, OCN and
Catanzano et al.89 fabricated porous scaffolds via inter- osteopontin expression compared to PCL/HA. Suspending
nal gelation, followed by gas foaming and freeze-drying. the HA nanoparticles in the polymer solution did not affect
Alginate was crosslinked with a mixture of strontium (Sr) the diameter of the fibres.
and calcium (Ca) ions. Sr2+ and Ca2+ have osteogenic Cheng et al.93 employed coaxial electrospinning to fab-
benefits.69 Scaffold porosities were around 50.74 ± 4.57 ricate platelet-rich plasma (PRP)-filled PVA nanofibres.
and had a pore size between 100 and 400 µm. Calcium Fibres had a diameter of 471.6 ± 210.8 nm. The fibres
ions were released quicker than the strontium ions. The exhibited a sustainable release of the PRP over a 30-day
scaffolds were seeded with human MSCs and after 7 days, period. In vitro, the growth factors from the PRP enhanced
the scaffolds demonstrated a high level of cellular attach- proliferation and induced mineralization. Chitosan/β-TCP
ment. Osteocalcin (OCN) expression was significantly scaffolds were seeded with MSCs and incubated for 1
higher with scaffolds crosslinked with Sr2+ and Ca2+ after week. After that, the scaffold was covered with electro-
21 days, compared to cells incubated on tissue culture spun PVA/PRP nanofibres and implanted subcutaneously
plates. in nude mice. After 30 days, the scaffolds exhibited a
higher collagen II expression and accumulated more cells
Electrospinning. Electrospinning creates fine fibres using when compared to a sham surgery.
electrostatic forces that accumulate in a mat on a collection Ao et al.94 fabricated nanofibres from cellulose and HA.
plate. The precursor material is either a polymer solution HA nanoparticles were incorporated with the polymer
or a molten polymer. Thicknesses of the fibres can range solution at different weight percentages: 3, 5 and 10 wt%.
from micrometres to nanometres. An electrospinning pro- Diameter of fibres increased with increasing HA content.
cess requires three essential components: (1) high voltage, For the highest HA content (10 wt%), the fibre diameters
(2) feeding capillary tube and (3) metal collector. The pol- ranged from 100 to 500 µm. Neat cellulose nanofibre
ymer liquid is fed to the capillary tube and a high voltage diameters ranged between 100 and 200 µm. The tensile
introduces a polarity that accelerates strings of the polymer strength and modulus reached a maximum at 5 wt% HA
from the tubes towards the collector. The continuous jet of with 70.6 MPa and 3.12 GPa, respectively. However, at 10
polymer is received on the collector that is either neutral or wt% HA, the mechanical properties deteriorated as a result
has the opposite polarity of the polymer. The process of of increased agglomerates. After 3 days seeded with
electrospinning is depicted in Figure 7. The process is human dental follicle cells, all scaffolds did not exhibit any
affected by parameters such as polymer type, solution con- cytotoxicity and increasing HA content increased cell via-
centration, polymer feed rate, distance between collector bility and proliferation rates.
Perić Kačarević et al. 11

Figure 8.  A flowchart of the different additive manufacturing techniques employed in tissue engineering indicating which methods
are suitable for cell printing.
SLA: stereolithography; LAB: laser-assisted bioprinting; SLS: selective laser sintering.

Electrospinning has great possibilities in BTE.95 The Although such high temperatures could degrade natural
fibrous structures produced closely mimic the fibrous polymers or biological molecules, studies have shown that
structure of natural tissue ECM, where collagen fibres during the duration of heating, the heat is very localized
have diameters ranging from 50 to 500 nm. The ability to and does not affect the quality or integrity of the biological
incorporate bioceramics particles at low concentrations materials.16 Piezoelectric printers use acoustic waves to
within the electrospun fibres improves scaffold mechani- print. The process enables a larger variety of materials to
cal properties as well as its bioactivity, however, can affect be printed compared with thermal inkjet printing, how-
fibre diameters and uniformity. Coaxial electrospinning ever, is limited by the viscosity of the ink, as high viscosi-
and electrospraying have shown great success in fabricat- ties will dampen out the acoustic waves before a droplet
ing bone tissue scaffolds that sustain growth factor deliv- can be ejected. The main advantages of inkjet printing
ery, enhancing the bone regeneration process as whole. include its high speed (up to 10,000 droplets per second)
and high cell viability (>90%). The main disadvantage of
inkjet printing is the low concentration of the inks, which
Additive manufacturing can influence the integrity of the printed structure, as well
Additive manufacturing (AM), or 3D printing, is a fabrica- as increase fabrication times.
tion process that builds structures from the bottom-up.16 It Ge et al.96 fabricated PLGA scaffolds using a binder of
has been successfully employed in regenerative medicine, ethanol and acetone that had a pore size of 1 mm, a poros-
especially in the fields of tissue engineering and drug ity of 50%, a compressive strength of 7.8 ± 3.1 MPa, and
delivery/screening. AM techniques fabricate structures or a Young’s modulus of 77.2 ± 10.8 MPa. The scaffolds
patterns in a pre-defined fashion via computer-aided were compared in vitro to a commercially available open
designs (CADs). The CADs can be geometrically tailored pore poly-lactic acid scaffold (OPLA®, Becton-Dickinson
for individual patient specifications and have macro- and (BD) Inc., Murray, UT, USA) and a collagen scaffold
microarchitecture that corresponds to the anatomical prop- (Becton-Dickinson (BD) Inc.). The mechanical properties
erties of the to-be-replaced tissue. Figure 8 provides a of the printed scaffold were 40 times higher than the
flowchart of the most common 3D printing techniques OPLA® scaffold and 1800 times higher than the BD col-
used in tissue engineering. lagen scaffold. Cell viability of human foetal osteoblasts
seeded onto the scaffold was 95% after 24 h. The results
Inkjet printing. Inkjet printing is a low-cost, simple and demonstrated that the 3D-printed PLGA scaffolds support
non-contact fabrication technique. Inkjet printing builds proliferation and differentiation of osteoblasts, compara-
up structures using droplets as individual building blocks. ble to commercially available scaffolds.
The ejection of each droplet is driven either by piezoelec-
tric or thermal–based processes. In thermal inkjet printing, Extrusion printing.  Extrusion bioprinting is the most com-
an air pocket is nucleated within the printhead via a ther- monly used printing technique for the extrusion of poly-
mal element, which causes a pressure increase within the mers or hydrogels through a micro-nozzle. Extrusion is
printhead leading to droplet ejection. The heating element controlled via pressure-based mechanisms, such as via a
can reach temperatures ranging between 100°C and 300°C. piston- or pneumatic-based.16 The main advantage of
12 The International Journal of Artificial Organs 00(0)

extrusion bioprinting is its fast fabrication times, however downwards after each subsequent layer. This technique
its resolution of around of 200 μm is considerably lower offers a limited range of materials, as the materials are
compared to other AM techniques. Pneumatic systems required to be photosensitive. To improve photosensitivity,
generally use compressed gas, which work better for the materials can be mixed with photoinitiators (PIs). PIs
highly viscous molten polymers but lack the same preci- are chemical compounds that form free radicals upon
sion as piston-driven mechanisms that offer more direct exposure to light energy, UV or visible.97 These radicals
control over the flow of the ink from the nozzle.97 Overall, initiate a chain polymer reaction between the monomers of
this technique provides excellent structural integrity due to the material, hardening the liquid resin into predesigned
the continuous deposition and high loading of the ink. structures. The type of PI and concentration are essential in
Boga et al.98 demonstrated bone regeneration in vitro determining the stiffness of the structure, fabrication time
with different ratios of TCP and alginic acid (AA) (60:40, as well as the cellular reaction with the 3D structure.101,102
70:30, 80:20). AA was crosslinked by immersing the scaf- For UV-based SLA, the most common and suitable PIs
folds in a solution of CaCl2. The printed scaffolds had a used are Irgacure 2959 and Irgacure 184 that polymerize at
high mechanical strength and elasticity and also shown wavelengths 257–276 nm and 246, 288 or 333 nm, respec-
bone-like structure. High loading of the scaffold with TCP tively.101 For visible light-based SLA, eosin Y and LAP PIs
deteriorated the mechanical properties. To improve the are the most commonly used, photocured at 514 and 405
mechanical properties further, graphene oxide was incor- nm, respectively.103,104
porated into the structure. By including a 0.5% (w/w) con- Visible light-based SLA was developed in order to ena-
centration of graphene oxide, Young’s modulus of scaffolds ble the incorporation of cells during fabrication and has
with a 60:40 TCP:AA ratio increased from 154 ± 8.7 MPa shown advantageous results over UV; as it rules out
to 188.3 ± 18.5 MPa. mutagenesis of cells, uses less cytotoxic PIs and produces
To test the effect of scaffold architecture on the mechan- higher mechanical integrity of the prints. This technique
ical properties of bone tissue scaffolds, Roohani-Esfahani can produce scaffolds in a short time, regardless of their
et al.99 fabricated BG ceramics, Sr-HT Gahnite scaffolds complexity as the fabrication time is instead dependent
with hexagonal, rectangular, curved and zig-zag architec- upon the height of the design. The main disadvantage of
tures. Scaffold porosities tested were: 50%, 55%, 60% and this technique is the limited choice of materials and bio-
70%. The scaffolds with hexagonal patterns had the high- compatibility issues caused by the PIs.
est compressive strengths of 122 ± 12 MPa, which is in Lan et al.105 fabricated BTE scaffolds from
the range of human cortical bone (100–150 MPa). The poly(propylene fumarate) (PPF) via SLA. PPF was mixed
compressive strength from 90 MPa at 70% porosity to 180 with diethyl fumarate (DEF) and 1 wt% bis-acyl phos-
MPa at 50% porosity, the result shows the application of phine oxide (BAPO) to prepare a photocurable resin. The
Sr-HT Gahnite–based scaffolds for load-bearing bone fabricated scaffolds had a lattice design with 65% porosity
defects. and 250 µm pore size. The scaffolds were further modified
Vozzi et al.100 demonstrated the fabrication of PLLA post-fabrication to enhance their osteo-potential. The scaf-
combined with multiwalled carbon nanotubes (MWCNTs) folds were coated subsequently with HA and arginine–
to enhance the mechanical properties. The scaffold was glycine–aspartic acid (RGD). These coatings improve
fabricated with an octagonal architecture and a porosity spreading, proliferation and osteogenic differentiation.
between 65% and 70%. In comparison to pure PLLA scaf- The coatings did not affect the bulk structure of the scaf-
folds, the inclusion of MWCNTs improved cell viability folds. After 14 days of seeding MC3T3-E1 preosteoblasts,
when seeded with human foetal osteoblasts. The inclusion the scaffolds with both RGD and HA coatings and only HA
of a small concentration (6.25 mg/mL) of MWCNTs coating had the same cell number.
within the ink, increased the elastic modulus and osteo- Lee et al.106 incorporated BMP-2-loaded PLGA spheres
blastic proliferation rates. with PPF/DEF/BAPO liquid resin to fabricate osteoinduc-
Altogether, extrusion printing is considered to have tive scaffolds and compared them to conventionally fabri-
great potential as it allows the fabrication of complex cated scaffolds. The scaffolds had 69.6% porosity. The
structures at clinically relevant sizes. It provides versatility 3D-printed scaffolds sustained the release of BMP-2 for 4
in the printing of multiple biomaterials and biomolecules. weeks, while conventional scaffolds only lasted up to 14
Of the different fabrication techniques, it has the capability days. In vitro MC3 T3-E1 proliferation was higher on SLA
of printing scaffolds with the most physiologically rele- scaffolds compared to conventional scaffolds, after 14
vant mechanical properties. days. Most importantly, the SLA scaffolds had signifi-
cantly higher expression of ALP, OCN and COL-I. The
Stereolithography. Stereolithography (SLA) is an additive SLA scaffolds were implanted in rat cranial defects and
technique based on the photopolymerization of layers in after 11 weeks, had developed new bone that made up
vat of photosensitive resin.16 Crosslinking occurs in a 90.90% ± 5.09% volume compared to 32.85% ± 8.27%
layer-by-layer fashion on a platform that moves in sham surgeries.
Perić Kačarević et al. 13

Ronca et al.107 prepared a photocurable resin from syn- Xia et al.113 fabricated PCL/HA scaffolds using an SLS
thesized dimethacrylate poly(d,l-lactic acid) (PDLLA) apparatus. The scaffolds had porosity of 70.31% and pore size
and 4 wt% of Lucirin 2,4,6-Trimethylbenzoyldi- between 600 and 800 µm. At 15 wt% of HA, compressive
Phenylphosphinate (TPO-L) (an PI). HA particles (<200 strength was 3.17 MPa while pure PCL scaffolds had a com-
µm) were dispersed in the liquid resin at 10 and 20 wt%. pressive strength of only 1.38 MPa. The PCL/HA and PCL
The scaffolds had porosities ~80% and pore sizes ~745 µm scaffolds were seeded with hBMSCs for 21 days. Attachment,
and demonstrated a high degree of similarity to the CAD. proliferation, mineralization and ALP expression increased
The incorporation of HA increased the mechanical strength with increasing HA content. For in vivo assessment, the PCL
from 0.61 ± 0.2 MPa to 1.23 ± 0.2 MPa for neat PDLLA and PCL/HA scaffolds were implanted in rabbit femur
scaffolds to PDLLA/20 wt% HA scaffolds, respectively. In defects. After 9 weeks, PCL with 15 wt% HA has fully
vitro assessment was carried out by seeding human bone degraded and replaced by new tissue, while defects implanted
marrow–derived stromal cells (hBMSCs) on scaffolds with pure PCL scaffolds still had remnants of the implanted
without HA, scaffolds with 10% HA and scaffolds with 20 graft. MGCs were present at the surface of the implants,
wt% HA. Scaffolds with 10 wt% HA exhibited the highest which were associated to the remodelling process at the site.
proliferation rate and osseous differentiation while a 20 Liao et al.114 fabricated PCL and PCL/β-TCP scaffolds
wt% HA content hindered the migration and attachment of by SLS. The β-TCP content was 30 wt%, increasing the
the BMSCs. compressive strength from 6.77 ± 0.19 MPa to 13.66 ±
SLA has a great potential for the fabrication of tissue 0.19 MPa. The porosity of the scaffolds was between 75%
engineering scaffolds; however, the mechanical properties and 77% and had a pore size between 300 and 500 µm. The
of the resulted structures might not be adequate for BTE scaffolds were coated with collagen I. The coating did not
applications, more specifically at load-bearing sites. This impact the structure nor the mechanical strength. In vitro,
technique can be combined with other AM techniques to human adipose-derived stem cells (hASCs) expressed
create biphasic structures. A rigid phase could provide the highest ALP, OCN and mineralization on collagen-coated
mechanical strength to support the structure, and a soft PCL/β-TCP scaffolds after 28 days. hASCs were seeded
phase that could facilitate vascularization and cellular on PCL/β-TCP/COL-I and PCL scaffolds for 21 days and
recruitment.108 subsequently implanted the gluteal muscle pockets in nude
mice. The PCL/β-TCP/COL-I formed woven bone con-
Selective laser sintering.  Selective laser sintering (SLS) is a taining a vascular structure and PCL scaffolds formed a
process where compacted powder setting on a piston plat- fibrous granulated tissue, all after 4 months.
form is exposed to a laser beam that melts the powder into SLS can produce scaffolds with architectures and
a pre-defined shape.16 Printing is performed in a layer-by- mechanical strengths that are relevant to cortical bone.
layer sequence, whereby for every layer; the powder is sin- However, SLS is limited by the choice of materials, and
tered, the piston platform moves downwards and a via this technique, drug delivery methods are not possible
cylindrical roller replenishes powder into the powder bed. due to the high thermal energies involved.16 With complex
Morphology and the size of the powder are crucial param- structures, powder could be entrapped within the structure
eters in this process and impact flowability of the powder which is problematic as well.
into the powder bed, as well as the quality of the print-
ing.109 Powder bed is usually heated just below the melting Bioprinting.  Bioprinting is the process of creating 3D struc-
point of the material, speeding up the process. However, tures from cell-laden precursor materials (bioinks). Bio-
SLS suffers from a limited choice of materials and warp- printing is commonly carried out by one or a combination
ing of the structure. Biomolecules and natural polymers of the four hydrogel-based printing techniques, which are
cannot be employed in this process as the process requires SLA, thermal or piezoelectric inkjet, mechanical or pneu-
high thermal energies. matic pressure-based extrusion and laser-assisted bioprint-
Feng et al.110 reinforced β-TCP-based SLS- ing (LAB).115,116 Bioprinting is advantageous over the
manufactured scaffolds with zinc oxide (ZnO). ZnO has manual seeding of cells in terms of cell viability and
shown to encourage angiogenesis and osteogensis,111 distribution.
which are both required and equally essential in BTE. The In SLA, cells are suspended in a photosensitive hydrogel
scaffolds with 2.5 wt% ZnO had porosity of 56.8% and positioned in a vat.97 However, the use of UV radiation is
compressive strength increased from 3.01 to 17.89 MPa. known to induce mutagenesis in mammalian cells, and
Increased content of ZnO slowed down the degradation therefore, visible light-based SLA has been adapted to avoid
rate and enhanced cellular response of MG-63 osteoblasts. the adverse effects of UV. Since PIs respond to certain
Deng et al.112 reinforced forsterite-based scaffolds with 20 wavelengths, visible light-sensitive PIs, which respond to
wt% nano-58S BG. Compressive strength increased from 400–700 nm wavelengths, have been used. The same
28.6 to 43.9 MPa. An increase in the BG enhanced cellular parameters of SLA apply on SLA with cell-laden hydrogels;
adhesion of MG-63 cells. however, encapsulation of the cells during the gelation
14 The International Journal of Artificial Organs 00(0)

process. A technical challenge of SLA is oxygen inhibition. mechanical competence and macro- and microarchitec-
O2 molecules can scavenge the free radicals produced by the ture is critical to produce scaffolds relevance to the
light exposure of PIs, hindering the crosslinking. This nega- nature of native bone tissue. Critical size bone defects,
tively impacts the print quality and could result in print col- as a result of trauma or disease, can be regenerated
lapse. Oxygen inhibition can be reduced by increasing light using a bone tissue scaffold. Currently, autografts are
intensity, concentration of PIs or carrying the process under the gold standard for BTE; however, extraction of auto-
inert conditions such as N2 or CO2; however, all of these grafts requires a second surgical site, hence increased
possibilities will negatively impact cell viability in case of morbidity and potential for infection.
SLA printing using bioinks. Visible light-based SLA over- Bone tissue scaffolds can provide an alternative to
comes issue as the increase in the light intensity or PI con- autologous bone grafts, and can offer highly tailorable
centration will not impact the cells as much, enhancing the characteristics. Many variables must be considered when
printing quality and integrity.117,118 deciding upon the materials and fabrication technique
Lim et al.117 demonstrated a novel SLA technique that used that is most suited for the end application of the
produced enhanced print qualities using a visible light–sen- bone tissue scaffold. The main consideration should be
sitive PI with high intensities, the PI was ruthenium (Ru)/ the positioning of the implant, as the requirements for
sodium persulfate. Immediate cell viability using this sys- load-bearing and non-load-bearing scaffolds can be sig-
tem was higher when UV-sensitive Irgacure 2959 was used, nificantly different. The aim of scaffolding material is to
which is the least toxic PI among the UV-sensitive options. preserve the existing bone and stimulate osteogenesis for
Inkjet bioprinting employs localized heat or acoustic pie- the regeneration of bone and functional restoration. For
zoelectric waves to propel bioink from a micro-nozzle.116 this purpose, it is important to consider the material to be
This process is very similar to conventional inkjet printing; used: polymeric scaffolds are more suited to non-load-
in fact, inkjet bioprinting was the first AM technique to be bearing sites, provide excellent ductility but have low
utilized for bioprinting by filling an ink cartridge of a con- strength; ceramics can maintain space for long periods of
ventional table-top printer with a bioink.115 However, this time and exhibit excellent osteoconductivity, but suffer
technique requires that cell suspensions/bioinks have a low from brittleness; biometals are ductile and strong, yet can
viscosity, and therefore a low concentration of materials and be expensive and difficult to process. For this purpose,
cells. Low viscosity also affects the quality of the print, espe- investigating novel biomaterials or combinations of bio-
cially in vertical configurations and structures with over- materials is essential for determining a suitable BTE
hangs. To overcome issues accompanied with overhanging approach.
structures, Christensen et al.119 synthesized inkjet-printed The manufacturing process used for the scaffold is also
structures in a vat of calcium chloride solution. The CaCl2 an important consideration as this influences the structural
acted as a mid-fabrication crosslinking agent and provided properties such as porosity, pore size and interconnectivity
buoyancy to support the print. Inkjet bioprinting has achieved of pores. Conventional manufacturing techniques offer an
accuracies of singular cells per droplet,120 enabling cell posi- established, low-cost and accessible method for manufac-
tioning that can be utilized in many applications in tissue turing scaffolds. They have demonstrated a high degree of
engineering such as prevascularization and innervation. success in vivo and clinically; however, the more recent
In extrusion-based bioprinting, mechanical or pneumatic development of 3D printing has led to improvements in
pressures are applied to eject continuous strand-like bioink scaffold design and repeatability. 3D printing has an
through a micro-nozzle in a predesigned layer-by-layer fash- advantage over conventional techniques as it can provide
ion.116 Mechanical-based processes are either piston- or control over spatial geometry and microarchitecture, as
screw-driven but screw-driven forces will produce deadly well as reproducibility between prints, yet usually requires
shear stresses on the cells; and therefore demonstrate the a high setup-cost. Overall, the techniques discussed in this
lowest cell viabilities of all the bioprinting techniques. review have demonstrated excellent potential for the appli-
Extrusion bioprinting offers versatile options (in terms of cation of BTE.
material choice) and enables the use of high cell-density
bioinks. Studying rheology of the printing bioink is very Author contributions
important in extrusion bioprinting as the more viscous the
S.A., S.R. and M.P. conducted a literature review to provide the
bioink, the higher the induced shear stress is during printing, information for this review article; Z.P.K., P.R. and M.B. wrote
resulting in higher cell apoptotic activity. Moreover, this the article; and R.S. and O.J. proof read the manuscript and
technique has been developed for in situ applications and helped with the final editing.
used for osteochondral repair with high success.121
Declaration of conflicting interests
Conclusion
The author(s) declared no potential conflicts of interest with
Bone regeneration is a complex and challenging appli- respect to the research, authorship and/or publication of this
cation of tissue engineering. The balance between article.
Perić Kačarević et al. 15

Ethical statement 13. Han Y, You X, Xing W, et al. Paracrine and endocrine
actions of bone – the functions of secretory proteins from
There are no animal or human experiments carried out for this
osteoblasts, osteocytes, and osteoclasts. Bone Res 2018; 6:
article.
16–11.
14. Charoenlarp P, Rajendran AK and Iseki S. Role of fibro-
Funding blast growth factors in bone regeneration. Inflamm Regen
The author(s) received no financial support for the research, 2017; 37: 10–17.
authorship and/or publication of this article. 15. Cui X, Breitenkamp K, Lotz M, et al. Synergistic action of
fibroblast growth factor-2 and transforming growth factor-
ORCID iDs beta1 enhances bioprinted human neocartilage formation.
Biotechnol Bioeng 2012; 109(9): 2357–2368.
Patrick Rider https://orcid.org/0000-0002-3620-7199
16. Rider P, Kacarevic ZP, Alkildani S, et al. Additive manu-
Mike Barbeck https://orcid.org/0000-0002-3001-1347
facturing for guided bone regeneration: a perspective for
alveolar ridge augmentation. Int J Mol Sci 2018; 19(11):
References 3308–3335.
1. Liu Y, Luo D and Wang T. Hierarchical structures of 17. Barradas A, Yuan H, van Blitterswijk CA, et al.
bone and bioinspired bone tissue engineering. Small 2016; Osteoinductive biomaterials: current knowledge of prop-
12(34): 4611–4632. erties, experimental models and biological mechanisms.
2. Piaia L, Salmoria GV and Hotza D. Additive manufac- Eur Cell Mater 2011; 21: 407–29; discussion 429.
turing of nanostructured bone scaffolds. In: Souza JCM, 18. O’Brien FJ. Biomaterials & scaffolds for tissue engineer-
Hotza D, Henriques B, et al. (eds) Nanostructured bio- ing. Mater Today 2011; 14(3): 88–95.
materials for Cranio-maxillofacial and oral applications. 19. Polo-Corrales L, Latorre-Esteves M and Ramirez-Vick
Amsterdam: Elsevier Inc, 2018, pp. 181–210. JE. Scaffold design for bone regeneration. J Nanosci
3. Wubneh A, Tsekoura E, Ayranci C, et al. Current state Nanotech; 2014; 14(1): 15–56.
of fabrication technologies and materials for bone tissue 20. Wang W and Yeung KWK. Bone grafts and biomateri-
engineering. Acta Biomater 2018; 80: 1–30. als substitutes for bone defect repair: a review. Bioact
4. Schemitsch EH. Size matters: defining critical in bone Mater 2017; 2(4): 224–247.
defect size! J Orthop Trauma 2017; 31: S20–S22. 21. Kloss FR, Offermanns V and Kloss-Brandstätter A. Com-
5. Sheikh Z, Brooks P, Barzilay O, et al. Macrophages, for- parison of allogeneic and autogenous bone grafts for aug-
eign body giant cells and their response to implantable mentation of alveolar ridge defects-A 12-month retrospective
biomaterials. Materials 2015; 8(9): 5671–5701. radiographic evaluation. Clin Oral Implants Res. Epub ahead
6. Hesketh M, Sahin KB, West ZE, et al. Macrophage pheno- of print 10 October 2018. DOI: 10.1111/clr.13380.
types regulate scar formation and chronic wound healing. 22. Schnettler R, Franke J, Rimashevskiy D, et al. Allogeneic
Int J Mol Sci 2017; 18(7): 1545–1510. bone grafting materials – update of the current scientific
7. Ghanaati S, Unger RE, Webber MJ, et al. Scaffold vascu- status. Travmatol Ortop Ross 2017; 23(4): 92–100.
larization in vivo driven by primary human osteoblasts in 23. Jelusic D, Zirk ML, Fienitz T, et al. Monophasic ß-TCP
concert with host inflammatory cells. Biomaterials 2011; vs. biphasic HA/ß-TCP in two-stage sinus floor augmenta-
32(32): 8150–8160. tion procedures – a prospective randomized clinical trial.
8. Barbeck M, Dard M, Kokkinopoulou M, et al. Small-sized Clin Oral Implants Res 2016; 28(10): e175–e183.
granules of biphasic bone substitutes support fast implant 24. Albrektsson T and Johansson C. Osteoinduction, osteo-
bed vascularization. Biomatter 2015; 5: e1056943. conduction and osseointegration. Eur Spine J 2001;
9. Jensen SS, Gruber R, Buser D, et al. Osteoclast-like cells 10(Suppl. 2): S96–S101.
on deproteinized bovine bone mineral and biphasic cal- 25. Cortizo MS and Belluzo MS. Biodegradable polymers for
cium phosphate: light and transmission electron micro- bone tissue engineering. In: Goyanes SN and D’Accorso
scopical observations. Clin Oral Implants Res 2015; NB (eds) Industrial applications of renewable biomass
26(8): 859–864. products. Cham: Springer International Publishing, 2017,
10. PericKacarevic Z, Kavehei F, Houshmand A, et al. pp. 47–74.
Purification processes of xenogeneic bone substitutes and 26. Parenteau-Bareil R, Gauvin R and Berthod F. Collagen-
their impact on tissue reactions and regeneration. Int J based biomaterials for tissue engineering applications.
Artif Organs 2018; 41(11): 789–800. Materials 2010: 3: 1863–1887.
11. Barbeck M, Unger RE, Booms P, et al. Monocyte pre- 27. Pina S, Oliveira JM and Reis RL. Natural-based nanocom-
seeding leads to an increased implant bed vascularization posites for bone tissue engineering and regenerative medi-
of biphasic calcium phosphate bone substitutes via vessel cine: a review. Adv Mater 2015; 27(7): 1143–1169.
maturation. J Biomed Mater Res A 2016; 104(12): 2928– 28. Pabst AM, Lehmann K-M, Walter C, et al. Influence of
2935. porcine-derived collagen matrix on endothelial progen-
12. Li B, Wang H, Qiu G, et al. Synergistic effects of vascu- itor cells: an in vitro study. Odontology 2014; 104(1):
lar endothelial growth factor on bone morphogenetic pro- 19–26.
teins induced bone formation in vivo: influencing factors 29. Lin S and Gu L. Influence of crosslink density and stiff-
and future research directions. Biomed Res Int 2016; 2016: ness on mechanical properties of type I collagen gel.
2869572. Materials 2015; 8(2): 551–560.
16 The International Journal of Artificial Organs 00(0)

30. Gentile P, McColgan-Bannon K, Gianone NC, et al. mechanical properties. Biomaterials 2009; 30(14): 2724–
Biosynthetic PCL-graft-collagen bulk material for tis- 2734.
sue engineering applications. Materials 2017; 10(7): 49. BaoLin G and Ma PX. Synthetic biodegradable functional
E693. polymers for tissue engineering: a brief review. Sci China
31. Tebyanian H, Norahan MH, Eyni H, et al. Effects of Chem 2014; 57(4): 490–500.
collagen/β-tricalcium phosphate bone graft to regener- 50. Nair LS and Laurencin CT. Biodegradable polymers as
ate bone in critically sized rabbit calvarial defects. J Appl biomaterials. Prog Polym Sci 2007; 32(8–9): 762–798.
Biomater Funct Mater 2019; 17(1): 2280800018820490. 51. Liu F, Huang B, Hinduja S, et al. Biofabrication tech-
32. Omenetto FG and Kaplan DL. New opportunities for an niques for ceramics and composite bone scaffolds. In:
ancient material. Science 2010; 329(5991): 528–531. Antoniac I (ed.) Bioceramics and biocomposites: from
33. Farokhi M, Mottaghitalab F, Samani S, et al. Silk fibroin/ research to clinical practice. Hoboken, NJ: John Wiley,
hydroxyapatite composites for bone tissue engineering. 2019, pp. 17–37.
Biotechnol Adv 2018; 36(1): 68–91. 52. Wu Y-H, Chiu Y-C, Lin Y-H, et al. 3D-printed bioac-
34. Kundu B, Rajkhowa R, Kundu SC, et al. Silk fibroin bio- tive calcium silicate/poly-ε-caprolactone bioscaffolds
materials for tissue regenerations. Adv Drug Deliv Rev modified with biomimetic extracellular matrices for bone
2013; 65(4): 457–470. regeneration. Int J Mol Sci 2019; 20(4): 942–919.
35. Vepari C and Kaplan DL. Silk as a biomaterial. Prog 53. Farah S, Anderson DG and Langer R. Physical and
Polym Sci 2007; 32(8–9): 991–1007. mechanical properties of PLA, and their functions in wide-
36. Kim SH, Yeon YK, Lee JM, et al. Precisely printable and spread applications – a comprehensive review. Adv Drug
biocompatible silk fibroin bioink for digital light process- Deliv Rev 2016; 107: 367–392.
ing 3D printing. Nature Communications 2018; 9: 1620. 54. Gremare A, Guduric V, Bareille R, et al. Characterization
37. Rider P, Brook I, Smith PJ, et al. Reactive inkjet printing of printed PLA scaffolds for bone tissue engineering. J
of regenerated silk fibroin films for use as dental barrier Biomed Mater Res A 2018; 106(4): 887–894.
membranes. Micromachines 2018; 9(2): 46. 55. Fairag R and Rosenzweig D. 3D-printed polylactic acid
38. Rider P, Brook IM, Smith PJ, et al. Reactive inkjet print- (PLA) scaffolds promote bone-like matrix deposition in-
ing of silk barrier membranes for dental applications. In: vitro. ACS Appl Mater Interfaces 2019; 11(17): 15306–
Smith PJ and Morrin A (eds) Reactive inkjet printing. 15315.
Cambridge: Royal Society of Chemistry, 2017, pp. 147– 56. Li T, Peng M, Yang Z, et al. 3D-printed IFN-γ-loading
168. calcium silicate-β-tricalcium phosphate scaffold sequen-
39. Ha YY, Park YW, Kweon HY, et al. Comparison of the tially activates M1 and M2 polarization of macrophages to
physical properties and in vivo bioactivities of silkworm- promote vascularization of tissue engineering bone. Acta
cocoon-derived silk membrane, collagen membrane, and Biomater 2018; 71: 96–107.
polytetrafluoroethylene membrane for guided bone regen- 57. Tanodekaew S, Channasanon S and Uppanan P.
eration. Macromol Res 2014; 22: 1018–1023. Preparation and degradation study of photocurable oligo-
40. Singh BN, Panda NN and Pramanik K. A novel electro- lactide-HA composite: a potential resin for stereolithog-
spinning approach to fabricate high strength aqueous silk raphy application. J Biomed Mater Res B Appl Biomater
fibroin nanofibers. Int J Biol Macromol 2016; 87: 201– 2014; 102(3): 604–611.
207. 58. Ginebra M-P, Espanol M, Maazouz Y, et al. Bioceramics and
41. Panilaitis B, Altman GH, Chen J, et al. Macrophage bone healing. EFORT Open Reviews 2018; 3(5): 173–183.
responses to silk. Biomaterials 2003; 24: 3079–3085. 59. Keaveny TM, Morgan EF and Yeh OC. Bone mechanics.
42. Aramwit P, Kanokpanont S, De-Eknamkul W, et al. In: Myer K (ed) Standard handbook of biomedical engi-
Monitoring of inflammatory mediators induced by silk neering and design, New York: McGraw-Hill, 2004, pp.
sericin. J Biosci Bioeng 2009; 107(5): 556–561. 8.1–8.23.
43. Croisier F and Jérôme C. Chitosan-based biomaterials 60. Markovic S, Veselinovic L, Lukic MJ, et al. Synthetical
for tissue engineering. European Polymer Journal 2013; bone-like and biological hydroxyapatites: a comparative
49(4): 780–792. study of crystal structure and morphology. Biomed Mater
44. Gadgey KK and Sharma GS. Investigation of mechanical 2011; 6(4): 045005.
properties of Chitosan based films: a review. Int J Adv Res 61. Zhou M, Geng Y-M, Li S-Y, et al. Nanocrystalline
Eng Technol 2017; 8(6): 93–102. hydroxyapatite-based scaffold adsorbs and gives sustained
45. Abraham A, Soloman PA and Rejini VO. Preparation of release of osteoinductive growth factor and facilitates
Chitosan-Polyvinyl alcohol blends and studies on thermal bone regeneration in mice ectopic model. J Nanomater
and mechanical properties. Procedia Technol 2016; 24: 2019; 2019(1): 1–10.
741–748. 62. Kamitakahara M, Ohtsuki C and Miyazaki T. Review
46. Lee J and Lee KY. Local and sustained vascular endothe- paper: behavior of ceramic biomaterials derived from tri-
lial growth factor delivery for angiogenesis using an calcium phosphate in physiological condition. J Biomater
injectable system. Pharm Res 2009; 26(7): 1739–1744. Appl 2008; 23(3): 197–212.
47. Lee KY and Mooney DJ. Alginate: properties and biomed- 63. Barrere F, van Blitterswijk CA and deGroot K. Bone
ical applications. Prog Polym Sci 2012; 37(1): 106–126. regeneration: molecular and cellular interactions with
48. Jeon O, Bouhadir KH, Mansour JM, et al. Photocrosslinked calcium phosphate ceramics. Int J Nanomed 2006; 1(3):
alginate hydrogels with tunable biodegradation rates and 317–332.
Perić Kačarević et al. 17

64. Gerhardt L-C and Boccaccini AR. Bioactive glass and mesenchymal stem cell proliferation and osteoblast differ-
glass-ceramic scaffolds for bone tissue engineering. entiation. Carbohydr Polym 2018; 195: 356–367.
Materials 2010; 3(7): 3867–3910. 82. Zeng W, Yan Y, Zhang F, et al. Chrysin promotes osteo-
65. Hench LL. The story of Bioglass®. J Mater Sci Mater genic differentiation via ERK/MAPK activation. Protein
Med 2006; 17(11): 967–978. Cell 2013; 4(7): 539–547.
66. Fu Q, Saiz E, Rahaman MN, et al. Bioactive glass scaf- 83. Akbarzadeh R and Yousefi A-M. Effects of processing
folds for bone tissue engineering: state of the art and future parameters in thermally induced phase separation tech-
perspectives. Mater Sci Eng C Mater Biol Appl 2011; nique on porous architecture of scaffolds for bone tissue
31(7): 1245–1256. engineering. J Biomed Mater Res B Appl Biomater 2014;
67. Jones JR. Review of bioactive glass: from Hench to 102(6): 1304–1315.
hybrids. Acta Biomater 2013; 9(1): 4457–4486. 84. Carfi Pavia F, Conoscenti G, Greco S, et al. Preparation,
68. Ghosh D, Singha PS and SFWR J. Biometals in health characterization and in vitro test of composites poly-lactic
and disease: a review. World J Pharm Res 2016; 5(12): acid/hydroxyapatite scaffolds for bone tissue engineering.
390–399. Int J Biol Macromol 2018; 119: 945–953.
69. Glenske K, Donkiewicz P, Kowitsch A, et al. Applications 85. Chen S, Zhao X and Du C. Macroporous poly (L-lactic
of metals for bone regeneration. Int J Mol Sci 2018; 19(3): acid)/chitosan nanofibrous scaffolds through cloud point
826. thermally induced phase separation for enhanced bone
70. Zheng YF, Gu XN and Witte F. Biodegradable metals. regeneration. European Polym J 2018; 109: 303–316.
Mater Sci Eng, R: Reports 2014; 77: 1–34. 86. Huang Y-C and Mooney DJ. Gas foaming to fabricate
71. Bornapour M, Celikin M, Cerruti M, et al. Magnesium polymer scaffolds in tissue engineering. In: Peter X M and
implant alloy with low levels of strontium and calcium: Jennifer E (eds) Scaffolds in tissue engineering. Florida:
the third element effect and phase selection improve bio- Taylor and Francis Group. CRC Press, 2006, pp. 155–167.
corrosion resistance and mechanical performance. Mater 87. Preethi Soundarya S, Haritha Menon A, Viji Chandran S, et al.
Sci Eng, C: Mater Biol Appl 2014; 35: 267–282. Bone tissue engineering: scaffold preparation using chitosan
72. Cheng M-Q, Wahafu T, Jiang G-F, et al. A novel open- and other biomaterials with different design and fabrication
porous magnesium scaffold with controllable microstruc- techniques. Int J Biol Macromol 2018; 119: 1228–1239.
tures and properties for bone regeneration. Sci Rep 2016; 88. Duarte RM, Correia-Pinto J, Reis RL, et al. Subcritical
6: 24134–24114. carbon dioxide foaming of polycaprolactone for bone tis-
73. Prasad A, Sankar MR and Katiyar V. State of art on solvent sue regeneration. J Supercrit Fluid 2018; 140: 1–10.
casting particulate leaching method for orthopedic scaffolds 89. Catanzano O, Soriente A, La Gatta A, et al. Macroporous
fabrication. Mater Today: Proc 2017; 4(Part A): 898–907. alginate foams crosslinked with strontium for bone tissue
74. Hutmacher DW. Scaffolds in tissue engineering bone and engineering. Carbohydr Polym 2018; 202: 72–83.
cartilage. Biomaterials 2006; 21: 2529–2878. 90. Turnbull G, Clarke J, Picard F, et al. 3D bioactive com-
75. Cao H and Kuboyama N. A biodegradable porous com- posite scaffolds for bone tissue engineering. Bioactive
posite scaffold of PGA/β-TCP for bone tissue engineer- Materials 2017; 3: 278–314.
ing. Bone 2010; 46(2): 386–395. 91. Sharifi F, Atyabi SM, Norouzian D, et al. Polycaprolactone/
76. Khang G, Park CS, Rhee JM, et al. Preparation and char- carboxymethyl chitosan nanofibrous scaffolds for bone
acterization of demineralized bone particle impregnated tissue engineering application. Int J Biol Macromol 2018;
poly(l-lactide) scaffolds. Korea Polym J/한국고분자학 115: 243–248.
회 2001; 9(5): 267–276. 92. Qin X. Coaxial electrospinning of nanofibers. In: Afshari
77. Wei J, Chen F, Shin J-W, et al. Preparation and characteri- M (ed.) Electrospun nanofibers (Woodhead Publishing
zation of bioactive mesoporous wollastonite – polycap- Series in Textiles). Duxford: Woodhead Publishing, 2017,
rolactone composite scaffold. Biomaterials 2009; 30(6): pp. 41–71.
1080–1088. 93. Cheng G, Ma X, Li J, et al. Incorporating platelet-rich
78. Fereshteh Z. Freeze-drying technologies for 3D scaffold plasma into coaxial electrospun nanofibers for bone tissue
engineering. In: Ying D and Jordan K (eds) Functional engineering. Int J Pharm 2018; 547(1–2): 656–666.
3D tissue engineering scaffolds. Cambridge: Woodhead 94. Ao C, Niu Y, Zhang X, et al. Fabrication and characteriza-
Publishing, 2017, p. 24. tion of electrospun cellulose/nano-hydroxyapatite nanofib-
79. Li X, Zhang S, Zhang X, et al. Biocompatibility and ers for bone tissue engineering. Int J Biol Macromol 2017;
physicochemical characteristics of poly(Ɛ-caprolactone)/ 97: 568–573.
poly(lactide-co-glycolide)/nano-hydroxyapatite compos- 95. Ramachandran K and Gouma P-I. Electrospinning for
ite scaffolds for bone tissue engineering. Mater Des 2017; bone tissue engineering. Recent Pat Nanotechnol 2008;
114: 149–160. 2(1): 1–7.
80. Sangkert S, Kamonmattayakul S, Lin CW, et al. A bio- 96. Ge Z, Wang L, Heng BC, et al. Proliferation and differen-
functional-modified silk fibroin scaffold with mimic tiation of human osteoblasts within 3D printed poly-lactic-
reconstructed extracellular matrix of decellularized pulp/ co-glycolic acid scaffolds. J Biomater Appl 2009; 23(6):
collagen/fibronectin for bone tissue engineering in alveo- 533–547.
lar bone resorption. Mater Lett 2015; 166: 30–34. 97. Kacarevic ZP, Rider PM, Alkildani S, et al. An introduc-
81. Menon AH, Soundarya SP, Sanjay V, et al. Sustained tion to 3D bioprinting: possibilities, challenges and future
release of chrysin from chitosan-based scaffolds promotes aspects. Materials 2018; 11(11): 1–21.
18 The International Journal of Artificial Organs 00(0)

98. Boga JC, Miguel SP, de Melo-Diogo D, et al. In vitro 109. Mazzoli A. Selective laser sintering in biomedical engi-
characterization of 3D printed scaffolds aimed at bone tis- neering. Med Biol Eng Comput 2012; 51(3): 245–256.
sue regeneration. Colloids Surf B Biointerfaces 2018; 165: 110. Feng P, Wei P, Shuai C, et al. Characterization of mechan-
207–218. ical and biological properties of 3-D scaffolds reinforced
99. Roohani-Esfahani S-I, Newman P and Zreiqat H. Design with zinc oxide for bone tissue engineering. PLoS ONE
and fabrication of 3D printed Scaffolds with a mechanical 2014; 9(1): e87755.
strength comparable to cortical bone to repair large bone 111. Laurenti M and Cauda V. ZnO nanostructures for tissue engi-
defects. Sci Rep 2016; 6: 19468. neering applications. Nanomaterials 2017; 7(11): 374–334.
100. Vozzi G, Corallo C and Daraio C. Pressure-activated 112. Deng J, Li P, Gao C, et al. Bioactivity improvement of
microsyringe composite scaffold of poly(L-lactic acid) forsterite-based scaffolds with nano-58S bioactive glass.
and carbon nanotubes for bone tissue engineering. J Appl Mater Manuf Process 2014; 29(7): 877–884.
Polym Sci 2012; 129(2): 528–536. 113. Xia Y, Xu S, Zhou P, et al. Selective laser sintering fab-
101. Mondschein RJ, Kanitkar A, Williams CB, et al. Polymer rication of nano-hydroxyapatite/poly-ε-caprolactone
structure-property requirements for stereolithographic 3D scaffolds for bone tissue engineering applications. Int J
printing of soft tissue engineering scaffolds. Biomaterials Nanomedicine 2013; 8: 4197–4213.
2017; 140: 170–188. 114. Liao H-T, Lee M-Y, Tsai W-W, et al. Osteogenesis of adi-
102. Mironi-Harpaz I, Wang DY, Venkatraman S, et al.
pose-derived stem cells on polycaprolactone-β-tricalcium
Photopolymerization of cell-encapsulating hydrogels: phosphate scaffold fabricated via selective laser sintering
crosslinking efficiency versus cytotoxicity. Acta Biomater and surface coating with collagen type I. J Tissue Eng
2012; 8(5): 1838–1848. Regen Med 2013; 10: E337–E353.
103. Wang Z, Abdulla R, Parker B, et al. A simple and high- 115. Rider P, Kačarević ŽP, Alkildani S, et al. Bioprinting
resolution stereolithography-based 3D bioprinting system of tissue engineering scaffolds. J Tissue Eng 2018; 9:
using visible light crosslinkable bioinks. Biofabrication 2041731418802090.
2015; 7(4): 045009. 116. Rider P, Kačarević ŽP, Retnasingh S, et al. Bioprinting.
104. Fairbanks BD, Schwartz MP, Bowman CN, et al.
In: Biomaterials in regenerative medicine. Intechopen.
Photoinitiated polymerization of PEG-diacrylate with lith- com, https://www.intechopen.com/books/biomaterial-sup-
ium phenyl-2,4,6-trimethylbenzoylphosphinate: polym- ported-tissue-reconstruction-or-regeneration/bioprinting
erization rate and cytocompatibility. Biomaterials 2009; 117. Lim KS, Schon BS, Mekhileri NV, et al. New visible-
30(35): 6702–6707. light photoinitiating system for improved print fidelity in
105. Lan PX, Lee JW, Seol Y-J, et al. Development of 3D PPF/ gelatin-based bioinks. ACS Biomater Sci Eng 2016; 2(10):
DEF scaffolds using micro-stereolithography and surface 1752–1762.
modification. J Mater Sci Mater Med 2008; 20(1): 271– 118. Tzeng J-J, Hsiao Y-T, Wu Y-C, et al. Synthesis, charac-
279. terization, and visible light curing capacity of polycaprol-
106. Lee JW, Kang KS, Lee SH, et al. Bone regeneration
actone acrylate. Biomed Res Int 2018; 2018: 8719624.
using a microstereolithography-produced customized 119. Christensen K, Xu C, Chai W, et al. Freeform inkjet print-
poly(propylene fumarate)/diethyl fumarate photopolymer ing of cellular structures with bifurcations. Biotechnol
3D scaffold incorporating BMP-2 loaded PLGA micro- Bioeng 2015; 112(5): 1047–1055.
spheres. Biomaterials 2011; 32(3): 744–752. 120. Xu T, Binder KW, Albanna MZ, et al. Hybrid printing of
107. Ronca A, Ambrosio L and Grijpma DW. Design of porous mechanically and biologically improved constructs for
three-dimensional PDLLA/nano-hap composite scaffolds cartilage tissue engineering applications. Biofabrication
using stereolithography. J Appl Biomater Funct Mater 2013; 5(1): 015001.
2012; 10(3): 249–258. 121. O’Connell CD, Di Bella C, Thompson F, et al.

108. Shanjani Y, Pan CC, Elomaa L, et al. A novel bioprinting Development of the Biopen: a handheld device for surgi-
method and system for forming hybrid tissue engineering cal printing of adipose stem cells at a chondral wound site.
constructs. Biofabrication 2015; 7(4): 045008. Biofabrication 2016; 8(1): 015019.

You might also like