Download as pdf or txt
Download as pdf or txt
You are on page 1of 59

Applied Materials Today 20 (2020) 10 070 0

Contents lists available at ScienceDirect

Applied Materials Today


journal homepage: www.elsevier.com/locate/apmt

Biodegradable Polymers for Biomedical Additive Manufacturing


Dario Puppi∗, Federica Chiellini∗
Department of Chemistry and Industrial Chemistry – University of Pisa, UdR INSTM Pisa, Via G. Moruzzi 13, 56124 Pisa (Italy)

a r t i c l e i n f o a b s t r a c t

Article history: The tremendous interest received by additive manufacturing (AM) within the biomedical community is a
Received 29 December 2019 consequence of the great versatility offered in terms of processing approach, materials selection, and cus-
Revised 7 May 2020
tomization of the resulting device. In particular the unparalleled control over structural and compositional
Accepted 9 May 2020
features at the macro- and microscale, as a result of the large design freedom and high reproducibility, is
making AM the technology of election for the fabrication of biodegradable medical devices. This article
Keywords: is aimed at providing an update overview of scientific literature on biodegradable polymers for AM ap-
biodegradable polymers plication in the biomedical field. The main AM techniques applied so far to biodegradable polymers are
additive manufacturing outlined by presenting relevant materials processing requirements. The different classes of biodegradable
natural polymers
polymers investigated for AM (i.e., proteins, polysaccharides, aliphatic polyesters of either natural or syn-
aliphatic polyesters
thetic origin, polyurethanes, as well as other synthetic polymers under AM implementation) are described
3D printing
by highlighting their source of extraction, chemical modification, or synthesis route, and their physical-
chemical and processing properties in relationship to AM. Relevant literature on their AM processing for
medical and pharmaceutical applications is accordingly reviewed.
© 2020 Elsevier Ltd. All rights reserved.

1. Introduction models. 3D computer solid models of the object can be directly


drawn by means of a CAD or math software. In addition, 3D model
Additive manufacturing (AM) was defined in 2012 by the Amer- data can be derived from medical imaging techniques used for di-
ican Society for Testing and Materials (ASTM) as “the process of agnostic purposes, e.g., computer tomography (CT) and magnetic
joining materials to make objects from three-dimensional (3D) resonance imaging (MRI). The digital model is converted into a
model data, usually layer upon layer, as opposed to subtractive standard tessellation language (STL) file containing the information
manufacturing methodologies” [1]. A diversified terminology has of the object’s surface geometry, and then expressed as a series of
been adopted over time to refer to AM by employing various terms cross-sectional layers with predefined thickness originating a slice
used interchangeably, such as 3D printing, rapid prototyping, solid file. A numerical control (NC) programming language file is finally
freeform fabrication, additive fabrication, additive processes, and loaded digitally in the machine to drive the motion of the fabricat-
additive layer manufacturing. Due to the increasing interest on ing parts.
AM industrialization, the ASTM and International Organization for The fabrication process involves starting from the bottom and
Standardization have been working also on relevant aspects other building layers up through a sequential delivery of energy and/or
than terminology, such as measuring the performance and safety materials, with each newly formed layer adhering to the previous
of relevant materials, process and equipment, ensuring the quality one. In laser-based AM techniques, a beam or projection of light is
of the finished parts and end products, and specifying procedures selectively directed to i) a photosensitive resin bath to form pat-
for the calibration of machines [2]. terns of photopolymerized/crosslinked material, in the case of vat
AM techniques are based on computer-controlled design (CAD) photopolymerization techniques, e.g., stereolithography (SLA) and
and manufacturing (CAM) approaches to process a wide range of microstereolithography (μSLA) [3] or ii) a powder bed to generate
materials (i.e., metals, polymers, ceramics, and composites) into fi- local heat and form patterns of sintered/fused material, in the case
nal products with geometry and size resembling those of digital of selective laser sintering (SLS).[4] Regarding binder jetting (BJ),
also referred to as 3D printing, a liquid binder is selectively de-
posited on a powder bed [5]. In extrusion-based AM techniques, a

Corresponding authors: Dario Puppi and Federica Chiellini, Department of
polymer as melt [6] or dissolved/suspended in a suitable solvent
Chemistry and Industrial Chemistry – University of Pisa, UdR INSTM Pisa, Via G.
Moruzzi 13, 56124 Pisa (Italy) [7] is extruded in a given environment, for instance in air at a
E-mail addresses: dario.puppi@unipi.it (D. Puppi), federica.chiellini@unipi.it (F.
Chiellini).

https://doi.org/10.1016/j.apmt.2020.10 070 0
2352-9407/© 2020 Elsevier Ltd. All rights reserved.
2 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

List of acronyms PLGA poly(lactide-co-glycolide)


PLA poly(lactide)
AM additive manufacturing PLATMC poly(l-lactide-co-trimethylene carbonate)
ASCs adipose-derived stem cells PLACL poly(l-lactide-co-ε -caprolactone)
ASTM American society for testing and materials PDLACL poly(d,l-lactide-co-ε -caprolactone) ()
BC bacterial cellulose PLLA poly(l-lactide)
BG bioactive glass PMMA poly(methyl methacrylate)
BJ binder jetting PPF poly(propylene fumarate)
BMP-2 bone morphogenetic protein-2 PPFDA poly(propylene fumarate)-diacrylate
BMP-7 bone morphogenetic protein-7 Pro proline
BMSCs bone marrow stromal cells PS polystyrene
Ca-P calcium phosphate PTMC poly(trimethylene carbonate)
CAD computer-controlled design PUs polyurethanes
CAM computer-controlled manufacturing PVA poly(vinyl alcohol)
CAWS computer-aided wet-spinning PVP poly(vinyl pyrrolidone)
CLSM confocal laser scanning microscopy RGD arginine-glycine-aspartic acid
CT computer tomography rhBMP-2 recombinant human bone morphogenetic
DEF diethyl fumarate protein-2
DLP digital light processing rhBMP-7 recombinant human bone morphogenetic
DMPA 2,2-bis(hydroxymethyl) propionic acid protein-7
EC ethyl cellulose ROP ring-opening polymerization
ECM extracellular matrix SLA stereolithography
EDA ethylenediamine SE-AM solution extrusion-additive manufacturing
EFDP electrofluidodynamic printing μSLA microstereolithography
FDM fused deposition modeling SLS selective laser sintering
GelMA gelatin methacryloyl SSLS surface selective laser sintering
Gly glycine S/O/W solid in oil in water
Hyp hydroxyproline STL standard tessellation language
ME-AM melt extrusion-additive manufacturing TE tissue engineering
HA hyaluronic acid TCP β -tricalcium phosphate
Hap hydroxyapatite TPP two-photon polymerization
HAMA hyaluronic acid methacryloyl TPUs thermoplastic polyurethanes
HA-pNIPAAM poly(N-isopropylacrylamide)- Tg glass transition temperature
grafted hyaluronic acid Tm melting temperature
HMDI hexamethylene diisocyanate Tdeg degradation temperature
IPDI isophorone diisocyanate UV ultraviolet
MEW melt-electrospinning writing Vis visible
MRI magnetic resonance imaging 2D two-dimensional
MSCs mesenchymal stem cells 3D three-dimensional
Mw molecular weight 3HB 3-hydroxybuytrate
NC numerical control 3HHx 3-hydroxyhexanoate
NIPS non-solvent-induced phase separation 3HV 3-hydroxyvalerate
NVP N-vinyl-2-pyrrolidone 4HB 4-hydroxybutyrate
PA11 polyamide 11
PA12 polyamide 12
PC polycarbonate PC controlled temperature or in a coagulating medium, and deposited
PCL poly(ε -caprolactone) with a predefined pattern on a fabrication platform.
∗ PCL star poly(ε -caprolactone) Overall the main challenging aspects for new developments are
PDLLA poly(d,l-lactide) related to build speed and resolution, biocompatibility and me-
PED precision extrusion deposition chanical properties of fabricated parts, controlled composition of
PEEK polyether ether ketone multi-material parts, as well as other peculiar requirements of spe-
PEG poly(ethylene glycol) cific biomedical applications [8]. AM has the potential of fastening
PEGDA polyethylene glycol diacrylate the product development process by reducing the time required
PEUs poly(ester urethanes) for design validation and functional prototypes development. How-
PGA poly(glycolic acid) ever, at the current stage AM processes are still slow in compari-
PHA polyhydroxyalkanoates son to those based on large scale production technologies, such as
PHB poly(3-hydroxybutyrate) injection molding. Even if this limitation is offset in customized
PHBHHx poly[3-hydroxybutyrate-co-3- medicine applications by AM flexibility, the perspective of broad-
hydroxyhexanoate) ening and optimizing its applicative spectrum encourages research
PHBV poly(3-hydroxybutyrate-co-3- on the enhancement of fabrication efficiency with a good compro-
hydroxyvalerate) mise between the spatial resolution and the time of production.
PHV poly(3-hydroxyvalerate) Insufficient resolution can result in a significant dimensional
P[3HB-co-4HB] poly(3-hydroxybutyrate-co-4- discrepancy between the CAD virtual model and the fabricated ob-
hydroxybutyrate) ject, with influence on its resulting quality, structural and func-
tional properties. Besides dimensional accuracy at the macroscale,
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 3

different biomedical applications require morphological features dition, solution-based AM is the election approach to functional-
structured at the micro/nanoscale, for instance as a means to ize materials with drugs and other bioactive agents, such as in the
steer cell behaviors in tissue engineering (TE). For these reasons, case of hydrogels with controlled porosity and external shape en-
μSLA and melt-electrospinning writing (MEW) have been attract- capsulating cells or growth factors [24], [25], and to endow the
ing great attention for the development of porous polymeric con- polymeric matrix with a nano/microporosity [7], [26]. Great ef-
structs structured at the submicrometric scale [3], [9]. In this op- forts have been also made to develop nanocomposite implants by
tic, combination of AM with other materials processing techniques AM in order to increase the mechanical properties and bioactiv-
at different integration levels (i.e., assembly, fabrication or tech- ity of polymers already in the clinical practice, such as poly(l-lactic
nique level) is a hot topic in the scientific community with the acid) (PLLA) and poly(ε -caprolactone) (PCL), as well as other poly-
aim of optimizing conflicting scaffold structural requirements, such meric materials currently under investigation for biomedical pur-
as high mechanical strength/multi-scale porosity, 3D structure/high poses [27].
resolution, high quality/high fabrication velocity, and structural This article aims to provide an updated overview of literature
role/bioactivity [10], [11]. focused on biodegradable polymers tailored to the application of
From a mechanical point of view, parts fabricated by AM have AM in biomedical science and technology. In particular, relevant
often lower properties than those produced by subtractive and applicative fields are introduced by highlighting the peculiar ad-
formative techniques. Depending on the AM technique employed, vantages and the most challenging issues of AM. The technolog-
such drawback can be the result of a limited choice among suitable ical and methodological aspects of the different AM techniques
materials (e.g., in the case of photosensitive polymers [3]), limited applied so far to biodegradable polymers are presented and their
materials engineering freedom (e.g., in the case of composite mate- specific processing requirements discussed. The different classes
rials processing by laser- or melt extrusion-based techniques [12]), of biodegradable polymers under investigation for AM are intro-
unwanted porosity and other structural defects (e.g., in the case duced by highlighting their source of extraction, chemical-physical
of uncomplete particles fusion in SLS or marked/insufficient fiber- modification or synthesis route, and their physical-chemical and
fiber fusion in extrusion AM [13]). The anisotropic mechanical be- processing properties in relationship to AM. Relevant literature on
havior of additive manufactured parts as a consequence of the lay- their AM processing for medical and pharmaceutical applications is
ered manufacturing process [14], [15] is particularly pronounced in accordingly reviewed. While the range of polymers for AM is be-
scaffolds for TE due to the highly porous structure necessary for ing increasing, the article is focused specifically on those that have
tissue growth processes. Ongoing research is aimed at developing been more extensively investigated showing promising outcomes
new software able to analyze the relationship between AM process for their commercial translation.
parameters and material performance [16]. In the next future, ma-
chine learning capabilities will allow the selection of the appropri- 2. Additive Manufacturing (AM) in Biomedical Science and
ate AM process parameters given a target mechanical performance, Technology
and the analysis in real time of material morphological features
and irregularities in order to quantify their relationship with the As previously introduced, AM of polymeric materials has found
resulting mechanical performance. application in different biomedical areas, including 3D anatomi-
Personalized medicine is emerging as one of the most success- cal models and surgical training, surgical equipment, prosthetics
ful and promising applications for AM. The possibility of using to- and implants, TE, in vitro tissue modelling, and drug discovery
mographic images from X-ray and MRI scan to manufacture ob- (Figure 1). The advanced technological level of AM, in terms of
jects customized to anatomical parts of the human body is revo- automation degree, reproducibility, design freedom of the man-
lutionizing the approach to surgical planning, building prosthetics, ufactured details, processing of medical digital images and their
dentistry, and TE. Tens of articles have reported the clinical ap- accurate reproduction as solid anatomical objects, is fastening its
plications of devices made from metals, polymers or their com- translation to clinical research and practice in different medical
bination, like in the case of maxillofacial surgery in which AM branches, including orthopedic, urogenital, cardiovascular, and neu-
is used for anatomic models, surgical guides, occlusal splints, im- rological surgery.
plants (e.g., osteosynthesis plates and skeletal reconstruction parts) 3D models for anatomical study and surgical training was one
and facial epithesis [17]. Research on biomedical polymers for AM the first successful biomedical applications of AM (Figure 1a). Dif-
is resulting in a tremendous impact on clinical treatment of hu- ferent articles have highlighted the potential of physical anatomical
man tissue failure defects, as well as on materials science and models developed by AM for understanding microsurgical anatomy
technology for polymeric devices development. Recently, a con- details and acquiring surgical skills without the ethical, legal and
siderable literature has grown up around polymeric materials for infection problems related to cadaver material. Since they are not
AM with the aim of optimize and broaden the range of applica- considered medical devices, anatomical models can be easily man-
tions of this class of techniques for biomedical purposes. Signifi- ufactured without following any regulation. Patient-specific phan-
cant progress has been achieved on the synthesis of biocompati- toms and surgical guides have been used increasingly in differ-
ble and biodegradable photoreactive macromers for SLA to replace ent surgery disciplines including among others craniofacial surgery
conventional lithographic acrylic resins which may cause toxic ef- [37–40], neurosurgery [41–43], spine surgery [44–46], cardiovas-
fects [8], [18], [19]. The decades-long history of clinical employ- cular surgery [47–49], and pelvic surgery [50–52]. Personalized
ment of polymeric implants made by conventional melt-processing anatomical models are used also as prototypes for designing medi-
techniques [20] is being exploited to implement in melt-based AM cal implants and surgical guides, assistive devices when planning
processes both polymers already approved for use in humans and and during surgery, as well as means for academic and clinical
those under development. In vivo outcomes on implantation in an- teaching, communication to patient, and dissemination [53].
imals and humans of polymeric devices made by fused deposi- Functional surgical instruments design and fabrication were
tion modeling (FDM) or SLS stimulate research for their perma- recently investigated as potential applications of AM to enhance
nent introduction in the interventional surgery routine [21–23]. customization and ease of modification according to a clinician’s
AM techniques based on polymers in solution or as suspension of- preference. As an example, Kondor et al. [54], [55] developed by
fer the advantages of processing naturally-derived polymers with FDM a generic surgical set that was functionally tested by laparo-
inherent bioactive and biomimetic properties to develop produc- tomy, splenectomy and suturing on a surgical simulator. A recent
tion processes with enhanced environmental sustainability. In ad- study showed that functional surgical instruments fabricated by
4 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 1. Representative examples of AM biomedical applications. (a) 3D anatomical models: picture of poly(d,l-lactide) (PDLLA) mandibular model by FDM [28]. (b) Surgical
instruments: picture of customized nylon surgical set by SLS (reproduced with permission [29]). (c) Exoprostheses: multifunctional prosthetic hand prototype made of
a fiber-reinforced nylon (Duraform® HST) and fabricated by SLS (reproduced with permission [30]). (d) Surgical implants: picture of porous poly(methyl methacrylate)
(PMMA) implants prototypes fabricated by computer-aided wet-spinning (CAWS) and submitted to different post-processing treatments (reproduced with permission [31]).
(e) Biodegradable surgical implants: picture of pediatric tracheal splints made of PCL and fabricated by SLS (reproduced with permission [32]). (f) Scaffold-guided tissue
engineering (TE): picture of a PCL/hydroxyapatite (Hap) bone scaffold by FDM modelled on a goat femoral head and equipped with an intramedullary stem (reproduced with
permission [33]). (g) 3D tissue modelling: confocal laser scanning microscopy (CLSM) micrograph of a spheroid of human pancreatic cancer cells grown in vitro on a 3D
microstructured polyelectrolyte complex by CAWS (reproduced with permission [34]). (h) Integrated organ-on-a-chip: CLSM micrograph of a 3D bioprinted hepatic construct
with patterns of hepatic cells (green) in gelatin methacryloyl (GelMA) and supporting cells (red) (scale bar, 500 μm) (reproduced with permission [35]). (i) Drug release
systems: picture of polypills with core-shell, multilayer, or concentration gradient configuration of drug loading (reproduced with permission [36]).

SLS offer some advantages compared to traditional manufacturing AM is bringing different advantages to this therapeutic field in-
methods, including no cost raising for increased complexity, accel- cluding the possibility of making products out of one part, easily
erated design to production times, and surgeon specific modifica- personalizing and customizing the device with large design free-
tions (Figure 1b) [29]. dom, and producing parts cheaply and quickly [56]. Besides dec-
Exoskeleton devices and exoprostheses that can easily fit up- orative devices, a technologically-advanced field of application in
per and lower limbs are designed and printed all over the world. this area is represented by functional prostheses. In particular, a
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 5

great number of upper limb prostheses with different types of ac- relevant composite materials is resulting in the customization of
tuation and control of hand kinematics are described in literature 3D scaffolds for engineering tissues with different structural and
and specialized internet websites [57]. Most of these prostheses functional features, such as complex solid organs (e.g., liver and
are available on the market and developed by recurring to different bone) and tubular structures (e.g., urethra and blood vessels) [66],
polymers such as acrylonitrile butadiene styrene (ABS), poly(d,l- [75], [76]. The great efforts spent on this research trend are giv-
lactide) (PDLLA), poly(ether ether ketone) (PEEK) and thermoplas- ing the first clinical results like in the case of PCL devices by FDM
tic polyurethanes (TPUs) processed by FDM, polyamides 12 and 11 implanted in humans for craniofacial and dentistry reconstruction
(PA12, PA11) processed by SLS, or photopolymer resins processed [21], [77].
by SLA. For instance, Andrianesis and Tzes [30] recently described 3D tissue modelling for tissue physiology, cancer research and
a SLS-manufactured anthropomorphic hand chassis made of fiber- drug evaluation is an emerging field of application of AM that is
reinforced PA12, that integrates a modular actuation system pro- benefiting of the great knowledge generated by TE research and
viding the prosthesis with the necessary dexterity, tactile sensing, the resulting powerful technological tools. In particular, the grow-
and controlled digits position (Figure 1c). ing interest of the AM community on experimental modelling of
Surgical implants are among the biomedical applications cur- healthy and pathological tissues stems from the evidence that 3D
rently benefiting of AM clinical translation. Virtual planning and scaffolds enable the development of culture microenvironments
guided surgery with additive manufactured patient-specific im- more closely resembling those found in native tissues in compari-
plants are becoming particularly common in mandibular recon- son to what achieved with two-dimensional (2D) cell culture sys-
struction.[58] For instance, Tarsitano et al. [59] demonstrated tems [78]. Seminal works on polymeric scaffolds for cancer mod-
that the employment of customized reconstructive titanium plates elling [79–86] showed that tumor cells grown in 3D tend to form
manufactured by laser sintering technology facilitated restoration proliferative masses or aggregates not detected in monolayer cul-
of native mandibular contour. The potential of AM for process- tures, resulting in different metabolic characteristics and gene ex-
ing biostable polymers already employed in reconstructive surgery pression profiles, as well as in increased tumor proliferation and
has been recently demonstrated, e.g., in an article describing FDM decreased sensitivity to induced apoptosis. Indeed, 3D scaffolds
fabrication of customized osteosynthesis plates as well as cranio- culture allow more accurate and reproducible information on drug
plasty and maxillofacial implants made of PEEK [60]. Other poly- toxicity and tissue chemoresistance for early-stage drug discovery
meric devices intended to optimize surgical intervention and plan- and high-throughput screening of drug candidates [87], [88]. Ow-
ning, such as occlusal splints and surgical guides, are widely em- ing the aforementioned advantages of AM in controlling and cus-
ployed by clinicians around the globe [17]. The evidence that a tomizing scaffold properties, a number of articles have been pub-
porous structure can favor implant integration, as consequence of lished on the investigation of additive manufactured systems to de-
tissue formation across the material creating a mechanical inter- velop 3D models of different pathologically-relevant tissues, such
locking that minimizes micromovements and stress shielding ef- as bone metastases [89] and pancreatic cancer [34] (Figure 1g).
fects, is propelling research on direct AM of biostable porous ar- In addition, bioprinting versatility has been exploited to recapitu-
chitectures made of biocompatible polymers, such as poly(methyl late certain features of solid tumor tissues, e.g., gradient distribu-
methacrylate) (PMMA) [31], [61], [62] and PEEK [63] (Figure 1d). tion of chemical and biological factors. In this optic, complex tissue
As in the case of endovascular stents, surgical implants intended models based on synthetic or natural polymers, cells, growth fac-
for children require to be made of a biodegradable material allow- tors, and well-defined vasculature were developed as systems scal-
ing for the physiological expansion of tissues and organs during able to mechanistic and high-throughput screening assays related
child growth (Figure 1e). Research on AM application to this as- to different biological phenomena, such as wound healing, vascu-
pect recently resulted in the clinical implantation of personalized larization and chondrogenesis [90], [91]. Humanized tumor tissue
3D printed tracheal and cardiovascular splints made of polymers models in animals were recently developed as alternative to the
(e.g., PCL) designed to be absorbed into the body [22], [23]. injection of human cancer cells in order to better mimic species-
Tissue engineering (TE) is by far the most extensively inves- specific mechanisms occurring in human diseases [92]. This ap-
tigated biomedical field of application of AM mainly as a con- proach involves the ectopic implantation of an additive manufac-
sequence of the strict requirements in terms of scaffold poros- tured scaffold seeded and cultured with human cells in combi-
ity, pore size and anatomical structure not attainable by means of nation with growth or differentiation factors, into an immunode-
other fabrication approaches [64], [65]. Indeed, TE relies on the ficient host. Partially humanized organs have been engineered in
implantation of a biodegradable scaffold with an interconnected vivo to study prostate or breast metastasis to bone by employing
porous structure, possibly in combination with cells and bioactive PCL/β -tricalcium phosphate (TCP) composite scaffolds produced by
molecules, which acts as a temporary template providing mechan- FDM [93] or PCL microfiber tubular scaffolds fabricated by MEW
ical support and cell adhesion substrate to guide the tissue re- and coated with calcium phosphate (Ca-P) [89], [94], [95].
generation process (Figure 1f) [66]. It is well known that scaffold Integrated organ-on-a-chip research is supported by the great
chemistry, topography, porosity, and mechanical stiffness influence progress made by AM in supplying advanced tools for fabricating
cell shape, cytoskeletal organization, function, protein expression, in a single continuous procedure the four key elements, i.e., a mi-
and differentiation [67–70]. AM techniques customized to TE pur- crofluidic chip, live cells/microtissues that will be cultured in this
poses enable an advanced control over scaffold composition and chip, components for stimulus loading to mature the microtissues,
architecture at different size scales (from macro- to micrometric and sensors for results readout.[96] In particular, the ability of bio-
scale) in terms of design freedom and resolution, with relevant printing to precisely positioning multiple materials, including hy-
effects on mechanical and biological properties [21], [65]. Struc- drogels, cells, growth factors, and even bacteria [97], in a sequen-
tural control can be pushed down to a sub-micrometer resolution tial order with a high spatial resolution enables the development
scale by employing hybrid AM approaches and μSLA with the aim of heterogenous and sophisticated microenvironments mimicking
of providing nanometric instructive cues to cells [21], [31], [71], those found in native tissues [98] (Figure 1h). Fabricating energy
[72]. In addition, cutting edge advancements on computer-aided actuators (e.g., electrical and mechanical transducers) and sensors
design and manufacturing approaches are leading to the fabrica- on chips to apply stimuli to cells and monitor their activities is an
tion of clinically-sized, anatomically-shaped scaffolds with a tai- effective way to make micronized organs mature and functional.
lored porous structure [73], [74]. Fast progress on novel technolog- A few articles described the possibility of integrating on the same
ical tools for processing a wide array of biomedical polymers and chip mechanical strain-inducing actuators and electrochemical sen-
6 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

sors, together with complex fluid handling modules [99]. Other tion. The integration of AM with other fabrication methods will be
microfluidic approaches explored to induce mechanical stimuli to properly presented as an effective means to develop hybrid poly-
cells on demand involves the employment of magnetic particles meric architectures with complementary structural features. The
[100] or actuators made from memory shape materials [101]. most successful attempts of AM combination with electrospinning
Drug release from additive manufactured implants is one of [112], salt leaching [113], gas foaming [114], freeze drying [115],
the most challenging aspects that could greatly influence the fu- and wet-spinning [7] will be described in this and the following
ture trend of AM applied to biomedical polymers [102]. Direct chapters, showing how the integration can be achieved at: i) the
drug loading of AM implantable devices during fabrication has assembly level by combining substructures fabricated by different
been intensively investigated for hydrogel-forming materials (e.g., technologies into a bi/multiphasic construct, ii) at the fabrication
crosslinked alginate) and other hydrophilic polymers that can be level by simultaneously incorporating multiple-length scale net-
processed by using aqueous solutions at room temperature [103], works (e.g., macro-, micro- and nanofeatures) within a single fi-
[104]. Indeed, most of relevant studies were based on solution nal bi/multimodal scaffold, iii) at the technique level by fusing the
extrusion-AM (SE-AM) and BJ, with few examples reporting on working principles of different fabrication techniques within a sin-
drug loading of implants fabricated by FDM, SLA or SLS, mainly gle, novel hybrid AM technology [10], [11].
by recurring to post-processing loading strategies because of the
tough processing conditions required (i.e., high temperatures, high 3.1. Stereolithography (SLA)
energy laser) that can lead to drug degradation. In particular, SE-
AM offers great opportunities for loading either hydrophilic or hy- SLA has been used for years in large-scale industrial processes
drophobic polymers with a drug by simply adding it as solute in the microelectronics sector as well as in the biomedical industry
or suspension to the solution to be processed at room tempera- for the production of custom hearing and dentistry devices. One of
ture [105], [106]. Advanced pharmaceutical AM technologies can the main advantages of SLA approaches over other AM techniques
be used to produce controlled drug delivery systems and person- is their higher resolution which is limited by the system’s optics
alized dosages for the future of personalized medicine [107]. Dif- rather than the extrudate or powder particles size.
ferent oral dosage forms by AM are currently under investigation SLA, also referred to as vat photopolymerization, is based on the
resulting in the recent approval of the first additive manufactured controlled polymerization and/or crosslinking of a photosensitive
drug-releasing tablet for oral administration, Spritam®. In partic- resin to fabricate an object layer-by-layer by using light, usually
ular the versatility of AM has been investigated to tune dosage in the ultraviolet (UV) and sometimes in the visible (Vis) spectrum
macroshape [108], drug concentration gradients [109], and com- [116]. Resin curing can be based on vector scanning SLA, mask pro-
plex inner porous structures [110] as a means to control drug re- jection SLA or two-photon polymerization (TPP). In all cases, a light
lease kinetics (Figure 1i). The first successful results suggest that source is irradiated over the surface of a vat filled with a photo-
AM has the potential to offer high versatility in controlling drug sensitive resin. Once a layer is photopolymerized, the build stage
release profiles, personalized customization, complex designs, and is submerged further into the photopolymer vat and the next layer
multi-drug incorporation, enabling a new paradigm of controlled is built directly on top of the previous one (Figure 2a). Alterna-
release and local delivery of drugs. The high technological readi- tive technological solutions developed for processing polysaccha-
ness level of AM techniques will facilitate the maturation of ad- ride derivatives into biocompatible hydrogels, involve pumping PBS
vanced fabrication approaches to drug delivery eventually adhering after the fabrication of each layer to rinse away partially-reacted
to GMP-standards and industrial scale-up. resin, and then dispensing fresh monomer to fabricate the next
layer [117].
3. Polymeric Materials Requirements for AM In the case of vector scanning SLA, a laser beam is scanned over
the vat surface, while in the case of mask projection SLA, often re-
In addition to fundamental requirements of polymeric bioma- ferred to as digital light processing (DLP), light is reflected off a
terials (e.g., biocompatibility), biomedical devices should assure set of dynamic micromirrors to irradiate simultaneously the entire
structural and functional roles tailored to the specific application surface of the photopolymer vat. In both approaches the resin is
they are intended to. As an example, those designed for inter- selectively solidified through photopolymerization reactions wher-
facing with cells and actively supporting a biological regenerative ever the laser is scanned. TPP relies on the simultaneous absorp-
process should provide biochemical and/or physical cues to influ- tion of two lower energy photons at high wavelength, typically in
ence cell behaviors. Moreover, each AM technique requires distinct the range 780–820 nm.[118] In this case, features on the nanome-
physical-chemical polymer properties in agreement with its pro- ter scale can be achieved, even if the time required to obtain phys-
cessing principles. The employment of polymers in the fabrica- iologically relevant sizes makes this technique in most of the cases
tion of ceramic-based devices (e.g., as binding/reinforcing agents in an impractical choice for 3D biomedical implant fabrication [119].
BJ, sacrificial materials in SLA, or molding systems in indirect AM μSLA approaches have the potential to create devices with fea-
[111]) further widens the relevant AM requirements window. ture sizes in the scale of tens of micrometers and overall dimen-
This chapter is dedicated to a synthetic description of the fun- sions of centimeters [3]. They are based on either high-resolution
damental scientific and technological aspects involved in the pro- vector scanning or mask projection systems using UV lamps, LEDs
cessing of biodegradable polymers by different AM techniques. Var- or lasers. A top-down approach in which the light is projected from
ious criteria have been adopted to classify AM techniques on the above the vat, or a bottom-up approach involving light projection
basis of different aspects, such as the processing principles, the from below can be either adopted. A layer of fresh resin with con-
technological solutions, or the physical state of the material. This trolled thickness is applied over the solidified layer by means of a
review specifically deals with techniques applied to biodegrad- process based on dipping, spreading, pumping or gravity.
able polymers by following a classification commonly used in the μSLA has been having a great impact on the biomedical mar-
biomedical field in agreement to their working principles, i.e., ket in large-scale production of customized removable teeth align-
stereolithography (SLA), selective laser sintering (SLS), binder jet- ers (Invisalign®) and hearing aid device parts [120], [121], as well
ting (BJ), melt extrusion-AM (ME-AM), solution extrusion-AM (SE- as in different surgery applications such as anatomical models for
AM), and bioprinting (Figure 2). Materials processing properties surgical planning, models and molds for customized implants, cus-
required by each AM technique will be discussed by highlight- tom surgical guides and templates [122]. The tremendous progress
ing advanced strategies for polymer structure design and formula- made on resolution increasing, and on the variety of photosensi-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 7

Figure 2. AM techniques commonly employed for biodegradable polymers processing [65,98]. (a) Stereolithography (SLA): a light source is irradiated over the surface of a vat
filled with a photosensitive resin. (b) Selective laser sintering (SLS): based on selective sintering of a powder bed made of polymer, ceramic or hybrid particles by means of
a computer-controlled laser beam. (c) Binder jetting (BJ): a liquid binder is selectively deposited on a bed of polymer, ceramic, metallic or composite powder by means of an
inkjet head. (d) Melt extrusion-AM (ME-AM): e.g., fused deposition modelling (FDM), based on the extrusion of a polymeric filament through a nozzle kept at a temperature
higher than the polymer Tg . (e) Solution extrusion-AM (SE-AM): a polymer, ceramic or composite solution is extruded through a translating nozzle by means of a pneumatic
or mechanical-driven dispensing system. (f) Extrusion bioprinting: based on pneumatics or mechanical force to continuously extrude cells suspended in a hydrogel solution;
inkjet bioprinting: based on a piezoelectric or thermal actuator to sequentially eject small droplets made of cells and a hydrogel; laser-assisted bioprinting: based on a laser
pulse to vaporize a region in the donor layer (top) forming a bubble that propels a suspended bioink to fall onto the substrate; stereolithography bioprinting: based on
projected light to selectively crosslink bioinks layer-by-layer.

tive materials for biodegradable scaffolds fabrication, encourages ity should be sufficiently low, typically below 5 Pa•s [18], [126],
research on SLA employment for TE [3]. Indeed, as it will be fur- to achieve the necessary flow properties. Low molecular weight
ther discussed, the great ability of SLA to control pore size, ge- (Mw) macromers with relatively low glass transition temperature
ometry, porosity and interconnectivity is being exploited to de- (Tg ), and a diluent when needed, are typically employed for this
velop advanced materials engineering strategies for regenerative reason.
medicine. In addition, the possibility of incorporating cells during The diluent can be reactive, like in the case of diethyl fu-
the photopolymerization treatment opens new possibilities for the marate (DEF) [127] or N-vinyl-2-pyrrolidone (NVP) [128] that are
development of living TE constructs and microscale cell patterning employed in the photoreaction of macromers containing fumarate
strategies [123], [124]. groups. When a non-reactive diluent is employed, for instance
Materials design and processing requirements. SLA resins are in the case of solid or highly viscous macromers functionalized
composed by reactive monomers, oligomers and/or polymers that with reactive groups (e.g., acrylates), the cured material can un-
polymerize and/or crosslink when exposed to light in the UV or dergo shrinkage upon diluent extraction and drying [129]. Since
Vis spectrum. The process is carried out in the presence of a suit- this shrinkage is usually isotropic and the designed architecture of
able photoinitiator and possible additives such as light absorbers, the object is preserved, this effect is to be accounted for in the de-
stabilizers, anti-foaming agents, and other functional components sign phase. Various photoinitiators with UV or Vis light absorbance
(e.g., ceramic particles and polypeptides) [125]. In order to ex- have been tested for applications requiring cell-material inter-
ploit the precise spatial and temporal control achievable by means actions, including the UV Irgacure photoinitiators, with Irgacure
of SLA techniques, it is necessary to employ photopolymers with 2959 showing the least toxicity to different cell lines, and the Vis
minimized shrinkage and distortion upon polymerization. At the light initiators lithium phenyl-2,4,6-trimethylbenzoylphosphinate
same time, the resin should not be thermosetting and its viscos- and eosin Y disodium salt with increased water solubility [130].
8 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

The most employed photopolymer resins for SLA are multifunc- and spreading process is repeated several times to build up the 3D
tional acrylates, methacrylates, and acrylamides submitted to free- object layer-by-layer. A high intensity laser beam (e.g., CO2 laser)
radical photopolymerization, as well as epoxides and vinyl ethers capable of raising the bed temperature and bonding the parti-
submitted to cationic photopolymerization [3], [8]. Photocuring cles within a layer and between adjacent layers is employed. Non-
of low-Mw, multi-functional monomers usually results in highly sintered powder serves as support for the subsequent layers, thus
cross-linked networks showing a rigid and brittle behavior. For this allowing the manufacturing of complex geometries without the
reason, a number of articles have focused on the development of need of implementing support structures and materials.
macromers with low Tg and relatively high Mw (1-5 kg/mol) to Further advantages of SLS over other AM techniques include
fabricate flexible and elastomeric porous constructs for biomedi- avoiding the use of solvents, diluents or other reactive agents that
cal applications [131–133]. Cutting edge advancements in SLA ma- can be toxic if not completely removed from the device [158]. On
terials science and technology deal with polymers with advanced the other hand, post-processing treatments (e.g., air blasting and
properties including shape memory [134], ionic conductivity [135], ultrasonic cleaning) are often required to eliminate non-sintered
high thermomechanical properties [136], biocompatibility for Class powder that may remain entrapped in porous architectures. SLS
IIA devices [137]. In addition, SLA synthetic materials are often resolution is typically in the range of 100 μm depending on laser
surface-treated by using bioactive coatings, such as calcium phos- parameters. As a consequence of the limited laser intensity applied
phates and biomimetic peptides, in order to increase their biocom- in order to avoid polymer thermal degradation, a poor control over
patibility and favor cell adhesion [138], [139]. surface roughness, which is defined by particle size and shape,
Biodegradable SLA materials can be obtained by employ- is generally achieved. For this reason, surface finishing treatments
ing oligomers with hydrolysable ester or carbonate linkages in (e.g., milling and coating) are typically applied.
the main chain, and functionalized with photocurable acrylate, SLS has found application in the biomedical field mainly for
methacrylate, or vinyl functional groups. As it will be dis- manufacturing patient-specific anatomical models, exoprostheses,
cussed in detail later, macromers based on poly(propylene fu- and biostable implantable devices with complex shapes and con-
marate) (PPF) [140], acrylate-endcapped poly(ε -caprolactone-co- trollable internal architectures [159]. Indeed, the great progress
trimethylene carbonate) [141], [142] or poly(trimethylene carbon- made on SLS in combination with medical imaging techniques is
ate) (PTMC) [143], [144], methacrylate [129] or fumaric acid mo- being resulting in fastening and improving the quality of surgi-
noethyl ester [145] end-functionalized PDLLA oligomers, or divinyl cal procedures, such as preoperative planning, implant design and
fumarate PCL [146] have been successfully employed to fabricate surgery operations. In addition, the development of tailored pow-
biodegradable scaffolds. The optimization of physical-chemical, ders made of biodegradable polymers, possibly loaded with os-
rheological and biological properties of new unsaturated polyester teoconductive ceramics, has widened the application of SLS to TE
resin formulations is a key aspect for SLA application in the TE scaffolds and drug delivery devices. The availability in the mar-
field [19]. Research on biostable photocurable resins for biomed- ket of biomaterial powders for SLS is still scarce and most of the
ical purposes is giving promising results as well, such as in the methodologies for their preparation are performed at a laboratory
case of polyethylene glycol diacrylate (PEGDA) hydrogels, which are scale. However, the successful implantation in humans of med-
widely investigated as scaffolds for cells growth [147–149]. Vari- ical devices fabricated by SLS witnessed the tremendous poten-
ous natural polymers, including gelatin [150], keratin [151], colla- tial of this AM technique for tissue reconstruction and pediatric
gen [152], chitosan [153], and hyaluronic acid (HA) [117] have been interventional surgery [21]. Indeed, Oxford Performance Materials
successfully modified with photopolymerizable pendant groups in has received FDA clearance to manufacture patient-specific PEEK
order to exploit their biocompatibility and high cellular adhesion implants (OsteoFab®) for cranial, maxilofaccial and spinal surgery
for photoreaction-based fabrication approaches. However, although [160,161], while PCL devices are surgically employed to treat in-
the great progress made on gelatin methacryloyl (GelMA) to fab- fants affected by tracheobronchomalacia [162].
ricate biocompatible scaffolds [124], [150], [154], [155], function- Materials design and processing requirements. SLS is based on
alized natural polymers for SLA still generally result in either 2D laser absorption-induced raising of the local temperature to a level
structures or 3D constructs with limited porosity in the direc- enough to soft and fuse the particles together, typically without
tion of printing (Z axis). Indeed, the high-water content and low causing an actual melting of the polymer. The material processing
crosslinking groups density in these resins typically cause high requirements of SLS concern different aspects including polymer
depth of penetration of light, not allowing the fabrication of high optical, thermal, viscosity, and surface tension properties, as well
resolution features in the print direction even by recurring to UV as particles shape and size distribution [159], [163], [164].
absorbers [3]. However, the great efforts spent by actors in this re- Since the CO2 laser produces a beam of infrared light, it is nec-
search area are leading to significant advances in the development essary that the polymer absorb energy at the laser wave length
of machines and resins for the next generation of naturally-derived (10.6 μm). Otherwise an increase of laser energy power can com-
SLA polymers. As an example, soybean oil epoxidized acrylate was pensate low energy absorption. In the case of semicrystalline poly-
recently polymerized by SLA into a highly compatible scaffold with mers, the indicative sintering window can be considered the tem-
shape memory properties suitable for 4D Printing at physiological perature range between the onset of polymer melting and that of
conditions [156]. crystallization, as obtained from differential scanning calorimetry
[165]. Indeed, it is essential to avoid polymer crystallization that
3.2. Selective laser sintering (SLS) often results in curling or warpage of SLS parts. The fabrication
chamber is typically maintained at a temperature just below the
SLS is extensively used across different industrial sectors, in- melting or softening point of the polymeric powder to speed up
cluding electronics, aerospace, and automotive industries, for pro- and optimize the sintering process by minimizing internal stresses
totyping devices, small components, and cost-effective consumer and curl distortions. An inert gas atmosphere in the chamber to
good parts. hinder oxidative degradation of the polymer during the process
SLS is based on the selective sintering of a powder bed made is therefore required. In comparison to semicrystalline polymers,
of polymer, ceramic or hybrid particles by means of a computer- amorphous polymers soften gradually over a broader range of tem-
controlled laser beam [157]. Once the fabrication of a layer is com- perature between their Tg and flow temperature. For this reason,
plete, a new powder bed is mechanically spread over it by means they typically present lower internal stresses and distortions upon
of a roller or a blade system (Figure 2b). The powder sintering solidification from melt. However, their melt viscosity tends to be
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 9

relatively high, resulting in limited coalescence of polymer parti- crease the manufacturing velocity. After the fabrication of each
cles. layer, the construction platform moves downward a given distance
Indeed, low values of melt zero-shear viscosity (η) and sur- and a new powder bed is placed on top of the previous one to
face tension are necessary to achieve an adequate coalescence of carry on with the layer-by-layer process until the 3D object build-
polymer particles during sintering [166]. This aspect is of par- ing is completed. Like in the case of SLS, the unbound powder acts
ticular importance considering that unlike other melt processing as a support for the building of the successive layer. Depending
techniques (e.g., injection molding) additional material compact- of the properties required, post processing treatments, such as de-
ing under pressure is not provided. Temperature effects together powering, sintering, infiltration, coating, polishing and machining,
with gravity and capillary forces are the only factors providing are carried out [183]. In particular, post processing sintering and
the driving force for particles consolidation in SLS [13]. A η not infiltration of a binder are the most common approaches to in-
low enough is the cause of imperfections and poor surface quality, crease the density and mechanical strength of printed parts.
resulting in brittleness and instability of sintered parts, as often The liquid binder can be deposited through either drop-on-
observed in the case of amorphous polymers [164]. For instance, demand or continuous-jet printheads. In the first case the most
semicrystalline PA12 (nylon-12) (η=100 Pas) can be fully densified common approaches involve ejecting a drop from a nozzle as a
by SLS but polycarbonate (PC) which is amorphous (η=50 0 0 Pas) consequence of a piezoelectric deformation of the chamber con-
cannot, even if they show similar surface tension in the melt state taining the binder or a confined thermal vaporization and sub-
[13]. sequent volume expansion of a small liquid volume. Continuous-
Particles shape, surface and granulometry are crucial parame- jet printheads involve pumping the liquid through a gun body
ters since affect powder flowability during spreading to form a bed equipped with a piezoelectric crystal that creates an acoustic wave
of a given thickness (usually around 100 μm) and with a density generating a constant droplet output.
suitable for the sintering process. The required powder properties BJ presents a high versatility in terms of materials selection
are generally assured by particles shape close to a spherical geom- since it can be applied to powders made of metals, ceramics, poly-
etry and a narrow size distribution between 20 and 80 μm, with mers, or composites. In addition, the high production rate and
a low share of fine particles below 10 μm to avoid their agglom- the tunable manufacturing scale ranging from millimeter objects
eration [167]. As reviewed by Schmid and Wegener [164], different that can be scaled up to large-sized models in the meter range,
particle shapes can be obtained by varying the preparation tech- make BJ a flexible technique adaptable to the industrial produc-
nique. Spherical particles are usually obtained by co-extrusion pro- tion [5]. Shape alterations and geometrical defects as consequence
cesses of immiscible mixtures (e.g., oil droplets in water), potato- of solvent evaporation or post-processing treatments (e.g., dipping
shaped particles by precipitation processes starting from a poly- in a binder solution [184] and sintering [185]) often required to
meric solution, irregularly-shaped particles by cryogenic milling. stabilize the printed structure, are the major peculiar drawbacks
Polymer/ceramic composite microspheres can be obtained by solid of BJ limiting its application. In addition, mechanical properties
in oil in water (S/O/W) emulsification solvent evaporation methods and surface resolution are lower in comparison to what achieved
[168]. with other AM techniques, such as SLA and FDM, even after post-
Industrial consumption of SLS powder is dominated by PA12 processing treatment.
and its blends, with a minor percentage of the market distributed The most investigated biomedical application of BJ is the fab-
on other polyamides (e.g., PA11) and few other polymers, such as rication of ceramic-based bone implants. Considering the limited
thermoplastic elastomers (i.e., TPUs and polyether block amides) ceramic processing versatility of other AM techniques, research on
and PEEK, not including the so-called commodity plastics, such as BJ has produced a large body of literature on both in vitro and in
poly(ethylene) and poly(vinyl chloride) [169]. The market offer of vivo investigation of implantable devices for applications extend-
amorphous polymers for SLS is limited to polystyrene (PS), with ing from tissue regeneration to bone tumor therapy [111]. In addi-
few investigations on other polymers such as PMMA [170] and PC tion, BJ represents the most exploited AM technique in pharma-
[171]. The reason behind this very limited range of materials is ceutical sciences, as witnessed by the launch on the market of
that only a successful combination of the aforementioned intrin- the oral antiepileptic medicine Spritam® [107]. The versatility of
sic and extrinsic polymer properties allows a new powder to be BJ in bioactive formulations processing has been also exploited to
implemented in SLS. fabricate polymeric implants made of either a hydrophobic or hy-
As previously mentioned, several attempts have been made to drophilic polymer and loaded with a drug that can be released in
process biodegradable polymers by SLS, mainly aliphatic polyesters situ with a controlled kinetics [186].
for TE scaffolds production.[4], [166], [172–174] In particular, SLS Materials design and processing requirements. In the case of
of PCL [4], [174–177], PDLLA [166], [172], [178], and poly(3- a ceramic or metallic powder, the binder is composed of a solu-
hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) [179–181] also in tion or dispersion of a polymer or an inorganic material capable
combination with osteoconductive calcium phosphates, has been to form a film with adhesive properties upon drying or to react
reported. with the particle material. For instance, when powders made of
a bioceramic or a bioactive glass (BG) are employed, an aqueous
3.3. Binder jetting (BJ) solution of a hydrophilic polymer (e.g., hydroxypropyl methylcel-
lulose and dextrin) or an acidic binder able to react with the ce-
Being one of the first AM techniques employed in the biomed- ramic (e.g., phosphoric acid and citric acid) is employed [111]. BJ
ical field [182], BJ has been widely investigated for processing bio- of a polymeric powder is based instead on material swelling un-
compatible ceramics and polymers. Although, originally referred to der the action of the binder, and the consequent polymer chains
as 3D printing, in this review it will be called BJ in order to avoid interdiffusion and entanglement at the interface between different
misinterpretations related to the frequent use in literature of the particles.
term ‘3D Printing’ as synonym of AM. Various BJ parameters, including binder composition, powder
BJ is based on the selective deposition of a binder, usually in bed thickness, powder particles size and shape, printing and post-
the liquid form, on a powder bed (Figure 2c). The binder acts as processing parameters, affect the surface morphology and me-
an adhesive between powder particles to form a solid layer or to chanical properties of the fabricated part. Although literature on
bond or fuse adjacent layers together as well [8]. A multi-channel BJ parameters/device properties relationship is scarce in the case
inkjet head can be employed to deposit different binders or in- of polymeric powder, general considerations on this topic can be
10 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

made relying on what experimentally observed for BJ processing materials selection versatility, design freedom, automation degree,
of ceramics [183], [187], as well as for polymer powder processing mechanical properties, and accuracy of deposition details.
by SLS [163]. Overall, for a given polymer most of the processing ME-AM comprises a set of techniques based on the controlled
requirements concerns the binder characteristics in relationship to extrusion through a nozzle of a polymer kept at a temperature
the final application. above its Tg [203–206]. The raw polymeric material can be in the
A polymer binder should have a strong solvating power to form of a continuous filament, pellets, or powder, depending of the
achieve effective particles fusion by quickly dissolving them during technological solution adopted. The computer-controlled motion of
the time required to print one layer [183]. It typically consists of a the extrusion head and/or the construction platform allows the de-
good solvent or solvents mixture, e.g., chlorinated solvents for hy- position of the extruded polymer following a predefined pattern.
drophobic polymers like aliphatic polyesters and aqueous binders After the fabrication of each layer, the distance between the ex-
for hydrophilic polymers like polysaccharides. The material fusion trusion head and the platform increases of a given distance corre-
degree depends also on particles size since by decreasing it the sponding to that of the designed layer thickness, to obtain a 3D
polymer is more easily dissolved. However, particles with a diame- object with a layer-by-layer process.
ter lower than 20 μm are difficult to spread and tend to agglomer- FDM or fused filament fabrication, the oldest and most diffuse
ate. In general, polymer particles shape, surface and granulometry ME-AM technique, is based on the extrusion of a polymeric fila-
should be taken under control to assure suitable flowability of the ment through a heated nozzle (Figure 2d). In the case of a com-
powder to form a bed of a given thickness and density, as previ- plex geometry of the object to be manufactured, two independent
ously considered for SLS. extrusion nozzles can be used to simultaneously deposit a sup-
The binder formulation should possess suitable viscosity and port material and fabricate the 3D object. Other AM techniques in-
surface tension to properly eject, spread, and infiltrate in the pow- volving the thermal processing of polymeric materials as powder
der bed without wicking out considerably from the impact area, or pellets are based on different principles of control over molten
leaving behind a too rough surface. These parameters can be opti- polymer flow (i.e., pressurized nozzle, force-controlled plunger, or
mized to enhance printing resolution by employing a polymer so- rotating screw), namely 3D fiber deposition [205], precision extru-
lution as a binder, as demonstrated by depositing a 10 wt. % PCL sion deposition (PED)[204], and bioextrusion [207]. The main ad-
solution in chloroform onto a PCL powder bed [188]. Reference val- vantages brought by these approaches are that they do not require
ues recommended for commercial BJ printers are a viscosity of 20 polymer pre-processing into a filament, as they offer the possibil-
cPs and a surface tension of 35 dynes/cm. Binder vapor pressure is ity of directly developing composite or blend scaffolds by simply
another key parameter since a too low evaporation rate can worsen feeding a mixture of the components to the machine. On the other
the process resolution, while a too fast evaporation rate will likely hand, the optimization of the fabrication process is often more dif-
result in warping of the printed component as a consequence of ficult than in the case of FDM, and a higher temperature and/or a
internal stresses. For instance, warpage effects in polyester parts longer residence at high temperature is required to achieve a uni-
are avoided by replacing methylene chloride with chloroform as form extrusion.
binder, or using a mixture of them. In the case of a binder based A number of articles have reported on the integration of ME-
on acetone, which has high volatility and a lower solvating power AM with electrospinning at a fabrication level in order to develop
for aliphatic polyesters in comparison to chlorinated solvents, most multi-scale structures by imparting nanoscale features to macro-
of the porosity of the original powder bed is likely to be main- porous scaffolds [112], [207–209]. This approach couples the 3D
tained after printing at room temperature. An effective approach shape and mechanical stability of macrostructured scaffolds by AM
to increase the evaporation rate of binders with low vapor pres- with the enhanced material/cells interaction of micro/nanosized
sure is represented by the employment of a warm powder bed. electrospun fibers. In this way, the hierarchically-organized archi-
Besides being the most employed technique for AM of biomed- tecture found in native tissues can be recreated to offer greater
ical ceramics, such as osteoconductive calcium phosphates [189– surface area for proteins absorption and more binding sites to cell
197], BJ has been applied to process also aliphatic polyesters membrane receptors [11]. Cutting edge advancements on techno-
[198] and hydrophilic polymers, such as poly(vinyl alcohol) (PVA), logical integration of AM with melt electrospinning recently led
starch, maltodextrin, and cellulose derivatives [8], [199]. In addition to the development of a hybrid technique, that is, melt electro-
to the binding component, the deposited liquid may contain addi- spinning in a direct writing way or melt electrospinning writing
tives such as surfactants, viscosity modifiers, and bioactive agents, (MEW) enabling the fabrication of 3D scaffolds with high resolu-
to improve the printing process or functionalize the final device. tion of the architectural features [210]. MEW relies on the depo-
As previously mentioned, this aspect has been widely investigated sition of a stable electrified molten jet onto a collector translating
to develop orally delivered drugs by adding a pharmaceutical ac- at a speed (~1 m min−1 ) approaching that of the jet [71], [211].
tive ingredient to the binder formulation. In addition, extensive re- The distance between the spinneret and the collector is typically
search has shown that BJ is suitable for the preparation of drug- below 5 cm. Advanced MEW approaches lately resulted in the de-
loaded implants based on different biodegradable polymers includ- velopment of 3D layered polymeric scaffolds composed by axially-
ing PCL, poly(lactic-co-glycolic acid) (PLGA), and their blends [198], aligned submicrometric fibers [212]. In addition, different studies
PLLA [200], and PDLLA [201]. Advanced approaches allow the com- have been carried out to endow a polymeric scaffold with a dual-
bination of multiple drugs within one implant with a relatively scale porous structure by a sequential combination of ME-AM and
long-term release profile [202]. either gas foaming [114], [213] or salt leaching [113].
As it will be described in section 4.2, ME-AM techniques have
3.4. Melt extrusion-AM (ME-AM) been extensively investigated for the development of bone scaf-
folds based on aliphatic polyesters, such as PCL [214], PDLLA [215],
The Melt-extrusion class of AM techniques (ME-AM) is find- and PLGA [215], [216] that were thoroughly characterized both in
ing increasing industrial exploitation particularly in the automo- vitro and in vivo. In particular, successful clinical trials indicate that
tive, aerospace, and medical sectors for rapid production of func- PCL implants fabricated by means of FDM meet all the require-
tional prototypes, models, and parts of civil aircrafts or vehicles ments needed to receive approval for use in clinical bone recon-
produced in small number. In particular, they are the most investi- struction [20]. Although the majority of scaffolds by ME-AM have
gated AM techniques for the fabrication of polymeric scaffolds for been designed for hard TE, a consistent body of literature has been
TE as a consequence of the good compromise offered in terms of
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 11

also dedicated to scaffolds for regenerating cartilage and other soft temperatures. However, PLLA has been found to partially degrade
tissues [217], [218]. during thermal processing, particularly when a long residence at
Materials design and processing requirements. ME-AM is high temperatures is required like in the case of pneumatic di-
generally based on polymer melt/softening and crystalliza- rect extrusion-based systems [225]. Although FDM allows mini-
tion/solidification to shape the material in the desired form. The mizing the thermal residence time in comparison to other melt
polymeric material is processed at a temperature above its Tg , and extrusion techniques, polymer processing for filament preparation
above its melting temperature (Tm ) in the case of semicrystalline can anyhow involve thermal degradation. In addition, commercial
polymers. As a direct consequence, polymers intended for this ap- PLA filaments may contain inorganic additives that could react dur-
plication should meet some basic requirements such as suitable ing processing causing changes in polymer macromolecular struc-
viscoelastic properties and good thermal stability at the process- ture and morphology [226]. Careful drying to remove residual wa-
ing conditions. For this reason, thermoplastic polymers that hold a ter that can induce melt hydrolytic degradation, purging with in-
long history of industrial melt processing have been rapidly imple- ert gas during extrusion, and thermal stabilizing additives are pre-
mented in ME-AM. cautions recommended whenever this risk exists [227]. PCL has
The polymer molecular structure and the resulting inter- and also received great interest as biomedical polymer for FDM due
intramolecular interactions determine the polymer Tg and chain to its superior rheological and viscoelastic properties over many
stiffness, which together with polymer Mw and polydispersity in- of its resorbable-polymer counterparts, which render it easy to
dex affect its rheological properties in the rubbery/melt state. In manufacture and manipulate into complex macroshapes and mi-
the case of FDM, filament viscoelastic properties are also funda- crostructures [228]. Although this class of AM techniques is not
mental for successful material processing. A too stiff filament will the election one for fabricating drug-loaded devices, a number of
not permit winding onto spools, a brittle filament can snap, and polymers of common use in pharmaceutical formulation, including
a soft filament can be squeezed between the driving gears, in any PCL, PLLA, PVA, poly(ethylene glycol) (PEG), cellulose derivatives,
case compromising polymer feeding [219]. Most of the commercial ethylene vinyl acetate, and poly(vinyl pyrrolidone) (PVP) have been
FDM machines on the market require a filament diameter of 1.75 evaluated for this application [229], [230]. In addition, a range of
or 2.85 mm. In the case of home-made filaments, dimensional con- biostable biomedical polymers including PEEK, poly(ether ketone
sistency (e.g., 1.75±0.05 mm) should be assured in order to have a ketone), poly(ether imide), polysulfone, ABS, and PMMA are com-
constant feed rate during fabrication. mercially available as filaments for FDM.
Common feedstock materials for FDM include amorphous ther-
moplastics, such as ABS. Extending FDM technology to semicrys- 3.5. Solution Extrusion-AM (SE-AM)
talline polymers has been more challenging due to warpage of
the deposited part, often occurring as a consequence of crystalliza- SE-AM represents the most versatile and employed processing
tion during cooling [220]. Indeed, polymer chains drawing to form approach to plot hydrogel scaffolds [231], possibly laden with cells
more dense, crystalline regions results in shrinkage of the printed or cells aggregates (see section 3.6). In addition, the possibility
object that can warp and become detached from the build plat- of avoiding material thermal treatment as well as developing ad-
form, reducing its final quality. Anyhow, residual stresses and part vanced structural and pharmacological functionalization strategies
warpage can be consequences of thermal gradients within FDM- widens the range of applications of SE-AM by including hydropho-
manufactured objects made of either amorphous or semicrystalline bic polyesters processing [7].
polymers. Manufacturing in a heated chamber often represents a This class of AM techniques is based on the controlled extru-
successful means to reduce thermal gradients within the printed sion through a nozzle of a polymeric solution or suspension by
material. Loading the polymeric matrix with fillers characterized means of a pneumatic or mechanically-driven dispensing system
by high thermal conductivity (e.g., carbon fibers) has been also (Figure 2e). The extruded material is generally deposited in the
employed to vary material thermal expansion coefficient. This ap- form of a continuous filament following a pattern defined by the
proach has been found to reduce distortion or warpage effects, die motion of the extrusion head and/or the building plane. Polymer
swell, and to influence polymer crystallization rate and crystallinity solidification/gelation can be achieved by means of different mech-
[220], [221]. anisms, including among others: drying through solvent casting
The key to achieve high mechanical strength of the final printed [232], physical coagulation in a non-solvent bath [26], ionotropic
part is a successful interdiffusion and re-entanglement of the poly- gelation [233], chemical cross-linking [234], photocrosslinking un-
mer chains across filament-filament interfaces, as the melt rapidly der UV light [235], and polyelectrolytes complexation [236].
cools towards its Tg [222]. Interface polymer welding is affected Mechanically-driven dispensing systems rely either on a piston
by the macromolecular structure and morphology (i.e., amorphous or a rotating screw. The piston-based approach generally results
or semicrystalline) and by different processing parameters, such in better control of material feeding rate in comparison with i)
as the deposition speed, extrusion temperature and environmen- pneumatically-driven systems in which a delay related to gas vol-
tal temperature. Indeed, thermally-driven filaments welding is af- ume compression can occur, ii) screw-driven systems which are
fected by polymer chains alignment under extrusion shear forces generally indicated only for viscous materials due to the high shear
as well as by post-extrusion anisotropic stresses acting on poly- stresses involved. Technological solutions based on arrays of noz-
mer melt during its deposition. Thermostatically-controlled print- zles that can deposit multiple filaments were recently designed to
ing chamber and deposition platform are technological solutions fabricate thick samples in short time [237].
effective to control the non-isothermal polymer chain diffusive Typical resolution achievable with these techniques is on the
process during FDM. While molecularly-aware models and exper- scale of hundreds of microns in the case of hydrogels, and can be
imental investigations of this aspect have been reported for amor- pushed down to tens of microns by employing aliphatic polyesters
phous polymers [222–224] there is still a gap in literature address- [238], [239]. However, the recent application of the layer-by-
ing tools to predict deformation and welding behavior of semi- layer stacking principle to solution electrospinning, is fastening the
crystalline melts in FDM. translation of AM resolution to a viable micro/nanoscale [240].
PLLA is the most used semicrystalline polymer for FDM fila- Indeed, advanced technological approaches commonly referred to
ments thanks to its Tm low enough to carry out melt extrusion as electrofluidodynamic printing (EFDP) or near-field electrospin-
outside of a dedicated facility and, at the same time, high enough ning, comprising tailored processing conditions (e.g., short work-
for manufacturing objects that retain their shape at average use ing distance, high translation speed of the fiber collector, low volt-
12 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

age, and high-precision solution feeding) allows enhanced control solution viscosity and surface tension suitable for maintaining the
over high-resolution polymer patterns whose large surface area 3D shape of the polymer strand after its deposition. A too high
can be exploited for instance to provide nano/microstructural in- viscosity can hinder a continuous solution feeding, besides causing
structive cues to cells [241–247]. In addition, in this way it is pos- a too fast solidification of the polymer [260]. Samples with lim-
sible to implement AM with the great versatility of electrospinning ited thickness are typically obtained by extrusion in air due to the
in terms of drug loading approach and control over drug release aforementioned constrains on solvent and polymer concentration
kinetics [248]. Similarly, research on the integration of AM with selection. In the case of integration of SE-AM with freeze drying,
electrofluidodynamic techniques for particles fabrication is aimed a solvent with a relatively high melting point (e.g., 1,4 dioxane
at the controlled deposition of assembled polymer particles struc- for hydrophobic polymers, and water for hydrophilic polymers) is
tures possibly encapsulating cells [249]. necessary to optimize the lyophilization process [255]. Once the
SE-AM avoids the shortcomings associated with high tempera- aforementioned polymer solution variables have been fixed, a set
ture processing, particularly the risk of materials thermal degrada- of deposition and environmental parameters, including deposition
tion. Other relevant advantages include the possibility of process- velocity, solution feed rate, and environment temperature, need to
ing natural polymers into 3D hydrogels with a controlled shape be optimized. In the case of EFDP approaches, the basic electro-
and porosity, loading the polymeric device with a bioactive agent spinning requirements related to solution, electric field and en-
through direct blending methods, carrying out proof of concept vironment variables, analyzed in depth by a vast literature [263],
studies with few amounts of polymer (e.g., less than 1 gram), ob- [264], should be specifically considered in relationship to the pe-
taining a dual-scale porosity through integration with other pro- culiar technological solutions adopted. For instance, the reduced jet
cessing techniques [10]. Indeed, hybrid AM techniques integrated travelling distance related to the short working gap typically em-
at a fabrication level with either wet-spinning [7] or freeze dry- ployed in EFDP (e.g., 500 μm to 3 mm to have a stable cone-jet
ing [250] enable the combination of an interconnected network of mode) [240] should be taken into account to select a solvent with
macropores determined by the designed lay-down pattern, with a a suitable volatility.
microporosity in the polymeric matrix that can be pushed down AM approaches relying on polymer solution physical coagula-
in some cases to the nanometer scale [31]. In particular, the local tion in a liquid medium are typically based on a non-solvent-
microporosity can be obtained by extruding an organic solution of induced phase separation (NIPS). Any hydrophobic or hydrophilic
a synthetic [26], [251], [252] or natural [238], [253], [254] aliphatic polymer suitable to be processed by wet-spinning for fibers pro-
polyester into an ethanol bath, or through the cryogenic extrusion duction can be in theory processed by CAWS [7]. High solvent/non-
of a polysaccharide [115], [250], polyester [255], [256] or protein solvent miscibility is a fundamental requirement for NIPS to take
[257] solution followed by lyophilization. The size and concentra- place since it is based on polymer desolvation caused by solvent–
tion of the resulting micropores can be tuned in a certain range, non-solvent counter-diffusion. In addition, by employing a polymer
particularly in the case of computer-aided wet-spinning (CAWS) concentration higher than a critical threshold, the polymer solu-
by acting on processing and post-processing parameters, in order tion can separate into a polymer-lean phase dispersed in a contin-
to tune specific material properties depending on porosity (e.g., uous polymer-rich phase, resulting in the formation of a microp-
biodegradation rate, material/cell interaction and release of loaded orous matrix after polymer solidification. Different variables such
drugs). On the other hand, it should be considered that the gen- as polymer concentration, coagulation bath temperature and com-
eration of a microporosity leads to a lower material stiffness as a position, and deposition parameters (e.g., deposition velocity and
consequence of the increased specific void volume. solution feed rate) affect thermodynamics and/or kinetics condi-
Major shortcomings of this class of techniques are related to tions of the NIPS process and, as consequence, can be exploited to
the employment of organic solvents with their inherent environ- tune the macro- and micromorphology of the resulting construct
mental and industrial impact. In addition, there are risks associ- [7]. Warping effects are often observed both for amorphous (e.g.,
ated to residual solvents which, if not completely removed, may PMMA) [31] and semicrystalline (e.g., PHBHHx) [238] polymers due
compromise the biocompatibility of the device, besides altering to free volume variations during coagulation. Variation of process-
bioactive factors loaded in the polymer matrix. However, proper ing parameters (e.g., warming up the coagulation bath) and post-
post-processing protocols based on vacuum treatment, washing processing treatments (e.g., slow drying) can be effective in mini-
and drying are effective in completely eliminating organic solvents mizing residual coagulation stresses in the layered structure. CAWS
from polymeric devices [31], [258]. has been widely investigated to process a range of polyesters ei-
Materials design and processing requirements. As a conse- ther synthetic [26], [105], [251], [252] or derived from natural re-
quence of the great variety of physical-chemical approaches in- sources [238], [239], [253], [254], [258], as well as polyelectrolyte
cluded in this class of techniques, peculiar materials processing re- complexes of chitosan and poly(γ -glutamic acid) [34], [236].
quirements need to be defined depending on the different solidifi- Besides complexation of polyelectrolytes, various specific
cation/gelation mechanisms adopted. crosslinking strategies can be pursued to confer additive manu-
Different technological designs based on extrusion in air have factured hydrogels with the physical stability necessary to main-
been developed to plot a broad range of materials including so- tain a 3D porous structure and shape. The ability of different
lutions of linear or highly branched polymers, acrylic dispersions, natural polymers to undergo physical crosslinking upon extru-
inorganic solutions, and inorganic pastes combined with polymeric sion in a liquid medium, through straightforward mechanisms
binders that are then sacrificed by sintering the green part [8]. Par- (e.g., temperature-dependent physical gelation of gelatin [265] and
ticularly referring to biodegradable polymers, tailored SE-AM ap- ionotropic gelation of alginate in the presence of divalent cations
proaches were optimized to process synthetic aliphatic polyesters [266]) can be exploited for AM. However, chemical crosslinking
[256], [259–261] and/or polysaccharides [115] through their extru- is often required to achieve acceptable morphology control and
sion and controlled deposition in air. An organic solvent that evap- reproducibility. Indeed, as consequence of their poor mechanical
orates upon printing eventually resulting in material solidification properties and hydrophilic behavior, in most of the cases natural
is typically employed when a hydrophobic polymer is processed. hydrogels cannot maintain a filament shape for the required time
If the solvent vapor pressure is too high or too low, processing and eventually collapse under the weight of upper layers during
shortcomings, such as needle clogging, layered structure distortion fabrication.
or fusion, and unwanted porosity defects can occur upon extrusion Different chemistries have been proposed for hydrogel
[262]. The polymer concentration should be tuned in order to have crosslinking, such as photoinitiated reactions giving good temporal
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 13

and spatial control with various naturally-derived macromolecules Bioprinting is based on the ejection from a reservoir of a
[124], [267]. Once grafted onto the functional residues of the bioink composed by living cells or cells aggregates suspended in
polymer chain, photoactive moieties can induce the formation a medium, possibly in combination with biomaterials and bioac-
of covalent crosslinks when exposed to a specific wavelength of tive molecules [281]. The different bioprinting techniques applied
light in the presence of a photoinitiator. In particular, free radical within the scientific community can be classified on the basis of
polymerization of acrylate derivatives of proteins and polysaccha- the mechanism adopted to control the deposition. In particular,
rides has been widely investigated to design novel photoreactive extrusion, inkjet and laser-assisted bioprinting are the most em-
inks also suitable for fabricating cell-laden constructs [268], [269]. ployed ones (Figure 2f). Analogous attempts of photopatterning
Synthetic polymers are also investigated for AM of biomedical cells-encapsulating hydrogels by means of SLA have been reported
hydrogels with enhanced processing and mechanical properties. [282]. In this case, Vis light-based systems are also investigated in
For instance, Liu and Li [270], [271] recently developed a novel order to prevent cells damage related to UV light irradiation [124].
strategy based on UV exposure of an extruded pre-gel solu- Extrusion bioprinting is based on pneumatically- or
tion to form a polymeric double network hydrogel composed of mechanically-controlled extrusion of the bioink through a nozzle,
covalently crosslinked polyacrylamide and ionically crosslinked k- as described in the previous section. Pneumatic and piston-based
carrageenan. In any case, concerns about the non-degradability of dispensing systems are commonly used to achieve high cell via-
the backbones resulting from free-radical chain-growth crosslink- bility, rather than screw-driven dispensing that generates larger
ing as well as the biocompatibility of the preparation process are pressure drops at the nozzle with potential harm to cells [231].
often raised [272]. Multiple-nozzle extrusion systems have been also developed
Photoinitiated thiol-ene chemistry has been recently imple- to process and combine different hydrogel materials into a so-
mented in AM as an alternative crosslinking strategy for natural phisticated living construct and exploit advanced crosslinking
polymers [268], [273]. It is based on dimerization of thiols with re- strategies. For instance, two-nozzle systems were employed to de-
active carbon–carbon double bonds. In this case the polymerization velop double-hydrogel crosslinking strategies (e.g., ionotropic and
follows a step-growth radical mechanism resulting in high con- enzymatic crosslinking) [283–286], and to combine natural and
version of functional groups and low polymerization shrinkage. In synthetic polymers with different processing requirements into
comparison to free radical polymerized diacrylates, a lower radical a hybrid scaffold [287–289]. Extrusion bioprinting processes can
initiator concentration is required, the reaction kinetics is generally rely on hydrogel shear-thinning properties, physically/chemically-
more rapid, and the photoinitiated thiol-ene can form more ho- triggered gelation inside a liquid bath, extrusion inside a pseudo-
mogeneous hydrogel networks [274], [275]. In addition, biodegrad- plastic or granular bath, or simultaneous dispensing of a bioink
able and bioactive molecules containing thiols can be straightfor- and a crosslinking agent using co-axial needles assemblies [290].
wardly conjugated to the network as pendant groups or crosslink- Inkjet bioprinting involves ejecting a bioink from a nozzle in
ing bridges. Examples of hydrogels fabrication based on thiolated the form of droplets (~10-50 μm in diameter) to a building surface
natural (e.g., alginate [274] and gelatin [273]) or synthetic poly- [291]. The bioink is forced through the nozzle under the action of a
mers, e.g., poly(glycidol) [276], are reported in studies focused on pressure commonly generated by either a piezoelectric actuator or
AM combined with UV exposure. a thermal element that superheats the ink and creates an expand-
Hydrogel solutions usually behave as a non-Newtonian shear ing vapor bubble. Alternative technological solutions are based on
thinning fluid, whereby an increase in shear rate leads to a de- electromagnetic pin actuators, or interdigitated gold rings gener-
crease in shear viscosity as consequence of chains disentanglement ating acoustic waves. The bioink is typically composed by a sus-
related to attenuation of molecular interactions [277]. Functional- pension of cells in a solution of a polymer that is either physically
ization of proteins and polysaccharides through acrylate derivati- or chemically crosslinked upon deposition. Other strategies involve
zation of pendant groups can lead to variation in their solution ejecting cells suspended in a solution of a crosslinking agent (e.g.,
rheological properties. This is often the case of GelMA showing CaCl2 or thrombin) over a biopaper substrate (e.g., alginate or fib-
increased viscosity in comparison to the native polypeptide as a rinogen) which is crosslinked upon bioink deposition [292], [293].
consequence of the increase in lysine side chain length, resulting Laser-based bioprinting involves coating a bionk layer with a
in increased chains entanglement and intermolecular interactions laser-absorption layer (e.g., titanium oxide or gold) which is fixed
[278]. Such rheological modifications can on one side help in sta- to an optically-transparent support (e.g., glass). Laser pulses are fo-
bilizing the extruded filament and on the other cause experimental cused through the glass side into the absorbing layer which heats
issues such as needle blockage or discontinuous extrusion. either evaporating or decomposing a small portion of the hydrogel
An effective strategy to process hydrogels with low elastic mod- in contact with it [294–296]. The generated high-pressure bubble
ulus and avoid their collapse is printing and embedding the mate- propels a small droplet of bioink towards a building stage through
rial within a second hydrogel support bath.[279] Indeed, by select- an orifice-free process.
ing a medium with a density close to that of the printing material, A vast literature is focused on the development of bioinks for
it is possible to achieve low-gravity conditions in which soft hy- the aforementioned techniques, based on either natural polymers
drogels can easily maintain the intended 3D geometry. and their derivatives (e.g., gelatin [124] and collagen [297]) or syn-
thetic polymers (e.g., PEG [298], pluronic [299], and PVP [300]).
They have been combined with a wide array of cells for the en-
3.6. Bioprinting gineering and in vitro modelling of health and pathological tissues
such as bone, cartilage, cardiac tissue, nerves, and lung tissue, as
Bioprinting, also referred to as organ printing, enables pattern- well as ovarian and cervical cancer [280].
ing and precisely placing biologics, including living cells, nucleic The main advantages of these techniques are related to the pos-
acids, drug particles, proteins and growth factors, to recapitulate sibility of directly fabricating customized living tissue engineered
tissue anatomy, biology and physiology. The tremendous scientific, constructs with timely and spatially-controlled deposition of cells
diagnostic and therapeutic potential of bioprinting technologies is and other biologics. This allows the development of controlled
witnessed by relevant fast-growing literature, together with the biomimetic microenvironments by exploiting the self-organizing
marketing of bioprinters and bioprinted tissues for different appli- properties of cells and tissues. Currently, the applicative potential
cations, ranging from TE and in vitro modelling to pharmaceutics of bioprinting is mainly related to in vitro applications rather than
and high-throughput screening [280]. surgical approaches. Indeed, before bioprinted organs are ready to
14 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

be transplanted into humans, several important ethical concerns constructs starting from bioinks with low viscosity [268]. In any
and regulatory issues need to be addressed [301]. Aspects like risks case, a good compromise among physical printability related to
of significant harm associated with human testing, lack of a spe- bioink rheology, mechanical stability given by polymer crosslink-
cific regulatory framework, combined with ethical questions of ir- ing, and biological functionality as a result of cells viability and mi-
reversibility, loss of treatment opportunity, and replicability still gration in the polymer network, is a challenging bioprinting aspect.
represent restrictions to bioprinting for humans. Representative examples of efforts spent on this topic are studies
Materials design and processing requirements. The strict pro- on bioprinted GelMA optimized through two-step thermo/photo-
cessing requirements related to printing living materials through a crosslinking strategies [269] or new Vis light photoinitiating ap-
mild and cell-friendly process, greatly limit the range of techno- proaches [307].
logical solutions and materials for bioprinting [291], [302]. FDM As it will be described in detail in section 4, various proteins
and SLS do not meet bioprinting requirements since high tem- including collagen, gelatin, fibrin and silk fibroin, and polysaccha-
perature processing conditions are unsuitable for cell viability. As rides including alginate, cellulose, chitosan and agarose, have been
previously mentioned, a few attempts on SLA-based bioprinting widely investigated for bioprinting resulting in the development of
have been made. Although Vis light employment allows preventing a large number of bioink formulations currently under investiga-
UV-related cell DNA damages, concerns related to cytotoxic pho- tion or on the biomaterials market. Multiple polymers bioinks (e.g.,
toinitiators, photoreactive oligomers, and crosslinking agents still alginate with gelatin/fibrin, cellulose with alginate) have been of-
exist. For these reasons, extrusion and inkjet bioprinting are the ten employed to meet different processing, mechanical, and func-
most exploited techniques along with a number of studies showing tional requirements necessary to develop biomimetic tissue-like
that laser-assisted bioprinting can be suitably employed to print constructs [308].
cell-laden hydrogels. As it will be discussed in detail throughout
the whole paper, various natural polymers, including proteins and 4. Biodegradable Polymeric Biomaterials for AM
polysaccharides from different sources, have been successfully em-
ployed in combination with crosslinking agents, cells, growth fac- The most widely investigated polymeric materials for AM
tors and other proteins to develop bioinks designed for bioprinting. biomedical applications are those susceptible to be degraded in
Bioink properties are generally tailored to meet the peculiar physiological environments. As previously discussed, this is justi-
technological requirements and constrains of a selected fabrication fied by the tremendous advantages of AM over other materials pro-
technique. In general, a non-Newtonian shear thinning behavior is cessing techniques in controlling key compositional, structural, and
fundamental for extrusion and inkjet bioprinting of cell-laden hy- functional parameters of polymeric systems designed for TE and
drogels in order to achieve high print fidelity and cells viability other advanced biomedical applications.
[303], [304]. In any case, bioink viscosity is a crucial parameter The following sections are dedicated to an overview of the most
that needs to be optimized considering that highly viscous poly- exploited classes of polymers in this context, obtained from natural
mer solutions on one side can hold the deposited shape for long sources or through synthetic routes. Critical aspects regarding their
time with high print fidelity, on the other side they require higher extraction or synthesis, purification, and physical-chemical modifi-
pressure to be processed resulting in higher stresses acting on cells cation to make them processable by means of specific AM tech-
and obvious limitations on gauge size and print resolution. This is- niques are highlighted. Cutting edge advancements on their de-
sue becomes relevant in the case of inkjet bioprinting that involves velopment into functional biomaterials are outlined by discussing
relatively high shear rates (105 -106 s−1 ), while in the case of ex- representative articles focused on relevant key aspects.
trusion bioprinting the processing viscosity window is much wider.
Reference bioink viscosity ranges are 1-10 mPas for inkjet bioprint- 4.1. Polymers from Natural Sources
ing, 30 to 6 × 107 mPas for extrusion bioprinting, and 1–300 mPas
for laser-assisted bioprinting [291]. After the oil crisis in the seventies, the use of polymers that
Another relevant limitation is on cell density which is typically could be obtained from renewable resources and eventually sus-
maintained lower than 106 cellsmL−1 to avoid constrains such as ceptible of degradation in the environment was seriously consid-
cell settling and sedimentation, nozzle clogging, droplet spread- ered as a reliable industrial alternative in agreement with the in-
ing, and worsened resolution [305]. Concerns about the deleterious creasing awareness of ecological issues. The parallel growing in-
effects on cells viability of high temperatures (~300°C) generated terest on regenerative therapeutic approaches, as well as on con-
in thermal inkjet have been overcome by modern technologies in trolled and targeted pharmacological strategies involving the em-
which thermal heating lasts few microseconds (~2 μs) [306]. In ployment of biodegradable materials has further encouraged re-
laser-assisted bioprinting the volume of the transferred material as search on polymers derived from natural sources. Specifically re-
consequence of laser pulse absorption is a function of the laser en- ferring to biomedical applications, natural polymers offer different
ergy and spot size, biomaterial composition, and coating thickness advantages over their synthetic counterparts, generally including
[294]. In general, only the small fraction of the bioink in the im- intrinsic biological signaling, surface chemistry promoting cell ad-
mediate vicinity of the absorbent layer is volatilized and the bulk hesion, and cell-responsive degradation and remodeling. In addi-
biomaterial undergoes only little to no heating. tion, the pendant functional groups present in the repeating units
The hydrogel molecular design strategies outlined in section 3.5 of proteins and polysaccharides provide many options for chemical
should take into account constraints related to the use of cells derivatization tailored to bioactive functionalization [309].
in the case of bioprinting. Many approaches exploit the capability One of the most challenging aspects involved in the biomed-
of different natural polymers to form physically-stable hydrogels ical application of naturally-derived polymers regards their shap-
under mild conditions, such as in the case of ionotropic gelation ing into a given geometry and a porous architecture that can be
of alginate or enzymatic crosslinking of fibrinogen. As previously maintained for predefined time under physiological, aqueous con-
discussed, photoinitiated radical chain polymerization of acrylate ditions. For this reason, a large body of literature has been dedi-
derivatives and step-growth thiol-ene reaction are the most ex- cated to physical-chemical modification of natural materials aimed
ploited strategies to chemical crosslink cell-laden hydrogels dur- at their processing into devices with adequate physical proper-
ing extrusion or after deposition, as well as for direct SLA biopat- ties. An appealing aspect of many natural polymers is the possi-
terning. Advanced approaches rely also on combined photoreac- bility of forming biocompatible hydrogels providing a highly hy-
tive acrylate and thiol-ene chemistries to develop complex living drated 3D environment for the cells. Additive manufactured hy-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 15

drogels made from proteins (e.g., collagen and gelatin) or polysac- Source of extraction. Collagen is a structural protein of native
charides (e.g., alginate and chitosan) are commonly employed for extracellular matrix (ECM) of various biological tissues, including
TE and other biomedical applications. As it will be discussed in skin, bone, cartilage, teeth, tendons, and blood vessels [231]. Types
depth in the following sections, the huge progress made by bio- I–IV are the most common among over twenty-two types of col-
materials researchers has enabled the successful application of lagen found in mammals. Collagen for biomedical applications is
different AM approaches to naturally-derived polymers, including extracted from bovine or porcine skin, as well as from Achilles ten-
SLA of photocrosslinkable derivatives of proteins and polysaccha- don of bovine or equine origin using neutral/slightly alkaline salt,
rides, BJ of hydrogels using water as a binding agent, and ex- acidic, alkaline, or acidic/proteolytic enzymes solutions followed by
trusion of hydrogel-forming aqueous solutions. Indirect AM has repeated salt precipitation [385]. Engineered recombinant bacterial
been also proposed as an effective tool for the production of self- collagen is under investigation as a reliable and cost-effective alter-
supporting porous hydrogels with a predefined shape. For instance, native to avoid immunogenicity, infection transmission, and batch
Van Hoorick et al. [310] employed an additive manufactured sac- variability [386].
rificial polyester scaffold that was selectively dissolved after UV Physical-chemical modification. Collagen is composed of three
crosslinking of the infiltrated hydrogel phase. polypeptide chains, with similar compositions at high content of
As previously introduced, natural polymers are more challeng- glycine (Gly) (near 33%) and imino acids (near 20%), that wrap
ing to build into scaffolds by AM in comparison to synthetic poly- around one another with a right-handed twist forming a tightly
mers, and generally result in poor resolution, particularly along packed triple helix (Figure 3a) [387], [388]. Collagen fibrils are
the print (build) direction. However, cutting edge advancements stabilized by covalent crosslinks between amino acid residues of
on hydrogels physical-chemical tailoring are showing the possibil- different chains. However, in the human body collagen undergoes
ity of integrating an enhanced fabrication reproducibility and reso- degradation via proteolytic enzymes (e.g., collagenases) causing a
lution with the superior biocompatibility and bioactivity of natural fast impairment of its structural role as implant biomaterial [389].
polymers. In addition, the recent application of AM to microbial Collagen stability in physiological environments can be increased
polyesters [311] is leading to the development of the next genera- through its combination with mineral crystals [390], or polymers
tion of load-bearing scaffolds with physical and mechanical proper- of either natural (e.g., elastin and glycosaminoglycans [391], HA
ties comparable to those of synthetic aliphatic polyesters, but com- [392]) or synthetic (e.g., acrylates [393], [394]) origin. In addi-
plying with the concept of sustainable development. tion, various chemical crosslinking strategies have been developed,
The most investigated proteins, polysaccharides and microbial to covalently bind the amine side groups of polypeptide chains.
polyesters in this field are following described by highlighting the Concerns raised towards the potential toxicity of the crosslinking
different strategies developed to optimize their AM processing into agents, e.g., glutaraldehyde [395], have encouraged research on al-
biocompatible and biofunctional devices (Table 1). Although also ternative approaches involving chemical modification of collagen
other naturally-derived macromolecules are employed as blend side groups to enable crosslinking by free radical polymerization
components for AM due to their biomechanical (e.g., elastin [312]) [396] or via enzymatic reaction [397].
and gelation properties (e.g., agarose [313]) this review specifically Biomedical use. Collagen in the form of sponges [398], gels
focuses on those polymers that have been giving the most promis- [399], [400], or electrospun ultrafine fibers [401] has been widely
ing results as main material constituent. investigated for bone and cartilage TE. In addition, a large body
of literature has been focused on collagen-based materials for
skin repair and blood vessel engineering to exploit its thrombo-
4.1.1. Proteins
genic properties. For instance, electrospinning has been widely in-
The rich landscape of proteins as a result of biological evolution
vestigated to fabricate micro/nanostructured collagen in combina-
offers a variety of genetically encoded structures and properties
tion with other natural [402] or synthetic polymers [403], [404] .
spanning many length scales. Indeed, the proteins repetitive na-
The tremendous progress achieved is witnessed by a number of
ture facilitates the formation of long-range ordered molecular sec-
collagen-based devices currently available in the biomedical mar-
ondary structures capable to self-assemble into complex 3D hierar-
ket worldwide, such as dermal membranes and sponges for local-
chical architectures characterized by peculiar mechanical, chemical,
ized delivery of antibiotics (e.g., Sulmycin® Implant, Collatamp®
electrical, electromagnetic, and optical properties [380]. As a con-
G, Septocoll E) [405], skin engineered substitutes (e.g., Apligraft®)
sequence of the aforementioned advantages, various proteins (e.g.,
[406], [407], as well as hemostatic sealants made from bovine col-
collagen, gelatin, silk fibroin, and keratin) have been investigated
lagen either in combination or not with thrombin (e.g., Sulzer-
for biomedical applications resulting in commercial products intro-
Spine® Tech, CoStasis® Surgical Hemostat, Floseal®, Gelfoam®,
duced in the surgical routine.
UltraFoamTM ) [408]. Two of the first TE clinical trials involved the
Because of their poor thermal stability, AM of proteins mostly
implantation of engineered autologous bladder [409] or vaginal
relies on extrusion of aqueous solutions or suspensions. Proteins
organs [410] based on scaffolds made from collagen or a colla-
physical stabilization in physiological environments is achieved
gen/poly(glycolic acid) (PGA) composite.
by exploiting their inherent gelation properties and/or applying
AM application. The aforementioned advantages in terms of
physical-chemical crosslinking. The high cell affinity of proteins
biocompatibility and bioactivity have been widely exploited in AM
is particularly exploited in bioprinting approaches. Indeed, as dis-
by employing collagen as main solution extrusion component, in
cussed in Section 3.6, most of the bioinks for AM are based on
the formulation of inks for bioprinting, and in BJ binders to fabri-
collagen, gelatin, and/or fibrin (Figure 3). Chemical modification of
cate collagen-Ca-P composites [320]. The low viscosity of collagen
proteins, as witnessed by the large body of literature on gelatin
solutions, gels, or dispersions commonly employed for biomedical
derivatives, has been also investigated as a means to endow pro-
purposes, and the slow gelation of collagen, are main disadvan-
tein with photocrosslinkable moieties exploitable for SLA or pho-
tages for direct fabrication of predesigned scaffolds [411]. Different
tocrosslinking of extruded polymers.
strategies have been adopted to increase the reproducibility, geo-
metrical accuracy, and control over structure of additive manufac-
4.1.1.1. Collagen. Collagen biodegradability, relatively low antigenic- tured collagen. These include among others extruding directly on
ity, cell-binding properties, and peculiar biomechanical behavior a cryogenic stage followed by lyophilization [257], [314], applying
have attracted great interest for different biomedical applications a crosslinking agent (e.g., aerosolized or nebulized NaHCO3 ) during
ranging from dermal dresses to TE [384]. extrusion [315], extruding hybrid inks containing polymers more
16 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Table 1
Naturally-derived biomedical polymers for AM.

Polymer AM technique Processing conditions and material design

Collagen SE-AM Extrusion and stabilization of collagen solutions through: cryogenic plotting [257,314], in situ crosslinking [315], hybrid
(bioprinting) inks (e.g., fibrin/alginate–collagen) [316,317], high density collagen dispersions at controlled pH [318,319], heated
printing chamber [297].
BJ Collagen dissolved into the binder solution plotted onto a ceramic bed [320].

Gelatin SE-AM Extrusion of: a warm solution containing gelatin into a cold environment and stabilized through chemical crosslinking
(bioprinting) [234], enzymatic crosslinking [321] or combination with other natural polymers (e.g., alginate [322,323], fibrinogen
[24], chitosan [324] and HA [325]) suitable to be more stably crosslinked; a solution of GelMA followed by
post-processing UV crosslinking [124,235,307]; a solution of norbornene-modified gelatin solutions followed by
photocrosslinking [273,326,327];
SLA Photocrosslinking of: GelMA [19,31–33]; a thiol–ene clickable derivative (e.g., allylated gelatin) [273].

Fibrin Bioprinting Controlled deposition of a thrombin solution containing cells or growth factors onto a fibrinogen substrate used as a
biopaper [293,328], [329].
Mixing fibrinogen/thrombin at low temperature in a dual-chamber single nozzle extrusion system just before their
deposition [330].
Controlled deposition of a solution containing fibrinogen and other hydrogel-forming materials (e.g., gelatin and HA)
followed by enzymatic crosslinking [331–333].
Silk fibroin SE-AM Controlled deposition of aqueous solutions of fibroin: stabilization with glycerol [334], blending with other natural
(bioprinting) polymers acting as (sacrificial) bulking agent amenable to be physically [335-337] or enzymatically [338] crosslinked.
SLA Photocrosslinking of methacrylate derivatives of fibroin [339].

Chitosan SE-AM Acetic acid solution of chitosan: NaOH pipetting on deposited fiber [340]; extrusion into a coagulation bath [341,342];
optimizing acid solution composition (acetic acid/lactic acid/citric acid) followed by post-processing NaOH immersion
[343,344]; cryogenic deposition followed by lyophilization/gelation [345–347].
Polyelectrolyte complexation: extrusion of a PEC suspension into ethanol [34,236]; extrusion of a chitosan suspension
in an alginate solution and post-processing pipetting of HCl [348].
SLA Combination with photocrosslinkable polymers (e.g., PEGDA) [153].

Alginate SE-AM Extrusion of: an (cell laden) aqueous solution of alginate stabilized through submerged crosslinking [233,349–353] or
(bioprinting) aerosol spraying [354]; norbornene functionalized alginate and UV-induced thiol-ene crosslinking [274].
SLA Photoreticolation of alginate derivatives with pendant phenolic hydroxyl groups [267].

Cellulose SE-AM Extrusion of aqueous solutions/suspensions containing cellulose: blending with physically-crosslinkable polymers (e.g.,
(bioprinting) alginate [353,355–358]), coagulation by means of a non-solvent [359], chemical crosslinking [360,361], extrusion of
cellulose acetate dissolvent in a highly volatile solvent followed by cellulose regeneration [362] .
ME-AM Cellulose fillers (e.g., nanocrystals, fibers and wood flour) for thermoplastic polymers (e.g., PLLA, PCL or PVA) [363].
SLA Cellulose nanofillers for stereolithographic resins [364,365].

Hyaluronic SE-AM Extrusion followed by photoreticolation of: HA solutions containing photocrosslinkable hydrogels (e.g., GelMA [366]
acid (bioprinting) and hydroxyethyl-methacrylate-derivatized dextran [367]); HAMA formulations containing other polymers (e.g., Gel-MA
[368] and poly(N-isopropylacrylamide)-grafted HA [369]); HAMA modified with moieties giving supramolecular
assembly [303,370]; thiolated HA formulation containing a suitable crosslinker [371–373].
SLA (bioprinting) Photoreticolation of HAMA-based formulations possibly containing cells and other photocrosslinkable polymers
[117,374].

PHA SLS Sintering of: PHB powder [375–377]; Ca-P-loaded PHBV microspheres [179–181].
ME-AM FDM of filaments made of PCL/PHBV blend [378] or PHB/PLA blend containing a plasticizer [379].
SE-AM CAWS extrusion into an ethanol bath of a PHBHHx-based solution in chloroform or THF [238,239,253,258], or a
PHBHHx/chloroform/ethanol mixture [254].

readily crosslinkable (e.g., fibrin and alginate) [316], [317], extrud- ing those of native auricular cartilage were fabricated by injecting
ing a high density collagen dispersion at a controlled pH [318], a chondrocytes-containing collagen hydrogel in an ABS mold fabri-
[319], and heating the printing chamber [297] (Figure 4a). The cated by FDM [416].
plotted structures can be further stabilized by freeze-drying or
chemical crosslinking. In this way the scaffold can exhibit high 4.1.1.2. Gelatin. Gelatin is widely employed in biomedical applica-
shape and dimensional fidelity with the designed model, being tions thanks to its biocompatibility, biodegradability, and versatil-
composed by fibrillar collagen strands with diameter of few hun- ity in forming physically-crosslinked hydrogels in physiological en-
dreds of micrometers that are stable after incubation in cell culture vironments [417].
medium. Source of extraction and physical-chemical modification.
The fast progress on AM of collagen has led to the success- Gelatin is derived from denaturation of collagen through hydrol-
ful free-form fabrication of complex living architectures. For in- ysis of amide groups into carboxyl groups through an alkaline or
stance, a proof of concept study on human skin bioprinting in- acid process, yielding to a higher and lower conversion, respec-
volved the use of keratinocytes and fibroblasts as constituent cells tively [418]. The proteins resulting from the two processes have
to reproduce the epidermis and dermis, and collagen to repro- different isoelectric points as a consequence of a different carboxyl
duce the dermal matrix of the skin [315]. Different bioprinting groups density. They can interact with charged molecules at a
processes and bioinks are currently marketed for drug screening physiological pH to form polyion complexes as positively- (Type A)
and in vitro modelling of soft and hard tissues. Some examples are or negatively-charged (Type B) basic gelatin. This aspect has been
the exVive3DTM Human Liver Model for drug screening [412], the widely exploited for the controlled release of charged biomolecules
BioInkTM [413], the Lifeink® 200 [415], and the OsteoInkTM [414]. including proteins and plasmid DNA, with potential application in
Living tissue engineered constructs based on collagen were also different biomedical sectors [419]. Differently to collagen, gelatin
developed by means of indirect AM. As an example, external ear from different sources typically show reduced structural variations
anatomical constructs with biomechanical properties recapitulat- as a result of the denaturation of the protein tertiary structure.
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 17

Figure 3. Structure of proteins commonly employed in biomedical AM. (a) Triple chain structure of collagen fibrils and chemical structure of the most common tripeptide
sequence found in collagen, composed of glycine (Gly), proline (Pro) and hydroxyproline (Hyp) sequences (reproduced with permission [66]). (b) Scheme for the preparation
of photocrosslinked GelMA through reaction of gelatin and methacrylic anhydride for grafting methacryloyl substitution groups (RGD domains illustrated as red segments,
and their chemical structure is depicted within the inset) (reproduced with permission [381]). (c) Fibrin polymerization (α chains shown in blue, β chains in green, and γ
chains in red; α C domains are in gray) (reproduced with permission [382]). (d) Schematic of silk fibroin structure (insets showing the fibril overall structure and the fine
β -sheet antiparallel alignment of polypeptide chains) (reproduced with permission [383]).

Figure 4. AM of proteins. (a) Digital model of print path (i) and printed collagen hydrogel (ii) developed starting from a CT scan of sheep meniscus (reproduced with
permission [297]). (b) Gel-MA-based hydrogel constructs fabricated by extrusion followed by either UV- or Vis-light photoreticolation (reproduced with permission [307]).
(c) Representative images of hydrogels by inkjet printing of a suspension of human microvascular endothelial cells in a thrombin solution onto a fibrinogen substrate,
showing a printed grid (i), minor defects (ii) and fiber alignment (iii) (reproduced with permission [328]). (d) Schematic of inkjet bioprinting strategy for 3D silk fibroin
cellular constructs using sacrificial alginate (reproduced with permission [337]).
18 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Such molecular and supramolecular modifications usually leads to In this way methacryloyl substitution groups are introduced on the
better solubility and less antigenicity of gelatin as compared to reactive amine and hydroxyl groups of the aminoacidic residues
collagen [420]. However, gelatin retains the polyaminoacid struc- (Figure 3b). GelMA photocrosslinking can be carried out in aque-
ture of collagen containing arginine-glycine-aspartic acid (RGD) se- ous environments under mild conditions with the presence of a
quences promoting cell attachment, together with the target se- photoinitiator. This inherent advantage makes GelMA the most
quences of enzymes involved in its degradation in vivo. Gelatin exploited naturally-derived polymer for photoinduced free-radical
can be either physically- or chemically-crosslinked to form hydro- crosslinking [381]. GelMA formulations enable good control over
gels with controlled stability in physiological environments. For their rheological properties and chemical reactivity depending on
instance, in the case of glutaraldehyde crosslinking aldehyde car- the degree of methacryloylation and polymer concentration. The
bonyl groups react with free amino groups of lysine and hydroxyly- resulting crosslinked hydrogels retain the intrinsic polypeptide bio-
sine aminoacidic residues to form covalent bonds between gelatin logical properties, such as enzyme-degradability and cell-adhesion
molecules at either intramolecular or intermolecular scale. In this promotion. Different microfabrication techniques have been used
case, by varying the crosslinking density it is possible to tailor in to microstructure photocrosslinked GelMA hydrogels, including
vivo gelatin degradation by matrix metalloproteinases (e.g., colla- photopatterning, AM approaches, micromolding, self-assembling,
genase) [421]. microfluidics, and biotextile [381]. Nichol et al. [150] were among
Biomedical use. Due to the aforementioned biocompatible and the first to micropattern functionalized gelatin with a top-down
bioactive properties, as well as the demonstrated hemostatic and approach involving UV irradiation of preformed arrays of GelMA,
adhesive behavior, research on gelatin eventually resulted on the resulting in a variety of biocompatible shapes and configurations
marketing and clinical use of various biomedical products, includ- with tunable hydration and mechanical properties.
ing biological glues, topical hemostatic agents, hemostatic sponges The first articles on layer-by-layer SLA crosslinking of GelMA
(e.g., Gelfoam®), films used in neurological, thoracic and ocular to fabricate 3D patterned structures were published in 2012 [154],
surgery (e.g., Gelfilm®), and microcarriers for cell culture (e.g., [155]. After that, several studies have reported the SLA processing
CultiSpher-G®).[422] In addition, a large body of literature has of resins based on GelMA to fabricate substrates with low toxic-
been published on gelatin in the form of disks, 3D hydrogels or ity and excellent cell-adhesion properties [124]. For instance, hu-
microparticles, as carriers for the delivery of growth factors (e.g., man meniscus cells-seeded GelMA scaffolds by SLA were placed
transforming growth factor beta-1, bone morphogenetic protein-2 ex vivo in surgically created defects in human meniscus tissue
and -7 (BMP-2 and BMP-7), and insulin-like growth factor [423– resulting in good integration and new bonding tissue at the in-
426]) or for encapsulating different cells including chondrocytes terface after 3 weeks of culture.[438] The fabrication of pat-
[427], [428], osteoblasts [429], mesenchymal stem cells (MSCs) terned hydrogels possibly encapsulating cells was also investi-
[430], [431], and preadipocytes [432]. In addition, gelatin has been gated by means of SE-AM of GelMA followed by post-processing
often used in combination with synthetic polymers to develop hy- UV crosslinking. As an example, Billiet et al. [235] proposed the
brid materials with enhanced bioactivity, like in the case of com- use of a photoinitiator with enhanced biocompatibility, i.e., 2,2 -
posite polyurethane (PUs)/gelatin constructs with optimized hu- Azobis[2-methyl-N-(2-hydroxyethyl)propionamide] (VA-086) com-
man umbilical vein endothelial cells adhesion and proliferation pared to the conventional Irgacure 2959, and successfully fabri-
[433]. cated GelMA scaffolds with fully-interconnected pores networks.
AM application. At the physiological temperature (37°C), gelatin This approach was exploited to fabricate mechanically stable hepa-
dissolves as a colloidal sol and it can reversibly form a gel when tocarcinoma cell line-laden GelMA scaffolds with high cell viability
cooled (< 29°C) because of a conformational change from coil to (>97%) through the optimization of printing pressure and needle
helix that leads to chain association and eventually the forma- shape. Vis-light-induced photoreticolation was also proposed as a
tion of a 3D network [265]. This temperature-dependent gelation means to crosslink GelMA in order to avoid cells damage risks re-
property represents a noteworthy advantage for SE-AM allowing lated to UV irradiation [124], [307]. Wang et al. [124] employed
extrusion of a warm gelatin solution into a cold environment to a mixture of PEGDA and GelMA with eosin Y as Vis-light pho-
form a gel with a customizable shape. Low mechanical resistance toinitiator to encapsulate NIH 3T3 fibroblast cells in 3D structures
and structural stability in aqueous environments are major short- with 50 μm resolution, achieving 85% cell viability for at least five
comings of additive manufactured gelatin. Indeed, even if physi- days. Analogously, Lim et al. [307] described a new Vis-light initi-
cally crosslinked, gelatin tends to dissolve again when exposed to ating system based on the water-soluble initiators ruthenium and
physiological conditions. Different strategies for gelatin stabiliza- sodium persulfate, which yielded higher viability of cells encap-
tion to form 3D hydrogels by AM has been followed, such as glu- sulated in Gel-MA in comparison to what achieved with conven-
taraldehyde crosslinking [234] and combination with other natu- tional UV systems based on Irgacure 2959 (Figure 4b). In any case,
ral polymers suitable to be more stably crosslinked (e.g., alginate extrusion of GelMA requires a strict control of the bioink, nozzle,
[322], [323], fibrinogen [24], chitosan [324], and HA [325]). Nanoa- and deposition platform temperature. Otherwise viscosity modi-
patite coating represents a further way to increase the mechanical fiers, such as HA [366] and gellan gum [439], should be employed.
properties of printed gelatin-based interpenetrating polymeric net- In particular, the addition of gellan gum was demonstrated to en-
works [434]. A successful example of native gelatin employment hance the processing properties of GelMA bioinks by increasing
for bioprinting is represented by a recent study on controlled ex- their viscosity, yield stress, and speed of gelation. Optimized for-
trusion of a suspension of human corneal epithelial cells in a so- mulations (10/0.5% w/v GelMA/gellan gum) resulted in improved
lution of gelatin/alginate/collagen followed by CaCl2 crosslinking of filament deposition and increased stiffness of UV-cured hydrogels
the polysaccharide to achieve well-defined porous hydrogels stable encapsulating chondrocytes able to produce cartilage matrix in
in PBS for more than 2 weeks [435]. An alternative AM approach vitro [440]. GelMA was also co-deposited with photocrosslinkable
for material patterning, proposed also for other biomedical poly- methacrylate poloxamer macromers, functioning as reinforcing gel,
mers (e.g., fibrin and alginate) is represented by inkjet printing a to fabricate 3D hydrogels with the shape of the femoral condyle or
crosslinker (e.g., glutaraldehyde) onto a gelatin bed [436]. the auricular cartilage [441].
Gelatin can be made susceptible to photocrosslinking into Thiol–ene click chemistry has been also successfully employed
a hydrogel stable in physiological conditions through a simple to crosslink norbornene-modified gelatin for cells encapsulation
and straightforward procedure involving its direct reaction with [326], [327]. A recent article described the application of allylated
methacrylic anhydride in phosphate buffer (pH 7.4) at 50°C [437]. gelatin as a thiol–ene clickable bioink for SLA or SE-AM combined
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 19

with light irradiation to fabricate porous constructs with relatively ronal precursor cells, onto a fibrinogen substrate used as biopa-
high shape fidelity to CAD models [273]. An alternative approach per [293]. Similarly, Cui and Boland [328] developed a process to
to photocuring is represented by enzymatic protein crosslinking as deposit a thrombin solution containing human microvascular en-
shown by Irvine et al. [321] who crosslinked gelatin as a blend dothelial cells onto a fibrinogen substrate to fabricate 3D channel
with PEG by employing microbial transglutaminase to print cell- structures forming a tubular microvasculature stable in vitro over
laden constructs. 3 weeks (Figure 4c). The high crosslinking versatility represents
the main advantage of fibrinogen for AM encapsulation of cells
4.1.1.3. Fibrin. Fibrin is a fibrous protein whose clinical relevance is and growth factors, as shown by Lee et al. [329] who printed a
related to its use for developing versatile biomaterials with inher- thrombin solution onto a printed fibrinogen solution, both contain-
ent biocompatibility, biodegradability and injectability, exploitable ing vascular endothelial growth factors.
for advanced applications in TE, drug delivery, cell encapsulation, Another AM strategy includes the employment of a dual-
and surgical adhesives [442]. chamber single nozzle extrusion system that allows blending fib-
Source of extraction and synthesis route. Fibrin is naturally rinogen and thrombin solutions at a low temperature just before
polymerized during blood clot formation starting from thrombin- their deposition, in order to prevent crosslinking in the needle
catalyzed proteolysis of fibrinogen, a protein present in the blood [330]. Attempts have been also made to combine fibrinogen with
as soluble component (Figure 3c) [443]. Fibrinogen is composed of other hydrogel-forming materials to improve its processing prop-
two sets of three polypeptide chains (α , β , and γ ) joined together erties and stability once extruded as droplet or filament. Indeed,
by six disulfide bridges. Cleavage of fibrinopeptide A from the Aα its fibrous nature represents a disadvantage for AM, since it of-
chains and fibrinopeptide B from the Bβ chains leads to fibrino- ten results in clogging issues and unstable printed geometry [333].
gen conformational changes and exposure of polymerization sites. For instance, fibrinogen can be combined with gelatin to endow
Fibrin monomer with great tendency to self-associate and to form the resulting bioink with thermoreversible gelation behavior ex-
insoluble fibrin, is thus formed [442], [444]. Once polymerized, the ploitable to print cell-laden hydrogels with predefined shape and
fibrin network is stabilized by covalent crosslinking in the presence porous structure. Fibrinogen/gelatin constructs fabricated by means
of the factor XIIIa. The resulting fibrous structure is characterized of pneumatic SE-AM were crosslinked through a dual-enzymatic
by extreme extensibility and compressibility, as well as an inter- strategy, involving thrombin and transglutaminase, to support mul-
connected porosity suitable for hemostasis, fibrinolysis, and wound tiple cell types, including human umbilical vein endothelial cells,
healing. human neonatal dermal fibroblasts, and human bone marrow-
Biomedical use. Various clinically-available formulations al- derived MSCs [332]. Laser bioprinting of fibrinogen/HA solutions
low the combination of fibrinogen and thrombin derived from containing a cells suspension, followed by thrombin crosslinking
plasma to form fibrin products for different applications, such as was also investigated as a suitable method to fabricate 3D multi-
hemostats in surgery, adhesives for skin graft attachment, and cellular arrays [331].
sealants in colostomy closure [445]. Surgically employed sealants
comprise those sourced from human pooled plasma (e.g., Tisseel 4.1.1.4. Silk Fibroin. Silk fibroin is widely investigated for differ-
VH®), autologous plasma (e.g., Cryoseal®), and bovine plasma (e.g., ent biomedical applications thanks to its processability in aque-
VitagelTM ). In addition, thanks to its inherent advantages, includ- ous environments at room temperature, biodegradability to amino
ing good cell seeding and cell encapsulation efficiency, combined acids, biocompatible crosslinking processes, high tensile strength,
with the possibility to be produced from the patient’s own blood, and elasticity [453], [454].
fibrin has been widely investigated in the form of 3D gels or mi- Source of extraction and chemical structure. Silks are proteins
crobeads to engineer different tissues (e.g., cardiac, cartilage, and produced in fiber form by silkworms and spiders. Differences in
adipose tissues), often in combination with growth factors [422], primary aminoacidic sequence among silks of different species, as
[442]. well as within a species, result in different functional properties,
Fibrin is degraded and remodeled in vivo by enzymatic activ- which are significantly affected also by the extraction, purification,
ity. Fibrinolysis is mediated by serine protease plasmin cleavage at processing and environmental conditions [455], [456]. Fibers from
specific sites on either fibrinogen or crosslinked fibrin to yield var- the filaments of silkworm (Bombyx mori) are composed by core
ious degradation products [446]. Rapid degradation of fibrin gels structural fibroin proteins held together by sericin, which is a fam-
can cause losing of the necessary material structural features, es- ily of glue-like proteins [457]. Adequate fibroin purification from
pecially in the case of TE in which a mechanical role is required sericin is necessary to avoid immune reaction and elicit accept-
before a sufficient formation of the growing tissue is achieved. able in vivo foreign body response [458]. Silk fibers are composed
Many studies have been addressed to tuning morphological, rhe- by two major fibroin proteins, light and heavy chains (25 and
ological, adhesive and other functional properties of fibrin gels by 325 kDa, respectively), consisting of polypeptides organized in lay-
optimizing the crosslinking conditions (i.e., pH, and concentration ers of antiparallel β sheets forming crystalline regions (Figure 3d)
of fibrinogen, thrombin, and calcium ion), using low cell density, [459], [460].
or adding specific protease inhibitors [382], [442]. For instance, Biomedical use. Silk fibroin materials in the form of hydrogels,
a challenging task is to develop an optimal fibrous network den- fibers, sponges, particles, and membranes have been widely in-
sity trough tailoring fibrinogen and thrombin concentration in or- vestigated for various biomedical applications, including sutures,
der to achieve a good balance between mechanical properties and wound dressing, hemostatic devices and scaffolds for the engineer-
cell infiltration.[447] Other strategies adopted to develop fibrin- ing of a wide array of tissues (e.g., vascular, neural, cartilage, liga-
based materials with advanced properties include PEGylation of ment, and bone tissue) [461]. Fibroin non-woven meshes, porous
either self-assembled peptides [448] or fibrinogen [449], genipin sponges, and electrospun nanofibers have been employed for in
crosslinking [450], and combination with synthetic [451] or natu- vitro growth of different human cell types, such as epithelial cells,
ral polymers [452]. keratinocytes, endothelial cells and bone marrow-derived MSCs
AM application. The use of fibrin in AM has been mainly re- [455], [462–470]. Various strategies have been pursued to biofunc-
lated to bioprinting of cell-laden constructs. One of the first ap- tionalize fibroin materials, including immobilization of growth fac-
proaches involved the alternate deposition by thermal inkjet print- tors and combination with osteoconductive ceramics [471–475].
ing of two bioinks, one composed of a thrombin solution and the AM application. The ability of fibroin to form physical
other one composed of a thrombin solution containing NT2 neu- crosslinks through intra- and intermolecular hydrogen bonding be-
20 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

tween hydrophobic domains can be exploited to stabilize materials rized in the next paragraphs by highlighting critical aspects for
manufactured by SE-AM. Self-assembled fibroin hydrogels have a each of the most investigated polymer in this area (Figure 5).
relatively high mechanical strength, even if low flexibility and brit-
tle behavior can represent major disadvantages depending on the 4.1.2.1. Alginate. Alginate is a natural polysaccharide widely used
application [476]. Glycerol was investigated as non-toxic additive to in different biomedical fields thanks to its high biocompatibility as
stabilize an intermediate conformation of crystallized fibroin with carrier for cell encapsulation and drug delivery, scaffold for TE, and
good flexibility [334]. Blending with other natural polymers acting anti-adhesion material [479].
as bulking agents and amenable to be physically crosslinked can be Source of extraction. Alginate is found as structural polysaccha-
also exploited. For instance, silk-based printing efficacy and resolu- rides in marine brown algae (e.g., Laminaria hyperborea, Ascophyl-
tion were enhanced by using gelatin as bulking agent and glycerol lum nodosum, and Macrocystis pyrifera) and as capsular polysaccha-
to induce physical crosslinking [335]. Silk fibroin-gelatin blend hy- ride in some soil bacteria (e.g., Azotobacter vinelandii) [480]. Algi-
drogels were also recently bioprinted and stabilized in physiologi- nate macromolecular structure is constituted by (1-4)-linked β -D-
cal conditions through either enzymatic crosslinking by mushroom mannuronic acid and α -L-guluronic acid units (Figure 5a), whose
tyrosinase or physical crosslinking via sonication [338]. The result- sequence changes along the polymer chain. Many authors referred
ing microstructured hydrogels supported the multilineage differ- to this polysaccharide as ‘alginates’ in the plural form to high-
entiation of encapsulated human nasal inferior turbinate tissue- light an extreme variability in material chemical-physical behav-
derived mesenchymal stromal cells. A recent study was focused on ior, as a consequence of batch-to-batch variations in composition,
inkjet printing of a fibroblasts-laden blend of alginate and silk fi- Mw, and extent of the residues sequence, related to environmen-
broin through a two-step gelation process [337]. First sodium al- tal conditions of the source and relevant climate changes. Indus-
ginate was crosslinked by means of CaCl2 during printing, then trial extraction processes are based on soaking acidified seaweeds
fibroin was chemically-crosslinked at tyrosine residues through in a sodium carbonate solution to convert insoluble alginic acid
post-printing horseradish peroxidase catalysis, and finally alginate into soluble sodium alginate [481]. Some in vivo studies on algi-
was sacrificed through its liquefaction as consequence of Ca ions nate have reported immunogenicity and inflammatory responses,
chelation (Figure 4d). likely attributed to impurities (heavy metals, endotoxins, proteins,
A novel strategy for 3D freeform fabrication of silk fibroin relies and polyphenolic compounds) [482], [483]. A proper purification is
on a one-step physical gelation process involving the extrusion into therefore necessary to consider alginate biocompatible and to guar-
a nanoclay colloidal suspension containing low-Mw PEG that acts antee an acceptable control over its chemical-physical properties,
as physical crosslinker by inducing the formation of the β sheet aqueous stability, and biodegradation rate [484].
secondary structure [336]. In addition, Kim et al. [339] chemically Physical-chemical modification and biomedical use. The
modified the primary amines of silk fibroin by glycidyl methacry- marked hydrophilicity of alginate, mainly imparted by its car-
late. The resulting photocurable formulation was processed by boxylic groups, does not favor the necessary proteins adsorption
means of DLP to fabricate complex structures reproducing the allowing for mediated biological phenomena, including immuno-
anatomical details of different organs (e.g., heart, trachea, and ear). genicity, but also cell adhesion [485]. A widely exploited approach
to increase alginate cell affinity is based on covalent linking of
4.1.2. Polysaccharides oligopeptides containing the RGD sequences, through straightfor-
Polysaccharides are the first choice of polymers meeting the ward chemical reactions mediated by carbodiimide-based reagents
principles of sustainability, eco-efficiency, and green chemistry for [486–492]. Although conventionally considered as a biodegradable
the development of biomedical materials. Various polysaccharides polymer, bioabsorbability of alginate can be limited due to the lack
with peculiar chemical nature and physical-chemical properties of specific enzymes in mammals as well as the relatively high
can be obtained from different sources (i.e., vegetable, animal, Mw of some commercial alginate batches, larger than the renal
and microbial sources) generally with lower impact and produc- clearance threshold [493]. Partial oxidation of backbone has been
tion costs in comparison with proteins. In this optic, great interest demonstrated as an effective means to make alginate degradable
is given to novel biotechnologies to recover polysaccharides from in physiological media [494], [495].
bioagro-wastes [477] and algal waste biomasses [478], and to pro- Alginate tends to form insoluble hydrogels in aqueous solutions
cess them into high value added materials. via ionotropic gelation based on ionic interactions between car-
Alginate, an anionic polymer typically extracted from brown boxylic acid groups and chelating cations [496]. In its native form,
algae, HA, an anionic polymer of animal source that can be alginate is mainly present as Ca2+ crosslinked gel, but it forms
also produced via microbial fermentation, and chitosan, a posi- physiologically stable hydrogels also in the presence of other mul-
tively charged polymer obtained from crustacean shell and fungal tivalent cations (i.e., Sr2+ and Ba2+ ). In general, Ca2+ crosslink-
biomass, can be mentioned among the most investigated biomed- ing is preferred because of the milder reaction conditions and
ical polysaccharides for AM. The well ascertained biocompatibil- less toxicity involved in comparison to the use of other cations
ity of polysaccharides and their high chemical versatility con- [266]. The biocompatibility and ease of gelation of alginate make
ferred by the functional groups present in the structural units, it attractive in a wide range of biomedical applications, includ-
label them as materials of election for various biomedical appli- ing wound healing, drug delivery, and tissue engineering. Various
cations.[309] Indeed, the wide plethora of chemical modification alginate-based wound dressings, such as AlgicellTM (Derma Sci-
reactions developed for this class of polymers provides powerful ences), AlgiSite MTM (Smith & Nephew) and Comfeel PlusTM (Colo-
tools to tune polymer bioactivity, biodegradability, processing, and plast) are commercially available [408], [479]. However, ionotropic
physical properties. gelation suffers poor reproducibility in physiological environments
The great knowledge acquired by the scientific community on due to monovalent cations that can interact with polymer car-
polysaccharides chemical-physical functionalization has been ex- boxylic groups [497]. The reversible nature of ionic bridges be-
ploited to optimize and widen their AM processing potential, for tween adjacent polymer chains represents one of the main short-
instance by tailoring solution rheological properties for extrusion- comings of alginate, resulting in weak hydrogels with poor phys-
based processing, introducing photoreactive moieties to stabilize iological stability. Another approach followed to stabilize alginate
extruded hydrogels, or optimizing novel SLA strategies. The in- exploited also for AM, as discussed in the following section rele-
creasingly rapid advances made in the past two decades on vant to chitosan-based materials (see section 4.1.2.2), is based on its
polysaccharides science and technology applied to AM is summa- complexation with a polycation. A number of covalent crosslinking
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 21

Figure 5. Representative chemical structure of the most investigated polysaccharides for AM. (a) Chain portion of alginates, linear copolymers consisting of blocks of continu-
ous mannuronate residues, guluronate residues or alternating guluronate/mannuronate residues. (b) Chemical structure of chitosan, commercially produced by deacetylation
of chitin yielding a heteropolymer. (c) Chemical structure of cellulose, a linear polymer of β -1,4-linked glucose residues. (d) Chemical structure of hyaluronic acid (HA),
consisting of alternating disaccharide units of α -1,4-D-glucuronic acid and β -1,3-N-acetyl-D-glucosamine.

strategies have been successfully pursued to enhance mechanical cent article reported on the development of viscoelastic, bioprint-
integrity and reproducibility of alginate hydrogels, even if in many able aldehyde-containing oxidized alginates that could be com-
cases they present different drawbacks, including lack of biological bined with different imine-type dynamic covalent chemistries (i.e.,
inertness and cross-reactivity with encapsulated cells and proteins oxime, semicarbazone, and hydrazone) to tune the rheological,
[498–503]. mechanical and self-healing properties of the resulting hydrogels
AM application. The most exploited AM strategy for the fab- [504].
rication of alginate hydrogels, possibly encapsulating cells, in-
volves the extrusion of an aqueous polymer solution and then its 4.1.2.2. Chitosan
crosslinking immediately after dispensing, for instance by means Different inherent properties make chitosan appealing for
of submerged crosslinking [233] or aerosol spraying [354]. By em- biomedical applications, such as a good biocompatibility, a hy-
ploying low sodium alginate concentrations (1 to 2% w/v), Khalil drophilic surface favoring cell adhesion, and a well-assessed an-
et al. [233] developed endothelial cells-laden hydrogels through tibacterial activity [505].
extrusion into a calcium-containing bath. Good cell viability was Source of extraction and physical-chemical properties. Chi-
achieved by this approach even if the elastic modulus was low and tosan is the deacetylated form of chitin, a polysaccharide found
decreased along time after incubation in a cell culture medium in the exoskeleton of crustaceans, insects, and some fungi [506].
bath. A recent study reported on fibroblasts-laden scaffolds with For this reason, it is often classified as semisynthetic rather
tunable stiffness by varying sodium alginate concentration in the than natural polymer. Chitosan is typically obtained by alka-
range 2-10% w/v, with a resulting significant influence on cell line hydrolysis of the acetated groups of chitin to produce on
migration and morphology (Figure 6a) [349]. It was shown that an industrial scale a linear copolymer of β -(1–4) linked 2-
at low concentration the 3D porous structure was compromised, acetamido-2-deoxy-β -D-glucopyranose and 2-amino-2-deoxy-β -D-
while at too high concentrations cell migration was hindered due glycopyranose (Figure 5b). It is commercially available with differ-
to high chain packing density. Other studies have shown decreased ent N-deacetylation degree, conventionally classified as low (55-
viability of various cell lines by increasing alginate concentration, 70%), medium (70-85%), high (85-95%) and ultra-high (>95%) de-
explainable with the increased shear stresses acting on cells dur- gree.
ing extrusion [350], [351]. Rheological modifiers, such as cellulose The source chitin, existing in three polymorphic crystalline
nanofibrils, have been investigated as suitable means to enhance structures (α , β , γ ) [507], the extraction and purification con-
viscosity and processing properties of alginate-based bioinks, with- ditions, and the resulting deacetylation degree greatly influence
out compromising cell viability [352], [353]. chitosan physical-chemical, morphological and functional proper-
Although the development of methacrylated alginate has been ties, including chemical reactivity, degree of crystallinity, solubility,
reported [501], its use for photocrosslinking-based AM has not hydrophilicity, solution viscosity, mucoadhesion, and antimicrobial
been extensively investigated mainly because of the high control activity [508]. After refinement, at the dry state chitosan macro-
over hydrogel properties achievable through ionotropic gelation. molecules can organize in a rigid polymorphic, semicrystalline
One of the few exceptions is represented by a recent study on structure through inter- and intra-molecular hydrogen bonding. In
Vis-light-induced SLA hydrogelation of an alginate derivative with general, crystallinity degree is maximum for both chitin (i.e., 0%
chemically incorporated phenolic hydroxyl groups, mediated by deacetylated form) and fully deacetylated chitosan [509]. As a con-
ruthenium II trisbipyridyl chloride ([Ru(bpy)3]2+) and sodium per- sequence of a wide variability in degree of deacetylation and Mw
sulfate [267]. Thiol-ene chemistry has been investigated as ap- between different commercially available batches, various chitosan
proach alternative to methacryloyl functionalities crosslinking also Tg values within a representative range of 140-200°C, have been
for alginate-derived AM materials. Ooi et al. [274] reported the reported.
development of a norbornene functionalized alginate system as a While chitin is insoluble in aqueous solutions and most of or-
cell-laden bioink for extrusion bioprinting with a rapid UV-induced ganic solvents, chitosan is readily soluble in acid aqueous solutions
crosslinking mechanism. The mechanical and swelling properties at pH below its pKa, as a consequence of amino groups protonation
of the developed hydrogels were tunable by varying the concen- into positively charged groups (NH3 + ). The viscosity of chitosan
tration, length, and structure of dithiol PEG crosslinkers. A re- solutions increases with an increase in polymer concentration or
deacetylation degree, while decreases with increasing the shear
22 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 6. AM of polysaccharides. (a) Schematic representation of influence of alginate concentration (2, 5 and 10% w/v) on cell migration and morphology: after 7 days
of in vitro culture, (i) in 2% constructs cells could stretch themselves showing a spindle like shape, (ii) in 5% constructs cells had a spherical shape due to the restrictions
from the surrounding polymer networks, (iii) in 10% constructs cells spheroids were clearly observed as a consequence of cells tightly packing (reproduced with permission
[349]). (b) Chitosan extrusion directly in air and at room temperature: top (i) and side view (ii) SEM images of a chitosan scaffold; fluorescent microscopy image of 3D
printed chitosan with starfish (iii) or leaf (iv) shape (inset images show relevant CAD models) (reproduced with permission [344]). (c) Cellulose ear model (i) and octopus
structure (ii) by extrusion (direct-ink-write): first column is SolidWorks models, second column is manufactured gel structures, and third column is resultant structures after
freeze-drying (displayed scale bars are 1 cm) (reproduced with permission [360]). (d) Schematic of fabrication, stabilization, and incubation processes for dual-crosslinking
of methacrylated HA hydrogels (i): (1) supramolecular hydrogel assembly with guest–host bonds within the syringe chamber; (2) guest–host bond disruption when extruded
through the narrow needle, due to shear; (3) rapid self-healing of the supramolecular hydrogel and guest–host bonds when shear is removed and the material is deposited;
(4) UV treatment to photo-crosslink methacrylates within the printed hydrogels; and (5) stabilization to enforce the polymerized network; (ii) representative images of the
printing process and a resulting multilayer hydrogel structure (reproduced with permission [303]).

rate, resulting in a pseudoplastic behavior [508]. From a chemi- biodegradation through hydrolysis of glucosamine–glucosamine,
cal point of view, chitosan behaves in solution as a polycationic glucosamine–N-acetyl-glucosamine, and N-acetyl-glucosamine–N-
base due to the presence of amino groups along its backbone, dif- acetyl-glucosamine linkages, mainly by lysozyme, an enzyme
ferently to other polysaccharides that are neutral (e.g., starch) or present in various human body fluids and tissues [513]. Besides af-
acidic (e.g., alginate, ulvan, carrageenan, fucoidan) owing the ex- fecting the previously mentioned chemical-physical properties, the
clusively presence of hydroxyl and acid groups along their struc- degree of deacetylation affects chitosan biodegradation rate and
ture [309]. This polycationic structure confers chitosan with a well- biocompatibility since the presence of free amino groups favors the
documented antimicrobial activity against many Gram-positive and interaction with cells [514], [515].
Gram-negative bacteria, whose mechanism is likely related to an Chitosan has been widely investigated in the form of mem-
electrostatic interaction with the anionic components of bacterial brane, gel, or micro/nanoparticles for the controlled release of var-
surface (e.g., peptidoglycan and lipopolysaccharide) leading to cell ious bioactive agents, ranging from growth factors, antibiotics and
damage or death [510]. anti-inflammatory drugs [508], [516]. A large and growing body
Biomedical use. Chitosan has been reported to be highly of literature has been dedicated to the development of chitosan-
biocompatible, displaying low immunogenicity when interfaced based scaffolds for engineering various human tissues, including
with living organisms, with acceptable host response upon im- bone, cartilage, skin, liver and nerves [517]. In order to enhance
plantation [511], [512]. In vivo it is susceptible to enzymatic its mechanical strength and ability to maintain a predefined shape
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 23

once processed, chitosan has been combined with numerous poly-


mers of synthetic (e.g., PVA [518] and PLLA [519]) or natural (e.g.,
alginate [520] and collagen [521]) origin as well as with bioac-
tive ceramics (e.g., Ca-P [522] and natural coralline [523]). In addi-
tion, the high chemical reactivity of the amino groups has been ex-
ploited to functionalize and tailor chitosan macromolecular struc-
ture, as shown by Park et al. [524] who developed thermosensitive
chitosan–g–pluronic copolymers as injectable hydrogels encapsu-
lating bovine chondrocytes.
AM application. SE-AM of a diluted acetic acid solution in air
or into a coagulation/crosslinking medium represents the most di-
rect approach to AM of chitosan-based hydrogels. One of the first
attempts in this context was made in 2002 by Ang et al. [341] who
extruded an acetic acid solution of chitosan into a bath composed
of NaOH and ethanol. By optimizing the composition of the coag-
ulation bath, they were able to fabricate 3D scaffolds that main-
tained square macropores after washing and freeze drying. The re-
producibility of the fabrication process was increased by Geng et
al. [340] who designed a dual dispensing method to simultane-
ously extrude a chitosan acid solution and dispense NaOH onto the
deposited polymeric strand. Chitosan constructs fabricated through
the same processing strategy were more recently investigated in
comparison with PDLLA-based constructs to evaluate in vitro the
inflammatory response elicited by them in terms of cytokine pro-
file of human monocytes/macrophages [525]. A recent study re-
ported on the extrusion of a silk particles dispersion (up to 300%
w/w) into a chitosan solution to fabricate scaffolds with enhanced
mechanical properties and printing accuracy, by manually pipet-
ting a NaOH solution onto each printed layer [526]. Extruding a
chitosan solution into a bath of isopropyl alcohol is a reliable al-
ternative for manufacturing scaffolds able to induce in vitro the
chondrogenesis of infrapatellar fat pad adipose-derived stem cells
(ASCs) cultured on them [342].
A different AM approach is based on freezing the extruded chi- Figure 7. In vivo investigation of additive manufactured chitosan. Representative
tosan solution during its deposition onto a low-temperature plat- picture of chitosan scaffold without (i) and with (ii) a film of chitosan at the base;
representative images (each photo corresponds to an area of 1 cm2 ) of wound heal-
form, followed by its lyophilization [345]. In this way, a defined
ing in treated and untreated groups (iii); histological staining Hematoxylin + picro-
open porosity along the Z-axis building direction can be achieved. sirius red of tissue excised from wounds treated with chitosan scaffolds (iv) or with
In order to avoid the expensive and time-consuming lyophiliza- a commercial product (v) for 14 days (v: blood vessels, g: sebaceous glands, p: hair
tion step, the frozen chitosan structure can be directly dipped into follicles, m: arrector pili muscles; scale bar 100 μm) (reproduced with permission
a KOH coagulation bath to induce polymer gelation, even if this [347]).

treatment can diminish the fabrication accuracy [346]. Anyhow,


scaffolds fabricated by this method were recently validated both
and positive molds by BJ. The method developed by Liu et al.
in vitro using human fibroblasts/keratinocytes co-culture and in
[348] involved extruding a suspension of chitosan in an alginate
vivo in a diabetic rat model resulting in improved wound heal-
solution and spraying a HCl solution on the deposited fiber in
ing in comparison to what observed with a commercial product
order to protonate the chitosan amino groups. In this case, scaf-
(Figure 7) [347]. High porous structure regularity was recently
fold structure reproducibility and manufacturing accuracy were en-
achieved through extrusion in air of concentrated chitosan solu-
hanced by increasing the molar concentration of chitosan in the
tions in a solvent mixture (acetic acid/lactic acid/citric acid) with
mixture.
optimized volatility (Figure 6b) [343], [344]. The resulting chi-
Chitosan was also recently added to a PEGDA-based resin up
tosan filaments showed relatively high strength and pronounced
to a 1:5 weight ratio for the SLA fabrication of ear-shaped scaf-
extensibility that were further increased after NaOH bath immer-
folds with optimized properties by controlling chitosan Mw and
sion (ultimate strain ~ 400%), as a consequence of polymer physical
photoinitiator concentration [153].
crosslinking through hydrophobic interactions and hydrogen bonds.
Ionic complexation with a polyanion is another effective way
to stabilize extruded chitosan, which represents one of the main 4.1.2.3. Plant and bacterial cellulose. Plant cellulose, particularly in
advantages of polycations for AM purposes. The first successful ex- micro- and nanostructured forms, has been widely investigated for
ample of this strategy applied to AM was represented by CAWS different biomedical applications to exploit its surface chemistry,
fabrication of hydrogels with defined porous structure and en- biocompatibility and biodegradability, in particular in the case of
hanced mechanical properties through the extrusion in an ethanol skin repair treatment thanks to its suitability in accelerating the
bath of an aqueous suspension of a chitosan/poly(γ -glutamic acid) process of epithelialization [528].
polyelectrolyte complex (PEC) [236]. The resulting PEC hydrogel Source of extraction and physical-chemical properties. Cellu-
scaffolds were successfully investigated in combination with the lose is the most abundant renewable polymer on earth, found
human cancer cell line BxPC-3 for long-term in vitro 3D modelling primarily in lignocellulosic material in forests. Algae, agriculture
of the pancreatic ductal carcinoma [34]. Reeds et al. [527] used residues, bacteria biosynthesis, and chemical synthesis represent
a chitosan-alginate PEC aqueous mixture and fabricated scaffolds other sources [529]. Cellulose is an unbranched polymer of β -1,4-
through indirect AM by employing negative molds made by FDM linked glucose residues (Figure 5c). In native cellulose (i.e., cellu-
24 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

lose I), hydroxyl groups tend to form hydrogen bonds resulting in AM application. The possibility of obtaining filaments suitable
monoclinic and triclinic crystallites interspersed with amorphous to be printed by FDM and based on different cellulose deriva-
regions [530]. Native cellulose can be converted through physical- tives commonly-used for pharmaceutical formulations (i.e., ethyl-
chemical and biological treatments to other crystalline forms with cellulose, hydroxypropyl methylcellulose acetate succinate, and hy-
different unit cell dimensions, such as in the case of cellophane drophilic cellulose derivatives) was recently demonstrated. How-
and rayon. ever, poor thermal stability represents one of the main shortcom-
Like other polysaccharides, cellulose is not thermoplastic and it ings of lignocellulose materials limiting their use in ME-AM as
undergoes degradation when thermally processed. In addition, the reinforcement components. As recently reviewed by Wang et al.
high degree of polymerization and the complex pattern of intra- [363], cellulose-containing filaments for FDM are typically com-
and intermolecular hydrogen bonding result in cellulose insolubil- posed of PLLA, PCL, or PVA reinforced with cellulose-based fillers
ity in water and in many organic solvents, making solution pro- in different forms, such as nanocrystals, fibers, and wood flour, at
cessing challenging. However, the possibility of dissolving cellulose weight percentages up to 50%. Few studies also involved cellulose
in ionic liquid solutions and then regenerating it in a non-solvent crystals as nanofillers for SLA manufacturing [364], [365].
after processing has been shown by several studies [531]. This en- Raw formulations for SE-AM have been optimized through
ables the application of different solution-based processing tech- combination of cellulose solutions or suspensions with other
niques, including casting, molding, wet-spinning, and electrospin- hydrogel-forming polymers (e.g., alginate) and proper additives in
ning. order to tailor their rheological and processing properties, as well
Plant cellulose derivatives from various sources and subjected as the stability of the printed hydrogel once in a physiological en-
to different treatments are available on the market as biomateri- vironment [363]. The resulting nanocellulose formulations show
als, including microfibrillated cellulose and nanocrystalline cellu- a shear-thinning behavior suitable for extrusion through a noz-
lose [532]. The first one is typically in the form of 5-60 nm wide zle into 3D shapes that can be retained as a result of different
and several microns long nanofibers, obtained by mechanical de- gelation/solidification methods (e.g., physical/chemical crosslink-
lamination of wood pulp followed by chemical or enzymatic treat- ing, air dry, and freeze-drying). A number of studies recently
ment. Nanocrystalline cellulose, in the form of nanofibrils with a focused on the controlled deposition of nanofibrillar cellulose-
cross-sectional size as low as 5 nm and hundreds of nm in length, alginate formulations that were crosslinked by using CaCl2 to
is obtained through wood pulp acid hydrolysis with concentrated develop 3D hydrogels possibly encapsulating cells [353], [355–
sulfuric acid to dissolve the cellulose non-crystalline domains, fol- 358]. The same crosslinking strategy was followed in the case of
lowed by high pressure treatment. methylcellulose-alginate bioinks [539]. A successful AM approach
Bacterial cellulose (BC) possesses peculiar physical-chemical involves printing cellulose dissolved in an ionic liquid (e.g., 1-
and structural features making it appealing for biomedical appli- ethyl-3-methylimidazolium acetate) followed by coagulation in a
cations, including high purity, high crystallinity, high Young’s mod- non-solvent bath to obtain spatially-structured hydrogels [359].
ulus, excellent biodegradability, high water holding capacity, and In this case, co-solvents (e.g., 1-butanol and dimethyl sulfoxide)
good biological affinity [533]. BC is synthesized by bacteria such have been investigated as rheological modifiers to ensure consis-
as Acetobacter xylinum, and its unique properties are the result of tent printing.[540] An alternative way to stabilize cellulose-based
a typical nanostructured fibrous architecture. BC fibril diameter is materials after extrusion relies on freeze drying followed by chem-
typically in the range of 30 nanometers, hundred times thinner ical crosslinking (Figure 6c) [360]. 3D structures printed adopt-
than that of plant cellulose fibers [534]. This nanosized structure, ing this strategy are characterized by a dual-scale porosity, due
together with the absence of residual contaminants typical of plant to the lyophilization process, facilitating cell growth. Free-standing
cellulose (e.g., lignin, hemicelluloses, and pectin), are the reasons gels can be also obtained by adding xylan conjugated with tyra-
of the aforementioned advantages. mine to cellulose nanofibrils suspensions to make them suscepti-
Biomedical use. Many attempts have been made to exploit BC ble to enzymatic crosslinking [361]. A recent article described an
properties for different biomedical applications. In particular, BC AM process based on the extrusion in air of an acetone solution of
porous structure and hydrophilicity have been shown to be suit- cellulose acetate [362]. After solvent evaporation and material so-
able for wound dressing by accelerating granulation, ensuring tis- lidification, cellulose acetate was reconverted to cellulose through
sue healing, absorbing exudates, and providing a physical barrier NaOH deacetylation.
from the surrounding environment to prevent microbial coloniza-
tion [535]. Wound dressings from BC are nowadays available on 4.1.2.4. Hyaluronic acid (HA). HA is widely exploited as injectable
the market (e.g., Biofill®, Bioprocess®, and XCell®) and applied or implantable biomedical polymer thanks to its inherent advan-
in patients affected by burns and chronic ulcers showing accel- tages, such as no immunogenic properties, widespread availability,
erated healing and pain relief in comparison with other commer- ease of chain size manipulation, and ability to interact with cell-
cial wound patches [536]. BC growing typically results in pellicles surface receptors [541].
rising to the surface of the reactor, and agglomerating to form a Source of extraction. HA is found in the ECM of different con-
membrane with high purity that can be either used as it is, pro- nective tissues as major structural and functional macromolecular
cessed into nanofibrils, solubilized, or dispersed in a liquid. BC can element that contributes to the water balance regulation, serves as
be also shaped and molded into 3D structures directly during in lubricant and shock absorber, and at the same time, acts as scav-
vitro cultures, depending on the bacterial culture conditions (i.e., enger for free radicals. Besides functioning as selective and protec-
static culture, agitated culture, or airlift reactor) that also affect tive coat around the cell membrane, HA interacts with bioactive
the resulting mechanical properties and micro/macrostructural fea- molecules and is recognized by specific cell receptors that regulate
tures [537]. This aspect has accelerated research on BC applica- cell behaviors, such as migration and ECM remodeling [541], [542].
tion for developing implantable scaffolds tailored to different tis- HA is a high molecular mass (103 to 107 Da) linear copolymer
sues, including blood vessels, cartilage, and bone. For instance, a consisting of alternating disaccharide units of α -1,4-D-glucuronic
number of bone engineering devices based on BC in combination acid and β -1,3-N-acetyl-D-glucosamine, linked by β (1→3) bonds
with other naturally-derived polymers, e.g., collagen, agarose, PHB, (Figure 5d) [543]. It is often referred to as ‘hyaluronan’, since it
and chitosan, or bioactive nanofillers, e.g., hydroxyapatite (Hap), exists in vivo as polyanion and not in the protonated acid form.
are available on the market [538]. It is produced on an industrial scale through extraction from ani-
mal tissues (e.g., umbilical cord and rooster comb) [422] or micro-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 25

bial fermentation [544]. Medical grade HA is obtained by means of native HA as thickener to optimize ink rheology [373]. By graft-
separation techniques, such as protease digestion, HA ion-pair pre- ing PLGA to HA, Park et al. [549] were able to obtain chloroform
cipitation, membrane ultrafiltration, non-solvent precipitation, and solutions that could be processed under a heated air stream to fab-
lyophilization, necessary for its purification from other biopoly- ricate 3D scaffolds encapsulating BMP-2/PEG nanosized complexes.
mers and molecules to which is complexed [545]. A recent strategy integrated photoreactive acrylate and thiol-ene
Biomedical use and physical-chemical modification. HA med- chemistries by developing HA functionalized with both methacry-
ical applications include, among others, viscosupplementation by late and norbornene, suitable to processed by AM into cell-laden
injection into joints to treat arthritis and relieve pain, ophthalmic hydrogels [268].
surgery, tissue augmentation and protection in cosmetic and recon-
struction surgery, and advanced applications, such as TE and cells 4.1.3. Microbial polyesters
encapsulation [546]. HA combination with other polysaccharides, Polyhydroxyalkanoates (PHA) are microbial polyesters widely
proteins, and ceramics has been investigated in order to provide investigated for biomedical applications thanks to their biocom-
artificial ECM environments to enhance cell interaction since the patibility and in vivo degradability into monomeric and oligomeric
hydrophilic, polyanionic nature of HA does not favor cell adhesion products not exerting toxic effects [309]. From an applicative and
[547]. technological perspective, the great interest raised on PHA is justi-
HA tends to aggregate in aqueous environment as a conse- fied by the possibility of combining the advantages of a potentially
quence of hydrophobic interactions and hydrogen bonding be- sustainable production, starting from renewable resources, with a
tween acetamido and carboxylate groups [548]. However, these in- thermoplastic behavior and processing versatility comparable to
teractions are fairly weak, thus polymer aggregates tend to disso- those of chemically synthesized aliphatic polyesters.
ciate under physiological conditions. For this reason, a large body Synthesis and source of extraction. PHA are produced as in-
of literature has been devoted to chemical modification of HA to tracellular carbon and energy storage compounds by many Gram-
reduce water solubility and control its biodegradation through, for positive and Gram-negative bacteria under unbalanced growth
instance, esterification. Various HA derivatives with different de- conditions [550], [551]. Extensive market penetration of PHA is
grees of esterification (e.g., HYAFF ® and ACPTM ) are currently hindered by production costs related to the need of maintaining
available on the biomedical market. A wide range of other HA axenic cultures and providing refined carbon substrates [552]. For
chemical modification approaches have been developed, includ- this reason, several attempts have been made to enhance the com-
ing hydrazide modifications, crosslinking with polyfunctional epox- petitiveness of PHA large-scale commercialization through novel
ides, glutaraldehyde crosslinking, reaction with divinyl sulfone, and metabolic engineering, synthetic biology, and bioinformatics ap-
crosslinking with carbodiimides [547]. proaches [553]. Ongoing research is investigating carbonaceous
AM application. Solutions of unmodified HA alone are not suit- (agro)industrial waste streams as alternative feedstocks for PHA
able for SE-AM owing their high viscosity, slow gelation behavior, production, such as waste glycerol from biofuel production, lig-
and low stability upon printing. These shortcomings justified re- nocellulose waste from the food industry and forestry, and even
search on HA combination with other hydrogels for AM, like in the petrochemical plastic waste [554]. Waste substrates coupled with
case of blending with photocrosslinkable hydrogels (e.g., GelMA mixed microbial consortia in three-stage systems, comprising feed-
[366] and hydroxyethyl-methacrylate-derivatized dextran [367]). stock fermentation, enrichment of PHA-producing bacteria, and
Chemical modification represents a powerful tool to widen the PHA production, have been investigated as means to optimize pro-
possibilities of HA processing by AM and optimize hydrogel stabil- cess commercial potential [555].
ity through crosslinking. Suri et al. [117] developed a novel method Chemical structure and physical-chemical properties. PHA
for the layered photoreticolation of HA methacryloyl (HAMA) into class is composed by aliphatic polyesters generally consisting
a set of 3D scaffolds with different geometry and porous structure. of 10 0 0-10 0 0 0 monomeric units with different pendant groups,
A similar approach allowed high survival of encapsulated MSCs af- or a hydrogen atom, at the beta-position (Figure 8). They can
ter HAMA photocrosslinking [374]. HAMA has been also employed be classified as short-chain-length or medium-chain-length PHA
in photocrosslinkable multimaterial ink formulations. Skardal et al. if they consist of 3–5 or 6–14 carbon monomers in the 3-
[368] developed a processing method based on partial photochem- hydroxyalkanoate unit.
ical co-crosslinking of GelMA with HAMA giving an extrudable Since the discovery in the Twenties of poly(3-hydroxybutyrate)
gel-like fluid, that was then printed under irradiation to fabricate (PHB) [556], which has a pendant methyl group, an array of
tubular constructs encapsulating cells. Kesti et al. [369] developed PHA macromolecular structures have been identified starting
bovine chondrocytes-laden hydrogels with a well-defined porous from poly(3-hydroxyvalerate) (PHV), with an ethyl pendant group,
structure by blending poly(N-isopropylacrylamide)-grafted HA (HA- and the copolyester poly(3-hydroxybutyrate-co-3-hydroxyvalerate)
pNIPAAM) with HAMA. HA-pNIPAAM provided fast gelation and (PHBV). The structural, processing and mechanical properties of
immediate post-printing structural fidelity, while HAMA ensured PHA depend on both the length of the pendant groups and the
long-term mechanical stability upon photocrosslinking. distance between ester linkages [311]. PHB has a high degree of
Separately coupling adamantane and β -cyclodextrin moieties to crystallization in the range 60-80%, as a consequence of the high
HA is an effective means to create two hydrogel-precursors that stereochemical regularity, resulting in a stiff and brittle mechanical
form a supramolecular assembly upon mixing [370]. By integrat- behavior (elongation at break<10%) (Table 2). The different molec-
ing this approach with photocrosslinkable methacrylate groups, ular structure of poly(4-hydroxybutyrate) (P4HB) generally results
Ouyang et al. [303] developed a dual-crosslinking strategy enabling in reduced degree of crystallization (~35%), higher flexibility, and
the fabrication of layered hydrogels with a predefined porous better processing properties in comparison to PHB. Indeed, P4HB
structure that were stable over a month in physiological conditions has a Tg (~-50°C) and a Tm (~60°C) much lower than those of
with no loss in mechanical properties (Figure 6d). Other attempts PHB (Tg ~0°C and Tm ~180°C) [557]. The copolymerization between
to optimize ink rheology and stability of the printed hydrogel, 3-hydroxybuytrate (3HB) and 4-hydroxybutyrate (4HB) monomers
were based on thiolated HA crosslinked by either poly(ethylene results in poly(3-hydroxybutyrate-co-4-hydroxybutyrate) (P[3HB-
glycol tetraacrylate) [371] or gold nanoparticles [372]. 3D double co-4HB]) co-polyesters with morphological, thermal and mechani-
printed constructs were recently developed by alternating strands cal parameters intermediate between those of the two relevant ho-
of PCL and a precursor solution composed of thiolated HA, allyl- mopolymers [558]. Since 3HB and 3-hydroxyvalerate (3HV) units
functionalized poly(glycidol) as cytocompatible cross-linker, and co-crystalize in a single crystalline unit cell as a consequence of
26 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Table 2
Reference data available in literature relevant to thermal transitions, mechanical properties, biodegradation rate, and suitable solvents of high-Mw natural and synthetic
aliphatic polyesters investigated for AM 557, 562, 566-578.

Tg Tm C Tensile Tensile Elongation In vivo Scaffold compressive properties


Polymer (°C) (°C) (%) modulus strength at break absorption Solvents
(GPa) (MPa) (%) timef AM technique Modulus Strength
(material) (MPa) (MPa)

PHB 0-5 170- 60-80 0.9-4.0 15-60 5-20 2-3 years SLS (PHB) [376] 0.4-24 0.1-1.7 Halogenates (e.g.,
180 chloroform and
P4HB -47 to 57-69 35 0.05-0.23 35-50 >1000 2-18 n/a n/a n/a dichloromethane),
-52 months dimethylformamide
PHBV 0-5 130- 35-70 0.5-3.5 10-40 5-50 > 1 year SLS (PHBV; PHBV/Ca-P) 2.4-6.5 0.2-0.6
180 [179,181] n/a 6.8-
FDM (PHBV/PCL) [378] 15.1
0-2 54-180 15-45 0.1-0.5 4-21 5-850 >6 CAWS (PHBHHx) 0.7-1.5 1.3-2.0
PHBHHx months [238,258] 0.2-0.4 0.4-0.6
CAWS (PHBHHx/PCL)
[239]

PCL -60 56-65 > 50 0.2-0.5 4-42 80-1100 > 2 years FDM (PCL) [579,580] 20-40 2.0- Halogenates, toluene,
FDM (PCL/TCP) [581] 124 6.8b cyclohexanone,
FDM (PCL/PLGA 1:1) 50 4.2-5.2 tetrahydrofuran,
[582] 27-56 6.1 acetone,
0.1-1.3 1.9-4.5 dimethylformamide
ME-AM (PCL/Hap) 1.4-7.3 0.3-5.2
[583] 52-67 0.5-1.5c
CAWS (PCL; PCL/Hap) 24-103 2.0-
[26,252] 0.1-14 3.2b
CAWS (∗ PCL; 2.5-

PCL/Hap) [251] 11.5
SLS (PCL) [4] n/a
SLS (PCL/Hap) [175]
SLS (PCL/TCP)
[584,585]

PLLA 55-80 170- 37-50 2.7-4.1 15-150 <10 2-5 years ME-AM (PLLA)[586] 195 8.3 Chloroform,
190 SE-AM/freeze drying 60 4.7 dichloromethane,
(PLLA/TCP) [255] 5.8-6.1 0.5-0.6 furan, dioxane,
SLS (PLLA; PLLA-Hap) dioxolane
[179]

PDLA 40-50 120- 30-45 3.4-3.8 58-67 <10 n/a n/a n/a n/a In addition to PLLA
160 solvents: acetone,
ethyl lactate,
PDLLAa 43-60 120- 15-40 1-3.5 28-50 <10 2-16 FDM (PDLLA) [218], 3-600 8.5-22
tetrahydrofuran, ethyl
170 months [587–589] 28-92 n/a
acetate, dimethylsul-
SE-AM (PDLLA/PEG) 44-100 n/a
foxide, N,N xylene,
[261], [590] 2.5-4.0 6-11
dimethylformamide
SE-AM 150- n/a
(PDLLA/PEG/BG) [261], 360 0.6-1.2
[590] 0.1
SSLS (PDLLA/TCP) 12-31
[591]
SLA (PDLLA derivative)
[592]
SLA(PDLLA-co-PCL
derivative) [593]
SLA (Hap-loaded
PDLLA derivative)
[594]
PGA 34-44 200- 45-55 6.1-7.2 12-70 <10 6 weeks n/a n/a n/a Highly fluorinated
231 organics, e.g.,
hexafluoro isopropanol

PLGA 44-55 — d
— d
1.4-2.8 40-55 <10 1-6 FDM (PLGA) [215] 2.6 n/a PDLLA solvents
months ME-AM (PCL/PLGA) 50-52 3.2-6.0
[582], [595] 51 8.4
ME-AM 18 0.7
(PCL/PLGA/TCP) [582] 23 0.8
SE-AM/freeze drying 130 12
(PLGA/TCP) [596]
SE-AM/freeze drying
(PLGA/pearl) [597]
SLS (PLGA/TCP) [598]

PPF -25-30 — d
— d
0.02-1e 20-70e <5 e Several μSLA (PPF/DEF) 8-50 77-290 Chloroform,
monthsg [599–601] 135 n/a dichloromethane,
DLP (PPF/DEF) [602] 50 n/a tetrahydrofuran,
SE-AM (Hap-loaded acetone, ethyl acetate
PPF/DEF) [603]
n/a: not available data. a Amorphous when d-lactide>20%.[604] b Values corresponding to yield strength. c Values calculated at stresses corresponding to 50% strain. d
Amorphous. e Values measured for crosslinked PPF/DEF samples. f Values relevant to full polymer resorption upon implantation either in human or animal, depending on
material morphology and site of application. g Data on in vivo full absorption of PPF not available in literature; based on in vitro studies, several months are needed for full
absorption depending on crosslinking degree.
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 27

Figure 8. Chemical structure of polyhydroxyalkanoates (PHA) investigated for biomedical AM. General structure of PHA (i); structure of poly(3-hydroxybutyrate) (PHB) (ii);
structure of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) (iii); structure of poly[3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx) (iv).

the small difference in size of the side group, PHBV crystallinity A large body of literature has been dedicated to combining PHA
is not much reduced in comparison to PHB, being around 60% with other bioactive materials in order to tailor their biodegra-
throughout a wide range of composition from 0 to 95% 3HV [559]. dation, thermal, mechanical and processing properties, as well
However, inhibition of secondary crystallization in PHBV results in as their hydrophilicity and cell interaction. Numerous osteoinduc-
increased flexibility and ductility (elongation at break up to 50%), tive ceramics in the form of nano/microsized fillers (e.g., HA, BG,
as well as a reduced Tm (down to 130°C) broadening its melt pro- TCP, and zirconia) have been loaded into PHA implants designed
cessing window in comparison to PHB. The longer alkyl side chain for bone repair [607]. Multi-block copolymerization with other
confers to poly[3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHB- acids/lactones (e.g., PLLA [608]) or ethers (e.g., PEG [609]) as well
HHx) even a broader processing window and higher elasticity as linking of functional proteins [610] have been also investigated
which can be exploited for tailored biomedical applications [560]. to tailor PHA physical-chemical, structural and functional prop-
Indeed, experimental investigations have demonstrated that by in- erties. In addition, the microbial production of novel oligomers
creasing 3-hydroxyhexanoate (3HHx) content, the Tm and degree were recently investigated by co-feeding Escherichia coli with di-
of crystallization can be decreased from 177 to 54°C and 60 to 18%, ethylene glycol, resulting in the formation of lactate-HB oligomers
respectively [561], [562]. The tensile strength can be correspond- that can undergo polyaddition reaction with diisocyanate to yield
ingly reduced from 43 to 20 MPa, while the elongation at break biodegradable PUs [611].
can be increased from 5 to 850%. The combination of different PHA, including PHB, PHBV and
In vivo PHA biodegradation is the result of two different mech- P[3HB-co-4HB] copolymers, with other blend polymers such as
anisms, i.e., nonenzymatic and enzymatic hydrolysis of the es- PLLA, PCL, starch, cellulose and chitosan has been also widely de-
ter bond, the latter by the action of nonspecific lipases and es- scribed [612]. However, PHA do not show the same blending ver-
terases being more extensive at the later stages [563], [564]. Be- satility of synthetic polyester counterparts, with which they gener-
sides polymer characteristics like chemical composition, Mw, and ally show poor miscibility, mainly due to processing issues. Short-
crystallinity, PHA biodegradation rate greatly depends on the spe- chain PHA, particularly PHB, are soluble in very few solvents, i.e.,
cific physiological environment. P4HB sutures complete biodegra- halogenated solvents (e.g., chloroform and dichloromethane) and
dation can require from 8 to 52 weeks, while no significant degra- dimethylformamide, often resulting only in a partial solubiliza-
dation of PHB implants was observed after 9 months in a rat tion at concentrations required by specific processing techniques
femoral defect [565]. (e.g., solvent casting and electrospinning) [613]. In addition, ther-
Biomedical use and physical-chemical modification. PHA mal processing is often challenging due to the narrow window be-
molecular structure similarity with synthetic aliphatic polyesters tween Tm and degradation temperature (Tdeg ). Indeed, PHA pro-
results in processing and mechanical properties generally superior cessing at temperatures above 150°C generally results in the pro-
to the other classes of natural polymers, suggesting their use for duction of trans-crotonic acids [606], [614].
the fabrication of devices with high resolution and reproducibility AM application. The limited PHA thermal processing proper-
intended to be used in load-bearing applications (e.g., bone TE). ties is a major shortcoming for AM application. A few articles re-
In addition, the numerous homopolymers and copolymers belong- ported on the employment of SLS for PHA scaffolds fabrication.
ing to this class of renewable materials, combined with the possi- In particular, PHB was successfully processed by SLS without any
bility of tuning their Mw and comonomers percentages by chang- variation in polymer thermal properties and chemical composition
ing microbial fermentation conditions, offer the possibility to tune [375–377]. However, although the resulting scaffolds macroscopi-
the resulting material biodegradation, mechanical and processing cally showed geometrical fidelity to the virtual model, they were
properties over a wide range. For these reasons, PHA have been characterized by poor resolution, high surface roughness, and un-
widely investigated for the development of various biodegradable wanted macropores in the polymeric matrix due to not completely
devices such as orthopedic screws, skin patches, implantable and sintered particles. Duan et al. [179–181] fabricated nanocompos-
injectable systems for drug delivery, scaffolds for the regeneration ite scaffolds through SLS of Ca-P-loaded PHBV microspheres (φ ~50
of different tissues, including bone, cartilage, nerves, and blood μm) prepared by means of a S/O/W emulsion-solvent evaporation
vessels [605]. Indeed, various biomedical products mainly based on method (Figure 9a). They developed PHBV scaffolds with inter-
PHB and P4HB, are commercially available for research and clinical connected porous geometries that were biofunctionalized by bind-
applications, such as sutures (e.g., TephaFlex®, Phantom FiberTM , ing the recombinant human BMP-2 (rhBMP-2) to a heparin-gelatin
and MonoMax®), scaffolds (e.g., BioFiberTM scaffold), and surgical conjugated coating that was physically entrapped onto the surface.
meshes (e.g., GalaFLEX and Tornier®) [606]. High surface roughness as a result of limited coalescence of com-
posite microspheres was observed also in this case.
28 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 9. AM of PHA scaffolds. (a) SLS of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV): picture of PHBV (i) and PHBV-CaP (ii) scaffolds; SEM micrographs 85 x (iii)
and 500x (iv) of PHBV-CaP scaffolds (reproduced with permission [179]). (b) FDM of PHBV: representative SEM micrographs and μ-CT images of top view (i) and (iii) and
cross-section (ii) and (iv) of PHBV/PCL blend scaffolds (50:50 weight ratio) (reproduced with permission [378]). (c) CAWS of poly[3-hydroxybutyrate-co-3-hydroxyhexanoate)
(PHBHHx): picture of PHBHHx scaffolds with different external size and porosity (i) and SEM micrographs of PHBHHx scaffold top view (ii) and cross-section (iii) (inset high
magnification micrographs show polymer microporosity) (reproduced with permission [238]). (d) Anatomical PHBHHx scaffold by CAWS with shape and dimensions resem-
bling those of a critical size segment of the New Zealand rabbit radio: representative pictures showing an implanted scaffold (i), the top view (ii) and the macrochanneled
edge (iii) of the scaffold prototypes (reproduced with permission [258]).

Few articles reported on successful application of FDM to PHA nanotubes [617], even if FDM processability was not tested in any
processing, in all cases through their blending with other polymers case.
and/or plasticizers. In particular, PCL/PHBV blend scaffolds at dif- The versatility of SE-AM for 3D PHA constructs fabrication has
ferent weight ratios (75:25, 50:50 and 25:75) were recently fab- been described by various articles reporting on different CAWS ap-
ricated by FDM (Figure 9b) [378]. The proliferative capacity and proaches to process polymeric solutions through their coagulation
chondrogenic potential of porcine chondrocytes cultured on these in an ethanol bath. The possibility of obtaining PHBHHx scaffolds
scaffolds were enhanced by increasing the PHBV content and by with tunable pores size and geometry as well as predefined ex-
treating the surface with low pressure oxygen plasma. A recent ternal shape and dimensions was shown by varying CAWS design
study reported on the development of a PHB/PLA blend in the and fabrication parameters (Figure 9c) [238]. Optimized PHBHHx
weight ratio 60/25, containing also 15% wt. of an ester of citric scaffolds developed through this approach supported the in vitro
acid (Citroflex®) acting as plasticizer, that was processed by FDM proliferation of MC3T3-E1 murine preosteoblast cells. A peculiar
into 3D dog bone shapes for tensile mechanical characterization feature of this kind of PHBHHx scaffolds is a homogenous microp-
[379]. Other studies dealt with the development of composite fil- orosity in the polymer matrix as a result of the NIPS process lead-
aments made of maleic anhydride-grafted PHBV reinforced with ing to polymer solidification [7]. A recent study showed the pos-
either palm fiber [615], wood flour [616], or multi-walled carbon sibility of tuning this local micromorphology in terms of concen-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 29

tration of micropores by processing ternary PHBHHx/solvent/non- ical properties, high permeability to many drugs, and good blend-
solvent mixtures with variable composition [254]. Anatomical PHB- compatibility.
HHx scaffolds with dimensions and shape resembling those of a Synthesis route and physical-chemical modification. PCL is
critical size segment of a New Zealand rabbit’s radius were en- a semicrystalline aliphatic polyester with thermoplastic behavior
dowed with a longitudinal macrochannel designed to support host (Figure 10a). High-Mw PCL is industrially synthesized by ROP of
tissue infiltration from the edges of the resected bone once im- ε-caprolactone monomer using tin bis(2-ethylhexanoate) (stannous
planted (Figure 9d) [258]. Mechanical characterization highlighted octoate) as catalyzer in the presence of a higher alcohol initiator
the anisotropic behavior under tension and compression of these (e.g., 1-dodecanol) [619]. PCL has a Tg of ca -60°C, a Tm in the
scaffolds whose compressive stiffness was increased by the pres- range 60-65°C, and a degree of crystallinity usually exceeding 50%
ence of the longitudinal channel. CAWS approach was also success- (Table 2). At the physiological temperature PCL shows an elastic
fully applied to fabricate scaffolds based on a blend of PHBHHx/PCL and ductile behavior since its amorphous domains are in a rub-
with tunable weight ratio between the two polymers.[239] In ad- bery state. As a consequence of a highly hydrophobic nature and
dition, by employing a rotating mandrel as auxiliary dynamic fiber a high crystallinity degree, PCL has a slow biodegradation (years
collector, tubular PHBHHx constructs were recently developed as for complete in vivo absorption [713]) suggesting its use for long-
potential biodegradable intravascular stents for small caliber blood term applications (e.g., load-bearing tissue scaffolds). Overall, PCL
vessels [253]. biodegradation in vivo involves first the non-enzymatic hydrolytic
cleavage of ester groups, and eventually intracellular degradation
once the Mw is decreased to less than 30 0 0 g/mol [228]. The
4.2. Synthetic Biodegradable Polymers
role and efficacy of enzymes, mainly esterases and lipases, in PCL
degradation is not clear and difficult to isolate from other contribu-
Chemically-synthesized biodegradable polymers present many
tions. This aspect greatly depends also on the specifics of the local
advantages over natural macromolecules favoring their process-
environment, like in the case of subcutaneous and intramuscular in
ing, application and industrialization. Indeed, physical-chemical,
vivo PCL degradation in which an absence of enzymatic activity is
mechanical, biodegradation, and functional properties of syn-
suggested [714]. Co-polymerization with other co-monomeric units
thetic polymers can be precisely tailored by changing the
(e.g., d,l-lactide, l-lactide, and ethyl ethylene phosphate) represents
synthesis/modification strategy and conditions to meet specific
an effective means to accelerate the biodegradation rate, and to
application-related requirements.
tune other physical-chemical and mechanical properties of PCL
A wide range of synthetic biodegradable polymers have been
[715–717]. The incorporation of bioactive ceramic particles, includ-
developed for medical and pharmaceutical applications, including
ing HA, TCP, CaCO3 , and Wollastonite, into PCL matrices has been
among others, polyesters, polyoxalates, polyanhydrides, PUs, poly-
also investigated to develop osteoconductive bone implants with
dioxanones, and polyphosphazenes [618]. They are susceptible of
enhanced mechanical properties, to increase polymer hydrophilic-
hydrolytic and/or enzymatic degradation in vivo through hydroly-
ity, as well as to create a pH buffer against the acidic degradation
sis of ester, urethane, urea, or anhydride linkage depending on the
products of the polymer [66].
molecular structure. The biodegradation rate of the polymer is di-
Processing properties and biomedical use. Thanks to its low
rectly related to the backbone linkage stability together with other
melting temperature, high thermal stability, broad processing win-
factors, such as local pH, degree of crystallinity, equilibrium water
dow (Tdeg >300°C), and suitable melt viscoelasticity, PCL can be
content, and material porosity.
easily processed by conventional melting techniques to develop
This section is mainly dedicated to aliphatic polyesters that rep-
composites and 3D devices [718]. PCL is soluble in different sol-
resent the most widely investigated biodegradable synthetic poly-
vents, including halogenates, toluene, cyclohexanone, tetrahydro-
mers for AM thanks to their decade-long history of clinical use
furan, acetone, and dimethylformamide, making it suitable to be
and processing as a melt, solution, or solid (Table 3). As nicely re-
processed also as an organic solution [719]. This aspect has moti-
viewed by Coulembier et al. [619], polyesters synthesis can be carry
vated a vast literature to investigate electrospinning as a suitable
out through either homopolycondensation of a hydroxycarboxylic
technique for the fabrication of PCL micro/nanofibrous meshes for
acid, hetero polycondensation of a diol with a dicarboxylic acid, or
different biomedical applications [720]. In addition, PCL insolubil-
ring-opening polymerization (ROP) of lactones and related cyclic
ity in water, ethyl alcohol, diethyl ether, and petroleum ether can
monomers. The latter method has become the most exploited one
be exploited to employ NIPS approaches [721].
to obtain high molar mass polyesters thanks to its simplicity and
The suitable rheological and viscoelastic properties of PCL, both
suitability to control Mw distribution. The successful application of
as melt or in solution, have been exploited to fabricate a wide
aliphatic polyesters in the biomedical field is related to a combina-
range of medical devices and drug release systems. These include
tion of different factors including biocompatibility of the polymer
among others polymeric rods, micro- and nanoparticles for drug
and relevant degradation products, tunable degradation kinetics in
delivery, sutures, wound dressings, contraceptive implants, fixa-
agreement to the envisaged application, high processing versatility,
tion devices, dentistry materials, and scaffolds for bone, cartilage,
and mechanical properties suitable to a large range of applications
blood vessels, skin, and nerve regeneration [228], [620]. In partic-
[571], [572], [574], [577], [620]. As a consequence, numerous fab-
ular, PCL-based sutures (e.g., Monocryl®) [722] and subcutaneous
rication processes based on different AM techniques, ranging from
contraceptive implants (e.g., Capronor®) [723] have been clinically
ME-AM and SLS to SE-AM and BJ, have been developed to pro-
used for years. In addition, PCL is employed in the production of
cess aliphatic polyesters and poly(ester urethanes) (PEUs). Together
other commercial products, such as tissue repair patches manufac-
with PPF, which was the first biodegradable polyester investigated
tured by Ethicon Inc. and filling agents for non-load bearing bone
for SLA, methacrylate-functionalized derivatives of PCL, PLA, and
cavities [606].
other biodegradable polymers, e.g., PTMC [143], [144], have been
AM application. PCL is the most widely employed biomedical
investigated as suitable photoreactive resin components for AM.
polymer in ME-AM thanks to its well-assessed melt processing
properties, as witnessed by its employment for the fabrication of
4.2.1. Poly(ε -caprolactone) (PCL) biodegradable devices used in the clinical practice (Figure 11a).
PCL represents one of the most investigated synthetic polymers PCL physical properties and thermoplastic behavior meet also
for biomedical applications thanks to its biocompatibility, inex- the basic requirements of SLS. Its solubility in different organic
pensive production routes, controlled biodegradation and mechan- solvents and the suitable rheological properties of the resulting
30 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 10. Chemical structure of biodegradable polyesters investigated for biomedical AM. (a) Poly(ε -caprolactone) (PCL). (b) poly(α -hydroxyacids), i.e., poly(d-lactide)
(PDLA), poly(l-lactide) (PLLA), poly(glycolide) (PGA), and poly(lactide-co-glycolide) (PLGA). (c) poly(propylene fumarate) (PPF).

Figure 11. AM of PCL scaffolds. (a) Clinical application of additive manufactured PCL scaffolds: pictures of PCL conical scaffold by FDM for bone healing (i), scaffold insertion
in a fresh extraction socket (ii), scaffold trimming off with a scalpel after insertion (iii) (reproduced with permission [729]). (b) FDM: top-view of 0/60/120° lay-down pattern
(i); top view (ii) and cross-section (iii) of 0/90° lay-down pattern (reproduced with permission [731]). (c) SLS: pictures and SEM micrographs of side view (i) and (iii) and top
view (ii) and (iv) of PCL/Hap scaffold (reproduced with permission [652]). (d) SLA: photograph (i), μCT visualization (ii) and SEM micrographs (iii) and (iv) of a PCL-based
scaffold (reproduced with permission [654]).
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 31

Table 3
Synthetic biodegradable polymers for biomedical AM.

Polymer AM technique Processing conditions and material design

PCL ME-AM FDM processing of filaments made of PCL possibly loaded with HA, TCP or a drug [203], [579], [581], [621].
Melt extrusion of PCL and relevant bioceramic-loaded composites, driven by pneumatic pressure [622], [623],
force-controlled plunger [583], [624], [625], or screw extruder [204], [207], [626–630].
Hybrid AM through: combination of melt-extruded strands with electrospun fibers [112], [207], [631–635], fully
integration of FDM and electrospinning at a technique level (i.e., MEW) [210], [211], [636–640], sequential
combination of FDM and pore-forming processes (e.g., thermally-induced phase separation [641], salt leaching
[113], [642]), MEW of a photocrosslinkable PCL derivative followed by UV exposure [643].
SE-AM Deposition onto a platform of a PCL solution extruded in air [259].
Hybrid AM through: extrusion and deposition of a PCL solution, possibly containing Hap particles or a drug, into a
non-solvent bath (i.e., CAWS) [26], [74], [105], [252], [253], [644], inkjet printing of a mixture of PCL
dimethylacrylate and PEGDA followed by UV curing [645].
SLS Processing of a PCL powder [4], [646–650], also in combination with polysaccharides (i.e., starch, dextran, or gellan)
[651], Hap [162], [168], [175], [652], or TCP [584], [585], possibly surface-coated with collagen [585] or gelatin
[653].
SLA Photocrosslinking of methacrylate [654] or divinyl-fumarate [655] PCL derivatives, or poly(ester amide) copolymers
[656].

Poly(α -hydroxy ME-AM FDM processing of filaments made of PLLA [14], [657–659], PDLLA [218], [587], [660–663] or PLGA [664], possibly
acids) loaded with TCP, Hap, or drugs.
Pneumatic-driven extrusion of PLLA [586], PLLA/Hap [665], PLGA [215], or PLGA/PCL blends [595]; plunger-driven
extrusion of PLGA/PCL blends loaded with TCP [582], [666–668].
Hybrid AM of PLA through combination of FDM and gas foaming [114], [213].
SE-AM Extrusion of: a solution of PDLLA, containing PEG as plasticizer and possibly a suspension of CaP or BG particles
[261], [262], [590], a concentrated PDLLA solution in a volatile solvent (e.g., dichloromethane) [232]; inkjet printing
of a PLGA solution to form 2D micropatterns [669].
Hybrid AM of: PLLA/TCP [255], [670], PLGA [671], PLGA/TCP [596] and PLGA/pearl [597] by sequential combination
of SE-AM and freeze drying; PLGA through a CAWS process involving the controlled deposition of a tetraglycol
solution into ethanol [672].
SLS Sintering of a powder made of: PLLA [673], PLLA/carbonated Hap [179], [674], [675], PDLLA [676], PDLLA/TCP [166],
[173], PLGA/TCP [598], carbon-loaded PDLLA [172], [677], [678], carbon-loaded PDLLA/Hap [178], or
TCP/carbon-loaded PDLLA [591].
BJ Hybrid AM through controlled deposition of chloroform, possibly containing Pluronic®, onto a PLGA/NaCl powder
bed, followed by salt leaching [679].
Controlled deposition of an acetone/ethanol mixture, containing a drug, onto a PLLA [200], [680] or PDLLA [201],
[202] powder bed.
SLA Photocrosslinking of PDLLA functionalized with fumaric acid [145] or methacrylate [129], [592], [681], possibly
loaded with Hap [594], [682], [683].

PPF SLA Photocrosslinking of PPF diluted in its monomer (i.e., DEF) [567], [599–601], [684], possibly functionalized with
ceramic and peptide coatings [138], [139], [685], [686] or with BMP-2-loaded PLGA microparticles that were
suspended in the photopolymer resin [687], [688].
SE-AM Hybrid AM through extrusion in air of a solution of PPF and DEF, containing a suspension of Hap, followed by UV
exposure [603].
PEUs ME-AM - FDM processing of PCL-based PEUs [689–692], also blended with PVA [693]
- Extrusion of: a dimethylformamide solution of a blend of two aromatic PEUs directly into a water bath [694], a
SE-AM
hexafluoro-2-propanol solution containing a poly(ester urethane urea) and paclitaxel [695], an organic PEU
(bioprinting)
solution onto a low T platform followed by freeze drying [696] possibly by means of a dual nozzle [[287–289],
[697–699], an aqueous dispersion of a thermoresponsive waterborne PEU for bioprinting [700–703].
- Inkjet printing of an acid aqueous medium onto a film of a pH-sensitive PU based on PCL diols [704], [705].

PTMC SLA - Photocrosslinking of formulations containing: PTMC functionalized with methacrylate end groups, possibly as
copolymer with PCL [141], [142], and either Hap [143], [144], [706] or β -TCP [707].

FDM - FDM processing of a copolymer with PLLA [708], [709].

PVA BJ - Deposition of PVA aqueous binders on a polysaccharide powder bed [8].

SE-AM - Extrusion of a drug-containing PVA aqueous solution [106], inkjet printing of PVA aqueous solutions containing
additives (e.g., humectant and pigments) [710].

ME-AM - FDM processing of PVA filaments loaded with either carbon nanotubes [711] or cellulose nanocrystals [712].

PCL solutions have also encouraged research on its processing by in vitro proliferation, differentiation, and ECM synthesis of primary
means of SE-AM techniques (e.g., CAWS). In addition, the well as- human fibroblasts and periosteal cells [579]. In vivo trials showed
certained melt- and solution-blending compatibility of PCL with a that this kind of PCL scaffolds promoted the formation of new bone
number of commercial polymers offers many possibilities to de- tissue in an orbit defect model surgically created in Yorkshire pigs
velop innovative AM materials for biomedical applications. [724], and in a defect on medial femoral condyle of adult New
Hutmacher et al. [203] were the first to develop PCL scaffolds Zealand White rabbits as well [580]. Afterwards, PCL plug scaffolds
by means of ME-AM (Figure 11b). In particular, they employed by FDM were tested in a clinical pilot study involving five patients
FDM to fabricate scaffolds made of PCL or PCL/Hap composites and needing cranioplasty surgery [725]. Twelve months after implanta-
endowed with a tailored porous architecture well supporting the tion, the scaffolds were well integrated in the surrounding calvarial
32 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

bone with new bony tissue filling the porous space. Clinical trials aments by melt extrusion without compromising the molecular
eventually led to FDA approval of a set of PCL scaffolds by FDM structure of the therapeutic agent. By following this approach, Teo
for craniofacial applications. They are currently marketed by Os- et al. [621] developed gentamicin sulphate-loaded PCL/TCP meshes
teopore International in the form of 3D implants (i.e., OsteoplugTM ) with effective antibacterial activity in an infected full thickness
or thin interwoven meshes (i.e., OsteomeshTM ) [726]. Implantation wound mouse model. Similarly, T-shaped PCL prototypes of an in-
of OsteoplugTM scaffolds into burr holes of twelve patients treated trauterine contraceptive device were fabricated by FDM and op-
for a chronic subdural hematoma, resulted in good osteointegration timized to release in vitro indomethacin under sink conditions
without any adverse events, with a mean follow-up of 16 months over a period of 30 days [743]. Kempin et al. [229] developed
[727]. OsteomeshTM implants were recently investigated for orbital hollow cylinder model implants made of PCL, PLLA, ethyl cellu-
floor fracture repair in 20 patients. The study led to good func- lose (EC), or the methacrylic polymer Eudragit® RS, by process-
tional and aesthetic outcomes with no signs of inflammation or ing quinine-loaded filaments prepared by solvent casting followed
reduction in limitation of ocular mobility, while neobone forma- by hot melt extrusion (Figure 12a). In particular, the PCL-based
tion was evident on CT scan performed 18 months after implan- formulation allowed an advanced control over in vitro quinine re-
tation [728]. A pilot randomized controlled clinical trial involving lease kinetics by varying the filament drug loading. A recent case
thirteen patients was aimed at evaluating the feasibility and ef- study was focused on custom-prepared PCL filaments to manu-
fectiveness of using a conical shape PCL scaffold, supplied by Os- facture disk-shaped model oral dosage forms [230]. In particu-
teopore International, in fresh extraction sockets for ridge preser- lar, feedstock indomethacin-loaded filaments with optimized me-
vation (Figure 11a) [729]. The implants were used as space fillers chanical and rheological properties for FDM were developed by
and resulted after 6 months in better bone healing and mainte- tuning the drug loading. PCL was also investigated as a blend
nance of ridge height as compared to extraction sockets without component in combination with hydrophilic polymers, i.e., poly(1-
the scaffold. PCL scaffolds by FDM were also recently shown to be vinylpyrrolidone-co-vinyl acetate) and PEG, to modulate material
good candidates for soft tissue regeneration upon in vitro culture processing properties and the release kinetics of the model drug
in combination with human adipose tissue-derived precursor cells caffeine from relevant FDM-printed tablets [219].
[730]. 2 and 4 weeks after implantation in nude mice, angiogenesis The successful application of FDM to PCL has encouraged re-
and adipose tissue formation were observed throughout the engi- search on alternative ME-AM technological solutions based on ei-
neered constructs by using a femoral arteriovenous flow-through ther pneumatic pressure-, piston- or screw-driven extrusion. As an
vessel loop. example, Yilgor et al. [622] developed PCL scaffolds with differ-
A large body of literature has investigated FDM manufactur- ent fibers stacking organization by using a Bioplotter® (Envisiontec
ing of PCL/TCP composite scaffolds with enhanced mechanical GmbH) with a pressurized cartridge unit heated at 140°C. Implan-
strength, osteoconductive potential, and tailored biodegradation ki- tation in a rat pelvis model showed that bone regeneration within
netics [581], [732–738]. Probst et al. [739] implanted in a hu- the scaffold was enhanced by increasing the pores size and en-
man patient an anatomical PCL/TCP scaffold shaped on the basis dowing the scaffold with a sequential release of BMP-2 and BMP-
of a CT scan of a complex calvaria defect, observing bone heal- 7 from polymeric nanoparticles. A multi-head deposition system
ing and no palpable defect area after 6 months. Different strate- equipped with pressure-assisted melt and solution syringes was
gies have been followed to enhance the bioactivity of PCL/TCP employed to alternatively plotting layers made of either PCL or
scaffolds, such as platelet rich plasma treatment [740], collagen chondrocytes-encapsulated alginate [623]. A ME-AM system using
coating [735], NaOH treatment [738], in vitro culture with animal a steel syringe was employed to fabricate 3D scaffolds made from
[733], or human MSCs [736], and combination with growth fac- a composite of PCL, PLGA, and TCP, and ornamented with cell-laid
tors [741], [742]. For instance, PCL/TCP scaffolds by FDM were cul- ECM that was shown to enhance bone formation in ectopic and
tured in vitro with human MSCs and then implanted in nude rat orthotopic rat models [624]. A similar approach was applied to
critical-sized femoral defects leading to cell survival for up to 3 fabricate PCL scaffolds loaded with synthetic [625] or bio-derived
weeks post-transplantation, as well as 50% of new bone grown Hap [583]. The screw-based PED technology was employed to de-
success rate [736]. Biphasic constructs composed by a PCL car- velop a set of PCL scaffolds with resolution at the microscale [204].
tilage scaffold coupled to a PCL/TCP osseous matrix, both fabri- Optimized PCL scaffold prototypes developed by PED were seeded
cated by FDM, were resurfaced with a PCL/collagen electrospun with primary fetal bovine osteoblasts and then subcutaneously im-
mesh, seeded with MSCs, and implanted into critically-sized os- planted in nude mice showing increased osseous ingrowth dur-
teochondral defects in pigs [737]. Bone ingrowth and remodeling ing a 8 weeks study period [626]. Cell attachment to this kind
with high mineralization rate, functional cartilage restoration, and of scaffolds can be increased by applying a coating made of a
low occurrence of fibrous tissue were observed after 6 months. polysaccharide (e.g., chitosan) after in situ plasma treatment [627].
Biphasic scaffolds were also investigated for the regeneration of Bioextruder is another screw-based system investigated to develop
the goat femoral head by combining an anatomical PCL/Hap scaf- PCL scaffolds with tunable pores size and mechanical properties
fold by FDM (Figure 1f) with a non-woven scaffold composed by varying processing parameters, such as screw rotation velocity
by PLA-coated PGA fibers [33]. These scaffolds were seeded with and nozzle translation velocity [207], [628]. This technology has
chondrocytes and bone marrow stromal cells (BMSCs) and subcu- been optimized to develop PCL scaffolds with high accuracy, pos-
taneously implanted in nude mice leading after 10 weeks to histo- sibly loaded with BGs (e.g., FastOs®BG) [629] or surface-modified
logical structures and biophysical properties similar to those of na- through plasma treatment during fabrication [630].
tive goat femoral head. Chhaya et al. [2] described a clinical case in PCL processing versatility has been greatly exploited to develop
which a 70-year old female patient received a customized scaffold hybrid AM techniques designed to combine hierarchical structural
loaded with autologous bone marrow and BMP-7 for the treatment features. A number of articles have reported on the alternating de-
of a non-union tibial defect of 4 cm. After 18 months of follow-up, position of PCL strand layers by ME-AM and ultrafine fibrous lay-
the scaffold was filled with 75% newly regenerated bone with no ers by electrospinning (Figure 13a). Indeed, the integration of PCL-
reported post-operative discomfort. based additive manufactured structures with electrospun fibers
As previously mentioned, FDM has lately come into focus as made of PCL [631], PCL/silk fibroin blend [632], PCL/collagen blend
potential fabrication technique for drug-releasing medical implants [112], or PLGA [207] has been reported to significantly influence
and pharmaceutical dosage forms. PCL is often investigated in this cell adhesion, proliferation, morphology, and migration. In addi-
field since its low Tm allows the preparation of drug-loaded fil- tion, intercalating collagen nanofibrous layers into a PCL scaffold
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 33

Figure 12. AM of drug-loaded biodegradable implants. (a) FDM manufacturing of quinine-loaded implants made of different polymers, i.e., the methacrylic polymer Eudrag-
it® RS, PCL, PLLA or EC: reflected light microscopic images of side view of the filaments employed (i) and the resulting implants (ii), fluorescence microscopic images of the
implants top view (iii); in vitro cumulative drug release profiles of implants made from different polymers, and of PCL implants loaded with different quinine percentages (iv)
(reproduced with permission [229]). (b) Levofloxacin-loaded PLLA implants by BJ: schematics of implant structures (i.e., matrix, capsule, and double-layer/capsule structures)
(i), picture of fabricated implants (ii), SEM micrographs of the manufactured implants before (iii) and after in vitro release for 40 days (iv) (reproduced with permission
[200]).

by ME-AM was shown to enhance the mechanical properties of porated a surface microporous structure into a FDM-manufactured
the resulting construct [633]. A more sophisticated fabrication pro- PCL scaffold by infusing it with a ternary mixture of PCL, 1,4-
cess was developed by alternating PCL strand layers with electro- dioxane, and water that underwent thermally-induced phase sep-
spun PCL and osteoblast-like cells-laden alginate struts in order aration followed by lyophilization. In order to fasten and optimize
to achieve homogeneous cell proliferation throughout the scaffold drug release from PCL scaffolds by FDM, Visscher et al. [113] in-
[634]. A multiscale hybrid AM approach was also investigated by troduced a tunable microporosity in the polymeric matrix through
Yoon and Kim [635] who integrated an electrospun mesh made of salt leaching, and then applied a Gel-MA coating achieving a sus-
rhodamine-B-loaded core/shell PEG fibers into a PCL scaffold fab- tain release of cefazolin for up to 3 days (Figure 13c). The micro-
ricated by ME-AM. In this case, drug release could be tailored by porosity was also shown to facilitate the scaffold degradation dur-
sandwiching the hydrophilic mesh between two electrospun PCL ing an accelerated test in NaOH. The combined FDM-salt leaching
meshes with adjustable thickness. approach was also recently employed to control the in vitro release
MEW represents a successful example of AM and electrospin- of different drug models (i.e., doxorubicin, paclitaxel, and cefazolin)
ning integration at a technique level to fabricate PCL structures from PCL scaffolds [642].
up to 10 mm in height over relatively large build areas, approach- PCL scaffolds with a dual-scale porosity can be also fabricated
ing single micron filament resolution and 20 μm placement reso- through a one-step CAWS process involving the extrusion of an
lution (Figure 13b) [210], [211]. PCL has been processed by MEW acetone polymeric solution into an ethanol bath (Figure 13d) [26].
into scaffolds with cuboid [211], [636] or tubular geometry [637], The scaffold can be functionalized by adding to the PCL solution
[638], and as fibers integrated in a hydrogel as pore-forming sacri- an osteoconductive ceramic [252] or a drug [105] that is then in-
ficial material [639] or reinforcing network [640]. Bicompartmen- corporated in the polymeric matrix upon coagulation. PCL scaf-
tal scaffolds were also developed by combining different PCL con- folds by CAWS were shown to support the adhesion, proliferation,
structs fabricated by FDM and MEW for periodontal TE [744] or by differentiation, and mineralized matrix deposition of murine pre-
combining a PCL construct by MEW with an alginate gel for os- osteoblast cells [26] and human ASCs [644]. Combining these PCL
teochondral TE [745]. In vitro studies have demonstrated that PCL scaffolds with an integrated chitosan-based hydrogel is an effec-
scaffolds developed via MEW can support cell attachment, prolifer- tive means to endow them with antimicrobial properties [252].
ation, ECM formation, and cell infiltration throughout the thickness PCL tubular constructs can be fabricated by using a rotating man-
of the scaffold, facilitated by the large pores and the high pores drel into the coagulation bath as an auxiliary fiber collector [253].
interconnectivity [746]. Different articles have reported animal ex- In addition, CAWS was employed to process a three-arm star PCL
periments involving the subcutaneous implantation in immuno- (∗ PCL) into dual-scale porous scaffolds with enhanced mechanical
compromised rodents of this kind of PCL scaffolds, often function- properties comparing to analogous constructs made from PCL with
alized with recombinant human BMP-7 and CaP coatings. In par- a linear molecular structure [251]. This kind of ∗ PCL scaffolds was
ticular, as previously mentioned, PCL tubular constructs by MEW endowed with an anatomical geometry, and implanted in critical
were investigated to develop humanized bone organ models in size defects created in a rabbit radius model, showing good inter-
mice for studying metastatic sites of prostate and breast cancer action with the host tissue without inflammatory response [74].
cells [89], [94], [95]. After 3 months, the defects were bridged with new bone tissue
The sequential combination of FDM with a pore-forming pro- covering the scaffolds and penetrating into the first fibrous layers.
cessing technique is an effective strategy to develop PCL scaffolds AM of PCL-based solutions through their extrusion and deposition
with a dual-scale porosity. For instance, Jensen et al. [641] incor- in air was also investigated to fabricate porous structures, achiev-
34 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 13. Hybrid AM of PCL. (a) Dual-scale fibers scaffolds by FDM and electrospinning integrated at a fabrication level: representative picture (i) and SEM micrographs
100x (ii) and 10 0 0x (iii) of scaffold composed of PCL macrostrands and PCL nanofibers (reproduced with permission [112]). (b) Microfiber scaffolds by MEW: representative
picture (i) and SEM micrographs (ii) and (iii) of scaffold composed by PCL microfibers (reproduced with permission [211]). (c) Dual-scale porous scaffolds by sequential FDM
and salt-leaching: representative cross-section SEM and μ-CT micrographs of dense PCL scaffold by FDM (i and ii) and microporous PCL scaffold by FDM/salt leaching (iii)
and (iv) (reproduced with permission [113]). (d) Dual-scale porous scaffolds by CAWS: representative picture (i) and SEM micrographs of top view (ii) and cross-section (iii)
of PCL scaffolds (inset high magnification micrographs show polymer microporosity) (reproduced with permission [26]).

ing a limited development along building direction (Z axis) [259]. surface-coated with collagen [585] or gelatin [653] for bone and
Overall, PCL scaffolds fabricated by CAWS show less accurate geo- cartilage TE. In general, these kinds of scaffold are characterized by
metrical and dimensional fidelity to the CAD model in comparison a lower resolution and a higher surface roughness than PCL scaf-
to what observed when ME-AM is applied to PCL because of dif- folds obtained by other AM techniques (Figure 11b). Smith et al.
ferent factors, such as volumetric shrinkage and geometrical dis- [646] computationally designed and fabricated a condylar ramus
tortion during coagulation, as well as fluid dragging forces acting unit PCL scaffold for temporomandibular joint reconstruction in a
on the depositing filament. On the other hand, the aforementioned Yucatan minipig animal model. Once implanted and fixed in the
advantages of CAWS in terms of versatility in microstructural and mandible using mini-plates, the scaffold could support masticatory
drug loading functionalization should be also taken into account. function as well as both osseous and cartilage regeneration. Nano-
SLS was first applied to PCL in 2005 for the fabrication of scaf- Hap inclusion into PCL scaffolds by SLS led to enhanced bone for-
folds that were subcutaneously implanted in an immunocompro- mation and increased biodegradation velocity after seeding with
mised mice model to study their suitability to support bone in- human MSCs and implantation in a critical femoral bone defect
growth [4]. After that, several studies investigated SLS to process in rabbits [652]. Composite microspheres made by S/O/W meth-
PCL also in combination with polysaccharides (i.e., starch, dextran, ods can be employed, in place of mechanically mixing PCL and
or gellan) [651], Hap [168], [175], [652], or TCP [584], possibly Hap particles, in order to obtain more uniform granulometry and
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 35

particles shape, even if mechanical properties worsening can oc- ferred to as poly(lactic acid), while those synthesized by ROP of
cur [168]. A recent study reported on the development of PCL/TCP lactide as poly(lactide).
scaffolds by loading the ceramic phase up to 30% wt. with a re- Considering that lactic acid is generally obtained by microbial
sulting significant increase in the compressive modulus [585]. The fermentation and that the resulting macromolecule in turn can
in vitro osteogenic differentiation of porcine ASCs seeded on these degrade down back to lactic acid, PLA is generally considered as
scaffolds was enhanced by applying a collagen type I coating that an environmentally friendly material [309]. Dilactide is synthe-
also favored in vivo bone formation and vascularization after intra- sized with high yield and selectivity by using weakly basic cat-
muscular implantation of the construct in nude mice. Analogously, alysts (e.g., tin and zinc oxides, organostannates and -titanates)
collagen type II coating favored the in vitro and in vivo prolifer- [748]. The l-isomer is the preferred feedstock for dilactide since
ation of porcine chondrocytes seeded on PCL scaffolds.[653] Zopf the majority of lactic acid from microbial fermentation exists in
et al. [22] described for the first time in 2013 the implantation of this form and it provides high dilactide yield for the synthesis
a PCL device in an infant suffering from tracheobronchomalacia to of high-Mw PLAs with high crystallinity and mechanical strength
prevent airway collapsibility (Figure 1e). From 2012 to 2018, 15 [604]. A large range of properties can be obtained by copolymer-
subjects with a median age of 8 months received 29 splints on ization with the d-isomer of lactic acid as well as with other oxy-
their trachea, right and/or left mainstem bronchi [747]. The tra- genated functional monomers, such as glycolide, ε -caprolactone,
cheobronchial splints were developed on the base of a 3D model and polyether polyols. A l-lactide content above 90% typically re-
of the patient’s airway and fabricated by SLS starting from a pow- sults in a semicrystalline structure with enough toughness and
der made of PCL [647] or a blend of PCL with Hap (4 wt.%), added high tensile strength. When the l-lactide percentage is below 80%,
as a flowing agent during the sintering process [162]. The same an amorphous morphology is obtained. Unfortunately, some com-
technology was investigated to develop patient-specific PCL scaf- panies do not clearly specify the composition and stereoisomeric
folds for craniofacial soft tissue reconstruction that were tested form of the marketed PLA batches. In some cases, commercially
in vitro [648], [649]. As an example, auricular scaffolds seeded available PLAs are blends of PLLA and PDLLA. Relevant informa-
with primary porcine auricular chondrocytes and cultured in a tion can be however deduced from polymer Tm , crystallinity, and
pro-chondrogenic medium were subcutaneously implanted in rats macroscopic appearance (i.e., translucent or opaque).
[650]. After 4 weeks, diffuse chondrogenic tissue formation was PLLA has been reported to have a Tg in the range 55-80°C, a
observed in both the peripheral and central aspects of the explants. Tm in the range 170-180°C, and a crystallinity degree in the range
Attempts to fabricate PCL-based constructs by photoretico- 40-50% (Table 2) [566], [569]. Since the amorphous domains of
lation AM were also reported. For instance, three-arm ∗ PCL PLLA are in a glassy state at the physiological temperature, the
oligomers of various Mw were end-functionalized with methacrylic polymer behaves as a brittle material when employed for clinical
anhydride, and processed by SLA to fabricate 3D biodegradable use. PDLA has typically lower Tg and Tm values than PLLA [568].
scaffolds with a predefined porous structure (Figure 11c) [654]. BG In addition, by increasing the d-stereoisomer content in PDLLA the
incorporation up to 20 wt. % into the polymer matrix, through its Tg and Tm are significantly decreased [749]. In general, commer-
dispersion in the photoreactive resin, allowed enhancement of fi- cial PLAs are soluble in different solvents, such as dioxane, ace-
broblasts proliferation. An alternative fabrication approach is based tonitrile, chloroform, methylene chloride, 1,1,2-trichloroethane, and
on inkjet printing of a mixture of PCL dimethylacrylate and PEGDA dichloroacetic acid, and partly soluble in ethyl benzene, toluene,
followed by UV curing to manufacture mesh structures with differ- acetone and tetrahydrofuran [570]. However, in the case of highly
ent patterns [645]. Divinyl-fumarate PCL was also recently inves- crystalline PLLA the list of suitable solvents is restricted. Water, al-
tigated as an alternative photocurable polymer to overcome prob- cohols (e.g., methanol, ethanol, and propylene glycol) and unsub-
lems related to substances leached out from acrylic resins, such as stituted hydrocarbons (e.g., hexane and heptane) are non-solvents
inflammation and allergic reactions [655]. The polymer was suc- exploited for PLA precipitation procedures.
cessfully employed in combination with NVP, as a reactive dilu- The development of new routes for the production of high-Mw
ent, to fabricate by μSLA porous scaffolds with gyroid and dia- polymer batches together with the growing environmental aware-
mond architectures supporting the adhesion and spreading of hu- ness in the general public have expanded the PLAs use for con-
man MSCs. SLA fabrication of 3D scaffolds through photocrosslink- sumer products and packaging applications [750]. In the biomed-
ing of poly(ester amide) copolymers based on ε -caprolactone and ical field PLLA has been investigated for different applications re-
l-alanine was also reported [656]. quiring a mechanical role, leading to various biodegradable prod-
ucts on the market and used in the clinical practice, including
4.2.2. Poly(α -hydroxy acids) long lasting sutures, suture reinforcements, suture anchors, menis-
Poly(α -hydroxy acids), including PLA, PGA, and their copoly- cal darts, and a wide range of devices for guided tissue regenera-
mers (Figure 10b), are widely investigated in the biomedical area, tion barrier, osteosynthesis, and orthopedic fixation (e.g., Atrisorb®
and clinically employed for various applications thanks to their FreeflowTM , Bio Interference Screw®, BioScrew®, Biotrak®, and
biocompatibility, controlled biodegradability into safe byproducts Bio-Anchor®) [573], [576], [606], [751], [752]. In addition, PLLA is
that can be resorbed through normal metabolic pathways, and me- considered as the reference biodegradable material for intravascu-
chanical properties suitable for different applications. lar biodegradable stents thanks to its relatively low Tg above the
Poly(lactide) (PLA). As a consequence of the chirality of lactic physiological temperature, which can be exploited for balloon ex-
acid (2-hydroxy propionic acid) that can exist in optically active d- pansion implantation procedures [753]. As a consequence of its
or l- isomers, different forms of PLA exist as poly(l-lactide) (PLLA), more brittle behavior, lower mechanical properties, and higher hy-
poly(d-lactide) (PDLA), poly(d,l-lactide) (PDLLA), and poly(l-lactide- drophilicity, amorphous PDLLA is mainly investigated for drug re-
co-d,l-lactide) (PLDLLA). The polymer stereochemical structure can lease applications [408].
be easily modified by polymerizing a controlled mixture of l- and Poly(α -hydroxy acids) are degraded in the human body mainly
d-isomers to yield semicrystalline or amorphous polymers. On an through hydrolysis into oligomers and monomeric acids, which can
industrial scale, high-Mw PLAs are commonly synthesized by ring- enter the tricarboxylic acid cycle and be excreted in the form of
opening polymerization of dilactide (3,6-dimethyl-p-dioxane-2,5- carbon dioxide and water via respiratory route and renal filtra-
dione), the cyclic dimer of lactic acid. The nomenclature adopted tion [754]. Depending on the application, the complete resorption
for PLAs is often confused and contradictory. As a rule of thumb of PLLA devices takes long time, such as in the case of interfer-
polymers obtained by polycondensation of lactic acid should be re- ence orthopedic screws, which persist in vivo for up to 5 years and
36 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

require 7 to 10 years for complete resorption [755], [756]. Amor- sorb®), bioresorbable fixation plates, screws, and pins for bone
phous PDLLA is in general subjected to a faster biodegradation fracture stabilization and craniofacial reconstruction (Rapidsorb®
than semicrystalline PLLA [757]. In addition, the methyl pending plates, LactoSorb®, RFSTM Screw and Pin System) [752], [776].
group confers to PLAs repeating unit a more hydrophobic character, AM application. Highly crystalline PLAs (e.g., PLLA) are among
generally causing a slower biodegradation in comparison to PGA. the most employed materials in FDM thanks to their relatively
The main biodegradation stage of poly(α -hydroxy acids) involves low Tm (<180°C) enabling their processing outside of a dedicated
a non-specific, bulk hydrolytic scission of ester bonds. After that, facility without the use of a heated construction platform.[777]
the cleavage of the chain end groups (chain unzipping) takes place However, as previously mentioned, PLAs can be susceptible to
together with the solubilization of the degradation products [620]. degradation during thermal processing [227]. In addition, draw-
The carboxylic acid end groups and the carboxylic degradation by- backs related to low melt elasticity and strength, often resulting
products can catalyze the hydrolytic process causing sometimes in melt rupture during processing, have been connected to PLLA
undesired effects, such as premature fail of the implant, tissue in- high chain stiffness and low entanglement density [778], [779].
flammation due to local acidification, denaturation of loaded drugs As discussed before, small percentages of d-lactide are enough
and/or poor control over their release kinetics [758], [759]. Inclu- to significantly decrease the crystallinity degree and change poly-
sion of bioceramics (e.g., Hap nanopowder) into polymeric matri- mer melt rheology. Macromolecular chain stiffness and morpho-
ces, besides enhancing material osteoconductivity and mechanical logical properties can be also modulated through copolymeriza-
strength [760], [761], can represent a strategy to counteract the tion with other co-monomeric units. For instance, poly(l-lactide-
acidic degradation by stabilizing the pH of the local environment co-ε -caprolactone) (PLACL) and poly(l-lactide-co-trimethylene car-
[762], [763]. bonate) (PLATMC) can be engineered in order to be endowed with
Poly(glycolide) (PGA). It is highly crystalline (45-55%), with a Tg a chain flexibility optimized for melt processing [708], [709]. In-
between 35°C and 40°C, relatively high Tm (> 200°C) and tensile deed, commercial PLACL and PLATMC are characterized by a sig-
strength (12.5 GPa) [575]. High-Mw PGA is typically synthesized by nificant lower Tg (e.g., 13 and 32°C, respectively) in comparison to
ROP of glycolide, the cyclic dimer of glycolic acid, commonly using PLAs [709].
as catalysts organo-tin, antimony, or zinc.[578] Although PGA was The fabrication of PLLA scaffolds by means of ME-AM was re-
used in the manufacturing of the first synthetic absorbable suture ported for the first time in 2001 by employing a pneumatic system
(DEXON®) in the sixties [764] and in the production of internal designed to process polymeric grains [586]. The possibility to ex-
bone pins (Biofix®) until 1996 [765], its rapid degradation in aque- trude PLLA filaments for FDM, possibly loaded with TCP, was sub-
ous environment and insolubility in common solvents have limited sequently shown [657]. Different studies demonstrated that FDM-
its investigation as main component of load-bearing devices and manufactured dog bone shaped samples made of different com-
drug release systems. Besides a quick decrease of scaffold mechan- mercial PLAs have tensile modulus and strength comparable to
ical properties, PGA fast biodegradation can lead to local glycolic those reported for PLLA-based films prepared by means of solvent
acid concentration and subsequent inflammation [766]. Anyhow, a casting or compression molding [14], [657], [658]. In particular,
few studies investigated PGA as a filler material in combination Chacón et al. [15] showed that PLA samples fabricated by FDM ex-
with other biodegradable polymers, or in the form of non-woven hibited anisotropic mechanical behavior that could be changed by
fabrics to support the regeneration of different tissues, including acting on build orientation, layer thickness, and feed rate. Chhaya
bone [767], [768], cartilage [769], [770], and tendon [771]. et al. [218] employed FDM to fabricate PDLLA scaffolds with a ge-
PLGA copolymers. They have been extensively employed in ometry modelled in silico via a laser scanning data set from a pa-
the biomedical field to exploit the possibility of tailoring their tient who underwent breast reconstruction surgery. These breast-
biodegradation, mechanical, and processing properties by varying shaped scaffolds were employed for in vivo modelling of angio-
the lactide/glycolide molar ratio, the end groups, and the lactide genesis and adipose tissue formation in athymic nude rats. FDM
isomeric form (i.e., l- or d,l). For instance, the degradation time was also recently employed to fabricate a porous PLLA screw-like
can be decreased from 5-6 months to 1-2 months by varying the scaffold that was coated with Hap and loaded with MSCs [659].
d,l-lactide/glycolide molar ratio from 85:15 to 50:50 [772]. Indeed, In vivo assessment in a rabbit anterior cruciate ligament model
as a consequence of their higher hydrophobicity, the lactic acid showed that PLLA screw constructs supported significant bone in-
moieties are hydrolyzed slower than the glycolic acid ones, result- growth and bone-graft interface formation. A recent article re-
ing in a variation of their ratio during biodegradation. The pro- ported on FDM fabrication of scaffolds composed of semicrystalline
cessing technique and the resulting macromolecular morphology PLA strands (~115 μm) and with different pore size (150, 200, 250
also play a key role in the resulting biodegradation kinetics, as re- μm), achieving high reproducibility and good dimensional fidelity
ported by an article demonstrating a significantly slower biodegra- to the CAD model [660]. However, thermal processing caused a sig-
dation of PLGA electrospun fibers in comparison to dense films nificant reduction of polymer Mw and Tdeg , even if the scaffolds
made from the same raw copolymer [773]. This finding was ex- well-supported the proliferation of human BMSCs. Low porosity
plained with the high packing density and axial orientation of PLA scaffolds with enhanced mechanical properties were recently
polymer chains during electrospinning, slowing down water dif- fabricated by FDM and investigated for bone TE [588]. Another
fusion in spite of the much higher surface area of electrospun study showed that by optimizing pore size (i.e., 750 μm) of PLA
fibers in comparison to monolithic films. PLGA copolymers have scaffolds fabricated by FDM, osteoblasts proliferation, metabolic ac-
been particularly investigated for different biomedical applications, tivity, and osteogenic matrix protein production were enhanced
such as electrospun fibers [773] or micro/nanoparticles [774] for [587].
drug release, and 3D porous scaffolds for TE [775]. Various PLGA- Lately, PLLA/Hap scaffolds fabricated by a piston ME-AM sys-
based products are available on the medical market, including in- tem were seeded with BMSCs and implanted in a rabbit model
jectable formulations for the treatment of prostate cancer and en- used as in vivo bioreactor to generate a large vascularized tissue
dometriosis (LUPRON DEPOT®), opioid and alcohol dependence engineered bone (Figure 14) [665]. In particular, the scaffold was
(Vivitrol®), human growth hormone deficiency (Nutropin Depot®), designed with a central channel and crossed with the saphenous
or macular edema and non-infectious uveitis (OZURDEX®). Other vessel bundle. Other attempts to biofunctionalize PLA scaffolds
commercial PLGA devices are absorbable sutures (Vicryl®, Vicryl fabricated by FDM involved coating them with gelatin/polylysine
Rapide®, Polysorb® and Purasorb®), patches for wound treatment interpenetrating polymer networks, designed to release differ-
and skin regeneration (Vicryl® mesh, Dermagraft®, CYTOPLAST Re- ent growth factors with a spatiotemporal controlled mechanism
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 37

Figure 14. Bone development research on poly(α -hydroxy acids) scaffolds by FDM: simplified diagram of vascularized tissue engineered bone development through in vivo
bioreactor culturing of scaffolds seeded with BMSCs (i), representative pictures of side view and top view of a channeled PLLA/Hap composite scaffold for in vivo cell
culturing (reproduced with permission from [665].

[589], or incorporating in the polymer matrix RGD-conjugated gold was increased and bone regeneration was enhanced after implan-
nanoparticles.[780] In addition, PLAs are among the pharmaceuti- tation in a rabbit calvarial defect model.[582] The same approach
cal grade polymers that can be extruded into filaments loaded with was used to manufacture PCL/PLGA/TCP membranes for guided
drugs to produce custom-made oral dosage forms or implantable bone regeneration in beagle implant models [666], [667]. Song
drug-releasing systems. For example, printing PDLLA model disk et al. [668] dispensed an alginate hydrogel, containing PLGA mi-
geometries incorporating the antimicrobial nitrofurantoin was de- crospheres loaded with the immunosuppressive drug cyclosporin,
scribed in details by different articles [661–663]. between strands made of a PLGA/PCL blend that were fabricated
Nanocomposite scaffolds by FDM were recently developed by ME-AM. The resulting 3 layers-integrated construct was seeded
through solvent casting blending of PLGA (lactide/glycolide ratio: with xenogeneic cells (i.e., human lung fibroblasts) and subcuta-
65/35) and TCP in a 75:25 weight ratio before melt extrusion into a neously implanted into the BALB/c mouse, resulting in significant
continuous filament.[664] The resulting scaffolds were coated with suppression of T-cell-mediated rejection for 4 weeks.
Hap and implanted in a rabbit femoral defect model showing good Zhou et al. [114] were the first to report the possibility of ob-
osteointegration, bone regeneration throughout the construct, and taining PLA scaffolds endowed with a dual-scale porosity by a se-
marked biodegradation after 12 weeks. Another study reported al- quential application of FDM and gas foaming. They successfully
most complete degradation of PLGA (lactide/glycolide ratio: 50:50) integrated macropores (10 0–80 0 μm) determined by the designed
scaffolds fabricated by pneumatic ME-AM after 8 weeks of their lay-down pattern, with interconnected micropores (1–10 μm) in
implantation in a rabbit tibia model (Figure 15a) [215]. Since this the polymer matrix formed by submitting the printed scaffold to
degradation rate is too fast for bone TE, a few articles described CO2 saturation and foaming. Marascio et al. [213] further developed
the processing of PLGA blended with PCL (typically in a 1:1 weight this approach by submitting PLA-based filaments to CO2 saturation
ratio) by means of pneumatic ME-AM. For instance, Hong et al. and then achieving polymer gas foaming during FDM due to the
[595] showed that the surface-functionalization of PCL/PLGA scaf- high processing temperature (Figure 16a). This method was vali-
folds by means of RGD fused on a coating of mussel adhesive dated by developing PLLA/TCP and PLACL scaffolds with macropore
proteins significantly enhanced the attachment, proliferation, and size of tens of microns and composed by strands with an intercon-
osteogenic differentiation of human ASCs. Another study demon- nected microporosity in their cross-section.
strated that by including TCP into PCL/PLGA scaffolds fabricated Xiong et al. [255] extruded a dioxane solution of PLLA in which
by mechanical pressure ME-AM, the resulting compressive strength TCP was suspended by using a low-temperature deposition SE-
38 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 15. AM of poly(α -hydroxy acids). (a) PLGA scaffolds by FDM: picture (i) and SEM micrograph of top view (inset high magnification micrograph shows scaffold lateral
view) (ii) of 3D scaffolds; histological specimens stained with hematoxylin and eosin (× 40) of PCL (iii) and PLGA (iv) constructs at 8 weeks after implantation (reproduced
with permission [215]). (b) PDLLA/TCP composites (50:50 weight ratio) by SLS: light microscope and SEM images of the top surfaces of test specimens (6 × 6 × 6 mm) for
different laser power settings; batch 1 with higher Mw, higher polymeric particles size, and lower ceramic particle size (i) and (ii) showed signs of carbonization (black dots)
and a more porous surface than batch 2 (iii) and (iv) (reproduced with permission [166]). (c) PDLLA derivative scaffolds by SLA: top-view μCT-visualization and thresholded
z-stacks of confocal microscopy images (500 μm thickness) showing the distribution of cells densities and wall shear rates in isotropic (i) and (iii) and gradient (ii) and (iv)
scaffolds (reproduced with permission [681]).

Figure 16. PLA scaffolds with dual-scale porosity. (a) PLLA/TCP foamed scaffold structure by sequential gas foaming and FDM: picture (i) and SEM micrographs of top-view
(ii) and fiber-fiber contact points (iii) and (iv) (reproduced with permission [213]). PDLLA scaffolds by SE-AM: SEM micrographs at different magnifications of top-view (i),
transversal cross-section (ii), fiber-fiber contact point (iii) and fiber surface microporosity (reproduced with permission [261]).
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 39

AM system. The subsequent sublimation of dioxane created a mi- planted into murine critical-sized femur segmental defects [677].
croporosity in the polymeric matrix forming a dual-scale porosity. Moreover, these scaffolds were seeded with human bone marrow
Similar technological approaches were adopted to fabricate PLGA MSCs and subcutaneously implanted in rats maintaining cells ad-
[671], PLGA/TCP [596], and PLGA/pearl composite scaffolds [597]. A hesion for 90 days without cytotoxicity signs [678]. SSLS approach
multi-nozzle low-temperature deposition system was employed to was also suitable to fabricate scaffolds made of composites with
obtain gradient compositions and morphologies in PLLA scaffolds, TCP/PDLLA weight ratio up to 80:20, showing compressive proper-
as well as to load them with BMP-2 [670]. 12 weeks after their im- ties comparable to those of trabecular bone [591].
plantation, the developed scaffolds were effective in repairing a 15- One of the earliest attempts to apply BJ to poly(α -hydroxyacids)
mm segment defect created in a rabbit radius model. However, the involved the controlled deposition of chloroform to pattern PLGA
described SE-AM approaches did not allow an accurate control over powder surfaces and to fabricate 3D porous scaffolds by combina-
deposition details and the resulting scaffold porous structure. Serra tion with salt leaching.[679] In addition, several efforts have been
et al. [261] achieved enhanced resolution of the deposition parts spent on the use of BJ for fabricating drug-eluting PLA implants
by employing PEG as a plasticizer to facilitate PDLLA solution ex- for the treatment of infected bone [783]. Notably, PLLA implant
trusion in air without any post-processing treatment (Figure 16b). prototypes loaded with a drug (e.g., antibiotic and antituberculo-
The scaffolds could be loaded with CaP particles after their suspen- sis agents) that was either dispersed in the polymeric matrix or
sion in the starting chloroform solution, leading to increase in sur- contained in an inner reservoir were developed [200], [784]. For
face roughness, hydrophilicity, and adhesion of primary MSCs. The instance, Huang et al. [200] added the antibiotic levofloxacin to
variation of PEG percentage in the solution could be exploited too a binder solution composed of a mixture of acetone and ethanol,
to vary scaffold surface roughness, wettability, mechanical proper- in order to fabricate PLLA implants with different loading design,
ties, and biodegradation rate [262]. This approach was further de- drug distribution, and in vitro release kinetics (Figure 12b). By fol-
veloped to fabricate a biphasic osteochondral construct, composed lowing similar approaches, a set of implants with complex lay-
of a PDLLA scaffold coupled to a PDLLA/BG composite scaffold, that ered structures were developed in order to achieve a sequential
was subcutaneously-implanted in a mice model for a preliminary release of different antibiotics [201], [202], [680]. As an example,
in vivo evaluation [590]. The potential of SE-AM for novel drug levofloxacin and tobramycin were recently incorporated into multi-
delivery systems development was shown by developing a multi- layered concentric cylinders made of PDLLA to achieve a sustained
layer system composed by a dexamethasone disodium salt-loaded and programmed multi-step release kinetics optimized for chronic
PVA layer sandwiched between PLGA layers [106]. Mironov et al. osteomyelitis treatment [202].
[672] recently described a novel CAWS method for scaffold fabri- One of the first published studies on SLA application to PLA
cation based on the controlled deposition of a tetraglycol solution derivatives involved photocrosslinking fumaric acid monoethyl
of PLGA directly into an ethanol bath. A solvent-cast AM approach ester-functionalized three-armed PDLLA oligomers using NVP as
was followed to fabricate different kinds of 3D self-supporting mi- diluent and comonomer [145]. Other studies were based on
crostructures (e.g., square spiral, circular spiral, micro-cup, and lay- methacrylate end-functionalized PDLLA and its copolymers. In
ered scaffolds) by extruding a concentrated PDLLA solution in a particular, PDLLA oligomers with 2, 3, or 6 arms were synthe-
highly volatile solvent (i.e., dichloromethane) [232]. In addition, sized, end-functionalized with methacryloyl chloride, and photo-
an inkjet printing process was lately employed to deposit a N,N- crosslinked in the presence of ethyl lactate as a non-reactive
dimethylformamide solution of PLGA on a PS substrate for pattern- diluent, to fabricate high-resolution 3D scaffolds supporting in
ing human ASCs adhesion [669]. vitro preosteoblast cells proliferation [129], [592]. These photo-
Various efforts have been spent on applying SLS to PLAs for polymerizable PDLLA macromers were investigated for the fabri-
the fabrication of porous scaffolds, possibly in combination with cation of scaffolds with isotropic or gradient pore size to con-
bioceramics [781]. In particular, 3D scaffolds made of PLLA [673], trol the distribution of cells seeded under perfusion (Figure 15c)
also loaded with carbonated Hap [179], [674], [675], or PLGA/TCP [681]. Porous scaffolds built by employing a poly(d,l-lactide-co-
[598] were successfully fabricated. However, limited polymer parti- ε-caprolactone) (PDLACL)-based resin showed excellent fidelity to
cles fusion generally resulted in marked porous defects that wors- the CAD design and controlled compressive properties ranging
ened the mechanical answer of this kind of scaffolds (compres- from rigid and strong to highly flexible and elastic [593]. Compos-
sion strength < 3MPa) [675], [782]. A high-density polymer ma- ite scaffolds with different pore sizes were also fabricated by mix-
trix could be manufactured when PDLLA was completely melted ing Hap particles to photocurable methacrylate end-functionalized
[676]. In general, with increasing laser power unwanted porosity PDLLA oligomers [594], [682], [683]. Hap nanoparticles inclusion
decreased until a fully coalesced structure was obtained, with the increased the scaffold mechanical properties and osteoconduc-
risk, however, of local carbonization. PDLLA/TCP composites (50:50 tive potential without affecting the morphology of the structure
weight ratio) with a close and dense surface morphology can be [594], [683]. Ultrafine fibers scaffolds for soft TE with high resis-
obtained by melting the polymer powder in order to glue the ce- tance to tensile dynamic loading were recently developed by pro-
ramic particles together [173]. In this case, a small polymer parti- cessing poly(l-lactide-co-ε -caprolactone-co-acryloyl carbonate) by
cle size, a large filler particle size, as well as a low polymer Mw means of MEW followed by UV photocrosslinking [643].
can allow melt viscosity and sintering rate suitable to fabricate
dense parts (Figure 15b) [166]. In 2004 Antonov et al. [172] re- 4.2.3. Polypropylene fumarate (PPF)
ported on an AM method called surface-SLS (SSLS) based on near- Owing the presence of a double carbon-carbon bond in its
infrared (λ=0.97 μm) laser radiation of PDLLA particles that were repeating unit, which allows crosslinking into a covalent poly-
loaded on the surface with carbon microparticles to enhance lo- mer network, PPF is the most investigated synthetic biodegradable
calized heating. The authors demonstrated that it was possible to polymer for photoreactive AM strategies.
load the scaffold with Hap or the ribonuclease A enzyme without Synthesis route. PPF is a linear polyester consisting of repeat-
compromising its activity, as well as to change scaffold topogra- ing units containing one unsaturated bond in trans configuration
phy by varying polymeric particles size and carbon content [178]. (Figure 10c). Optimized protocols to synthesize PPF on a lab scale
The use of carbon does raise issues of concern regarding safety and are based on a two-step reaction of DEF and propylene glycol
biocompatibility. However, optimized PDLLA scaffolds fabricated by through a bis(hydroxypropyl) fumarate diester intermediate [785].
SSLS sustained human fetal femur-derived cells viability, growth, Since its development in the late 1980’s [786], [787], various lab
and osteogenesis in vitro, and favored bone formation when im- procedures for the synthesis of PPF for biomedical applications
40 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

have been developed. However, many shortcomings related to the experimental investigation showed that changes in DEF concentra-
step-growth polymerization of PPF are hindering its moving into tion and pore size significantly affected scaffold mechanical stiff-
clinical materials market. Indeed, issues influencing the final mate- ness and osteogenic signal expression of rat BMSCs [601].
rial properties, such as low conversion, poor control over molecular PPF scaffolds with a 3D porous architecture obtained by al-
mass distribution, conjugate-addition side reactions, and unwanted ternating layers composed of either lattices or columns were dip
crosslinking, are reported in literature [788]. coated with biomimetic apatite and RGD to biofunctionalize the
Photoreticolation and biomedical use. Photochemical crosslink- surface [138]. In vitro investigations showed that preosteoblast cells
ing of PPF has been broadly explored to obtain biomedical ma- proliferation in these biofunctionalized scaffolds was improved
terials with suitable mechanical and degradation properties. The compared to non-coated samples. Similarly, Shin et al. [139] sub-
crosslinked network can be tailored by varying the molecular mitted PPF/DEF scaffolds to three different peptide coating modi-
properties of the starting macromers, such as the Mw, which is fications that altered the material hydrophobic surface properties
typically in the range 50 0-40 0 0 Da [789], [790]. PPF photore- and promoted cell adhesion. This kind of scaffolds were also func-
action can be achieved in the presence of different crosslink- tionalized by suspending BMP-2-loaded PLGA microspheres in the
ing agents, such as NVP [789], PPF-diacrylate (PPFDA) [791], and PPF/DEF resin (Figure 17c) [687]. The effective release of BMP-
methyl methacrylate [792]. Since PPF oligomers are in a liquid 2 was assessed both in vitro and in vivo in a rat calvarial defect
state before crosslinking, custom-shaped implants can be easily ob- model, leading after 11 weeks to significantly increased bone for-
tained by molding before crosslinking. mation in the growth factor-loaded group compared to the non-
Given its relatively high stiffness (i.e., compressive modulus loaded one. BMP-2-loaded PPF scaffolds were also seeded with hu-
up to 200 MPa and tensile modulus up to 1 GPa [793], [794]), man ASCs and implanted in rat crania showing that the synergistic
crosslinked PPF has been considered for hard tissue repair. In ad- effect of growth factor release and cell differentiation led to en-
dition, its copolymerization with other biodegradable monomeric hanced bone formation [688]. Dadsetan et al. [685] evaluated the
units (e.g., l-lactide) was investigated as an effective route to tune role of a coating made of a calcium phosphate (i.e., either TCP, car-
the processing, morphological, thermal, and mechanical properties bonate HA, or biphasic CaP) and the simultaneous surface loading
of the resulting biodegradable construct [795]. PPF biocompatibil- with rhBMP-2 on the in vivo bone regeneration capacity of porous
ity to bone-forming cells was demonstrated both in vitro and in PPF scaffolds. After implantation in a critical-size calvarial defect
vivo [796], [797]. PPF undergoes in vivo bulk degradation through model, enhanced bone healing was observed in the case of rhBMP-
hydrolysis of the ester linkages into propylene glycol and fumaric 2-loaded scaffolds. By using a different approach, PPF scaffolds by
acid, which are readily removed from the body, even if the acidic SLA were either spray-coated or dip-coated with a PLGA solution
degradation products can elicit inflammatory response [790]. Fac- containing a dispersion of Hap or TCP. The hybrid coating enabled
tors affecting crosslinked PPF biodegradation are, among others, new bone ingrowth within 4 weeks after implantation of scaffolds
the Mw of PPF macromer, the crosslinking agent, the crosslink- in a canine femoral multi-defect model [686].
ing density, the scaffold porosity and pore size, the environmental PPF was also recently employed to validate a continu-
pH, and the presence of additives. For instance, the biodegrada- ous DLP system for the fabrication of scaffolds through the
tion rate of crosslinked PPF-diacrylate was shown to decrease by use of a dye-initiator package, i.e., TiO2 and bis (2,4,6-
increasing the crosslinking density [798]. Like in the case of PLAs, trimethylbenzoyl)phenylphosphine oxide, facilitating high accuracy
ceramic fillers inclusion can act as a pH buffer inhibiting polymer along the Z dimension [602]. In this way, PPF/DEF scaffolds with
autocatalytic degradation [799]. Composites based on PPF loaded high resolution, accurate pore geometries, and high compressive
with bioactive ceramics (e.g., Hap and TCP) were investigated as modulus (i.e., 135 MPa) were fabricated. In addition, Dilla et
scaffolding materials with improved osteoconductivity, mechanical al.[806] developed diblock and triblock copolymers of PEG-PPF that
properties, and hydrophilicity [792], [799–802]. Different studies were processed by DLP into hydrogels with a 10-fold increase in
have investigated both in vitro and in vivo the combination of PPF elongation at break (>150%) compared to those fabricated by tra-
matrices with PLGA microspheres loaded with the osteogenic pep- ditional DEF-based AM. A novel approach was developed to fabri-
tide TP508 [790], [803–805] In particular, composite PPF scaffolds cate PPF scaffolds loaded with a layered gradient of Hap by means
with optimized TP508 release kinetics resulted in 80% of defect of SE-AM coupled with photoreticolation [603]. For this purpose,
length bridging after 12 weeks of implantation in a rabbit radius Hap was suspended in the presence of a surfactant in a solution of
model [804]. PPF, DEF, and Irgacure, and UV crosslinking was carried out after
AM application. Most of the first attempts to fabricate extrusion of each layer.
biodegradable devices by SLA focused on processing PPF diluted in
its monomer (i.e., DEF) to reduce resin viscosity (Figure 17a) [684]. 4.2.4. Polyurethanes (PUs)
Both vector scanning and DLP techniques have been investigated PUs have been used over the past 50 years in the biomedical
for PPF reticulation under selective UV irradiation by employing a industry as flexible and rigid materials for different applications,
free radical photoinitiator (e.g., Irgacure 819), typically without in- including cardiovascular devices, spinal grafts, breast implants, and
cluding a light absorber. Cooke et al. [567] were the first to em- more recently, biodegradable tissue scaffolds.
ploy SLA for processing PPF into implants with a predefined shape. Synthesis and chemical structure. PUs are a class of poly-
The employment of μSLA vector or projection scanning allowed the mers characterized by urethane groups (—NHCOO—) in their chem-
fabrication of high resolution PPF structures (Figure 17b) [599], ical structure, obtained by reaction of an aliphatic or aromatic
[793]. Lee et al. [599] systematically investigated the effect of μSLA diisocyanate, a macrodiol (i.e., a polyether, polyester, or polycar-
vector scanning speed and working temperature on solidification bonate, with 10 0 0 < Mw < 50 0 0 Da), and a chain extender
depth of PPF/DEF mixtures with 75:25 or 50:50 weight ratio. They which can be a diol or a diamine with a low Mw (typically <
eventually fabricated 3D scaffolds with different pore size charac- 400 Da). Poly(ether urethanes), e.g., those obtained from PEG or
terized by a compressive ultimate strength (20-40 MPa) and an poly(tetramethylene ether) glycol, show high flexibility, hydrolytic
elastic modulus (20 0-30 0 MPa) comparable to those of trabecu- resistance, low oxidative and thermal stability [807]. Poly(ester
lar bone. Various studies investigated PPF scaffold porous structure urethanes) (PEUs), e.g., those synthesized from poly(ethylene adi-
tuning to affect cell behaviors. For instance, the possibility of con- pate) diols or poly(butylene adipate) diols, show higher mechani-
trolling the proliferation of pre-osteoblast cells by varying PPF scaf- cal strength, and are susceptible to hydrolysis of the ester groups
fold pores size and architecture was demonstrated [600]. Another [808]. Poly(carbonate urethanes), e.g., those obtained from reac-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 41

Figure 17. (a) Schematics of polyaddition reactions that occur between PPF and DEF, including the direct addition between two PPF chains and the crosslinking of two PPF
chains by polymerized DEF (reproduced with permission [793]). (b) Customized PPF structures by SLA techniques: SEM micrographs of kidney scaffold (i) (reproduced with
permission [684]), cylindrical scaffold with continuous channel geometry (ii) (reproduced with permission [601]). (c) Biofunctionalized PPF scaffold by SLA: SEM micrographs
at different magnifications of PPF scaffold incorporating BMP-2-loaded PLGA microparticles (i) and (ii) (protrusion in (ii) are microparticles embedded in the PPF matrix);
micro-CT images of rat cranial bone at 11 weeks after scaffold implantation: negative control (iii), PPF scaffold by a combined particulate leaching/gas foaming method (iv),
PPF scaffold by SLA (v), and BMP-2-loaded PPF scaffold by SLA (vi) (reproduced with permission [687]).

tion of poly(hexamethylene carbonate) diols, have good mechani- In the case of biodegradable PUs, the soft segment is typically
cal properties and hydrolytic resistance, even if they can undergo represented by an aliphatic polyester, (e.g., PCL, PGA, or PDLLA),
long-term in vivo enzymatic and oxidative degradation [809]. a copolymer between two aliphatic polyesters, or a poly(ester-
PUs can be synthesized through either a one-step or a two-step ether) copolymer (e.g., PCL-co-PEG), while a linear diisocyante, e.g.,
method. In the first one, a diisocyanate, a macrodiol, and a chain 1,6 hexamethylene diisocyanate (HMDI), is employed as the hard
extender are mixed to react together. The second method, which is segment to avoid carcinogenic products from degradation of aro-
most often used thanks to the possibility of achieving a better con- matic units (Figure 18). The resulting copolymers are also referred
trol on the resulting macromolecular structure, involves first react- to as thermoplastic PUs (TPUs), since they can soften, melt, and
ing a macrodiol with a slight excess of a diisocyanate to obtain a be processed under heat application, in addition to the possibil-
prepolymer, which is then reacted with a chain extender. In this ity of being processed upon dissolution in a proper organic sol-
way, a linear copolymer with alternating blocks of soft and hard vent. So-called waterborne PUs are synthesized by incorporating
segments is eventually synthesized [810]. in the chemical structure hydrophilic monomers containing ionic
functionality, including quaternary ammonium, carboxylate, or sul-
42 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Figure 18. General chemical structure of biodegradable poly(ester urethanes) (PEUs) (i); chemical structure of representative macrodiols and a linear diisocyanate commonly
used for PEUs synthesis.

fonate groups, in order to achieve stable dispersions of copoly- the first attempts to apply AM to PUs was made in 2004 and re-
mer particles in a continuous aqueous phase, also without recur- lied on FDM to process PCL-based PEUs into porous scaffolds [689].
ring to emulsifiers [811]. Thermosetting PUs have been also syn- However, the preparation of continuous PU filaments with a con-
thesized by using a multifunctional chain extender, or introducing sistent diameter suitable for reproducible FDM processing is still
a crosslinking agent [812]. a challenge nowadays [690]. This is due to some inherent short-
Soft segments are composed by reacted macrodiols with a low comings related to thermal extrusion of PUs, including pirohydrol-
Tg , in a rubbery state at room temperature, while hard segments ysis of polyester segments, low thermal and oxidative polyether
are composed by diisocyanates reacted with the chain extender, stability, as well as the elastomeric behavior of PEUs, which can
organized in semicrystalline or highly ordered domains with high hinder a steady extrusion process for producing and processing an
Tg . Microphase separation due to incompatibility between soft and FDM filament (see section 3.4). Anyway, Güney et al. [691] recently
hard segments occurs as a consequence of their different polarity developed a TPU based on a PCL-b-PEG-b-PCL triblock copolymer
and free energy [813]. Hard segments form hydrogen bonds and and HMDI, that behaved like a tough hydrogel and could be pro-
act as dispersed rigid fillers and physical crosslinks, conferring the cessed by FDM. Blending with a more rigid polymer is an effective
material with high material elastic recovery. By varying different way to achieve filament viscoelastic properties suitable for FDM.
factors, such as the length and chemical structure of macrodiols, By adopting this strategy, Castro et al. [693] processed filaments
the nature of the chain extender, and the NHCOO/OH ratio, ma- made from a PEU blended with PVA, the latter leached out from
terial mechanical response can be tuned from low-modulus elas- the scaffolds by water immersion, leaving behind a porous micro-
tomeric to rigid behavior [814]. Other polymer properties, such as filamentous surface topography exploitable for directional cell mi-
thermal stability, hydrophilicity, biodegradation rate, and solvent gration. FDM was also recently applied to process a PEU based on
solubility, can be varied accordingly. a PCL-co-PDLLA soft segment and a block urethane hard segment,
Biomedical use. The tunable elastomeric properties of PUs composed by 1,4-butanediisocyanate and 1,4-butanediol, to fabri-
have attracted great interest in different biomedical areas re- cate scaffolds with two different structures (500 μm pore size and
quiring advanced mechanical performance, such as high flexibil- 90° or 60° deposition angle) [692]. The scaffolds were shown to
ity and fatigue resistance. For instance, PUs are used for the support the dynamic compression-relaxation loads physiologically
manufacturing of elastics and cavity liners for dentistry, cardio- applied onto the knee, and to promote the chondrogenic differen-
vascular catheters and cardiac valves, breast implants, wound tiation of human BMSCs (Figure 19a).
dressing membranes, bone adhesives, and condoms [815]. Cardio- SE-AM represents the most exploited approach in this context.
thane, a silicone-urethane copolymer invented by Kontron Inc., Agrawal et al. [694] employed a CAWS technique to extrude a
Chronoflex®, a class of poly(carbonate urethanes) marketed by Ad- dimethylformamide solution of a blend of two aromatic PEUs di-
vanSource Biomaterials, Tecoflex®, aliphatic polyether-based TPUs rectly into a water bath. The fabricated 3D porous scaffold was
from The Lubrizol Corporation, are only few examples of the wide then impregnated with an epoxy-based hydrogel to form a fiber-
variety of biomedical PUs available in the market. Biomedical ap- reinforced gel with optimized mechanical strength, modulus, and
plications of biodegradable PUs are mainly related to tissue engi- toughness. Another article described the layer-by-layer deposition
neering and drug release systems [816]. As a consequence, their on metallic substrates of a hexafluoro-2-propanol solution contain-
chemistry has been optimized to meet relevant requirements in ing a poly(ester urethane urea) and paclitaxel, to develop novel
terms of biocompatibility, degradation kinetics tailored to specific drug-releasing coatings for surgical implant devices [695]. Xu et al.
tissue formation rate or drug release profile, nontoxicity of ab- [696] deposited at a low temperature (-28°C) a dioxane solution of
sorbable degradation products, and tissue-mimicking mechanical a PU based on PCL, PEG, and HMDI, to fabricate 3D macrochan-
properties. neled and porous scaffolds. The fabricated structures were then
The overall in vivo degradation rate of PEUs is governed mainly submitted to freeze drying for solvent removal, resulting in the for-
by the hydrolysis of the ester groups, yielding α -hydroxy acids mation of a microporous matrix morphology. This approach was
degradation products [817]. Hydrolysis of the urethane groups in further developed through the use of a double nozzle AM sys-
the hard segments, resulting in urethane and urea fragments with tem allowing the simultaneous deposition of the PU organic solu-
terminal acid groups, occurs at a slower rate. The concentration tion and an aqueous solution of gelatin, to fabricate a hybrid con-
of acidic degradation products related to bulk hydrolysis can cause struct engineered as a model for bioartificial liver manufacturing
autocatalyzed degradation and adverse tissue reaction [818]. [287]. The double nozzle system was also exploited for double-
AM application. Different AM technologies have been investi- layer nerve conduits fabrication through a sequential deposition
gated to process biodegradable PUs and their composites with syn- of an acetic acid solution of collagen and a dioxane solution of
thetic polymers, polysaccharides, or proteins [819], [820]. One of PU, then stabilized by means of chemical crosslinking and freeze-
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 43

drying, respectively [697], [698]. Another application of this dual-


deposition strategy was the fabrication of tubular PU constructs
sandwiching a gelatin/alginate/fibrinogen hydrogel laden with ASCs
[699]. In this case, a cell-compatible organic solvent (i.e., tetragly-
col) was employed for PU, while a cryoprotectant (i.e., DMSO or
glycerol) was incorporated in the hydrogel phase to guarantee cell
survival during fabrication and freeze-thawing steps. More com-
plex sandwiched geometries endowed with an intrinsic grid net-
work and branched channels for oxygen and cell nutrients supply
were also developed [288], [289]. Merceron et al. [821] used a four
cartridges system to fabricate a single integrated muscle–tendon
unit by co-printing a TPU with a C2C12 myoblasts-laden hydrogel,
and PCL with a hydrogel encapsulating NIH3T3 fibroblasts.
The employment of aqueous dispersions of waterborne PUs en-
ables direct cell-compatible bioprinting approaches. Some studies
focused on the copolymerization of polyester diols with isophorone
diisocyanate (IPDI), as a hard segment, and two chain extenders,
i.e., ethylenediamine (EDA), and 2,2-bis(hydroxymethyl) propionic
acid (DMPA), to synthesize thermoresponsive PUs that could be
dispersed in water as nanoparticles and form a gel when printed at
physiological temperature [700–702]. By varying the composition
of the soft segment (i.e., PCL, PCL-co-PLLA, PCL-co-PDLLA, PLLA-
co-PEG, or PDLLA-co-PEG) it was possible to tune nanoparticles
size and zeta potential, the rheological properties of the disper-
sion, and the mechanical properties of the printed cell-laden hy-
drogel. Stacked gel fibers encapsulating either murine neural stem
cells [701] or human umbilical cord derived MSCs [702] were de-
veloped by following this approach. Blending soy protein with this
kind of thermoresponsive PU dispersions was proved to be an ef-
fective strategy for fastening gel formation process and enhancing
structural integrity during the layer-by-layer deposition [703]. The
liquid-frozen deposition manufacturing previously described was
applied also to waterborne PUs. For instance, a PCL diol and a
polyethylene butylene adipate diol (2:3 molar ratio) were reacted
as soft segment with the previously described IPDI/EDA/DMPA
hard segment chemistry to obtain a water dispersion that was
printed on a platform maintained at -30°C. The addition of PEG
as a water-leachable viscosity enhancer [822], or HA as a thick-
ener and constitutive blending material [823], enabled the opti-
mization of PU dispersion rheological properties. In this way, the
porous structure of the resulting scaffolds, possibly loaded with
growth factors, was tailored to chondrogenic tissue engineering
(Figure 19b).
An Inkjet printing approach to pattern pH-sensitive PUs based
on PCL diols was developed by employing an ionizable chain ex-
tender. It involves the controlled deposition of an acid aqueous
medium onto a film of an alkaline solution of the neutralized poly-
mer that precipitates due to a local pH change. 2D patterns were
fabricated with this method by employing HMDI as a hard segment
Figure 19. AM of biodegradable PUs. (a) CLSM micrographs of human BMSCs cul-
and bicine as an ionizable chain extender [704], or methylene di-p- tured in vitro for 14 and 28 days in basal (control) (i) and (ii) and chondrogenic
phenyl-diisocyanate and N,N-bis (2-hydorxyethyl)-2-aminoethane- medium (iii) and (iv) on FDM-fabricated PEU scaffolds, BMSCs were immunostained
sulfonic acid as a hard segment and an ionizable chain extender, for F-actin (Phalloidin, red), nucleus (Hoechst, yellow), collagen I (green) and colla-
respectively [705]. gen II (blue), scale bar is 100 μm; μCT images of PEU scaffolds after 28 days of
culture in differentiation (v) and basal medium (vi), scale bar is 1 mm and in-
sets are 700 × 700 μm (reproduced with permission [692]). (b) Representative
4.2.5. Other synthetic polymers pictures showing the high flexibility and excellent shape recovery after compres-
Poly(trimethylene carbonate) (PTMC) (Figure 20a) has been sion of waterborne PU/HA scaffolds fabricated by SE-AM (i); histological images
recently employed as a macromer functionalized with methacry- based on safranin O/fast green stained sections from the regenerated cartilage in
New Zealand white rabbit MSCs-seeded scaffolds 1 month after their implanta-
late end groups to enable SLA photocrosslinking. In addition,
tion into an osteochondral defect created in the rabbit knee joints: PU/HA scaffold
as previously mentioned, PTMC has been also investigated as loaded with the chondrogenic induction factor Y27632 in comparison with PU/HA
a copolymer either with PCL [141], [142] or trimethylolpropane and PLGA scaffolds, the box indicates the repair region, S: scaffold debris (repro-
[142] to develop acrylate-endcapped macromers for SLA, as well as duced with permission [823]).
with PLA to achieve optimized melt rheological properties for FDM
[708], [709]. Biocompatible PTMC is typically synthesized by ring-
opening polymerization of trimethylene carbonate [824]. PTMC un-
dergoes slow enzyme-mediated surface erosion enabling the main-
tenance of mechanical properties for long time, without sudden
44 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

MA/Hap ratio in this kind of formulations allowed the fabrica-


tion of porous scaffolds with tunable surface roughness, wettabil-
ity, stiffness, in vitro cell adhesion, and in vivo bone healing in
rabbits calvarial defects [144], [706]. By following the same ap-
proach, PTMC-based composite scaffolds loaded with up 51 wt.%
β -TCP were endowed with a patient-specific shape for the regener-
ation of large mandibular defects (Figure 21a) [707]. PTMC porous
constructs by SLA were also investigated in vitro as scaffolds for
hASCs differentiation towards annulus fibrosus cells when exposed
Figure 20. Chemical structure of potential biodegradable polymers under biomedi- to growth factors under optimized cell seeding [828].
cal AM implementation. (a) Poly(trimethylene carbonate) (PTMC). (b) Poly(vinyl al- Poly(vinyl alcohol) (PVA) (Figure 20b) is a hydrophilic vinyl
cohol) (PVA). polymer, typically manufactured on an industrial scale by free-
radical polymerization of vinyl acetate, followed by alkaline hy-
drolysis (alcoholysis) of poly(vinyl acetate) [829]. PVA commercial
implant failure related to bulk degradation [825]. In addition, un- batches can have a different deacetylation degree, in the range 70
like poly(α -hydroxy acids), acidic moieties responsible of autocat- to 99%, which significantly affects polymer water solubility, chem-
alytic degradation and tissue necrosis are not produced upon PTMC ical properties, and crystallinity. Besides being employed in differ-
degradation. PTMC is an amorphous polymer with a Tg ~ −17°C, ent industrial sectors, such as food packaging, cleaning and deter-
low Young’s modulus (~3 MPa) and relatively high strain at break gent products, and water treatment, PVA has FDA approval for clin-
(~160%) [826]. As a consequence of its low stiffness, PTMC has ical uses in humans [830]. It can be enzymatically degraded in aer-
been mainly employed in the biomedical area for soft tissue en- obic and anaerobic conditions under the action of specific microor-
gineering, and in the development of copolymers, generally with ganisms [829], [831]. Although its biodegradability and/or elimina-
glycolides, for applications as flexible sutures (BiosynTM ) tissue fix- tion in animals is still under debate [832], PVA has been exten-
ation devices (SuretacTM ) and orthopedic screws (Acufex®) [827]. sively investigated as a potential biodegradable polymer to develop
Genev et al. [143] synthesized three-armed PTMC macromers biomedical hydrogels, often in combination with a wide range of
by ring-opening polymerization and then reacted them with natural or synthetic polymers [833].
methacrylic anhydride to prepare a photocrosslinkable resin. They As previously discussed, PVA has been used in aqueous binders
processed by SLA reactive formulations, composed of PTMC for BJ of polysaccharides [8], as a hydrophilic matrix in multilayer
macromers with methacrylate end-groups (PTMC-MA), Hap parti- drug-loaded systems by SE-AM [106], or as a water-leachable blend
cles, and propylene carbonate as a non-reactive diluent, to fab- component in PEU filaments for FDM [693]. In addition, few recent
ricate customized orbital floor implants. The variation of PTMC- studies focused on PVA as the main polymer matrix component

Figure 21. AM of PTMC: (a) engineering route from imaging data of a large mandibular defect to SLA manufacturing of a porous individualized implant made of PTMC/β -
TCP [707]. AM of PVA: (b) representative SEM micrographs of fracture surface after tensile test of FDM-printed samples made of neat PVA (i) or PVA loaded with 5 wt.%
nanocellulose (ii) [712].
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 45

for AM. Two recent articles by the same research group described increasing automation and digital control of the design and fabri-
the development of PVA filaments loaded with either carbon nan- cation processes, in relationship to the resulting properties of the
otubes [711] or cellulose nanocrystals [712], through blending in fabricated items, are expected in the near future to tremendously
aqueous solution followed by drying and single screw extrusion. In impact the biomedical industry.
both cases, a variation of the organic nanofiller content resulted in
significant changes of Tg and mechanical properties of nanocom- Declaration of Competing Interest
posites samples fabricated by FDM (Figure 21b). Another study
dealt with the optimization of PVA-based aqueous inks formulated The authors declare that they have no known competing finan-
with the addition of additives (e.g., humectant and pigments), for cial interests or personal relationships that could have appeared to
inkjet printing of multilayer structures [710]. influence the work reported in this paper.

5. Conclusions Acknowledgments

AM is revolutionizing the approach to a wide range of biomed- The financial support of the University of Pisa PRA-2016-50
ical applications, including, among others, exoprostheses, surgical and PRA-2018-23 projects entitled “Functional Materials”, the Tus-
implants, tissue engineering, in vitro tissue modelling, and con- cany Region (Italy) funded Project “Nuovi Supporti Bioattivi a Ma-
trolled drug release. The interest of the scientific and industrial trice Polimerica per la Rigenerazione Ossea in Applicazioni Odon-
communities is justified by the great versatility and tremendous toiatriche (R.E.O.S.S.)” as part of the program POR CReO FESR
technological potential of advanced AM strategies to customiza- 2007–2013—Le ali alle tue idee, and the European Eranet+ project,
tion of biodegradable systems composition, micro/nanostructure, BI-TRE—BIophotonic technologies for Tissue Repair (2014–2016), is
macroshape, mechanical behavior, and functional properties. gratefully acknowledged.
Recent progress in the design and development of biodegrad- References
able polymers and relevant processing approaches is providing
unique means for AM development of biomedical devices. Preclin- [1] International A. ASTM, Standard Terminology for Additive Manufacturing
ical and clinical trials are giving promising results for the next Technologies: Designation F2792-12a. West Conshohocken, PA20122012.
[2] MP Chhaya, PSP Poh, ER Balmayor, M van Griensven, J-T Schantz, DW Hut-
generation of implantable medical devices that will likely improve macher, Additive manufacturing in biomedical sciences and the need for def-
and save the life of thousands of adults and pediatric patients all initions and norms, Expert Rev Med Devices 12 (2015) 537–543.
around the world. In parallel, AM is supplying always new in vitro [3] NA Chartrain, CB Williams, AR Whittington, A review on fabricating tissue
scaffolds using vat photopolymerization, Acta Biomater 74 (2018) 90–111.
technological tools needed in the modern healthcare ecosystem
[4] JM Williams, A Adewunmi, RM Schek, CL Flanagan, PH Krebsbach, SE Fein-
for advanced diagnostics, pharmaceutical development, and tissue berg, et al., Bone tissue engineering using polycaprolactone scaffolds fabri-
physiology studies. AM impact is also consolidating in the phar- cated via selective laser sintering, Biomaterials 26 (2005) 4817–4827.
maceutical industry, as witnessed by the presence in the market of [5] A Butscher, M Bohner, S Hofmann, L Gauckler, R Müller, Structural and ma-
terial approaches to bone tissue engineering in powder-based three-dimen-
relevant drug formulations and the growing scientific literature on sional printing, Acta Biomater 7 (2011) 907–920.
this topic. [6] S A. Gold, R Strong, B N. Turner, A review of melt extrusion additive manufac-
To date, many studies have reported the successful applica- turing processes: I. Process design and modeling, Rapid Prototyp J 20 (2014)
192–204.
tion of experimental protocols for processing natural polymers and [7] D Puppi, F Chiellini, Wet-spinning of biomedical polymers: from single-fibre
their functional derivatives by means of different AM techniques. production to additive manufacturing of three-dimensional scaffolds, Polym
Industrial processing methods, as well as consolidated AM ap- Int 66 (2017) 1690–1696.
[8] SC Ligon, R Liska, J Stampfl, M Gurr, R Mülhaupt, Polymers for 3D Printing
proaches, have been adapted and extended for fabricating 3D con- and Customized Additive Manufacturing, Chem Rev 117 (2017) 10212–10290.
structs based on proteins and/or polysaccharides, possibly encapsu- [9] M Wunner Felix, ML Wille, G Noonan Thomas, O Bas, D Dalton Paul,
lating cells and other biologics. Various SLA and SE-AM technolo- M De-Juan-Pardo Elena, et al., Melt Electrospinning Writing of Highly Ordered
Large Volume Scaffold Architectures, Adv Mater 30 (2018) 1706570.
gies, together with relevant ad-hoc processing protocols are cur- [10] SM Giannitelli, P Mozetic, M Trombetta, A Rainer, Combined additive manu-
rently under industrial implementation to this purpose, and some facturing approaches in tissue engineering, Acta Biomater 24 (2015) 1–11.
of them are likely to be integrated in the next future in approved [11] D Puppi, F. Chiellini, Biofabrication via integrated additive manufacturing and
electrofluidodynamics. In: Guarino V, Ambrosio L, editors. Electrofluidody-
diagnostic and therapeutic procedures.
namic Technologies (EFDTs) for Biomaterials and Medical Devices, Woodhead
AM application to PHA is solving the great challenge of coupling Publishing (2018) 71–85.
the sustainable development concept, peculiar of natural polymers, [12] F Ning, W Cong, J Qiu, J Wei, S Wang, Additive manufacturing of carbon fiber
with the advanced control over the fabrication accuracy and re- reinforced thermoplastic composites using fused deposition modeling, Com-
pos Part B Eng 80 (2015) 369–378.
producibility achievable with aliphatic polyesters. In addition, the [13] JP Kruth, G Levy, F Klocke, Childs THC Consolidation phenomena in laser and
wide experience gained on synthetic polymers processing by con- powder-bed based layered manufacturing, CIRP Annals 56 (2007) 730–759.
ventional techniques is fastening the translation to clinic of ad- [14] J Kotlinski, Mechanical properties of commercial rapid prototyping materials,
Rapid Prototyp J 20 (2014) 499–510.
ditive manufactured devices made of thermoplastic polyesters. In [15] JM Chacón, MA Caminero, E García-Plaza, PJ Núñez, Additive manufacturing
particular, decades-long history of clinical exploitation of polymer of PLA structures using fused deposition modelling: Effect of process param-
thermal processing is likely to be further implemented in AM. eters on mechanical properties and their optimal selection, Mater Des 124
(2017) 143–157.
FDM, MEW, and SLS would consequently find increasing industrial [16] YW Wang, F Yang, Q Wu, YC Cheng, PH Yu, J Chen, et al., Effect of composi-
application to consolidate their ongoing biomedical translation for tion of poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) on growth of fibrob-
the fabrication, among others, of surgical devices and exoprosthe- last and osteoblast, Biomaterials 26 (2005) 755–761.
[17] A Louvrier, P Marty, A Barrabé, E Euvrard, B Chatelain, E Weber, et al., How
ses. Biostable polymers, such as those engineered for advanced ap-
useful is 3D printing in maxillofacial surgery? J Stomatol Oral Maxi 118 (2017)
plications (e.g., PEEK), are also fully involved in this technological 206–212.
revolution. In this context, SE-AM will offer advanced technological [18] FPW Melchels, J Feijen, DW Grijpma, A review on stereolithography and its
applications in biomedical engineering, Biomaterials 31 (2010) 6121–6130.
tools for polymer structuring at multi-scale levels, and for device
[19] FAMM Gonçalves, AC Fonseca, M Domingos, A Gloria, AC Serra, JFJ Coelho,
biofunctionalization through drug loading strategies. The potential of unsaturated polyesters in biomedicine and tissue engineer-
Cutting-edge advancements on 4D printing, ing: Synthesis, structure-properties relationships and additive manufacturing,
nano/microfabrication, and smart drug release are leading to Prog Polym Sci 68 (2017) 1–34.
[20] BD Ratner, AS Hoffman, FJ Schoen, JE Lemons, Introduction - Biomaterials
the next frontiers of biomedical AM. These, together with the Science: An Evolving, Multidisciplinary Endeavor, in: BD Ratner, AS Hoffman,
development of advanced materials engineering tools, the always FJ Schoen, JE Lemons (Eds.), Biomater Sci., Academic Press, 2013, pp. 25–39.
46 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

[21] A Youssef, SJ Hollister, PD Dalton, Additive manufacturing of polymer melts Technology for Surgeries of the Pediatric Spine and Pelvis: Benefits Analysis,
for implantable medical devices and scaffolds, Biofabrication 9 (2017) 012002. Journal of Pediatric Orthopaedics 27 (2007).
[22] DA Zopf, SJ Hollister, ME Nelson, RG Ohye, GE Green, Bioresorbable Airway [51] C Hurson, A Tansey, B O’Donnchadha, P Nicholson, J Rice, J McElwain, Rapid
Splint Created with a Three-Dimensional Printer, N Engl J Med 368 (2013) prototyping in the assessment, classification and preoperative planning of ac-
2043–2045. etabular fractures, Injury 38 (2007) 1158–1162.
[23] Y Han, Q Yin, Y Wang, H Zhao, J He, C Gu, Three-dimensional printed degrad- [52] X-B Wu, J-Q Wang, C-P Zhao, X Sun, Y Shi, Z-A Zhang, et al., Printed Three-
able splint in the treatment of pulmonary artery sling associated with severe -dimensional Anatomic Templates for Virtual Preoperative Planning Before
bilateral bronchus stenosis, Cardiol Young 28 (2018) 1477–1480. Reconstruction of Old Pelvic Injuries: Initial Results, Chin Med J (Engl) 128
[24] W Xu, X Wang, Y Yan, W Zheng, Z Xiong, F Lin, et al., Rapid Prototyping (2015) 477–482.
Three-Dimensional Cell/Gelatin/Fibrinogen Constructs for Medical Regenera- [53] N Martelli, C Serrano, H van den Brink, J Pineau, P Prognon, I Borget, et al.,
tion, J Bioact Compatible Polym 22 (2007) 363–377. Advantages and disadvantages of 3-dimensional printing in surgery: A sys-
[25] MR Blatchley, S Gerecht, Acellular implantable and injectable hydrogels for tematic review, Surgery 159 (2016) 1485–1500.
vascular regeneration, Biomed Mater 10 (2015) 034001. [54] S Kondor, CG Grant, P Liacouras, MAJJR Schmid, LTC Michael Parsons, VK Ras-
[26] D Puppi, C Mota, M Gazzarri, D Dinucci, A Gloria, M Myrzabekova, et al., Ad- togi, et al., On Demand Additive Manufacturing of a Basic Surgical Kit, J Med
ditive manufacturing of wet-spun polymeric scaffolds for bone tissue engi- Devices 7 (2013) pp. 030916–2.
neering, Biomed Microdevices 14 (2012) 1115–1127. [55] S Kondor, CG Grant, P Liacouras, MAJJR Schmid, LTC Michael Parsons,
[27] A Wubneh, EK Tsekoura, C Ayranci, H Uludağ, Current state of fabrication B Macy, et al., Personalized Surgical Instruments, J Med Devices 7 (2013) pp.
technologies and materials for bone tissue engineering, Acta Biomater 80 030934–2.
(2018) 1–30. [56] L Resnik, MR Meucci, S Lieberman-Klinger, C Fantini, DL Kelty, R Disla, et al.,
[28] D Puppi, A Russo, G Parrini, F Chiellini, Development of customized poly- Advanced Upper Limb Prosthetic Devices: Implications for Upper Limb Pros-
meric scaffolds with multiscale porosity by a hybrid additive manufacturing thetic Rehabilitation, Arch Phys Med Rehabil 93 (2012) 710–717.
technique, IXth ECNP International Conference on Nanostructured Polymers [57] J Ten Kate, Smit G, Breedveld P, 3D-printed upper limb prostheses: a review,
and Nanocomposites. Rome, 2016 ItalySeptember 19 to 21. Disabil Rehabil Assist Technol 12 (2017) 300–314.
[29] M George, KR Aroom, HG Hawes, BS Gill, J Love, 3D Printed Surgical Instru- [58] GJC van Baar, T Forouzanfar, NPTJ Liberton, HAH Winters, Leusink FKJ Accu-
ments: The Design and Fabrication Process, World J Surg 41 (2017) 314–319. racy of computer-assisted surgery in mandibular reconstruction: A systematic
[30] K Andrianesis, A Tzes, Development and Control of a Multifunctional Pros- review, Oral Oncol 84 (2018) 52–60.
thetic Hand with Shape Memory Alloy Actuators, J Intell Robotic Syst 78 [59] A Tarsitano, L Ciocca, R Scotti, C Marchetti, Morphological results of cus-
(2015) 257–289. tomized microvascular mandibular reconstruction: A comparative study, J
[31] D Puppi, A Morelli, F Bello, S Valentini, F Chiellini, Additive Manufacturing Craniomaxillofac Surg 44 (2016) 697–702.
of Poly(methyl methacrylate) Biomedical Implants with Dual-scale Porosity, [60] P Honigmann, N Sharma, B Okolo, U Popp, B Msallem, FM Thieringer, Patien-
Macromol Mater Eng 303 (2018) 1800247. t-Specific Surgical Implants Made of 3D Printed PEEK: Material, Technology,
[32] 3D-Printed Tracheal Splints Used in Groundbreaking Pediatric Surgery, https: and Scope of Surgical Application, Biomed Res Int (2018) (2018) 4520636-.
//www.news.gatech.edu; 2018 [September 18, 2018]. [61] D Espalin, K Arcaute, D Rodriguez, F Medina, M Posner, R Wicker, Fused de-
[33] C Ding, Z Qiao, W Jiang, H Li, J Wei, G Zhou, et al., Regeneration of a position modeling of patient-specific polymethylmethacrylate implants, Rapid
goat femoral head using a tissue-specific, biphasic scaffold fabricated with Prototyp J 16 (2010) 164–173.
CAD/CAM technology, Biomaterials 34 (2013) 6706–6716. [62] R Velu, S Singamneni, Evaluation of the influences of process parameters
[34] F Chiellini, D Puppi, AM Piras, A Morelli, C Bartoli, C Migone, Modelling while selective laser sintering PMMA powders, Proc Inst Mech Eng, Part C
of pancreatic ductal adenocarcinoma in vitro with three-dimensional mi- 229 (2015) 603–613.
crostructured hydrogels, RSC Adv 6 (2016) 54226–54235. [63] M Vaezi, S Yang, Extrusion-based additive manufacturing of PEEK for biomed-
[35] X Ma, X Qu, W Zhu, Y-S Li, S Yuan, H Zhang, et al., Deterministically pat- ical applications, Virtual Phys Prototyp 10 (2015) 123–135.
terned biomimetic human iPSC-derived hepatic model via rapid 3D bioprint- [64] FPW Melchels, MAN Domingos, TJ Klein, J Malda, PJ Bartolo, DW Hutmacher,
ing, Proc Natl Acad Sci 113 (2016) 2206. Additive manufacturing of tissues and organs, Prog Polym Sci 37 (2012)
[36] AP Haring, Y Tong, J Halper, BN Johnson, Programming of Multicomponent 1079–1104.
Temporal Release Profiles in 3D Printed Polypills via Core–Shell, Multilayer, [65] C Mota, D Puppi, F Chiellini, E Chiellini, Additive manufacturing techniques
and Gradient Concentration Profiles, Adv Healthcare Mater 7 (2018) 1800213. for the production of tissue engineering constructs, J Tissue Eng Regener Med
[37] PS D’Urso, DJ Effeney, WJ Earwaker, TM Barker, MJ Redmond, RG Thompson, 9 (2015) 174–190.
et al., Custom cranioplasty using stereolithography and acrylic, Br J Plast Surg [66] D Puppi, F Chiellini, AM Piras, E Chiellini, Polymeric materials for bone and
53 (20 0 0) 20 0–204. cartilage repair, Prog Polym Sci 35 (2010) 403–440.
[38] A Müller, KG Krishnan, E Uhl, G Mast, The Application of Rapid Prototyp- [67] MM Stevens, JH George, Exploring and engineering the cell surface interface,
ing Techniques in Cranial Reconstruction and Preoperative Planning in Neu- Science 310 (2005) 1135–1138.
rosurgery, J Craniofac Surg 14 (2003). [68] AJ Engler, Sen S, Sweeney HL, Discher DE, Matrix Elasticity Directs Stem Cell
[39] ME Mavili, HI Canter, B Saglam-Aydinatay, S Kamaci, I Kocadereli, Use of Lineage Specification, Cell 126 (2006) 677–689.
Three-Dimensional Medical Modeling Methods for Precise Planning of Or- [69] M Nikkhah, F Edalat, S Manoucheri, A Khademhosseini, Engineering mi-
thognathic Surgery, J Craniofac Surg 18 (2007). croscale topographies to control the cell–substrate interface, Biomaterials 33
[40] M Wanibuchi, M Ohtaki, T Fukushima, AH Friedman, K Houkin, Skull base (2012) 5230–5246.
training and education using an artificial skull model created by selective [70] HN Kim, A Jiao, NS Hwang, MS Kim, DH Kang, D-H Kim, et al., Nanotopogra-
laser sintering, Acta Neurochir (Wien) 152 (2010) 1055–1060. phy-guided tissue engineering and regenerative medicine, Adv Drug Del Rev
[41] W Gabriele, T Berndt, P Peter, H Kurt, T Johannes, Cerebrovascular stere- 65 (2013) 536–558.
olithographic biomodeling for aneurysm surgery, J Neurosurg 10 0 (20 04) [71] PD Dalton, C Vaquette, BL Farrugia, TR Dargaville, TD Brown, DW Hutmacher,
139–145. Electrospinning and additive manufacturing: converging technologies, Bio-
[42] FL Giesel, AR Hart, HK Hahn, E Wignall, F Rengier, R Talanow, et al., 3D Re- mater Sci 1 (2013) 171–185.
constructions of the Cerebral Ventricles and Volume Quantification in Chil- [72] T Weiß, G Hildebrand, R Schade, K Liefeith, Two-Photon polymerization for
dren with Brain Malformations, Acad Radiol 16 (2009) 610–617. microfabrication of three-dimensional scaffolds for tissue engineering appli-
[43] Q Lan, A Chen, T Zhang, G Li, Q Zhu, X Fan, et al., Development of Three-Di- cation, Eng Life Sci 9 (2009) 384–390.
mensional Printed Craniocerebral Models for Simulated Neurosurgery, World [73] JP Temple, DL Hutton, BP Hung, PY Huri, CA Cook, R Kondragunta, et al.,
Neurosurg 91 (2016) 434–442. Engineering anatomically shaped vascularized bone grafts with hASCs and
[44] WS Paiva, R Amorim, DAF Bezerra, M Masini, Aplication of the stereolithog- 3D-printed PCL scaffolds, J Biomed Mater Res A 102 (2014) 4317–4325.
raphy technique in complex spine surgery, Arq Neuropsiquiatr 65 (2007) [74] F Dini, G Barsotti, D Puppi, A Coli, A Briganti, E Giannessi, et al., Tailored
443–445. star poly (ε -caprolactone) wet-spun scaffolds for in vivo regeneration of long
[45] B Wilcox, RJ Mobbs, A-M Wu, K Phan, Systematic review of 3D printing in bone critical size defects, J Bioact Compatible Polym 31 (2016) 15–30.
spinal surgery: the current state of play, J Spine Surg 3 (2017) 433–443. [75] T Lu, Y Li, T Chen, Techniques for fabrication and construction of three-
[46] Q Ling, E He, H Ouyang, J Guo, Z Yin, W Huang, Design of mulitlevel OLF ap- -dimensional scaffolds for tissue engineering, Int J Nanomedicine 8 (2013)
proach (“V”-shaped decompressive laminoplasty) based on 3D printing tech- 337–350.
nology, Eur Spine J 27 (2018) 323–329. [76] Atala A, Kasper FK, Mikos AG, Engineering Complex Tissues, Sci Transl Med,
[47] A Armillotta, P Bonhoeffer, G Dubini, S Ferragina, F Migliavacca, G Sala, et al., 4 (2012), pp. 160rv12.
Use of rapid prototyping models in the planning of percutaneous pulmonary [77] MA Woodruff, C Lange, J Reichert, A Berner, F Chen, P Fratzl, et al., Bone
valved stent implantation, Proc Inst Mech Eng, Part H 221 (2007) 407–416. tissue engineering: from bench to bedside, Mater Today 15 (2012) 430–435.
[48] L Olivieri, A Krieger, MY Chen, P Kim, JP Kanter, 3D heart model guides com- [78] DE Ingber, Can cancer be reversed by engineering the tumor microenviron-
plex stent angioplasty of pulmonary venous baffle obstruction in a Mustard ment? Semin Cancer Biol 18 (2008) 356–364.
repair of D-TGA, Int J Cardiol 172 (2014) e297–e2e8. [79] DW Hutmacher, D Loessner, S Rizzi, DL Kaplan, DJ Mooney, JA Clements, Can
[49] B Ripley, T Kelil, MK Cheezum, A Goncalves, MF Di Carli, FJ Rybicki, et al., tissue engineering concepts advance tumor biology research? Trends Biotech-
3D printing based on cardiac CT assists anatomic visualization prior to tran- nol 28 (2010) 125–133.
scatheter aortic valve replacement, J Cardiovasc Comput Tomogr 10 (2016) [80] N Arya, V Sardana, M Saxena, A Rangarajan, DS Katti, Recapitulating tumour
28–36. microenvironment in chitosan–gelatin three-dimensional scaffolds: an im-
[50] J Guarino, S Tennyson, G McCain, L Bond, K Shea, H King, Rapid Prototyping proved in vitro tumour model, J Royal Soc Interface 9 (2012) 3288–3302.
[81] S Talukdar, SC Kundu, A Non-Mulberry Silk Fibroin Protein Based 3D In Vitro
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 47

Tumor Model for Evaluation of Anticancer Drug Activity, Adv Funct Mater 22 [111] H Ma, C Feng, J Chang, C Wu, 3D-printed bioceramic scaffolds: From bone
(2012) 4778–4788. tissue engineering to tumor therapy, Acta Biomater 79 (2018) 37–59.
[82] Grzesiak JJ, Bouvet M, Determination of the Ligand-Binding Specificities of [112] SH Park, TG Kim, HC Kim, D-Y Yang, TG Park, Development of dual scale scaf-
the α 2β 1 and α 1β ;1 Integrins in a Novel 3-Dimensional In Vitro Model of folds via direct polymer melt deposition and electrospinning for applications
Pancreatic Cancer, Pancreas, 34 (2007), pp. in tissue regeneration, Acta Biomater 4 (2008) 1198–1207.
[83] TB Deramaudt, M Takaoka, R Upadhyay, MJ Bowser, J Porter, A Lee, et al., N– [113] Visscher LE, Dang HP, Knackstedt MA, Hutmacher DW, Tran PA, 3D printed
Cadherin and Keratinocyte Growth Factor Receptor Mediate the Functional Polycaprolactone scaffolds with dual macro-microporosity for applications in
Interplay between Ki-RAS(G12V) and p53(V143A) in Promoting Pancreatic local delivery of antibiotics, Mater Sci Eng, C, 87 (2018), pp. 78-89.
Cell Migration, Invasion, and Tissue Architecture Disruption, Mol Cell Biol 26 [114] C Zhou, K Yang, K Wang, X Pei, Z Dong, Y Hong, et al., Combination of fused
(20 06) 4185–420 0. deposition modeling and gas foaming technique to fabricated hierarchical
[84] AM Gutierrez-Barrera, DG Menter, JL Abbruzzese, Reddy SAG Establishment macro/microporous polymer scaffolds, Mater Des 109 (2016) 415–424.
of three-dimensional cultures of human pancreatic duct epithelial cells, [115] CB Pham, KF Leong, TC Lim, KS Chian, Rapid freeze prototyping technique
Biochem Biophys Res Commun 358 (2007) 698–703. in bio-plotters for tissue scaffold fabrication, Rapid Prototyp J 14 (2008)
[85] Q He, X Wang, X Zhang, H Han, B Han, J Xu, et al., A tissue-engineered 246–253.
subcutaneous pancreatic cancer model for antitumor drug evaluation, Int J [116] B Mueller, Additive Manufacturing Technologies – Rapid Prototyping to Direct
Nanomedicine 8 (2013) 1167–1176. Digital Manufacturing, Assembly Autom 32 (2012).
[86] X Wang, X Zhang, Z Fu, H Yin, A bioengineered metastatic pancreatic tumor [117] S Suri, L-H Han, W Zhang, A Singh, S Chen, CE Schmidt, Solid freeform fab-
model for mechanistic investigation of chemotherapeutic drugs, J Biotechnol rication of designer scaffolds of hyaluronic acid for nerve tissue engineering,
166 (2013) 166–173. Biomed Microdevices 13 (2011) 983–993.
[87] J Vanderburgh, JA Sterling, SA Guelcher, 3D Printing of Tissue Engineered [118] U Hinze, B. Chichkov, Light Sources and Systems for Multiphoton Lithog-
Constructs for in vitro Modeling of Disease Progression and Drug Screening, raphy, in: J Stampfl, R Liska, A Ovsianikov (Eds.), Multiphoton Lithography:
Ann Biomed Eng 45 (2017) 164–179. Techniques, Materials and Applications, 2017 Wiley&VCH Verlag GmbH & Co.
[88] X Xu, MC Farach-Carson, X Jia, Three-dimensional in vitro tumor models for KGaA, 2016.
cancer research and drug evaluation, Biotechnol Adv 32 (2014) 1256–1268. [119] H-B Sun, S Kawata, in: Two-photon Photopolymerization and 3D Lithographic
[89] LC Martine, BM Holzapfel, JA McGovern, F Wagner, VM Quent, P Hesami, Microfabrication. NMR; 3D Analysis; Photopolymerization, Springer-Verlag,
et al., Engineering a humanized bone organ model in mice to study bone Berlin Heidelberg, New York, 2004, pp. 169–273.
metastases, Nat Protoc 12 (2017) 639. [120] BH Wong, Invisalign A to Z, Am J Orthod Dentofacial Orthop 121 (2002)
[90] UA Gurkan, R El Assal, SE Yildiz, Y Sung, AJ Trachtenberg, WP Kuo, et al., 540–541.
Engineering Anisotropic Biomimetic Fibrocartilage Microenvironment by Bio- [121] N Jacquel, CW Lo, HS Wu, YH Wei, S Wang Shaw, Solubility of polyhy-
printing Mesenchymal Stem Cells in Nanoliter Gel Droplets, Mol Pharm 11 droxyalkanoates by experiment and thermodynamic correlations, AlChE J 53
(2014) 2151–2159. (2007) 2704–2714.
[91] DB Kolesky, RL Truby, AS Gladman, TA Busbee, KA Homan, JA Lewis, 3D [122] AM Christensen, K Weimer, C Beaudreau, M Rensberger, B Johnson, The Dig-
Bioprinting of Vascularized, Heterogeneous Cell-Laden Tissue Constructs, Adv ital Thread for Personalized Craniomaxillofacial Surgery, in: AM Greenberg
Mater 26 (2014) 3124–3130. (Ed.), Digital Technologies in Craniomaxillofacial Surgery, Springer New York,
[92] BM Holzapfel, F Wagner, L Thibaudeau, J-P Levesque, DW Hutmacher, Con- New York, NY, 2018, pp. 23–45.
cise Review, Humanized Models of Tumor Immunology in the 21st Century: [123] H Lin, D Zhang, PG Alexander, G Yang, J Tan, AW-M Cheng, et al., Application
Convergence of Cancer Research and Tissue Engineering, Stem Cells 33 (2015) of visible light-based projection stereolithography for live cell-scaffold fabri-
1696–1704. cation with designed architecture, Biomaterials 34 (2013) 331–339.
[93] MC Quent Verena, C Theodoropoulos, W Hutmacher Dietmar, C Reichert [124] Z Wang, Abdulla R, Parker B, Samanipour R, Ghosh S, Kim K, A simple and
Johannes, Differential osteogenicity of multiple donor-derived human mes- high-resolution stereolithography-based 3D bioprinting system using visible
enchymal stem cells and osteoblasts in monolayer, scaffold-based 3D culture light crosslinkable bioinks, Biofabrication 7 (2015) 045009.
and in vivo, Biomed Eng (NY) 61 (2016) 253–266. [125] JV Crivello, E Reichmanis, Photopolymer Materials and Processes for Ad-
[94] BM Holzapfel, F Wagner, D Loessner, NP Holzapfel, L Thibaudeau, R Crawford, vanced Technologies, Chem Mater 26 (2014) 533–548.
et al., Species-specific homing mechanisms of human prostate cancer metas- [126] C Hinczewski, S Corbel, T Chartier, Ceramic suspensions suitable for stere-
tasis in tissue engineered bone, Biomaterials 35 (2014) 4108–4115. olithography, J Eur Ceram Soc 18 (1998) 583–590.
[95] L Thibaudeau, AV Taubenberger, BM Holzapfel, VM Quent, T Fuehrmann, [127] K-W Lee, S Wang, BC Fox, EL Ritman, MJ Yaszemski, L Lu, Poly(propylene
P Hesami, et al., A tissue-engineered humanized xenograft model of human fumarate) Bone Tissue Engineering Scaffold Fabrication Using Stereolithogra-
breast cancer metastasis to bone, Dis Model Mech 7 (2014) 299. phy: Effects of Resin Formulations and Laser Parameters, Biomacromolecules
[96] Q Yang, Q Lian, F Xu, Perspective: Fabrication of integrated organ-on-a-chip 8 (2007) 1077–1084.
via bioprinting, Biomicrofluidics 11 (2017) 031301-. [128] IK Kwon, T Matsuda, Photo-polymerized microarchitectural constructs
[97] M Schaffner, PA Rühs, F Coulter, S Kilcher, AR Studart, 3D printing of bacteria prepared by microstereolithography (μSL) using liquid acrylate-end–
into functional complex materials, Sci Adv 3 (2017) eaao6804. capped trimethylene carbonate-based prepolymers, Biomaterials 26 (2005)
[98] C Mandrycky, Z Wang, K Kim, Kim D-H 3D bioprinting for engineering com- 1675–1684.
plex tissues, Biotechnol Adv 34 (2016) 422–434. [129] FPW Melchels, J Feijen, DW Grijpma, A poly(d,l-lactide) resin for the prepa-
[99] A Junaid, A Mashaghi, T Hankemeier, P Vulto, An end-user perspective on Or- ration of tissue engineering scaffolds by stereolithography, Biomaterials 30
gan-on-a-Chip: Assays and usability aspects, Curr Opin Biomed Eng 1 (2017) (2009) 3801–3809.
15–22. [130] RJ Mondschein, A Kanitkar, CB Williams, SS Verbridge, TE Long, Polymer
[100] Y Li, Poon CT, Li M, Lu TJ, Pingguan-Murphy B, Xu F, Chinese-Noodle-Inspired structure-property requirements for stereolithographic 3D printing of soft tis-
Muscle Myofiber Fabrication, Adv Funct Mater 25 (2015) 5999–6008. sue engineering scaffolds, Biomaterials 140 (2017) 170–188.
[101] B Gao, Q Yang, X Zhao, G Jin, Y Ma, F Xu, 4D Bioprinting for Biomedical Ap- [131] A Pansky, C Tille, H Seitz, M Emons, A Bens, B Roitzheim, et al., Non-toxic
plications, Trends Biotechnol 34 (2016) 746–756. flexible photopolymers for medical stereolithography technology, Rapid Pro-
[102] S Bose, D Ke, H Sahasrabudhe, A Bandyopadhyay, Additive Manufacturing of totyp J 13 (2007) 38–47.
Biomaterials, Prog Mater Sci 93 (2017) 45–111. [132] B Dhariwala, E Hunt, T Boland, Rapid Prototyping of Tissue-Engineering Con-
[103] SJ Buwalda, T Vermonden, WE Hennink, Hydrogels for Therapeutic Delivery: structs, Using Photopolymerizable Hydrogels and Stereolithography, Tissue
Current Developments and Future Directions, Biomacromolecules 18 (2017) Eng 10 (2004) 1316–1322.
316–330. [133] G Mapili, Y Lu, S Chen, K Roy, Laser-layered microfabrication of spatially pat-
[104] GS van Tienderen, M Berthel, Z Yue, M Cook, X Liu, S Beirne, et al., Advanced terned functionalized tissue-engineering scaffolds, J Biomed Mater Res B Appl
fabrication approaches to controlled delivery systems for epilepsy treatment, Biomater 75B (2005) 414–424.
Expert Opin Drug Deliv 15 (2018) 915–925. [134] Q Ge, AH Sakhaei, H Lee, CK Dunn, NX Fang, ML Dunn, Multimaterial 4D
[105] D Puppi, AM Piras, A Pirosa, S Sandreschi, F Chiellini, Levofloxacin-loaded Star Printing with Tailorable Shape Memory Polymers, Sci Rep 6 (2016) 31110.
Poly(ε -caprolactone) Scaffolds by Additive Manufacturing, J Mater Sci - Mater [135] AR Schultz, PM Lambert, NA Chartrain, DM Ruohoniemi, Z Zhang, C Jangu,
Med 27 (2016) 44. et al., 3D Printing Phosphonium Ionic Liquid Networks with Mask Projection
[106] P Rattanakit, SE Moulton, KS Santiago, S Liawruangrath, GG Wallace, Extru- Microstereolithography, ACS Macro Lett 3 (2014) 1205–1209.
sion printed polymer structures: A facile and versatile approach to tailored [136] M Hegde, V Meenakshisundaram, N Chartrain, S Sekhar, D Tafti, CB Williams,
drug delivery platforms, Int J Pharm 422 (2012) 254–263. et al., 3D Printing All-Aromatic Polyimides using Mask-Projection Stere-
[107] J Zhang, AQ Vo, X Feng, S Bandari, MA Repka, Pharmaceutical Additive Manu- olithography: Processing the Nonprocessable, Adv Mater 29 (2017) 1701240.
facturing, a Novel Tool for Complex and Personalized Drug Delivery Systems, [137] W-S Lin, BT Harris, J Pellerito, D Morton, Fabrication of an interim com-
AAPS PharmSciTech 19 (2018) 3388–3402. plete removable dental prosthesis with an in-office digital light processing
[108] A Goyanes, P Robles Martinez, A Buanz, AW Basit, S Gaisford, Effect of geome- three-dimensional printer: A proof-of-concept technique, J Prosthet Dent 120
try on drug release from 3D printed tablets, Int J Pharm 494 (2015) 657–663. (2018) 331–334.
[109] J Zhang, W Yang, AQ Vo, X Feng, X Ye, DW Kim, et al., Hydroxypropyl methyl- [138] PX Lan, JW Lee, Y-J Seol, Cho D-W Development of 3D PPF/DEF scaffolds us-
cellulose-based controlled release dosage by melt extrusion and 3D printing: ing micro-stereolithography and surface modification, J Mater Sci Mater Med
Structure and drug release correlation, Carbohydr Polym 177 (2017) 49–57. 20 (2009) 271–279.
[110] M Sadia, B Arafat, W Ahmed, RT Forbes, MA Alhnan, Channelled tablets: An [139] JH Shin, JW Lee, JH Jung, D-W Cho, G Lim, Evaluation of cell proliferation
innovative approach to accelerating drug release from 3D printed tablets, J and differentiation on a poly(propylene fumarate) 3D scaffold treated with
Controlled Release 269 (2018) 355–363. functional peptides, J Mater Sci 46 (2011) 5282–5287.
48 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

[140] MN Cooke, JP Fisher, D Dean, C Rimnac, AG Mikos, Use of stereolithography crostructure and excellent biocompatibility, Colloids Surf B Biointerfaces 135
to manufacture critical-sized 3D biodegradable scaffolds for bone ingrowth, J (2015) 81–89.
Biomed Mater Res B Appl Biomater 64B (2002) 65–69. [169] M Schmid, A Amado, K Wegener, Polymer powders for selective laser sinter-
[141] T Matsuda, Mizutani M, Liquid acrylate-endcapped biodegradable ing (SLS), AIP Conf Proc (2015) (1664) 160 0 09.
poly( -caprolactone-co-trimethylene carbonate). II. Computer-aided stere- [170] R Velu, S Singamneni, Selective laser sintering of polymer biocomposites
olithographic microarchitectural surface photoconstructs, J Biomed Mater Res based on polymethyl methacrylate, J Mater Res 29 (2014) 1883–1892.
62 (2002) 395–403. [171] Y Shi, J Chen, Y Wang, Z Li, S Huang, Study of the selective laser sintering of
[142] S-J Lee, H-W Kang, JK Park, J-W Rhie, SK Hahn, Cho D-W Application of mi- polycarbonate and postprocess for parts reinforcement, Proc Inst Mech Eng,
crostereolithography in the development of three-dimensional cartilage re- Part L 221 (2007) 37–42.
generation scaffolds, Biomed Microdevices 10 (2008) 233–241. [172] EN Antonov, VN Bagratashvili, MJ Whitaker, JJA Barry, KM Shakesheff,
[143] MA Geven, V Varjas, L Kamer, X Wang, J Peng, D Eglin, et al., Fabrication of AN Konovalov, et al., Three-Dimensional Bioactive and Biodegradable Scaf-
patient specific composite orbital floor implants by stereolithography, Polym folds Fabricated by Surface-Selective Laser Sintering, Adv Mater 17 (2004)
Adv Technol 26 (2015) 1433–1438. 327–330.
[144] O Guillaume, MA Geven, CM Sprecher, VA Stadelmann, DW Grijpma, TT Tang, [173] M Lindner, S Hoeges, W Meiners, K Wissenbach, R Smeets, R Telle, et al.,
et al., Surface-enrichment with hydroxyapatite nanoparticles in stereolithog- Manufacturing of individual biodegradable bone substitute implants using se-
raphy-fabricated composite polymer scaffolds promotes bone repair, Acta Bio- lective laser melting technique, J Biomed Mater Res A 97A (2011) 466–471.
mater 54 (2017) 386–398. [174] P-H Lee, E Chang, S Yu, SW Lee, IW Kim, S Park, et al., Modification and
[145] J Jansen, FPW Melchels, DW Grijpma, J Feijen, Fumaric Acid Monoethyl Ester– characteristics of biodegradable polymer suitable for selective laser sintering,
Functionalized Poly(d,l-lactide)/N-vinyl-2-pyrrolidone Resins for the Prepara- Int J, Precis, Eng, Manuf 14 (2013) 1079–1086.
tion of Tissue Engineering Scaffolds by Stereolithography, Biomacromolecules [175] FE Wiria, KF Leong, CK Chua, Y Liu, Poly-ε -caprolactone/hydroxyapatite for
10 (2009) 214–220. tissue engineering scaffold fabrication via selective laser sintering, Acta Bio-
[146] A Ronca, S Ronca, G Forte, S Zeppetelli, A Gloria, R De Santis, et al., Synthe- mater 3 (2007) 1–12.
sis and characterization of divinyl-fumarate poly-ε -caprolactone for scaffolds [176] S Eosoly, S Lohfeld, D Brabazon, Effect of Hydroxyapatite on Biodegradable
with controlled architectures, J Tissue Eng Regener Med 12 (2016) e523–ee31. Scaffolds Fabricated by SLS, Key Eng Mater 396-398 (2009) 659–662.
[147] C Xia, NX Fang, 3D microfabricated bioreactor with capillaries, Biomed Mi- [177] S Eshraghi, S Das, Mechanical and microstructural properties of polycapro-
crodevices 11 (2009) 1309–1315. lactone scaffolds with one-dimensional, two-dimensional, and three-dimen-
[148] K Arcaute, B Mann, R Wicker, Stereolithography of spatially controlled mul- sional orthogonally oriented porous architectures produced by selective laser
ti-material bioactive poly(ethylene glycol) scaffolds, Acta Biomater 6 (2010) sintering, Acta Biomater 6 (2010) 2467–2476.
1047–1054. [178] EN Antonov, VN Bagratashvili, SM Howdle, AN Konovalov, VK Popov,
[149] SJ Leigh, HTJ Gilbert, IA Barker, JM Becker, SM Richardson, JA Hoyland, et al., VY Panchenko, Fabrication of polymer scaffolds for tissue engineering using
Fabrication of 3-Dimensional Cellular Constructs via Microstereolithography surface selective laser sintering, Laser Phys 16 (2006) 774–787.
Using a Simple, Three-Component, Poly(Ethylene Glycol) Acrylate-Based Sys- [179] B Duan, M Wang, WY Zhou, WL Cheung, ZY Li, Lu WW Three-dimensional
tem, Biomacromolecules 14 (2013) 186–192. nanocomposite scaffolds fabricated via selective laser sintering for bone tis-
[150] JW Nichol, ST Koshy, H Bae, CM Hwang, S Yamanlar, Khademhosseini A sue engineering, Acta Biomater 6 (2010) 4495–4505.
Cell-laden microengineered gelatin methacrylate hydrogels, Biomaterials 31 [180] B Duan, M Wang, Customized Ca–P/PHBV nanocomposite scaffolds for bone
(2010) 5536–5544. tissue engineering: design, fabrication, surface modification and sustained re-
[151] JK Placone, J Navarro, GW Laslo, MJ Lerman, AR Gabard, GJ Herendeen, et al., lease of growth factor, J Royal Soc Interface 7 (2010) S615–SS29.
Development and Characterization of a 3D Printed, Keratin-Based Hydrogel, [181] B Duan, WL Cheung, M Wang, Optimized fabrication of Ca–P/PHBV nanocom-
Ann Biomed Eng 45 (2017) 237–248. posite scaffolds via selective laser sintering for bone tissue engineering, Bio-
[152] W Zhang, D Li, K Wang, W Bian, X Li, Q Lian, et al., Fabrication of a bio-in- fabrication 3 (2011) 015001.
spired beta-Tricalcium phosphate/collagen scaffold based on ceramic stere- [182] E Sachs, M Cima, P Williams, D Brancazio, Cornie J Three Dimensional Print-
olithography and gel casting for osteochondral tissue engineering, Rapid Pro- ing, Rapid Tooling and Prototypes Directly from a CAD Model, J Eng Ind 114
totyp J 18 (2012) 68–80. (1992) 481–488.
[153] VB Morris, S Nimbalkar, M Younesi, P McClellan, O Akkus, Mechanical Proper- [183] B Utela, D Storti, R Anderson, M Ganter, A review of process development
ties, Cytocompatibility and Manufacturability of Chitosan:PEGDA Hybrid-Gel steps for new material systems in three dimensional printing (3DP), J Manuf
Scaffolds by Stereolithography, Ann Biomed Eng 45 (2017) 286–296. Process 10 (2008) 96–104.
[154] AP Zhang, X Qu, P Soman, KC Hribar, JW Lee, S Chen, et al., Rapid Fabrication [184] U Gbureck, T Hölzel, I Biermann, JE Barralet, LM Grover, Preparation of tri-
of Complex 3D Extracellular Microenvironments by Dynamic Optical Projec- calcium phosphate/calcium pyrophosphate structures via rapid prototyping, J
tion Stereolithography, Adv Mater 24 (2012) 4266–4270. Mater Sci Mater Med 19 (2008) 1559–1563.
[155] R Gauvin, Y-C Chen, JW Lee, P Soman, P Zorlutuna, JW Nichol, et al., Micro- [185] Y Shanjani, JNA De Croos, RM Pilliar, RA Kandel, E Toyserkani, Solid freeform
fabrication of complex porous tissue engineering scaffolds using 3D projec- fabrication and characterization of porous calcium polyphosphate structures
tion stereolithography, Biomaterials 33 (2012) 3824–3834. for tissue engineering purposes, J Biomed Mater Res B Appl Biomater 93B
[156] S Miao, W Zhu, NJ Castro, M Nowicki, X Zhou, H Cui, et al., 4D printing smart (2010) 510–519.
biomedical scaffolds with novel soybean oil epoxidized acrylate, Sci Rep 6 [186] S Pravin, A Sudhir, Integration of 3D printing with dosage forms: A new per-
(2016) 27226. spective for modern healthcare, Biomed Pharmacother 107 (2018) 146–154.
[157] L Froyen, JP Kruth, T Laoui, X Wang, Lasers and materials in selective laser [187] EO Garzón, JL Alves, RJ Neto, Post-process Influence of Infiltration on Binder
sintering, Assembly Autom 23 (2003) 357–371. Jetting Technology, in: LFMd Silva (Ed.), Materials Design and Applications,
[158] F Fina, A Goyanes, S Gaisford, AW Basit, Selective laser sintering (SLS) 3D Springer International Publishing, Cham, 2017, pp. 233–255.
printing of medicines, Int J Pharm 529 (2017) 285–293. [188] L Cima, M. Cima, Preparation of medical devices by solid free-form fabrica-
[159] A Mazzoli, Selective laser sintering in biomedical engineering, Med Biol Eng tion methods, US Patent and Trademark Office. United States: Massachusetts
Comput 51 (2013) 245–256. Institute of Technology (1996).
[160] http://oxfordpm.com, May , 2019 [189] Q Wei, Y Wang, W Chai, Y Zhang, X Chen, Molecular dynamics simulation and
[161] C Adamzyk, P Kachel, M Hoss, F Gremse, A Modabber, F Hölzle, et al., Bone experimental study of the bonding properties of polymer binders in 3D pow-
tissue engineering using polyetherketoneketone scaffolds combined with au- der printed hydroxyapatite bioceramic bone scaffolds, Ceram Int 43 (2017)
tologous mesenchymal stem cells in a sheep calvarial defect model, J Cran- 13702–13709.
iomaxillofac Surg 44 (2016) 985–994. [190] Q Wei, Y Wang, X Li, M Yang, W Chai, K Wang, et al., Study the bonding
[162] RJ Morrison, SJ Hollister, MF Niedner, MG Mahani, AH Park, DK Mehta, et al., mechanism of binders on hydroxyapatite surface and mechanical properties
Mitigation of tracheobronchomalacia with 3D-printed personalized medical for 3DP fabrication bone scaffolds, J Mech Behav Biomed Mater 57 (2016)
devices in pediatric patients, Sci Transl Med 7 (2015) pp. 285ra64-ra64. 190–200.
[163] M Schmid, A Amado, K Wegener, Materials perspective of polymers for ad- [191] H Shao, M Sun, F Zhang, A Liu, Y He, J Fu, et al., Custom Repair of Mandibu-
ditive manufacturing with selective laser sintering, J Mater Res 29 (2014) lar Bone Defects with 3D Printed Bioceramic Scaffolds, J, Dent Res 97 (2017)
1824–1832. 68–76.
[164] M Schmid, K Wegener, Additive Manufacturing, Polymers Applicable for Laser [192] M Seidenstuecker, L Kerr, A Bernstein, OH Mayr, PN Suedkamp, R Gadow,
Sintering (LS), Procedia Eng 149 (2016) 457–464. et al., 3D Powder Printed Bioglass and β -Tricalcium Phosphate Bone Scaffolds,
[165] M Schmid, K Wegener, Thermal and molecular properties of polymer pow- Materials 11 (2018).
ders for Selective Laser Sintering (SLS), AIP Conf Proc (2016) (1779) 10 0 0 03. [193] Y Deng, C Jiang, C Li, T Li, M Peng, J Wang, et al., 3D printed scaffolds of cal-
[166] C Gayer, J Abert, M Bullemer, S Grom, L Jauer, W Meiners, et al., Influence cium silicate-doped β -TCP synergize with co-cultured endothelial and stro-
of the material properties of a poly(D,L-lactide)/β -tricalcium phosphate com- mal cells to promote vascularization and bone formation, Sci Rep 7 (2017)
posite on the processability by selective laser sintering, J Mech Behav Biomed 5588.
Mater 87 (2018) 267–278. [194] C Miguel, D Marta, G Uwe, G Jürgen, F Paulo, P Inês, et al., Fabrication of
[167] D Drummer, D Rietzel, F Kühnlein, Development of a characterization ap- computationally designed scaffolds by low temperature 3D printing, Biofab-
proach for the sintering behavior of new thermoplastics for selective laser rication 5 (2013) 035012.
sintering, Phys Procedia 5 (2010) 533–542. [195] R Trombetta, JA Inzana, EM Schwarz, SL Kates, HA Awad, 3D Printing of Cal-
[168] Y Du, H Liu, J Shuang, J Wang, J Ma, S Zhang, Microsphere-based selective cium Phosphate Ceramics for Bone Tissue Engineering and Drug Delivery,
laser sintering for building macroporous bone scaffolds with controlled mi- Ann Biomed Eng 45 (2017) 23–44.
[196] A Barba, A Diez-Escudero, Y Maazouz, K Rappe, M Espanol, EB Montufar,
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 49

et al., Osteoinduction by Foamed and 3D-Printed Calcium Phosphate Scaf- hesion development in fused deposition techniques, J Mater Process Technol
folds: Effect of Nanostructure and Pore Architecture, ACS Appl Mater Inter- 245 (2017) 167–179.
faces 9 (2017) 41722–41736. [225] V Taubner, R Shishoo, Influence of processing parameters on the degradation
[197] A Khalyfa, S Vogt, J Weisser, G Grimm, A Rechtenbach, W Meyer, et al., Devel- of poly(L-lactide) during extrusion, J Appl Polym Sci 79 (2001) 2128–2135.
opment of a new calcium phosphate powder-binder system for the 3D print- [226] MA Cuiffo, J Snyder, AM Elliott, N Romero, S Kannan, GP Halada, Impact of
ing of patient specific implants, J Mater Sci Mater Med 18 (2007) 909–916. the Fused Deposition (FDM) Printing Process on Polylactic Acid (PLA) Chem-
[198] S Lin, PY Chao, YW Chien, S Sayani, S Kuma, M Mason, et al., In vitro and istry and Structure, Applied Sciences 7 (2017).
in vivo evaluations of biodegradable implants for hormone replacement ther- [227] F Carrasco, P Pagès, J Gámez-Pérez, OO Santana, Maspoch ML Processing of
apy: effect of system design and PK-PD relationship, AAPS PharmSciTech 2 poly(lactic acid): Characterization of chemical structure, thermal stability and
(2001) E16. mechanical properties, Polym Degrad Stab 95 (2010) 116–125.
[199] CXF Lam, XM Mo, SH Teoh, DW Hutmacher, Scaffold development using 3D [228] MA Woodruff, DW Hutmacher, The return of a forgotten polymer—Polycapro-
printing with a starch-based polymer, Mater Sci Eng, C 20 (2002) 49–56. lactone in the 21st century, Prog Polym Sci 35 (2010) 1217–1256.
[200] W Huang, Q Zheng, W Sun, H Xu, X Yang, Levofloxacin implants with prede- [229] W Kempin, C Franz, L-C Koster, F Schneider, M Bogdahn, W Weitschies, et al.,
fined microstructure fabricated by three-dimensional printing technique, Int Assessment of different polymers and drug loads for fused deposition mod-
J Pharm 339 (2007) 33–38. eling of drug loaded implants, Eur J Pharm Biopharm 115 (2017) 84–93.
[201] W Wu, Q Zheng, X Guo, J Sun, Y Liu, A programmed release multi-drug im- [230] J Aho, JP Bøtker, N Genina, M Edinger, L Arnfast, J Rantanen, Roadmap to
plant fabricated by three-dimensional printing technology for bone tubercu- 3D-Printed Oral Pharmaceutical Dosage Forms: Feedstock Filament Properties
losis therapy, Biomed Mater 4 (2009) 065005. and Characterization for Fused Deposition Modeling, J Pharm Sci 108 (2019)
[202] W Wu, C Ye, Q Zheng, G Wu, Z Cheng, A therapeutic delivery system for 26–35.
chronic osteomyelitis via a multi-drug implant based on three-dimensional [231] H Li, C Tan, L Li, Review of 3D printable hydrogels and constructs, Mater Des
printing technology, J Biomater Appl 31 (2016) 250–260. 159 (2018) 20–38.
[203] DW Hutmacher, Scaffolds in tissue engineering bone and cartilage, Biomate- [232] S-Z Guo, F Gosselin, N Guerin, A-M Lanouette, M-C Heuzey, D Therriault, Sol-
rials 21 (20 0 0) 2529–2543. vent-Cast Three-Dimensional Printing of Multifunctional Microsystems, Small
[204] F Wang, L Shor, A Darling, S Khalil, W Sun, S Güçeri, et al., Precision ex- 9 (2013) 4118–4122.
truding deposition and characterization of cellular poly-ε -caprolactone tissue [233] S Khalil, W Sun, Bioprinting Endothelial Cells With Alginate for 3D Tissue
scaffolds, Rapid Prototyp J 10 (2004) 42–49. Constructs, J Biomech Eng 131 (2009) 111002–111008.
[205] A Gloria, T Russo, R De Santis, L Ambrosio, 3d Fiber Deposition Technique to [234] X Wang, Y Yan, Y Pan, Z Xiong, H Liu, J Cheng, et al., Generation of Three-
Make Multifunctional and Tailor-Made Scaffolds for Tissue Engineering Appli- -Dimensional Hepatocyte/Gelatin Structures with Rapid Prototyping System,
cations, J Appl Biomater Biomech 7 (2009) 141–152. Tissue Eng 12 (2006) 83–90.
[206] R Landers, R Mülhaupt, Desktop manufacturing of complex objects, proto- [235] T Billiet, E Gevaert, T De Schryver, M Cornelissen, P Dubruel, The 3D printing
types and biomedical scaffolds by means of computer-assisted design com- of gelatin methacrylamide cell-laden tissue-engineered constructs with high
bined with computer-guided 3D plotting of polymers and reactive oligomers, cell viability, Biomaterials 35 (2014) 49–62.
Macromol Mater Eng 282 (20 0 0) 17–21. [236] D Puppi, C Migone, A Morelli, C Bartoli, M Gazzarri, D Pasini, et al., Mi-
[207] C Mota, D Puppi, D Dinucci, C Errico, P Bártolo, F Chiellini, Dual-Scale crostructured chitosan/poly(γ -glutamic acid) polyelectrolyte complex hydro-
Polymeric Constructs as Scaffolds for Tissue Engineering, Materials 4 (2011) gels by computer-aided wet-spinning for biomedical three-dimensional scaf-
527–542. folds, J Bioact Compatible Polym 31 (2016) 531–549.
[208] G Kim, J Son, S Park, W Kim, Hybrid process for fabricating 3D hierarchical [237] CJ Hansen, R Saksena, DB Kolesky, JJ Vericella, SJ Kranz, GP Muldowney, et al.,
scaffolds combining rapid prototyping and electrospinning, Macromol Rapid High-Throughput Printing via Microvascular Multinozzle Arrays, Adv Mater
Commun 29 (2008) 1577–1581. 25 (2013) 96–102.
[209] L Moroni, Schotel R, Hamann D, de Wijn JR, van Blitterswijk CA, 3D Fiber-De- [238] C Mota, SY Wang, D Puppi, M Gazzarri, C Migone, F Chiellini, et al., Additive
posited Electrospun Integrated Scaffolds Enhance Cartilage Tissue Formation, manufacturing of poly[(R)-3-hydroxybutyrate-co-(R)-3-hydroxyhexanoate]
Adv Funct Mater 18 (2008) 53–60. scaffolds for engineered bone development, J Tissue Eng Regener Med 11
[210] TD Brown, PD Dalton, DW Hutmacher, Melt electrospinning today: An op- (2017) 175–186.
portune time for an emerging polymer process, Prog Polym Sci 56 (2016) [239] D Puppi, A Morelli, F Chiellini, Additive Manufacturing of
116–166. Poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)/poly(ε -caprolactone) Blend
[211] TD Brown, PD Dalton, DW Hutmacher, Direct Writing By Way of Melt Elec- Scaffolds for Tissue Engineering, Bioengineering 4 (2017) 49.
trospinning, Adv Mater 23 (2011) 5651–5657. [240] B Zhang, J He, X Li, F Xu, D Li, Micro/nanoscale electrohydrodynamic printing:
[212] H Gernot, J Tomasz, DB Toby, H Kathrin, M Claus, J Franz, et al., Additive from 2D to 3D, Nanoscale 8 (2016) 15376–15388.
manufacturing of scaffolds with sub-micron filaments via melt electrospin- [241] N Bu, Y Huang, X Wang, Z Yin, Continuously Tunable and Oriented
ning writing, Biofabrication 7 (2015) 035002. Nanofiber Direct-Written by Mechano-Electrospinning, Mater Manuf Pro-
[213] MGM Marascio, J Antons, DP Pioletti, Bourban P-E 3D Printing of Polymers cesses 27 (2012) 1318–1323.
with Hierarchical Continuous Porosity, Adv Mater Technol 2 (2017) 1700145. [242] Y Huang, Y Duan, Y Ding, N Bu, Y Pan, N Lu, et al., Versatile, kinetically con-
[214] D Rohner, W Hutmacher Dietmar, K Cheng Tan, M Oberholzer, B Hammer, trolled, high precision electrohydrodynamic writing of micro/nanofibers, Sci
In vivo efficacy of bone-marrow-coated polycaprolactone scaffolds for the re- Rep 4 (2014) 5949.
construction of orbital defects in the pig, J Biomed Mater Res B Appl Biomater [243] Y Huihua, Z Qihui, L Biyun, B Min, L Xiangxin, Z Yanzhong, Direct printing of
66B (2003) 574–580. patterned three-dimensional ultrafine fibrous scaffolds by stable jet electro-
[215] SH Park, DS Park, JW Shin, YG Kang, HK Kim, TR Yoon, et al., Scaffolds for spinning for cellular ingrowth, Biofabrication 7 (2015) 045004.
bone tissue engineering fabricated from two different materials by the rapid [244] G Luo, KS Teh, Y Liu, X Zang, Z Wen, L Lin, Direct-Write, Self-Aligned Elec-
prototyping technique: PCL versus PLGA, J Mater Sci Mater Med 23 (2012) trospinning on Paper for Controllable Fabrication of Three-Dimensional Struc-
2671–2678. tures, ACS Appl Mater Interfaces 7 (2015) 27765–27770.
[216] K Jinku, M Sean, T Brandi, A-U Pedro, S Young-Hye, DD David, et al., [245] M Lee, Kim H-Y Toward Nanoscale Three-Dimensional Printing, Nanowalls
Rapid-prototyped PLGA/β -TCP/hydroxyapatite nanocomposite scaffolds in a Built of Electrospun Nanofibers, Langmuir 30 (2014) 1210–1214.
rabbit femoral defect model, Biofabrication 4 (2012) 025003. [246] J Lee, SY Lee, J Jang, YH Jeong, Cho D-W Fabrication of Patterned Nanofibrous
[217] DO Adeola, I Zohreh, LS Jennifer, MLC David, BF Eames, Daniel XBC Modu- Mats Using Direct-Write Electrospinning, Langmuir 28 (2012) 7267–7275.
lating mechanical behaviour of 3D-printed cartilage-mimetic PCL scaffolds: [247] J-C Wang, M-W Chang, Z Ahmad, Li J-S Fabrication of patterned polymer-an-
influence of molecular weight and pore geometry, Biofabrication 8 (2016) tibiotic composite fibers via electrohydrodynamic (EHD) printing, J Drug Deliv
025020. Sci Technol 35 (2016) 114–123.
[218] MP Chhaya, FPW Melchels, BM Holzapfel, JG Baldwin, DW Hutmacher, Sus- [248] D Puppi, F. Chiellini, 12 - Drug release kinetics of electrospun fibrous sys-
tained regeneration of high-volume adipose tissue for breast reconstruction tems, in: ML Focarete, A Tampieri (Eds.), Core-Shell Nanostructures for Drug
using computer aided design and biomanufacturing, Biomaterials 52 (2015) Delivery and Theranostics, Woodhead Publishing, 2018, pp. 349–374.
551–560. [249] R Altobelli, V Guarino, L Ambrosio, Micro- and nanocarriers by electrofludo-
[219] E Fuenmayor, M Forde, AV Healy, DM Devine, JG Lyons, C McConville, dynamic technologies for cell and molecular therapies, Process Biochem 51
et al., Material Considerations for Fused-Filament Fabrication of Solid Dosage (2016) 2143–2154.
Forms, Pharmaceutics 10 (2018) 44. [250] B Dorj, J-H Park, Kim H-W Robocasting chitosan/nanobioactive glass dual–
[220] ER Fitzharris, N Watanabe, DW Rosen, ML Shofner, Effects of material proper- pore structured scaffolds for bone engineering, Mater Lett 73 (2012) 119–122.
ties on warpage in fused deposition modeling parts, Int J Adv Manuf Technol [251] C Mota, D Puppi, D Dinucci, M Gazzarri, F Chiellini, Additive manufacturing
95 (2018) 2059–2070. of star poly(ε -caprolactone) wet-spun scaffolds for bone tissue engineering
[221] LJ Love, V Kunc, O Rios, CE Duty, AM Elliott, BK Post, et al., The importance applications, J Bioact Compatible Polym 28 (2013) 320–340.
of carbon fiber to polymer additive manufacturing, J Mater Res 29 (2014) [252] D Puppi, C Migone, L Grassi, A Pirosa, G Maisetta, G Batoni, et al., Integrated
1893–1898. three-dimensional fiber/hydrogel biphasic scaffolds for periodontal bone tis-
[222] C McIlroy, PD Olmsted, Disentanglement effects on welding behaviour of sue engineering, Polym Int 65 (2016) 631–640.
polymer melts during the fused-filament-fabrication method for additive [253] D Puppi, A Pirosa, G Lupi, PA Erba, G Giachi, F Chiellini, Design and fabrica-
manufacturing, Polymer 123 (2017) 376–391. tion of novel polymeric biodegradable stents for small caliber blood vessels
[223] JE Seppala, KD Migler, Infrared thermography of welding zones produced by by computer-aided wet-spinning, Biomed Mater 12 (2017) 035011.
polymer extrusion additive manufacturing, Addit Manuf 12 (2016) 71–76. [254] D Puppi, S Braccini, A Ranaudo, C Federica,
[224] SF Costa, FM Duarte, JA Covas, Estimation of filament temperature and ad- Poly(3-hydroxybutyrate-co-3-hydroxyexanoate) scaffolds with tunable macro-
50 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

and microstructural features by additive manufacturing, J Biotechnol 308 rication of a Hybrid Cell/Hydrogel Construct by a Double-nozzle Assembling
(2020) 96–107. Technology, J Bioact Compatible Polym 24 (2009) 249–265.
[255] Z Xiong, Y Yan, S Wang, R Zhang, C Zhang, Fabrication of porous scaffolds [285] M Xu, Y Van, H Liu, R Yag, X Wang, Controlled Adipose-derived Stromal
for bone tissue engineering via low-temperature deposition, Scripta Mater 46 Cells Differentiation into Adipose and Endothelial Cells in a 3D Structure Es-
(2002) 771–776. tablished by Cell-assembly Technique, J Bioact Compatible Polym 24 (2009)
[256] N Zhu, MG Li, D Cooper, XB Chen, Development of novel hybrid 31–47.
poly(l-lactide)/chitosan scaffolds using the rapid freeze prototyping tech- [286] M Xu, X Wang, Y Yan, R Yao, Y Ge, An cell-assembly derived physiological
nique, Biofabrication 3 (2011) 034105. 3D model of the metabolic syndrome, based on adipose-derived stromal cells
[257] G Kim, S Ahn, H Yoon, Y Kim, W Chun, A cryogenic direct-plotting system for and a gelatin/alginate/fibrinogen matrix, Biomaterials 31 (2010) 3868–3877.
fabrication of 3D collagen scaffolds for tissue engineering, J Mater Chem 19 [287] W Xu, X Wang, Y Yan, R Zhang, A Polyurethane-Gelatin Hybrid Construct for
(2009) 8817–8823. Manufacturing Implantable Bioartificial Livers, J Bioact Compatible Polym 23
[258] D Puppi, A Pirosa, A Morelli, F Chiellini, Design, fabrication and characteriza- (2008) 409–422.
tion of tailored poly[(R)-3-hydroxybutyrate-co-(R)-3-hydroxyexanoate] scaf- [288] X Wang, K He, W Zhang, Optimizing the fabrication processes for manu-
folds by Computer-aided Wet-spinning, Rapid Prototyp J 24 (2018) 1–8. facturing a hybrid hierarchical polyurethane–cell/hydrogel construct, J Bioact
[259] G Vozzi, A Previti, D De Rossi, A Ahluwalia, Microsyringe-Based Deposition of Compatible Polym 28 (2013) 303–319.
Two-Dimensional and Three-Dimensional Polymer Scaffolds with a Well-De- [289] Y Huang, K He, X Wang, Rapid prototyping of a hybrid hierarchical
fined Geometry for Application to Tissue Engineering, Tissue Eng 8 (2002) polyurethane-cell/hydrogel construct for regenerative medicine, Mater Sci
1089–1098. Eng, C 33 (2013) 3220–3229.
[260] G Vozzi, C Flaim, A Ahluwalia, S Bhatia, Fabrication of PLGA scaffolds us- [290] M Costantini, C Colosi, W Świeszkowski
˛ , Barbetta A Co-axial wet-spinning in
ing soft lithography and microsyringe deposition, Biomaterials 24 (2003) 3D bioprinting: state of the art and future perspective of microfluidic inte-
2533–2540. gration, Biofabrication 11 (2018) 012001.
[261] T Serra, Planell JA, Navarro M, High-resolution PLA-based composite scaffolds [291] S Ji, M Guvendiren, Recent Advances in Bioink Design for 3D Bioprinting of
via 3-D printing technology, Acta Biomater 9 (2013) 5521–5530. Tissues and Organs, Front Bioeng Biotechnol 5 (2017) 23-.
[262] Serra T, Ortiz-Hernandez M, Engel E, Planell JA, Navarro M, Relevance of PEG [292] T Xu, W Zhao, J-M Zhu, MZ Albanna, JJ Yoo, Atala A Complex heterogeneous
in PLA-based blends for tissue engineering 3D-printed scaffolds, Mater Sci tissue constructs containing multiple cell types prepared by inkjet printing
Eng, C, 38 (2014), pp. 55-62. technology, Biomaterials 34 (2013) 130–139.
[263] JM Deitzel, J Kleinmeyer, D Harris, NC Beck Tan, The effect of processing vari- [293] T Xu, CA Gregory, P Molnar, X Cui, S Jalota, SB Bhaduri, et al., Viability and
ables on the morphology of electrospun nanofibers and textiles, Polymer 42 electrophysiology of neural cell structures generated by the inkjet printing
(2001) 261–272. method, Biomaterials 27 (2006) 3580–3588.
[264] SH Tan, R Inai, M Kotaki, S Ramakrishna, Systematic parameter study for [294] JA Barron, BJ Spargo, BR Ringeisen, Biological laser printing of three dimen-
ultra-fine fiber fabrication via electrospinning process, Polymer 46 (2005) sional cellular structures, Appl Phys A 79 (2004) 1027–1030.
6128–6134. [295] Koch L, Kuhn S, Sorg H, Gruene M, Schlie S, Gaebel R, et al. Laser Printing of
[265] JHY Chung, S Naficy, Z Yue, R Kapsa, A Quigley, SE Moulton, et al., Bio-ink Skin Cells and Human Stem Cells2009.
properties and printability for extrusion printing living cells, Biomater Sci 1 [296] A Doraiswamy, RJ Narayan, T Lippert, L Urech, A Wokaun, M Nagel, et al.,
(2013) 763–773. Excimer laser forward transfer of mammalian cells using a novel triazene ab-
[266] KI Draget, G Skjåk-Bræk, O Smidsrød, Alginate based new materials, Int J Biol sorbing layer, Appl Surf Sci 252 (2006) 4743–4747.
Macromol 21 (1997) 47–55. [297] S Rhee, JL Puetzer, BN Mason, CA Reinhart-King, LJ Bonassar, 3D Bioprinting
[267] S Sakai, H Kamei, T Mori, T Hotta, H Ohi, M Nakahata, et al., Visible Light-In- of Spatially Heterogeneous Collagen Constructs for Cartilage Tissue Engineer-
duced Hydrogelation of an Alginate Derivative and Application to Stereolitho- ing, ACS Biomater Sci Eng 2 (2016) 1800–1805.
graphic Bioprinting Using a Visible Light Projector and Acid Red, Biomacro- [298] G Gao, T Yonezawa, K Hubbell, G Dai, X Cui, Inkjet-bioprinted acrylated pep-
molecules 19 (2018) 672–679. tides and PEG hydrogel with human mesenchymal stem cells promote robust
[268] L Ouyang, CB Highley, W Sun, JA Burdick, A Generalizable Strategy for the bone and cartilage formation with minimal printhead clogging, Biotechnol J
3D Bioprinting of Hydrogels from Nonviscous Photo-crosslinkable Inks, Adv 10 (2015) 1568–1577.
Mater 29 (2016) 1604983. [299] M Michael, B Jana, S Matthias, Marcy Z-W Nanostructured Pluronic hydrogels
[269] Z Wang, X Jin, R Dai, JF Holzman, K Kim, An ultrafast hydrogel photocrosslink- as bioinks for 3D bioprinting, Biofabrication 7 (2015) 035006.
ing method for direct laser bioprinting, RSC Adv 6 (2016) 21099–21104. [300] WL Ng, WY Yeong, MW Naing, Polyvinylpyrrolidone-Based Bio-Ink Improves
[270] S Liu, L Li, Recoverable and Self-Healing Double Network Hydrogel Based on Cell Viability and Homogeneity during Drop-On-Demand Printing, Materials
κ -Carrageenan, ACS Appl Mater Interfaces 8 (2016) 29749–29758. 10 (2017) 190.
[271] S Liu, L Li, Ultrastretchable and Self-Healing Double-Network Hydrogel for 3D [301] F Gilbert, CD O’Connell, T Mladenovska, S Dodds, Print Me an Organ? Ethi-
Printing and Strain Sensor, ACS Appl Mater Interfaces 9 (2017) 26429–26437. cal and Regulatory Issues Emerging from 3D Bioprinting in Medicine, Sci Eng
[272] JE Kim, SH Kim, Y Jung, Current status of three-dimensional printing inks for Ethics 24 (2018) 73–91.
soft tissue regeneration, Tissue Eng Regen Med 13 (2016) 636–646. [302] IT Ozbolat, KK Moncal, H Gudapati, Evaluation of bioprinter technologies, Ad-
[273] S Bertlein, G Brown, KS Lim, T Jungst, T Boeck, T Blunk, et al., Thiol–Ene Click- dit Manuf 13 (2017) 179–200.
able Gelatin: A Platform Bioink for Multiple 3D Biofabrication Technologies, [303] L Ouyang, CB Highley, CB Rodell, W Sun, JA Burdick, 3D Printing of
Adv Mater 29 (2017) 1703404. Shear-Thinning Hyaluronic Acid Hydrogels with Secondary Cross-Linking, ACS
[274] HW Ooi, C Mota, AT ten Cate, A Calore, L Moroni, MB Baker, Thiol–Ene Al- Biomater Sci Eng 2 (2016) 1743–1751.
ginate Hydrogels as Versatile Bioinks for Bioprinting, Biomacromolecules 19 [304] W Liu, MA Heinrich, Y Zhou, A Akpek, N Hu, X Liu, et al., Extrusion Bioprint-
(2018) 3390–3400. ing of Shear-Thinning Gelatin Methacryloyl Bioinks, Adv Healthcare Mater 6
[275] Hoyle CE, Bowman CN, Thiol–Ene Click Chemistry, Angew Chem, 49 (2010), (2017), doi:10.1002/adhm.201601451.
pp. 1540-73. [305] RF Pereira, A Sousa, CC Barrias, A Bayat, PL Granja, PJ Bártolo, Advances in
[276] S Stichler, T Jungst, M Schamel, I Zilkowski, M Kuhlmann, T Böck, et al., Thi- bioprinted cell-laden hydrogels for skin tissue engineering, Biomanufact Rev
ol-ene Clickable Poly(glycidol) Hydrogels for Biofabrication, Ann Biomed Eng 2 (2017) 1.
45 (2017) 273–285. [306] RF Pereira, PJ Bártolo, 3D bioprinting of photocrosslinkable hydrogel con-
[277] E Hoch, C Schuh, T Hirth, GEM Tovar, K Borchers, Stiff gelatin hydrogels can structs, J Appl Polym Sci 132 (2015).
be photo-chemically synthesized from low viscous gelatin solutions using [307] KS Lim, BS Schon, NV Mekhileri, GCJ Brown, CM Chia, S Prabakar, et al.,
molecularly functionalized gelatin with a high degree of methacrylation, J New Visible-Light Photoinitiating System for Improved Print Fidelity in Gelat-
Mater Sci Mater Med 23 (2012) 2607–2617. in-Based Bioinks, ACS Biomater Sci Eng 2 (2016) 1752–1762.
[278] BH Lee, N Lum, LY Seow, PQ Lim, LP Tan, Synthesis and Characterization [308] N Ashammakhi, S Ahadian, C Xu, H Montazerian, H Ko, R Nasiri, et al., Bioinks
of Types A and B Gelatin Methacryloyl for Bioink Applications, Materials 9 and bioprinting technologies to make heterogeneous and biomimetic tissue
(2016) 797. constructs, Materials Today Bio 1 (2019) 10 0 0 08.
[279] TJ Hinton, Q Jallerat, RN Palchesko, JH Park, MS Grodzicki, H-J Shue, et al., [309] A Morelli, D Puppi, F Chiellini, Polymers from Renewable Resources, J Renew-
Three-dimensional printing of complex biological structures by freeform re- able Mater 1 (2013) 83–112.
versible embedding of suspended hydrogels, Sci Adv 1 (2015) pp. e1500758-e. [310] J Van Hoorick, H Declercq, A De Muynck, A Houben, L Van Hoorebeke, R Cor-
[280] IT Ozbolat, W Peng, V Ozbolat, Application areas of 3D bioprinting, Drug Dis- nelissen, et al., Indirect additive manufacturing as an elegant tool for the pro-
cov Today 21 (2016) 1257–1271. duction of self-supporting low density gelatin scaffolds, J Mater Sci Mater
[281] J Groll, JA Burdick, DW Cho, B Derby, M Gelinsky, SC Heilshorn, et al., A def- Med 26 (2015) 247.
inition of bioinks and their distinction from biomaterial inks, Biofabrication [311] D Puppi, G Pecorini, F Chiellini, Biomedical Processing of Polyhydroxyalka-
11 (2018) 013001. noates, Bioengineering 6 (2019) 108.
[282] V Chan, P Zorlutuna, JH Jeong, H Kong, R Bashir, Three-dimensional photopat- [312] D Miranda-Nieves, EL Chaikof, Collagen and Elastin Biomaterials for the Fab-
terning of hydrogels using stereolithography for long-term cell encapsulation, rication of Engineered Living Tissues, ACS Biomater Sci Eng 3 (2017) 694–711.
Lab Chip 10 (2010) 2062–2070. [313] J Gopinathan, I Noh, Recent trends in bioinks for 3D printing, Biomater Res
[283] S Li, Y Yan, Z Xiong, CWR Zhang, X Wang, Gradient Hydrogel Construct 22 (2018) 11.
Based on an Improved Cell Assembling System, J Bioact Compatible Polym [314] A SeungHyun, K Young Ho, K GeunHyung, A three-dimensional hierarchical
24 (2009) 84–99. collagen scaffold fabricated by a combined solid freeform fabrication (SFF)
[284] L Shengjie, X Zhuo, W Xiaohong, Y Yongnian, L Haixia, Z Renji, Direct Fab- and electrospinning process to enhance mesenchymal stem cell (MSC) prolif-
eration, J Micromech Microeng 20 (2010) 065015.
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 51

[315] V Lee, G Singh, JP Trasatti, C Bjornsson, X Xu, TN Tran, et al., Design and [345] H Shan-hui, Chen-Huan L, Ching-Shiow T, Air plasma treated chitosan fiber-
Fabrication of Human Skin by Three-Dimensional Bioprinting, Tissue Eng Part s-stacked scaffolds, Biofabrication 4 (2012) 015002.
C 20 (2013) 473–484. [346] E Lisa, F Ruben, B Carlo, Z Francesca, M Cinzia, B Annalisa, et al., Highly de-
[316] X Tao, WB Kyle, ZA Mohammad, D Dennis, Z Weixin, JY James, et al., Hybrid fined 3D printed chitosan scaffolds featuring improved cell growth, Biomed
printing of mechanically and biologically improved constructs for cartilage Mater 12 (2017) 045009.
tissue engineering applications, Biofabrication 5 (2013) 015001. [347] C Intini, L Elviri, J Cabral, S Mros, C Bergonzi, A Bianchera, et al., 3D-printed
[317] Yang X, Lu Z, Wu H, Li W, Zheng L, Zhao J, Collagen-alginate as bioink for chitosan-based scaffolds: An in vitro study of human skin cell growth and an
three-dimensional (3D) cell printing based cartilage tissue engineering, Mater in-vivo wound healing evaluation in experimental diabetes in rats, Carbohydr
Sci Eng, C, 83 (2018), pp. 195-201. Polym 199 (2018) 593–602.
[318] L Anja, M Michael, B Sophie, P Birgit, B Hagen, S Michaela, et al., Additive [348] Q Liu, Q Li, S Xu, Q Zheng, X Cao, Preparation and Properties of 3D Printed Al-
manufacturing of collagen scaffolds by three-dimensional plotting of highly ginate–Chitosan Polyion Complex Hydrogels for Tissue Engineering, Polymers
viscous dispersions, Biofabrication 8 (2016) 015015. 10 (2018).
[319] AD Nocera, R Comín, NA Salvatierra, MP Cid, Development of 3D printed fib- [349] P Shi, A Laude, WY Yeong, Investigation of cell viability and morphology in
rillar collagen scaffold for tissue engineering, Biomed Microdevices 20 (2018) 3D bio-printed alginate constructs with tunable stiffness, J Biomed Mater Res
26. A 105 (2016) 1009–1018.
[320] JA Inzana, D Olvera, SM Fuller, JP Kelly, OA Graeve, EM Schwarz, et al., 3D [350] G Marchioli, Gurp Lv, Krieken PPv, D Stamatialis, M Engelse, Blitterswijk CAv,
printing of composite calcium phosphate and collagen scaffolds for bone re- et al., Fabrication of three-dimensional bioplotted hydrogel scaffolds for islets
generation, Biomaterials 35 (2014) 4026–4034. of Langerhans transplantation, Biofabrication 7 (2015) 025009.
[321] SA Irvine, A Agrawal, BH Lee, HY Chua, KY Low, BC Lau, et al., Printing [351] L Ning, A Guillemot, J Zhao, G Kipouros, X Chen, Influence of Flow Behavior
cell-laden gelatin constructs by free-form fabrication and enzymatic protein of Alginate–Cell Suspensions on Cell Viability and Proliferation, Tissue Eng
crosslinking, Biomed Microdevices 17 (2015) 16. Part C 22 (2016) 652–662.
[322] Y Yan, X Wang, Z Xiong, H Liu, F Liu, F Lin, et al., Direct Construction of [352] J Leppiniemi, P Lahtinen, A Paajanen, R Mahlberg, S Metsä-Kortelainen,
a Three-dimensional Structure with Cells and Hydrogel, J Bioact Compatible T Pinomaa, et al., 3D-Printable Bioactivated Nanocellulose–Alginate Hydro-
Polym 20 (2005) 259–269. gels, ACS Appl Mater Interfaces 9 (2017) 21959–21970.
[323] Y Rui, Z Renji, Y Yongnian, W Xiaohong, In Vitro Angiogenesis of 3D Tissue [353] K Markstedt, A Mantas, I Tournier, H Martínez Ávila, D Hägg, P Gatenholm,
Engineered Adipose Tissue, J Bioact Compatible Polym 24 (2009) 5–24. 3D Bioprinting Human Chondrocytes with Nanocellulose–Alginate Bioink
[324] Y Yan, X Wang, Y Pan, H Liu, J Cheng, Z Xiong, et al., Fabrication of viable for Cartilage Tissue Engineering Applications, Biomacromolecules 16 (2015)
tissue-engineered constructs with 3D cell-assembly technique, Biomaterials 1489–1496.
26 (2005) 5864–5871. [354] S Ahn, H Lee, G Kim, Functional cell-laden alginate scaffolds consisting of
[325] T Zhang, Y Yan, X Wang, Z Xiong, F Lin, R Wu, et al., Three-dimensional core/shell struts for tissue regeneration, Carbohydr Polym 98 (2013) 936–942.
Gelatin and Gelatin/Hyaluronan Hydrogel Structures for Traumatic Brain In- [355] H Martínez Ávila, S Schwarz, N Rotter, P Gatenholm, 3D bioprinting of hu-
jury, J Bioact Compatible Polym 22 (2007) 19–29. man chondrocyte-laden nanocellulose hydrogels for patient-specific auricular
˜
[326] Z Munoz , H Shih, C-C Lin, Gelatin hydrogels formed by orthogonal thi- cartilage regeneration, Bioprinting 1-2 (2016) 22–35.
ol–norbornene photochemistry for cell encapsulation, Biomater Sci 2 (2014) [356] KMO Håkansson, IC Henriksson, C de la Peña Vázquez, V Kuzmenko, K Mark-
1063–1072. stedt, P Enoksson, et al., Solidification of 3D Printed Nanofibril Hydrogels into
[327] T Greene, C-C Lin, Modular Cross-Linking of Gelatin-Based Thiol–Norbornene Functional 3D Cellulose Structures, Adv Mater Technol 1 (2016) 160 0 096.
Hydrogels for in Vitro 3D Culture of Hepatocellular Carcinoma Cells, ACS Bio- [357] M Müller, E Öztürk, Arlov Ø, P Gatenholm, M Zenobi-Wong, Alginate Sulfate—
mater Sci Eng 1 (2015) 1314–1323. Nanocellulose Bioinks for Cartilage Bioprinting Applications, Ann Biomed Eng
[328] X Cui, T Boland, Human microvasculature fabrication using thermal inkjet 45 (2017) 210–223.
printing technology, Biomaterials 30 (2009) 6221–6227. [358] D Nguyen, DA Hägg, A Forsman, J Ekholm, P Nimkingratana, C Brantsing,
[329] Y-B Lee, S Polio, W Lee, G Dai, L Menon, RS Carroll, et al., Bio-printing of et al., Cartilage Tissue Engineering by the 3D Bioprinting of iPS Cells in a
collagen and VEGF-releasing fibrin gel scaffolds for neural stem cell culture, Nanocellulose/Alginate Bioink, Sci Rep 7 (2017) 658.
Exp Neurol 223 (2010) 645–652. [359] Markstedt K, Sundberg J, Gatenholm P, 3D Bioprinting of Cellulose Structures
[330] IT Ozbolat, M Hospodiuk, Current advances and future perspectives in extru- from an Ionic Liquid, 3D Print Addit Manuf, 1 (2014), pp. 115-21.
sion-based bioprinting, Biomaterials 76 (2016) 321–343. [360] VCF Li, A Mulyadi, CK Dunn, Y Deng, HJ Qi, Direct Ink Write 3D Printed
[331] M Gruene, M Pflaum, C Hess, S Diamantouros, S Schlie, A Deiwick, et al., Cellulose Nanofiber Aerogel Structures with Highly Deformable, Shape Re-
Laser Printing of Three-Dimensional Multicellular Arrays for Studies of Cell— coverable, and Functionalizable Properties, ACS Sustain Chem Eng 6 (2018)
Cell and Cell–Environment Interactions, Tissue Eng Part C 17 (2011) 973–982. 2011–2022.
[332] DB Kolesky, KA Homan, MA Skylar-Scott, JA Lewis, Three-dimensional bio- [361] K Markstedt, A Escalante, G Toriz, P Gatenholm, Biomimetic Inks Based on
printing of thick vascularized tissues, Proc Natl Acad Sci 113 (2016) 3179. Cellulose Nanofibrils and Cross-Linkable Xylans for 3D Printing, ACS Appl
[333] M Hospodiuk, M Dey, D Sosnoski, IT Ozbolat, The bioink: A comprehensive Mater Interfaces 9 (2017) 40878–40886.
review on bioprintable materials, Biotechnol Adv 35 (2017) 217–239. [362] SW Pattinson, AJ Hart, Additive Manufacturing of Cellulosic Materials with
[334] RR Jose, JE Brown, KE Polido, FG Omenetto, DL Kaplan, Polyol-Silk Bioink For- Robust Mechanics and Antimicrobial Functionality, Adv Mater Technol 2
mulations as Two-Part Room-Temperature Curable Materials for 3D Printing, (2017) 160 0 084.
ACS Biomater Sci Eng 1 (2015) 780–788. [363] Q Wang, J Sun, Q Yao, C Ji, J Liu, Q Zhu, 3D printing with cellulose materials,
[335] MJ Rodriguez, J Brown, J Giordano, SJ Lin, FG Omenetto, DL Kaplan, Silk based Cellulose 25 (2018) 4275–4301.
bioinks for soft tissue reconstruction using 3-dimensional (3D) printing with [364] S Kumar, M Hofmann, B Steinmann, EJ Foster, C Weder, Reinforcement of
in vitro and in vivo assessments, Biomaterials 117 (2017) 105–115. Stereolithographic Resins for Rapid Prototyping with Cellulose Nanocrystals,
[336] MJ Rodriguez, TA Dixon, E Cohen, W Huang, FG Omenetto, DL Kaplan, 3D ACS Appl Mater Interfaces 4 (2012) 5399–5407.
freeform printing of silk fibroin, Acta Biomater 71 (2018) 379–387. [365] NB Palaganas, JD Mangadlao, ACC de Leon, JO Palaganas, KD Pangilinan,
[337] AM Compaan, K Christensen, Y Huang, Inkjet Bioprinting of 3D Silk Fibroin YJ Lee, et al., 3D Printing of Photocurable Cellulose Nanocrystal Composite for
Cellular Constructs Using Sacrificial Alginate, ACS Biomater Sci Eng 3 (2017) Fabrication of Complex Architectures via Stereolithography, ACS Appl Mater
1519–1526. Interfaces 9 (2017) 34314–34324.
[338] S Das, F Pati, Y-J Choi, G Rijal, J-H Shim, SW Kim, et al., Bioprintable, cel- [366] W Schuurman, PA Levett, MW Pot, PR van Weeren, WJA Dhert, DW Hut-
l-laden silk fibroin–gelatin hydrogel supporting multilineage differentiation macher, et al., Gelatin-Methacrylamide Hydrogels as Potential Biomaterials
of stem cells for fabrication of three-dimensional tissue constructs, Acta Bio- for Fabrication of Tissue-Engineered Cartilage Constructs, Macromol Biosci 13
mater 11 (2015) 233–246. (2013) 551–561.
[339] SH Kim, YK Yeon, JM Lee, JR Chao, YJ Lee, YB Seo, et al., Precisely printable [367] L Pescosolido, W Schuurman, J Malda, P Matricardi, F Alhaique, T Coviello,
and biocompatible silk fibroin bioink for digital light processing 3D printing, et al., Hyaluronic Acid and Dextran-Based Semi-IPN Hydrogels as Biomaterials
Nat Commun 9 (2018) 1620. for Bioprinting, Biomacromolecules 12 (2011) 1831–1838.
[340] JYH Fuh, W Feng, L Geng, H Tong Loh, Y San Wong, DW Hutmacher, Di- [368] A Skardal, J Zhang, L McCoard, X Xu, S Oottamasathien, GD Prestwich, Pho-
rect writing of chitosan scaffolds using a robotic system, Rapid Prototyp J 11 tocrosslinkable hyaluronan-gelatin hydrogels for two-step bioprinting, Tissue
(2005) 90–97. Eng Part A 16 (2010) 2675–2685.
[341] Ang TH, Sultana FSA, Hutmacher DW, Wong YS, Fuh JYH, Mo XM, et al. Fab- [369] M Kesti, M Müller, J Becher, M Schnabelrauch, M D’Este, D Eglin, et al.,
rication of 3D chitosan–hydroxyapatite scaffolds using a robotic dispensing A versatile bioink for three-dimensional printing of cellular scaffolds based
system, Mater Sci Eng, C, 20 (2002), pp. 35-42. on thermally and photo-triggered tandem gelation, Acta Biomater 11 (2015)
[342] K Ye, R Felimban, K Traianedes, SE Moulton, GG Wallace, J Chung, et al., 162–172.
Chondrogenesis of Infrapatellar Fat Pad Derived Adipose Stem Cells in 3D [370] CB Highley, CB Rodell, JA Burdick, Direct 3D Printing of Shear-Thinning Hy-
Printed Chitosan Scaffold, PLOS ONE 9 (2014) e99410. drogels into Self-Healing Hydrogels, Adv Mater 27 (2015) 5075–5079.
[343] Q Wu, M Maire, S Lerouge, D Therriault, M-C Heuzey, 3D Printing of Mi- [371] A Skardal, J Zhang, GD Prestwich, Bioprinting vessel-like constructs us-
crostructured and Stretchable Chitosan Hydrogel for Guided Cell Growth, Adv ing hyaluronan hydrogels crosslinked with tetrahedral polyethylene glycol
Biosyst 1 (2017) 170 0 058. tetracrylates, Biomaterials 31 (2010) 6173–6181.
[344] Q Wu, D Therriault, M-C Heuzey, Processing and Properties of Chitosan Inks [372] A Skardal, J Zhang, L McCoard, S Oottamasathien, GD Prestwich, Dynamically
for 3D Printing of Hydrogel Microstructures, ACS Biomater Sci Eng 4 (2018) Crosslinked Gold Nanoparticle – Hyaluronan Hydrogels, Adv Mater 22 (2010)
2643–2652. 4736–4740.
[373] S Stichler, T Böck, N Paxton, S Bertlein, R Levato, V Schill, et al.,
52 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

Double printing of hyaluronic acid/poly(glycidol) hybrid hydrogels with [402] ED Boland, JA Matthews, KJ Pawlowski, DG Simpson, GE Wnek, GL Bowlin,
poly(ε -caprolactone) for MSC chondrogenesis, Biofabrication 9 (2017) 044108. Electrospinning collagen and elastin: preliminary vascular tissue engineering,
[374] MT Poldervaart, B Goversen, M de Ruijter, A Abbadessa, FPW Melchels, Front Biosci 9 (2004) 1422–1432.
FC Öner, et al., 3D bioprinting of methacrylated hyaluronic acid (MeHA) hy- [403] J Venugopal, LL Ma, T Yong, S Ramakrishna, In vitro study of smooth muscle
drogel with intrinsic osteogenicity, PLOS ONE 12 (2017) e0177628. cells on polycaprolactone and collagen nanofibrous matrices, Cell Biol Int 29
[375] MF Oliveira, IA Maia, PY Noritomi, GC Nargi, JVL Silva, BMP Ferreira, et al., (2005) 861–867.
Construção de Scaffolds para engenharia tecidual utilizando prototipagem [404] J Stitzel, J Liu, SJ Lee, M Komura, J Berry, S Soker, et al., Controlled fabrication
rápida, Matéria 12 (2007) 373–382. of a biological vascular substitute, Biomaterials 27 (2006) 1088–1094.
[376] TF Pereira, MAC Silva, MF Oliveira, IA Maia, JVL Silva, MF Costa, et al., [405] NM Blanco, J Edwards, WA Zamboni, Dermal Substitute, (Integra) for Open
Effect of process parameters on the properties of selective laser sintered Nasal Wounds, Plast Reconstr Surg 113 (2004) 2224–2225.
Poly(3-hydroxybutyrate) scaffolds for bone tissue engineering AU - Pereira, [406] SG Priya, H Jungvid, A Kumar, Skin Tissue Engineering for Tissue Repair and
T.F, Virtual Phys Prototyp 7 (2012) 275–285. Regeneration, Tissue Eng Part B 14 (2008) 105–118.
[377] TF Pereira, MF Oliveira, IA Maia, JVL Silva, MF Costa, RMSM Thiré, 3D Printing [407] YM Bello, AF Falabella, The role of Graftskin (Apligraf) in difficult-to-heal ve-
of Poly(3-hydroxybutyrate) Porous Structures Using Selective Laser Sintering, nous leg ulcers, J Wound Care 11 (2002) 182–183.
Macromol Symp 319 (2012) 64–73. [408] LS Nair, CT Laurencin, Biodegradable polymers as biomaterials, Prog Polym
[378] Kosorn W, Sakulsumbat M, Uppanan P, Kaewkong P, Chantaweroad S, Jitsaard Sci 32 (2007) 762–798.
J, et al. PCL/PHBV blended three dimensional scaffolds fabricated by fused [409] A Atala, SB Bauer, S Soker, JJ Yoo, AB Retik, Tissue-engineered autologous
deposition modeling and responses of chondrocytes to the scaffolds, J Biomed bladders for patients needing cystoplasty, The Lancet 367 (2006) 1241–1246.
Mater Res B Appl Biomater, ( 2016 ), pp. [410] AM Raya-Rivera, D Esquiliano, R Fierro-Pastrana, E López-Bayghen, P Valen-
[379] P Menčík, R Přikryl, I Stehnová, V Melčová, S Kontárová, S Figalla, et al., Ef- cia, R Ordorica-Flores, et al., Tissue-engineered autologous vaginal organs in
fect of Selected Commercial Plasticizers on Mechanical, Thermal, and Mor- patients: a pilot cohort study, The Lancet 384 (2014) 329–336.
phological Properties of Poly(3-hydroxybutyrate)/Poly(lactic acid)/Plasticizer [411] SV Murphy, A Skardal, A Atala, Evaluation of hydrogels for bio-printing appli-
Biodegradable Blends for Three-Dimensional (3D) Print, Materials 11 (2018). cations, J Biomed Mater Res A 101A (2012) 272–284.
[380] X Hu, P Cebe, AS Weiss, F Omenetto, DL Kaplan, Protein-based composite [412] M Vaidya, Startups tout commercially 3D-printed tissue for drug screening,
materials, Mater Today 15 (2012) 208–215. Nat Med 21 (2015) 2.
[381] K Yue, G Trujillo-de Santiago, MM Alvarez, A Tamayol, N Annabi, [413] M Rimann, S Laternser, H Keller, O Leupin, U Graf-Hausner, 3D Bioprinted
A Khademhosseini, properties Synthesis, and biomedical applications of Muscle and Tendon Tissues for Drug Development, CHIMIA 69 (2015) 65–67.
gelatin methacryloyl (GelMA) hydrogels, Biomaterials 73 (2015) 254–271. [414] L Hockaday, 3D Bioprinting: A Deliberate Business, Genetic Engineering &
[382] AC Brown, TH Barker, Fibrin-based biomaterials, Modulation of macroscopic Biotechnology News 35 (2014) 14–17.
properties through rational design at the molecular level, Acta Biomater 10 [415] https://www.bioprintabm.com/, 2018 [11/2018].
(2014) 1502–1514. [416] AJ Reiffel, C Kafka, KA Hernandez, S Popa, JL Perez, S Zhou, et al., High-Fi-
[383] V Volkov, AV Ferreira, A Cavaco-Paulo, On the Routines of Wild-Type Silk Fi- delity Tissue Engineering of Patient-Specific Auricles for Reconstruction of Pe-
broin Processing Toward Silk-Inspired Materials: A Review, Macromol Mater diatric Microtia and Other Auricular Deformities, PLOS ONE 8 (2013) e56506.
Eng 300 (2015) 1199–1216. [417] Y Li, J Rodrigues, H Tomás, Injectable and biodegradable hydrogels: gela-
[384] CH Lee, A Singla, Y Lee, Biomedical applications of collagen, Int J Pharm 221 tion, biodegradation and biomedical applications, Chem Soc Rev 41 (2012)
(2001) 1–22. 2193–2221.
[385] MD Luis, S Naledi, F Kieran, IZ Dimitrios, Acetic acid and pepsin result in [418] Y Tabata, Y Ikada, Protein release from gelatin matrices, Adv Drug Del Rev 31
high yield, high purity and low macrophage response collagen for biomedical (1998) 287–301.
applications, Biomed Mater 12 (2017) 065009. [419] S Young, M Wong, Y Tabata, AG Mikos, Gelatin as a delivery vehicle for the
[386] B An, DL Kaplan, B Brodsky, Engineered recombinant bacterial collagen as an controlled release of bioactive molecules, J Controlled Release 109 (2005)
alternative collagen-based biomaterial for tissue engineering, Front Chem 2 256–274.
(2014) 40-. [420] PH II Maurer, Antigenicity of Gelatin in Rabbits and other Species, J Exp Med
[387] B Brodsky, J Werkmeister, JA. Ramshaw, Collagens and Gelatins. In: Fahne- 100 (1954) 515.
stock SR, Steinbüchel A, editors. Biopolymers, Weinheim: Wiley-VCH (2003) [421] D Hellio, M Djabourov, Physically and Chemically Crosslinked Gelatin Gels,
119–153. Macromol Symp 241 (2006) 23–27.
[388] K Gelse, E Pöschl, T Aigner, Collagens–structure, function, and biosynthesis, [422] PB Malafaya, GA Silva, RL Reis, Natural-origin polymers as carriers and scaf-
Adv Drug Del Rev 55 (2003) 1531–1546. folds for biomolecules and cell delivery in tissue engineering applications,
[389] P Angele, J Abke, R Kujat, H Faltermeier, D Schumann, M Nerlich, et al., Adv Drug Del Rev 59 (2007) 207–233.
Influence of different collagen species on physico-chemical properties of [423] K Yamada, Y Tabata, K Yamamoto, S Miyamoto, I Nagata, H Kikuchi, et al., Po-
crosslinked collagen matrices, Biomaterials 25 (2004) 2831–2841. tential efficacy of basic fibroblast growth factor incorporated in biodegradable
[390] MB Yaylaoglu, C Yıldız, F Korkusuz, V Hasırcı, Novel osteochondral implant, hydrogels for skull bone regeneration, J Neurosurg 86 (1997) 871–875.
Biomaterials 20 (1999) 1513–1520. [424] Y Tabata, A Nagano, M Muniruzzaman, Y Ikada, In vitro sorption and desorp-
[391] WF Daamen, HTB van Moerkerk, T Hafmans, L Buttafoco, AA Poot, tion of basic fibroblast growth factor from biodegradable hydrogels, Biomate-
JH Veerkamp, et al., Preparation and evaluation of molecularly-defined col- rials 19 (1998) 1781–1789.
lagen-elastin-glycosaminoglycan scaffolds for tissue engineering, Biomaterials [425] M Yamamoto, Y Takahashi, Y Tabata, Controlled release by biodegradable hy-
24 (2003) 4001–4009. drogels enhances the ectopic bone formation of bone morphogenetic protein,
[392] L-S Liu, AY Thompson, MA Heidaran, JW Poser, RC Spiro, An osteoconductive Biomaterials 24 (2003) 4375–4383.
collagen/hyaluronate matrix for bone regeneration, Biomaterials 20 (1999) [426] ZS Patel, M Yamamoto, H Ueda, Y Tabata, AG Mikos, Biodegradable gelatin
1097–1108. microparticles as delivery systems for the controlled release of bone mor-
[393] S Woerly, R Marchand, G Lavallée, Interactions of copolymeric poly(glyceryl phogenetic protein-2, Acta Biomater 4 (2008) 1126–1138.
methacrylate)-collagen hydrogels with neural tissue: effects of structure and [427] H Park, JS Temenoff, TA Holland, Y Tabata, AG Mikos, Delivery of TGF-[beta]1
polar groups, Biomaterials 12 (1991) 197–203. and chondrocytes via injectable, biodegradable hydrogels for cartilage tissue
[394] JK Rao, DV Ramesh, KP Rao, Implantable controlled delivery systems for pro- engineering applications, Biomaterials 26 (2005) 7095–7103.
teins based on collagenpHEMA hydrogels, Biomaterials 15 (1994) 383–389. [428] J Malda, E Kreijveld, JS Temenoff, Blitterswijk CAv, J Riesle, Expansion of hu-
[395] MD Harriger, AP Supp, GD Warden, ST Boyce, Glutaraldehyde crosslinking of man nasal chondrocytes on macroporous microcarriers enhances redifferen-
collagen substrates inhibits degradation in skin substitutes grafted to athymic tiation, Biomaterials 24 (2003) 5153–5161.
mice, J Biomed Mater Res 35 (1997) 137–145. [429] RG Payne, MJ Yaszemski, AW Yasko, AG Mikos, Development of an injectable,
[396] WT Brinkman, K Nagapudi, BS Thomas, EL Chaikoff, Photo-cross-linking of in situ crosslinkable, degradable polymeric carrier for osteogenic cell pop-
type I collagen gels in the presence of smooth muscle cells: Mechanical prop- ulations. Part 1. Encapsulation of marrow stromal osteoblasts in surface
erties, cell viability, and function, Biomacromolecules 4 (2003) 890–895. crosslinked gelatin microparticles, Biomaterials 23 (2002) 4359–4371.
[397] WM Elbjeirami, EO Yonter, BC Starcher, JL West, Enhancing mechanical prop- [430] MS Ponticiello, RM Schinagl, S Kadiyala, FP Barry, Gelatin-based resorbable
erties of tissue-engineered constructs via lysyl oxidase crosslinking activity, J sponge as a carrier matrix for human mesenchymal stem cells in cartilage
Biomed Mater Res A 66A (2003) 513–521. regeneration therapy, J Biomed Mater Res 52 (20 0 0) 246–255.
[398] M Fujioka-Kobayashi, B Schaller, N Saulacic, BE Pippenger, Y Zhang, RJ Miron, [431] Y Liu, XZ Shu, GD Prestwich, Osteochondral Defect Repair with Autologous
Absorbable collagen sponges loaded with recombinant bone morphogenetic Bone Marrow–Derived Mesenchymal Stem Cells in an Injectable, In Situ,
protein 9 induces greater osteoblast differentiation when compared to bone Cross-Linked Synthetic Extracellular Matrix, Tissue Eng 12 (2006) 3405–3416.
morphogenetic protein 2, Clin, Exp, Dent Res 3 (2017) 32–40. [432] Y Kimura, M Ozeki, T Inamoto, Y Tabata, Adipose tissue engineering based on
[399] HJ Pulkkinen, V Tiitu, P Valonen, JS Jurvelin, MJ Lammi, I Kiviranta, Engineer- human preadipocytes combined with gelatin microspheres containing basic
ing of cartilage in recombinant human type II collagen gel in nude mouse fibroblast growth factor, Biomaterials 24 (2003) 2513–2521.
model in vivo, Osteoarthritis Cartilage 18 (2010) 1077–1087. [433] N Detta, C Errico, D Dinucci, D Puppi, D Clarke, G Reilly, et al., Novel electro-
[400] F Chamieh, A-M Collignon, BR Coyac, J Lesieur, S Ribes, J Sadoine, et al., Ac- spun polyurethane/gelatin composite meshes for vascular grafts, J Mater Sci
celerated craniofacial bone regeneration through dense collagen gel scaffolds Mater Med 21 (2010) 1761–1769.
seeded with dental pulp stem cells, Sci Rep 6 (2016) 38814. [434] Y Luo, Y Li, X Qin, Q Wa, 3D printing of concentrated alginate/gelatin scaf-
[401] C Dhand, ST Ong, N Dwivedi, SM Diaz, JR Venugopal, B Navaneethan, et al., folds with homogeneous nano apatite coating for bone tissue engineering,
Bio-inspired in situ crosslinking and mineralization of electrospun collagen Mater Des 146 (2018) 12–19.
scaffolds for bone tissue engineering, Biomaterials 104 (2016) 323–338. [435] Z Wu, X Su, Y Xu, B Kong, W Sun, S Mi, Bioprinting three-dimensional
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 53

cell-laden tissue constructs with controllable degradation, Sci Rep 6 (2016) siou, Electrospun silk fibroin fibers for storage and controlled release of hu-
24474. man platelet lysate, Acta Biomater 73 (2018) 365–376.
[436] C Tse, Y Zhang, PJ Smith, The use of reactive inkjet printing in tissue engi- [468] L Meinel, S Hofmann, V Karageorgiou, L Zichner, R Langer, D Kaplan, et al.,
neering, in: PJ Smith, A Morrin (Eds.), Reactive Inkjet Printing: A Chemical Engineering cartilage-like tissue using human mesenchymal stem cells and
Synthesis Tool, CPI Group, Croydon, UK, 2018. silk protein scaffolds, Biotechnol Bioeng 88 (2004) 379–391.
[437] AI Van Den Bulcke, B Bogdanov, N De Rooze, EH Schacht, M Cornelissen, [469] L Meinel, S Hofmann, O Betz, R Fajardo, HP Merkle, R Langer, et al., Osteoge-
H Berghmans, Structural and Rheological Properties of Methacrylamide Mod- nesis by human mesenchymal stem cells cultured on silk biomaterials: Com-
ified Gelatin Hydrogels, Biomacromolecules 1 (20 0 0) 31–38. parison of adenovirus mediated gene transfer and protein delivery of BMP-2,
[438] SP Grogan, PH Chung, P Soman, P Chen, MK Lotz, S Chen, et al., Digital mi- Biomaterials 27 (2006) 4993–5002.
cromirror device projection printing system for meniscus tissue engineering, [470] X Zhang, C Cao, X Ma, Y Li, Optimization of macroporous 3-D silk fibroin scaf-
Acta Biomater 9 (2013) 7218–7226. folds by salt-leaching procedure in organic solvent-free conditions, J Mater
[439] FPW Melchels, WJA Dhert, DW Hutmacher, J Malda, Development and char- Sci Mater Med 23 (2012) 315–324.
acterisation of a new bioink for additive tissue manufacturing, J Mater Chem [471] V Karageorgiou, L Meinel, S Hofmann, A Malhotra, V Volloch, D Kaplan, Bone
B 2 (2014) 2282–2289. morphogenetic protein-2 decorated silk fibroin films induce osteogenic dif-
[440] VHM Mouser, FPW Melchels, J Visser, WJA Dhert, D Gawlitta, J Malda, Yield ferentiation of human bone marrow stromal cells, J Biomed Mater Res A 71A
stress determines bioprintability of hydrogels based on gelatin-methacryloyl (2004) 528–537.
and gellan gum for cartilage bioprinting, Biofabrication 8 (2016) 035003. [472] L Uebersax, HP Merkle, L Meinel, Insulin-like growth factor I releasing silk
[441] FPW Melchels, MM Blokzijl, R Levato, QC Peiffer, Ruijter Md, WE Hennink, fibroin scaffolds induce chondrogenic differentiation of human mesenchymal
et al., Hydrogel-based reinforcement of 3D bioprinted constructs, Biofabrica- stem cells, J Controlled Release 127 (2008) 12–21.
tion 8 (2016) 035004. [473] M Farokhi, F Mottaghitalab, S Samani, MA Shokrgozar, SC Kundu, RL Reis,
[442] TAE Ahmed, EV Dare, M Hincke, Fibrin: A Versatile Scaffold for Tissue Engi- et al., Silk fibroin/hydroxyapatite composites for bone tissue engineering,
neering Applications, Tissue Eng Part B (2008). Biotechnol Adv 36 (2018) 68–91.
[443] RI Litvinov, JW Weisel, What Is the Biological and Clinical Relevance of Fib- [474] HJ Kim, U-J Kim, HS Kim, C Li, M Wada, GG Leisk, et al., Bone tissue engi-
rin? Semin Thromb Hemost 42 (2016) 333–343. neering with premineralized silk scaffolds, Bone 42 (2008) 1226–1234.
[444] E Ryan, L Mockros, J Weisel, L Lorand, Structural origins of fibrin clot rheol- [475] C Li, C Vepari, H-J Jin, HJ Kim, DL Kaplan, Electrospun silk-BMP-2 scaffolds
ogy, Biophys J 77 (1999) 2813–2826. for bone tissue engineering, Biomaterials 27 (2006) 3115–3124.
[445] WD Spotnitz, Fibrin Sealant, Past, Present, and Future: A Brief Review, World [476] C Holland, K Numata, J Rnjak-Kovacina, FP Seib, The Biomedical Use of Silk:
J Surg 34 (2010) 632–634. Past, Present, Future, Adv Healthcare Mater 8 (2019) 1800465.
[446] JC Chapin, KA Hajjar, Fibrinolysis and the control of blood coagulation, Blood [477] P Di Donato, A Poli, V Taurisano, B Nicolaus, Polysaccharides from Bioa-
Rev 29 (2015) 17–24. gro-Waste for New Biomolecules, in: KG Ramawat, J-M Mérillon (Eds.),
[447] W Ho, B Tawil, JCY Dunn, BM Wu, The Behavior of Human Mesenchymal Polysaccharides: Bioactivity and Biotechnology, Springer International Pub-
Stem Cells in 3D Fibrin Clots: Dependence on Fibrinogen Concentration and lishing, Cham, 2015, pp. 603–637.
Clot Structure, Tissue Eng 12 (2006) 1587–1595. [478] A Morelli, D Puppi, F Chiellini, Chapter 16 - Perspectives on Biomedical Appli-
[448] P Jing, JS Rudra, AB Herr, JH Collier, Self-Assembling Peptide-Polymer Hydro- cations of Ulvan, in: J Venkatesan, S Anil, S-K Kim (Eds.), Seaweed Polysaccha-
gels Designed From the Coiled Coil Region of Fibrin, Biomacromolecules 9 rides. Radarweg 29, PO Box 211, 10 0 0 AE Amsterdam, Elsevier, Netherlands,
(2008) 2438–2446. 2017, pp. 305–330.
[449] G Zhang, X Wang, Z Wang, J Zhang, L Suggs, A PEGylated Fibrin Patch for [479] KY Lee, DJ Mooney, Alginate: Properties and biomedical applications, Prog
Mesenchymal Stem Cell Delivery, Tissue Eng 12 (2006) 9–19. Polym Sci 37 (2012) 106–126.
[450] TC Gamboa-Martínez, V Luque-Guillén, C González-García, JL Gómez Ribelles, [480] M George, TE Abraham, Polyionic hydrocolloids for the intestinal delivery of
G Gallego-Ferrer, Crosslinked fibrin gels for tissue engineering: Two ap- protein drugs: Alginate and chitosan – a review, J Controlled Release 114
proaches to improve their properties, J Biomed Mater Res A 103 (2014) (2006) 1–14.
614–621. [481] P Vauchel, K Leroux, R Kaas, A Arhaliass, R Baron, J Legrand, Kinetics model-
[451] MV Lieshout, G Peters, M Rutten, F Baaijens, A Knitted, Fibrin-Covered Poly- ing of alginate alkaline extraction from Laminaria digitata, Bioresour Technol
caprolactone Scaffold for Tissue Engineering of the Aortic Valve, Tissue Eng 10 0 (20 09) 1291–1296.
12 (2006) 481–487. [482] M. Otterlei, K. Ostgaard, G. Skjakbraek, O. Smidsrod, P. Soonshiong, T Espevik,
[452] RR Rao, AW Peterson, J Ceccarelli, AJ Putnam, JP Stegemann, Matrix composi- Induction of cytokine production from human monocytes stimulated with al-
tion regulates three-dimensional network formation by endothelial cells and ginate, J Immunother 10 (1991) 286.
mesenchymal stem cells in collagen/fibrin materials, Angiogenesis 15 (2012) [483] U. Zimmermann, G. Klock, K. Federlin, K. Haning, M. Kowaslski, R.G. Bretzel,
253–264. et al., Production of mitogen contamination free alginates with variable ratios
[453] C Vepari, DL Kaplan, Silk as a biomaterial, Prog Polym Sci 32 (2007) of mannuronic to guluronic acid by free flow electrophoresis, Electrophoresis
991–1007. 13 (1992) 269.
[454] DN Rockwood, RC Preda, T Yücel, X Wang, ML Lovett, DL Kaplan, Materials [484] HK Holme, L Davidsen, A Kristiansen, O Smidsrød, Kinetics and mechanisms
fabrication from Bombyx mori silk fibroin, Nat Protoc 6 (2011) 1612. of depolymerization of alginate and chitosan in aqueous solution, Carbohydr
[455] C Vepari, DL Kaplan, Silk as a biomaterial, Prog Polym Sci 32 (2007) Polym 73 (2008) 656–664.
991–1007. [485] M Machluff, Protein therapeutic delivery using encapsulated cell platform, in:
[456] F Vollrath, DP Knight, Liquid crystalline spinning of spider silk, Nature 410 V. Nedovic, R. Villaert (Eds.), Application of cell immobilization Biotechnol-
(2001) 541–548. ogy, Springer, Dordrecht, 2005, p. 200.
[457] B Panilaitis, Altman GH, Chen J, Jin H-J, Karageorgiou V, Kaplan DL, [486] JA Rowley, G Madlambayan, DJ Mooney, Alginate hydrogels as synthetic ex-
Macrophage responses to silk, Biomaterials 24 (2003) 3079–3085. tracellular matrix materials, Biomaterials 20 (1999) 45–53.
[458] A Glisovic, T Vehoff, R Davies, T Salditt, Correlation between structure and [487] O. Jeon, C. Powell, S.M. Ahmed, Alsberg Biodegradable E, photocrosslinked al-
mechanical properties of spider silk, Comp Biochem Physiol, A: Mol Integr ginate hydrogels with independently tailorable physical properties and cell
Physiol 146 (2007) pp. S138-S. adhesivity, Tissue Eng Part A 16 (2010) 2915.
[459] H-J Jin, DL Kaplan, Mechanism of silk processing in insects and spiders, Na- [488] S Degala, WR Zipfel, LJ Bonassar, Chondrocyte calcium signaling in response
ture 424 (2003) 1057–1061. to fluid flow is regulated by matrix adhesion in 3-D alginate scaffolds, Arch
[460] GH Altman, F Diaz, C Jakuba, T Calabro, RL Horan, J Chen, et al., Silk-based Biochem Biophys 505 (2011) 112–117.
biomaterials, Biomaterials 24 (2003) 401–416. [489] MB Evangelista, SX Hsiong, R Fernandes, P Sampaio, H-J Kong, CC Barrias,
[461] B Kundu, R Rajkhowa, SC Kundu, X Wang, Silk fibroin biomaterials for tissue et al., Upregulation of bone cell differentiation through immobilization within
regenerations, Adv Drug Del Rev 65 (2013) 457–470. a synthetic extracellular matrix, Biomaterials 28 (2007) 3644–3655.
[462] RE Unger, M Wolf, K Peters, A Motta, C Migliaresi, C James Kirkpatrick, [490] PK Kreeger, JW Deck, TK Woodruff, LD Shea, The in vitro regulation of ovarian
Growth of human cells on a non-woven silk fibroin net: a potential for use follicle development using alginate-extracellular matrix gels, Biomaterials 27
in tissue engineering, Biomaterials 25 (2004) 1069–1075. (2006) 714–723.
[463] F Zhang, Z Zhang, X Zhu, E-T Kang, K-G Neoh, Silk-functionalized titanium [491] KY Lee, E Alsberg, S Hsiong, W Comisar, J Linderman, R Ziff, et al., Nanoscale
surfaces for enhancing osteoblast functions and reducing bacterial adhesion, Adhesion Ligand Organization Regulates Osteoblast Proliferation and Differ-
Biomaterials 29 (2008) 4751–4759. entiation, Nano Lett 4 (2004) 1501–1506.
[464] RE Unger, K Peters, M Wolf, A Motta, C Migliaresi, CJ Kirkpatrick, Endothe- [492] WA Comisar, SX Hsiong, H-J Kong, DJ Mooney, JJ Linderman, Multi-scale mod-
lialization of a non-woven silk fibroin net for use in tissue engineering: eling to predict ligand presentation within RGD nanopatterned hydrogels,
growth and gene regulation of human endothelial cells, Biomaterials 25 Biomaterials 27 (2006) 2322–2329.
(2004) 5137–5146. [493] A Al-Shamkhani, R Duncan, Radioiodination of Alginate via Covalently-Bound
[465] S Fuchs, A Motta, C Migliaresi, CJ Kirkpatrick, Outgrowth endothelial cells iso- Tyrosinamide Allows Monitoring of its Fate In Vivo, J Bioact Compatible
lated and expanded from human peripheral blood progenitor cells as a poten- Polym 10 (1995) 4–13.
tial source of autologous cells for endothelialization of silk fibroin biomateri- [494] KH Bouhadir, KY Lee, E Alsberg, KL Damm, KW Anderson, DJ Mooney, Degra-
als, Biomaterials 27 (2006) 5399–5408. dation of Partially Oxidized Alginate and Its Potential Application for Tissue
[466] H-J Jin, J Chen, V Karageorgiou, GH Altman, DL Kaplan, Human bone mar- Engineering, Biotechnol Prog 17 (2001) 945–950.
row stromal cell responses on electrospun silk fibroin mats, Biomaterials 25 [495] A Lueckgen, DS Garske, A Ellinghaus, RM Desai, AG Stafford, DJ Mooney,
(2004) 1039–1047. et al., Hydrolytically-degradable click-crosslinked alginate hydrogels, Bioma-
[467] C Pignatelli, G Perotto, M Nardini, R Cancedda, M Mastrogiacomo, A Athanas- terials 181 (2018) 189–198.
54 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

[496] GT Grant, Morris ER, Rees DA, Smith PJC, Thom D, Biological interactions be- three-dimensional printing and directional freezing, Biofabrication 8 (2016)
tween polysaccharides and divalent cations: the egg box model, FEBS Lett 32 015003.
(1973) 195–198. [528] GF Picheth, CL Pirich, MR Sierakowski, MA Woehl, CN Sakakibara, CF de
[497] NC Hunt, AM Smith, U Gbureck, RM Shelton, LM Grover, Encapsulation of Souza, et al., Bacterial cellulose in biomedical applications: A review, Int J
fibroblasts causes accelerated alginate hydrogel degradation, Acta Biomater 6 Biol Macromol 104 (2017) 97–106.
(2010) 3649–3656. [529] D Klemm, H-P Schmauder, T Heinze, Cellulose, in: A Steinbüchel (Ed.),
[498] P Eiselt, KY Lee, DJ Mooney, Rigidity of Two-Component Hydrogels Pre- Biopolymers, WILEY-VCH, Weinheim, 2002, pp. 275–319.
pared from Alginate and Poly(ethylene glycol)−Diamines, Macromolecules 32 [530] S Park, JO Baker, ME Himmel, PA Parilla, DK Johnson, Cellulose crystallinity
(1999) 5561–5566. index: measurement techniques and their impact on interpreting cellulase
[499] KH Bouhadir, DS Hausman, DJ Mooney, Synthesis of cross-linked performance, Biotechnol Biofuels 3 (2010) 10.
poly(aldehyde guluronate) hydrogels, Polymer 40 (1999) 3575–3584. [531] K Jedvert, T Heinze, Cellulose modification and shaping – a review, J Polym
[500] KY Lee, JA Rowley, P Eiselt, EM Moy, KH Bouhadir, DJ Mooney, Control- Eng 37 (2017) 845.
ling Mechanical and Swelling Properties of Alginate Hydrogels Independently [532] JC Courtenay, RI Sharma, JL Scott, Recent Advances in Modified Cellulose for
by Cross-Linker Type and Cross-Linking Density, Macromolecules 33 (20 0 0) Tissue Culture Applications, Molecules 23 (2018) 654.
4291–4294. [533] M Shoda, Y Sugano, Recent advances in bacterial cellulose production,
[501] O Jeon, KH Bouhadir, JM Mansour, E Alsberg, Photocrosslinked alginate hy- Biotechnol Bioprocess Eng 10 (2005) 1.
drogels with tunable biodegradation rates and mechanical properties, Bioma- [534] P Gatenholm, D Klemm, Bacterial Nanocellulose as a Renewable Material for
terials 30 (2009) 2724–2734. Biomedical Applications, MRS Bull 35 (2010) 208.
[502] D Seliktar, Designing Cell-Compatible Hydrogels for Biomedical Applications, [535] I Sulaeva, U Henniges, T Rosenau, A Potthast, Bacterial cellulose as a material
Science 336 (2012) 1124. for wound treatment: Properties and modifications, A review, Biotechnol Adv
[503] RM Desai, ST Koshy, SA Hilderbrand, DJ Mooney, NS Joshi, Versatile click algi- 33 (2015) 1547–1571.
nate hydrogels crosslinked via tetrazine–norbornene chemistry, Biomaterials [536] J Kucińska-Lipka, I Gubanska, H Janik, Bacterial cellulose in the field of wound
50 (2015) 30–37. healing and regenerative medicine of skin: recent trends and future prospec-
[504] S Hafeez, WH Ooi, LF Morgan, C Mota, M Dettin, C Van Blitterswijk, et al., tives, Polym Bull 72 (2015) 2399–2419.
Viscoelastic Oxidized Alginates with Reversible Imine Type Crosslinks: Self- [537] TR Stumpf, X Yang, J Zhang, X Cao, In situ and ex situ modifications of bacte-
-Healing, Injectable, and Bioprintable Hydrogels, Gels 4 (2018). rial cellulose for applications in tissue engineering, Mater Sci Eng, C 82 (2018)
[505] A Di Martino, M Sittinger, MV Risbud, Chitosan: A versatile biopolymer for 372–383.
orthopaedic tissue-engineering, Biomaterials 26 (2005) 5983–5990. [538] S Torgbo, P Sukyai, Bacterial cellulose-based scaffold materials for bone tissue
[506] MG Peter, Chitin and Chitosan from Animal Sources, in: A Steinbüchel (Ed.), engineering, Appl Mater Today 11 (2018) 34–49.
Biopolymers, WILEY-VCH, Weinheim, 2002, pp. 481–574. [539] K Schütz, A-M Placht, B Paul, S Brüggemeier, M Gelinsky, A Lode, Three-
[507] M-K Jang, B-G Kong, Y-I Jeong, CH Lee, J-W Nah, Physicochemical characteri- -dimensional plotting of a cell-laden alginate/methylcellulose blend: towards
zation of α -chitin, β -chitin, and γ -chitin separated from natural resources, J biofabrication of tissue engineering constructs with clinically relevant dimen-
Polym Sci, Part A: Polym Chem 42 (2004) 3423–3432. sions, J Tissue Eng Regener Med 11 (2017) 1574–1587.
[508] M Dash, F Chiellini, RM Ottenbrite, E Chiellini, Chitosan—A versatile [540] DHAT Gunasekera, S Kuek, D Hasanaj, Y He, C Tuck, AK Croft, et al., Three di-
semi-synthetic polymer in biomedical applications, Prog Polym Sci 36 (2011) mensional ink-jet printing of biomaterials using ionic liquids and co-solvents,
981–1014. Faraday Discuss 190 (2016) 509–523.
[509] I Aranaz, M Mengibar, R Harris, I Panos, B Miralles, N Acosta, et al., Functional [541] DD Allison, KJ Grande-Allen, Review. Hyaluronan: A Powerful Tissue Engi-
Characterization of Chitin and Chitosan, Curr Chem Biol 3 (2009) 203–230. neering Tool, Tissue Eng 12 (2006) 2131–2140.
[510] M Kong, XG Chen, K Xing, HJ Park, Antimicrobial properties of chitosan and [542] D Campoccia, P Doherty, M Radice, P Brun, G Abatangelo, DF Williams,
mode of action: A state of the art review, Int J Food Microbiol 144 (2010) Semisynthetic resorbable materials from hyaluronan esterification, Biomate-
51–63. rials 19 (1998) 2101–2127.
[511] K Kurita, Chitin and Chitosan: Functional Biopolymers from Marine Crus- [543] TC Laurent, UG Laurent, JE Fraser, Functions of hyaluronan, Ann Rheum Dis
taceans, Mar Biotechnol 8 (2006) 203–226. 54 (1995) 429–432.
[512] S Hirano, Chitin and chitosan as novel biotechnological materials, Polym Int [544] L Liu, Y Liu, J Li, G Du, J Chen, Microbial production of hyaluronic acid: cur-
48 (1999) 732–734. rent state, challenges, and perspectives, Microb Cell Fact 10 (2011) 99.
[513] T Kean, M Thanou, Biodegradation, biodistribution and toxicity of chitosan, [545] G Kogan, L Šoltés, R Stern, P Gemeiner, Hyaluronic acid: a natural biopolymer
Adv Drug Del Rev 62 (2010) 3–11. with a broad range of biomedical and industrial applications, Biotechnol Lett
[514] C Chatelet, O Damour, A Domard, Influence of the degree of acetylation on 29 (2007) 17–25.
some biological properties of chitosan films, Biomaterials 22 (2001) 261–268. [546] CB Highley, GD Prestwich, JA Burdick, Recent advances in hyaluronic acid hy-
[515] D Ren, H Yi, W Wang, X Ma, The enzymatic degradation and swelling prop- drogels for biomedical applications, Curr Opin Biotechnol 40 (2016) 35–40.
erties of chitosan matrices with different degrees of N-acetylation, Carbohydr [547] MN Collins, C Birkinshaw, Hyaluronic acid based scaffolds for tissue engineer-
Res 340 (2005) 2403–2410. ing—A review, Carbohydr Polym 92 (2013) 1262–1279.
[516] K Divya, MS Jisha, Chitosan nanoparticles preparation and applications, Envi- [548] JE Scott, F Heatley, Biological Properties of Hyaluronan in Aqueous Solution
ronmental Chemistry Letters 16 (2018) 101–112. Are Controlled and Sequestered by Reversible Tertiary Structures, Defined by
[517] F Croisier, C Jérôme, Chitosan-based biomaterials for tissue engineering, Eur NMR Spectroscopy, Biomacromolecules 3 (2002) 547–553.
Polym J 49 (2013) 780–792. [549] JK Park, J-H Shim, KS Kang, J Yeom, HS Jung, JY Kim, et al., Solid Free-Form
[518] B Duan, X Yuan, Y Zhu, Y Zhang, X Li, Y Zhang, et al., A nanofibrous compos- Fabrication of Tissue-Engineering Scaffolds with a Poly(lactic-co-glycolic acid)
ite membrane of PLGA-chitosan/PVA prepared by electrospinning, Eur Polym Grafted Hyaluronic Acid Conjugate Encapsulating an Intact Bone Morpho-
J 42 (2006) 2013–2022. genetic Protein–2/Poly(ethylene glycol) Complex, Adv Funct Mater 21 (2011)
[519] X Cai, H Tong, X Shen, W Chen, J Yan, J Hu, Preparation and characteriza- 2906–2912.
tion of homogeneous chitosan-polylactic acid/hydroxyapatite nanoco,mposite [550] F Schellauf, E Grillo Fernandes, G Braunegg, E Chiellini, Properties of PHAs
for bone tissue engineering and evaluation of its mechanical properties, Acta and Their Correlation to Fermentation Conditions in Biorelated Polymers, in:
Biomater (2009). E Chiellini, H Gil, G Braunegg, J Buchert, P Gatenholm, M van der Zee (Eds.),
[520] Z Li, HR Ramay, KD Hauch, D Xiao, M Zhang, Chitosan-alginate hybrid scaf- Sustainable Polymer Science and Technology, Kluwer Academic/Plenum Pub-
folds for bone tissue engineering, Biomaterials 26 (2005) 3919–3928. lishers, New York, 2001, p. 115.
[521] JL Cuy, BL Beckstead, CD Brown, AS Hoffman, CM Giachelli, Adhesive protein [551] M Zinn, B Witholt, T Egli, Occurrence, synthesis and medical application of
interactions with chitosan: Consequences for valve endothelial cell growth on bacterial polyhydroxyalkanoate, Adv Drug Del Rev 53 (2001) 5–21.
tissue-engineering materials, J Biomed Mater Res A 67A (2003) 538–547. [552] C Fernández-Dacosta, JA Posada, R Kleerebezem, MC Cuellar, A Ramirez,
[522] Y Zhang, M Zhang, Microstructural and mechanical characterization of chi- Microbial community-based polyhydroxyalkanoates (PHAs) production from
tosan scaffolds reinforced by calcium phosphates, J Non-Cryst Solids 282 wastewater: Techno-economic analysis and ex-ante environmental assess-
(2001) 159–164. ment, Bioresour Technol 185 (2015) 368–377.
[523] M Gravel, R Vago, M Tabrizian, Use of Natural Coralline Biomaterials As Re- [553] M Koller, L Maršálek, MM de Sousa Dias, G Braunegg, Producing micro-
inforcing and Gas-Forming Agent for Developing Novel Hybrid Biomatrices: bial polyhydroxyalkanoate (PHA) biopolyesters in a sustainable manner, New
Microarchitectural and Mechanical Studies, Tissue Eng 12 (2006) 589–600. Biotechnology 37 (2017) 24–38.
[524] KM Park, YK Joung, JS Na, MC Lee, KD Park, Thermosensitive chitosan– [554] M Koller, Advances in Polyhydroxyalkanoate (PHA) Production, Bioengineer-
Pluronic hydrogel as an injectable cell delivery carrier for cartilage regenera- ing 4 (2017) 88.
tion, Acta Biomater 5 (2009) 1956–1965. [555] ER Coats, BS Watson, CK Brinkman, Polyhydroxyalkanoate Synthesis by Mixed
[525] CR Almeida, T Serra, MI Oliveira, JA Planell, MA Barbosa, M Navarro, Im- Microbial Consortia Cultured on Fermented Dairy Manure: Effect of Aeration
pact of 3-D printed PLA- and chitosan-based scaffolds on human mono- on Process Rates/Yields and the Associated Microbial Ecology, Water Res 106
cyte/macrophage responses: Unraveling the effect of 3-D structures on in- (2016) 26–40.
flammation, Acta Biomater 10 (2014) 613–622. [556] M Lemoigne, Produits de deshydration et de polymerisation de l’acide
[526] J Zhang, BJ Allardyce, R Rajkhowa, Y Zhao, RJ Dilley, SL Redmond, et al., 3D b-oxobutyrique, Bull Soc Chem Biol 8 (1926) 770–782.
Printing of Silk Particle-Reinforced Chitosan Hydrogel Structures and Their [557] DP Martin, SF Williams, Medical applications of poly-4-hydroxybutyrate: a
Properties, ACS Biomater Sci Eng 4 (2018) 3036–3046. strong flexible absorbable biomaterial, Biochem Eng J 16 (2003) 97–105.
[527] R Stephanie, L Grace, D Benjamin, L David Don, PT Antoni, MW Ben- [558] M Miranda De Sousa Dias, M Koller, D Puppi, A Morelli, F Chiellini,
jamin, Macro- and micro-designed chitosan-alginate scaffold architecture by G Braunegg, Fed-Batch Synthesis of Poly(3-Hydroxybutyrate) and
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 55

Poly(3-Hydroxybutyrate-co-4-Hydroxybutyrate) from Sucrose and 4-Hy- [585] H-T Liao, M-Y Lee, W-W Tsai, H-C Wang, W-C Lu, Osteogenesis of adi-
droxybutyrate Precursors by Burkholderia sacchari Strain DSM 17165, pose-derived stem cells on polycaprolactone–β -tricalcium phosphate scaffold
Bioengineering 4 (2017). fabricated via selective laser sintering and surface coating with collagen type
[559] IK Radecka, G Jiang, DJ Hill, MM Kowalczuk, Poly(hydroxyalkanoates) com- I, J Tissue Eng Regener Med 10 (2016) E337–EE53.
posites and their applications, in: Inamuddin (Ed.), Green Polymer Compos- [586] Z Xiong, Y Yan, R Zhang, L Sun, Fabrication of porous poly(l-lactic acid)
ites Technology: Properties and Applications, CRC Press, Boca Raton, 2016, scaffolds for bone tissue engineering via precise extrusion, Scripta Mater 45
pp. 163–175. (2001) 773–779.
[560] A Padermshoke, Y Katsumoto, H Sato, S Ekgasit, I Noda, Y Ozaki, Sur- [587] R Fairag, DH Rosenzweig, JL Ramirez-Garcialuna, MH Weber, L Haglund,
face melting and crystallization behavior of polyhydroxyalkanoates stud- Three-Dimensional Printed Polylactic Acid Scaffolds Promote Bone-like Ma-
ied by attenuated total reflection infrared spectroscopy, Polymer 45 (2004) trix Deposition in Vitro, ACS Appl Mater Interfaces 11 (2019) 15306–15315.
6547–6554. [588] A Gregor, E Filová, M Novák, J Kronek, H Chlup, M Buzgo, et al., Designing of
[561] L Feng, T Watanabe, Y Wang, T Kichise, T Fukuchi, G-Q Chen, PLA scaffolds for bone tissue replacement fabricated by ordinary commercial
et al., Studies on Comonomer Compositional Distribution of Bacterial 3D printer, J Biol Eng 11 (2017) 31-.
Poly(3-hydroxybutyrate-co-3-hydroxyhexanoate)s and Thermal Characteris- [589] H Cui, W Zhu, B Holmes, LG Zhang, Biologically Inspired Smart Release Sys-
tics of Their Factions, Biomacromolecules 3 (2002) 1071–1077. tem Based on 3D Bioprinted Perfused Scaffold for Vascularized Tissue Regen-
[562] Q Yang, J Wang, S Zhang, X Tang, G Shang, Q Peng, et al., The Properties of eration, Adv Sci 3 (2016) 160 0 058.
Poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) and its Applications in Tis- [590] M Barbeck, T Serra, P Booms, S Stojanovic, S Najman, E Engel, et al., Analy-
sue Engineering, Curr Stem Cell Res Ther 9 (2014) 215–222. sis of the in vitro degradation and the in vivo tissue response to bi-layered
[563] H Abe, Y Doi, Side-Chain Effect of Second Monomer Units on Crystalline 3D-printed scaffolds combining PLA and biphasic PLA/bioglass components –
Morphology, Thermal Properties, and Enzymatic Degradability for Random Guidance of the inflammatory response as basis for osteochondral regenera-
Copolyesters of (R)-3-Hydroxybutyric Acid with (R)-3-Hydroxyalkanoic Acids, tion, Bioact Mater 2 (2017) 208–223.
Biomacromolecules 3 (2002) 133–138. [591] EN Antonov, SM Barinov, IV Vakhrushev, VS Komlev, VK Popov, AY Fedotov,
[564] A Shrivastav, H-Y Kim, Y-R Kim, Advances in the Applications of Polyhydrox- et al., Selective laser sintering of bioactive composite matrices for bone tissue
yalkanoate Nanoparticles for Novel Drug Delivery System, Biomed Res Int engineering, Inorg Mater Appl Res 6 (2015) 171–178.
(2013) (2013) 12. [592] FPW Melchels, AH Velders, J Feijen, DW Grijpma, Photo-Cross-Linked
[565] M Koller, Biodegradable and Biocompatible Polyhydroxy-alkanoates (PHA): Poly(dl-lactide)-Based Networks. Structural Characterization by HR-MAS NMR
Auspicious Microbial Macromolecules for Pharmaceutical and Therapeutic Spectroscopy and Hydrolytic Degradation Behavior, Macromolecules 43
Applications, Molecules 23 (2018). (2010) 8570–8579.
[566] A Södergård, Stolt M Properties of lactic acid based polymers and their cor- [593] FPW Melchels, K Bertoldi, R Gabbrielli, AH Velders, J Feijen, DW Grijpma,
relation with composition, Prog Polym Sci 27 (2002) 1123–1163. Mathematically defined tissue engineering scaffold architectures prepared by
[567] MN Cooke, JP Fisher, D Dean, C Rimnac, AG Mikos, Use of stereolithography stereolithography, Biomaterials 31 (2010) 6909–6916.
to manufacture critical-sized 3D biodegradable scaffolds for bone ingrowth, J [594] A Ronca, L Ambrosio, DW Grijpma, Design of Porous Three-Dimensional
Biomed Mater Res B Appl Biomater 64B (2003) 65–69. PDLLA/nano-hap Composite Scaffolds Using Stereolithography, J Appl Bio-
[568] JR Sarasua, AL Arraiza, P Balerdi, I Maiza, Crystallinity and mechanical proper- mater Func 10 (2012) 249–258.
ties of optically pure polylactides and their blends, Polym Eng Sci 45 (2005) [595] JM Hong, BJ Kim, J-H Shim, KS Kang, K-J Kim, JW Rhie, et al., Enhancement
745–753. of bone regeneration through facile surface functionalization of solid freeform
[569] K Madhavan Nampoothiri, NR Nair, RP John, An overview of the recent fabrication-based three-dimensional scaffolds using mussel adhesive proteins,
developments in polylactide (PLA) research, Bioresour Technol 101 (2010) Acta Biomater 8 (2012) 2578–2586.
8493–8501. [596] D Yu, Q Li, X Mu, T Chang, Z Xiong, Bone regeneration of critical calvarial
[570] AJR Lasprilla, GAR Martinez, BH Lunelli, AL Jardini, RM Filho, Poly-lactic acid defect in goat model by PLGA/TCP/rhBMP-2 scaffolds prepared by low-tem-
synthesis for application in biomedical devices — A review, Biotechnol Adv perature rapid-prototyping technology, Int J Oral Maxillofac Surg 37 (2008)
30 (2012) 321–328. 929–934.
[571] E Rudnik, 13 - Compostable Polymer Properties and Packaging Applications. [597] M Xu, Y Li, H Suo, Y Yan, L Liu, Q Wang, et al., Fabricating a pearl/PLGA com-
In: Ebnesajjad S, editor. Plastic Films in Food Packaging, Oxford: William An- posite scaffold by the low-temperature deposition manufacturing technique
drew Publishing (2013) 217–248. for bone tissue engineering, Biofabrication 2 (2010) 025002.
[572] JS Bergström, D Hayman, An Overview of Mechanical Properties and Material [598] RL Simpson, FE Wiria, AA Amis, CK Chua, KF Leong, UN Hansen, et al.,
Modeling of Polylactide (PLA) for Medical Applications, Ann Biomed Eng 44 Development of a 95/5 poly(L-lactide-co-glycolide)/hydroxylapatite and
(2016) 330–340. β -tricalcium phosphate scaffold as bone replacement material via selective
[573] S Farah, DG Anderson, R Langer, Physical and mechanical properties of PLA, laser sintering, J Biomed Mater Res B Appl Biomater 84B (2008) 17–25.
and their functions in widespread applications — A comprehensive review, [599] JW Lee, PX Lan, B Kim, G Lim, D-W Cho, Fabrication and characteristic anal-
Adv Drug Del Rev 107 (2016) 367–392. ysis of a poly(propylene fumarate) scaffold using micro-stereolithography
[574] M Biron, 4 - Renewable Plastics Derived From Natural Polymers, In: Biron M, technology, J Biomed Mater Res B Appl Biomater 87B (2008) 1–9.
editor. Industrial Applications of Renewable Plastics, William Andrew Pub- [600] JW Lee, G Ahn, JY Kim, D-W Cho, Evaluating cell proliferation based on in-
lishing (2017) 115–154. ternal pore size and 3D scaffold architecture fabricated using solid freeform
[575] OS Manoukian, N Sardashti, T Stedman, K Gailiunas, A Ojha, A Penalosa, fabrication technology, J Mater Sci Mater Med 21 (2010) 3195–3205.
et al., Biomaterials for Tissue Engineering and Regenerative Medicine, in: [601] K Kim, D Dean, J Wallace, R Breithaupt, AG Mikos, JP Fisher, The influence of
R Narayan (Ed.), Encyclopedia of Biomedical Engineering, Elsevier, Oxford, stereolithographic scaffold architecture and composition on osteogenic sig-
2019, pp. 462–482. nal expression with rat bone marrow stromal cells, Biomaterials 32 (2011)
[576] G Narayanan, VN Vernekar, EL Kuyinu, CT Laurencin, Poly (lactic acid)-based 3750–3763.
biomaterials for orthopaedic regenerative engineering, Adv Drug Del Rev 107 [602] J Wallace, MO Wang, P Thompson, M Busso, V Belle, N Mammoser, et al.,
(2016) 247–276. Validating continuous digital light processing (cDLP) additive manufacturing
[577] M. Niaounakis, 2 - Properties, in: M Niaounakis (Ed.), Biopolymers: Applica- accuracy and tissue engineering utility of a dye-initiator package, Biofabrica-
tions and Trends, William Andrew Publishing, Oxford, 2015, pp. 91–138. tion 6 (2014) 015003.
[578] PA Gunatillake, R Adhikari, Biodegradable synthetic polymers for tissue engi- [603] JE Trachtenberg, JK Placone, BT Smith, JP Fisher, AG Mikos, Extrusion-based
neering, eCM Journal 5 (2003) 1–16. 3D printing of poly(propylene fumarate) scaffolds with hydroxyapatite gradi-
[579] DW Hutmacher, T Schantz, I Zein, KW Ng, SH Teoh, KC Tan, Mechanical ents, J Biomater Sci Polym Ed 28 (2017) 532–554.
properties and cell cultural response of polycaprolactone scaffolds designed [604] LT Lim, R Auras, M Rubino, Processing technologies for poly(lactic acid), Prog
and fabricated via fused deposition modeling, J Biomed Mater Res 55 (2001) Polym Sci 33 (2008) 820–852.
203–216. [605] R Nigmatullin, P Thomas, B Lukasiewicz, H Puthussery, I Roy, Polyhydrox-
[580] XX Shao, DW Hutmacher, ST Ho, JCH Goh, EH Lee, Evaluation of a hybrid yalkanoates, a family of natural polymers, and their applications in drug de-
scaffold/cell construct in repair of high-load-bearing osteochondral defects in livery, J Chem Technol Biotechnol 90 (2015) 1209–1221.
rabbits, Biomaterials 27 (2006) 1071–1080. [606] I Manavitehrani, A Fathi, H Badr, S Daly, A Negahi Shirazi, Dehghani F
[581] Y Zhou, F Chen, ST Ho, MA Woodruff, TM Lim, DW Hutmacher, Combined Biomedical Applications of Biodegradable Polyesters, Polymers 8 (2016).
marrow stromal cell-sheet techniques and high-strength biodegradable com- [607] G-Q Chen, J Zhang, Microbial polyhydroxyalkanoates as medical implant bio-
posite scaffolds for engineered functional bone grafts, Biomaterials 28 (2007) materials AU - Chen, Guo-Qiang, Artif Cells Nanomed Biotechnol 46 (2018)
814–824. 1–18.
[582] J-H Shim, T-S Moon, M-J Yun, Y-C Jeon, C-M Jeong, D-W Cho, et al., Stimula- [608] DC Aluthge, C Xu, N Othman, N Noroozi, SG Hatzikiriakos, P Mehrkhoda-
tion of healing within a rabbit calvarial defect by a PCL/PLGA scaffold blended vandi, PLA–PHB–PLA Triblock Copolymers, Synthesis by Sequential Addition
with TCP using solid freeform fabrication technology, J Mater Sci Mater Med and Investigation of Mechanical and Rheological Properties, Macromolecules
23 (2012) 2993–3002. 46 (2013) 3965–3974.
[583] W Jiang, J Shi, W Li, K Sun, Three dimensional melt-deposition of polycapro- [609] M Winnacker, B Rieger, Copolymers of polyhydroxyalkanoates and polyethy-
lactone/bio-derived hydroxyapatite composite into scaffold for bone repair, J lene glycols: recent advancements with biological and medical significance,
Biomater Sci Polym Ed 24 (2013) 539–550. Polym Int 66 (2017) 497–503.
[584] S Lohfeld, S Cahill, V Barron, P McHugh, L Dürselen, L Kreja, et al., Fabri- [610] H-S Kwon, S-G Jung, H-Y Kim, SA Parker, CA Batt, Y-R Kim, A multi-functional
cation, mechanical and in vivo performance of polycaprolactone/tricalcium polyhydroxybutyrate nanoparticle for theranostic applications, J Mater Chem
phosphate composite scaffolds, Acta Biomater 8 (2012) 3446–3456. B 2 (2014) 3965–3971.
56 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

[611] C Utsunomia, T Saito, Matsumoto Ki, C Hori, T Isono, T Satoh, et al., Syn- J Groll, et al., Melt electrospinning onto cylinders: effects of rotational ve-
thesis of lactate (LA)-based poly(ester-urethane) using hydroxyl-terminated locity and collector diameter on morphology of tubular structures, Polym Int
LA-based oligomers from a microbial secretion system, J Polym Res 24 (2017) 64 (2015) 1086–1095.
167. [639] JN Haigh, Chuang Y-m, B Farrugia, R Hoogenboom, PD Dalton, TR Dargav-
[612] H Ramachandran, S Kannusamy, K-H Huong, R Mathava, AA Amirul, Blends of ille, Hierarchically Structured Porous Poly(2-oxazoline) Hydrogels, Macromol
Polyhydroxyalkanoates (PHAs), in: I Roy, PM Visakh (Eds.), Polyhydroxyalka- Rapid Commun 37 (2016) 93–99.
noate (PHA) based Blends, Composites and Nanocomposites. Thomas Graham [640] J Visser, FPW Melchels, JE Jeon, EM van Bussel, LS Kimpton, HM Byrne, et al.,
House, Science Park, Milton Road, Cambridge CB4 0WF, The Royal Society of Reinforcement of hydrogels using three-dimensionally printed microfibres,
Chemistry, UK, 2015, pp. 66–97. Nat Commun 6 (2015) 6933.
[613] MH Madkour, D Heinrich, MA Alghamdi, II Shabbaj, A Steinbüchel, PHA Re- [641] J Jensen, JHD Rölfing, DQ Svend Le, AA Kristiansen, JV Nygaard, LB Hokland,
covery from Biomass, Biomacromolecules 14 (2013) 2963–2972. et al., Surface-modified functionalized polycaprolactone scaffolds for bone re-
[614] E Leroy, I Petit, JL Audic, G Colomines, R Deterre, Rheological characterization pair: In vitro and in vivo experiments, J Biomed Mater Res A 102 (2014)
of a thermally unstable bioplastic in injection molding conditions, Polym De- 2993–3003.
grad Stab 97 (2012) 1915–1921. [642] HP Dang, T Shabab, A Shafiee, QC Peiffer, K Fox, N Tran, et al., 3D printed dual
[615] C-S Wu, H-T Liao, Y-X Cai, Characterisation, biodegradability and applica- macro-, microscale porous network as a tissue engineering scaffold with drug
tion of palm fibre-reinforced polyhydroxyalkanoate composites, Polym De- delivering function, Biofabrication 11 (2019) 035014.
grad Stab 140 (2017) 55–63. [643] F Chen, G Hochleitner, T Woodfield, J Groll, PD Dalton, BG Amsden, Additive
[616] C-S Wu, H-T Liao, characterization Fabrication, and application of Manufacturing of a Photo-Cross-Linkable Polymer via Direct Melt Electrospin-
polyester/wood flour composites, J Polym Eng 37 (2017) 689. ning Writing for Producing High Strength Structures, Biomacromolecules 17
[617] CS Wu, HT Liao, Interface design of environmentally friendly carbon nan- (2016) 208–214.
otube-filled polyester composites: Fabrication, characterisation, functionality [644] C Romagnoli, R Zonefrati, D Puppi, C Rosati, A Aldinucci, G Palmini, et al.,
and application, Express Polym Lett 11 (2017) 187–198. Human Adipose Tissue-Derived Stem Cells and a Poly(ε -Caprolactone) Scaf-
[618] H Tian, Z Tang, X Zhuang, X Chen, X Jing, Biodegradable synthetic polymers: fold Produced by Computer-Aided Wet Spinning for Bone Tissue Engineering,
Preparation, functionalization and biomedical application, Prog Polym Sci 37 Journal of Biomaterials and Tissue Engineering 7 (2017) 622–633.
(2012) 237–280. [645] Y He, CJ Tuck, E Prina, S Kilsby, SDR Christie, S Edmondson, et al., A new
[619] O Coulembier, P Degée, JL Hedrick, P Dubois, From controlled ring-opening photocrosslinkable polycaprolactone-based ink for three-dimensional inkjet
polymerization to biodegradable aliphatic polyester: Especially poly(β -malic printing, J Biomed Mater Res B Appl Biomater 105 (2017) 1645–1657.
acid) derivatives, Prog Polym Sci 31 (2006) 723–747. [646] MH Smith, CL Flanagan, JM Kemppainen, JA Sack, H Chung, S Das, et al.,
[620] D Puppi, F Chiellini, M Dash, E Chiellini, Biodegradable Polymers for Biomed- Computed tomography-based tissue-engineered scaffolds in craniomaxillofa-
ical Applications, in: GP Felton (Ed.), Biodegradable Polymers: Process- cial surgery, Int J Med Robot Comp 3 (2007) 207–216.
ing, Degradation & Applications, Nova Science Publishers, New York, 2011, [647] SJ Hollister, CL Flanagan, DA Zopf, RJ Morrison, H Nasser, JJ Patel, et al., De-
pp. 545–560. sign Control for Clinical Translation of 3D Printed Modular Scaffolds, Ann
[621] EY Teo, S-Y Ong, MS Khoon Chong, Z Zhang, J Lu, S Moochhala, et al., Poly- Biomed Eng 43 (2015) 774–786.
caprolactone-based fused deposition modeled mesh for delivery of antibacte- [648] RJ Morrison, HB Nasser, KN Kashlan, DA Zopf, DJ Milner, CL Flanangan, et al.,
rial agents to infected wounds, Biomaterials 32 (2011) 279–287. Co-culture of adipose-derived stem cells and chondrocytes on three-dimen-
[622] P Yilgor, G Yilmaz, MB Onal, I Solmaz, S Gundogdu, S Keskil, et al., An in vivo sionally printed bioscaffolds for craniofacial cartilage engineering, The Laryn-
study on the effect of scaffold geometry and growth factor release on the goscope 128 (2018) E251–E2E7.
healing of bone defects, J Tissue Eng Regener Med 7 (2013) 687–696. [649] DA Zopf, AG Mitsak, CL Flanagan, M Wheeler, GE Green, SJ Hollister, Com-
[623] J Kundu, J-H Shim, J Jang, S-W Kim, D-W Cho, An additive manufactur- puter aided-designed, 3-dimensionally printed porous tissue bioscaffolds for
ing-based PCL–alginate–chondrocyte bioprinted scaffold for cartilage tissue craniofacial soft tissue reconstruction, Otolaryngol Head Neck Surg 152 (2015)
engineering, J Tissue Eng Regener Med 9 (2015) 1286–1297. 57–62.
[624] F Pati, T-H Song, G Rijal, J Jang, SW Kim, D-W Cho, Ornamenting 3D printed [650] DA Zopf, CL Flanagan, AG Mitsak, JR Brennan, SJ Hollister, Pore architec-
scaffolds with cell-laid extracellular matrix for bone tissue regeneration, Bio- ture effects on chondrogenic potential of patient-specific 3-dimensionally
materials 37 (2015) 230–241. printed porous tissue bioscaffolds for auricular tissue engineering, Int J Pe-
[625] W Jiang, J Shi, W Li, K Sun, wettability Morphology, and mechanical prop- diatr Otorhinolaryngol 114 (2018) 170–174.
erties of polycaprolactone/hydroxyapatite composite scaffolds with intercon- [651] G Ciardelli, V Chiono, G Vozzi, M Pracella, A Ahluwalia, N Barbani, et al.,
nected pore structures fabricated by a mini-deposition system, Polym Eng Sci Blends of Poly-(ε -caprolactone) and Polysaccharides in Tissue Engineering
52 (2012) 2396–2402. Applications, Biomacromolecules 6 (2005) 1961–1976.
[626] L Shor, S Güçeri, R Chang, J Gordon, Q Kang, L Hartsock, et al., Precision ex- [652] Y Xia, P Zhou, X Cheng, Y Xie, C Liang, C Li, et al., Selective laser sintering fab-
truding deposition (PED) fabrication of polycaprolactone (PCL) scaffolds for rication of nano-hydroxyapatite/poly-ε -caprolactone scaffolds for bone tissue
bone tissue engineering, Biofabrication 1 (2009) 015003. engineering applications, Int J Nanomedicine 8 (2013) 4197–4213.
[627] K Lee, G Jin, CH Jang, W-K Jung, G Kim, Preparation and characterization of [653] Chen C-H, Lee M-Y, Shyu VB-H, Chen Y-C, Chen C-T, Chen J-P, Surface mod-
multi-layered poly(ε -caprolactone)/chitosan scaffolds fabricated with a com- ification of polycaprolactone scaffolds fabricated via selective laser sintering
bination of melt-plotting/in situ plasma treatment and a coating method for for cartilage tissue engineering, Mater Sci Eng, C, 40 (2014), pp. 389-97.
hard tissue regeneration, J Mater Chem B 1 (2013) 5831–5841. [654] L Elomaa, S Teixeira, R Hakala, H Korhonen, DW Grijpma, JV Seppälä, Prepa-
[628] M Domingos, F Chiellini, A Gloria, L Ambrosio, P Bartolo, E Chiellini, Effect ration of poly(ε -caprolactone)-based tissue engineering scaffolds by stere-
of process parameters on the morphological and mechanical properties of 3D olithography, Acta Biomater 7 (2011) 3850–3856.
Bioextruded poly(ε -caprolactone) scaffolds, Rapid Prototyp J 18 (2012) 56–67. [655] A Ronca, S Ronca, G Forte, S Zeppetelli, A Gloria, R De Santis, et al., Synthe-
[629] Fiedler T, Videira AC, Bártolo P, Strauch M, Murch GE, Ferreira JMF, On the sis and characterization of divinyl-fumarate poly-ε -caprolactone for scaffolds
mechanical properties of PLC–bioactive glass scaffolds fabricated via BioEx- with controlled architectures, J Tissue Eng Regener Med 12 (2018) e523–ee31.
trusion, Mater Sci Eng, C, 57 (2015), pp. 288-93. [656] L Elomaa, Y Kang, JV Seppälä, Y Yang, Biodegradable photocrosslinkable
[630] F Liu, W Wang, W Mirihanage, S Hinduja, A Bartolo PJ, plasma-assisted bioex- poly(depsipeptide-co-ε -caprolactone) for tissue engineering: Synthesis, char-
trusion system for tissue engineering, CIRP Annals 67 (2018) 229–232. acterization, and In vitro evaluation, J Polym Sci, Part A: Polym Chem 52
[631] A Martins, S Chung, AJ Pedro, RA Sousa, AP Marques, RL Reis, et al., Hierar- (2014) 3307–3315.
chical starch-based fibrous scaffold for bone tissue engineering applications, [657] D Drummer, D Rietzel, S Cifuentes-Cuéllar, Suitability of PLA/TCP for fused
J Tissue Eng Regener Med 3 (2009) 37–42. deposition modeling, Rapid Prototyp J 18 (2012) 500–507.
[632] H Yoon, S Ahn, G Kim, Three-Dimensional Polycaprolactone Hierarchical Scaf- [658] A Lanzotti, M Grasso, M Martorelli, G Staiano, The impact of process parame-
folds Supplemented with Natural Biomaterials to Enhance Mesenchymal Stem ters on mechanical properties of parts fabricated in PLA with an open-source
Cell Proliferation, Macromol Rapid Commun 30 (2009) 1632–1637. 3-D printer, Rapid Prototyp J 21 (2015) 604–617.
[633] H Lee, M Yeo, S Ahn, D-O Kang, CH Jang, H Lee, et al., Designed hybrid scaf- [659] A Liu, Xue G-h, M Sun, Shao H-f, Ma C-y, Q Gao, et al., 3D Printing Surgical
folds consisting of polycaprolactone microstrands and electrospun collagen– Implants at the clinic: A Experimental Study on Anterior Cruciate Ligament
nanofibers for bone tissue regeneration, J Biomed Mater Res B Appl Biomater Reconstruction, Sci Rep 6 (2016) 21704.
97B (2011) 263–270. [660] A Grémare, V Guduric, R Bareille, V Heroguez, S Latour, N L’Heureux, et al.,
[634] M Yeo, G Kim, Cell-printed hierarchical scaffolds consisting of micro-sized Characterization of printed PLA scaffolds for bone tissue engineering, J
polycaprolactone (PCL) and electrospun PCL nanofibers/cell-laden alginate Biomed Mater Res A 106 (2017) 887–894.
struts for tissue regeneration, J Mater Chem B 2 (2014) 314–324. [661] N Sandler, I Salmela, A Fallarero, A Rosling, M Khajeheian, R Kolakovic, et al.,
[635] H Yoon, G Kim, A three-dimensional polycaprolactone scaffold combined with Towards fabrication of 3D printed medical devices to prevent biofilm forma-
a drug delivery system consisting of electrospun nanofibers, J Pharm Sci 100 tion, Int J Pharm 459 (2014) 62–64.
(2011) 424–430. [662] JJ Water, A Bohr, J Boetker, J Aho, N Sandler, HM Nielsen, et al., Three-Di-
[636] G Hochleitner, T Jüngst, TD Brown, K Hahn, C Moseke, F Jakob, et al., Additive mensional Printing of Drug-Eluting Implants: Preparation of an Antimicrobial
manufacturing of scaffolds with sub-micron filaments via melt electrospin- Polylactide Feedstock Material, J Pharm Sci 104 (2015) 1099–1107.
ning writing, Biofabrication 7 (2015) 035002. [663] J Boetker, JJ Water, J Aho, L Arnfast, A Bohr, Rantanen J Modifying release
[637] TD Brown, A Slotosch, L Thibaudeau, A Taubenberger, D Loessner, C Vaquette, characteristics from 3D printed drug-eluting products, Eur J Pharm Sci 90
et al., Design and Fabrication of Tubular Scaffolds via Direct Writing in a Melt (2016) 47–52.
Electrospinning Mode, Biointerphases 7 (2012) 13. [664] J Kim, S McBride, B Tellis, P Alvarez-Urena, Y-H Song, DD Dean, et al.,
[638] T Jungst, ML Muerza-Cascante, TD Brown, M Standfest, DW Hutmacher,
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 57

Rapid-prototyped PLGA/β -TCP/hydroxyapatite nanocomposite scaffolds in a for fused deposition modelling, in: 7th World Biomaterials Congress, Curran
rabbit femoral defect model, Biofabrication 4 (2012) 025003. Associates, Inc., Sidney, Australia, 2004, p. 885.
[665] H Zhang, X Mao, D Zhao, W Jiang, Z Du, Q Li, et al., Three dimensional printed [690] A Haryńska, J Kucinska-Lipka, A Sulowska, I Gubanska, M Kostrzewa, H Janik,
polylactic acid-hydroxyapatite composite scaffolds for prefabricating vascular- Medical-Grade PCL Based Polyurethane System for FDM 3D Printing—Charac-
ized tissue engineered bone: An in vivo bioreactor model, Sci Rep 7 (2017) terization and Fabrication, Materials 12 (2019) 887.
15255. [691] A Güney, C Gardiner, A McCormack, J Malda, DW Grijpma, Thermoplastic
[666] J-H Shim, J-Y Won, S-J Sung, D-H Lim, W-S Yun, Y-C Jeon, et al., Comparative PCL-b-PEG-b-PCL and HDI Polyurethanes for Extrusion-Based 3D-Printing of
Efficacies of a 3D-Printed PCL/PLGA/β -TCP Membrane and a Titanium Mem- Tough Hydrogels, Bioengineering 5 (2018) 99.
brane for Guided Bone Regeneration in Beagle Dogs, Polymers 7 (2015). [692] S Camarero-Espinosa, C Tomasina, A Calore, L Moroni, Additive Manufactured,
[667] JY Won, CY Park, JH Bae, G Ahn, C Kim, DH Lim, et al., Evaluation of 3D Highly Resilient, Elastic and Biodegradable Poly(ester)urethane Scaffolds with
printed PCL/PLGA/β -TCP versus collagen membranes for guided bone regen- Chondroinductive Properties for Cartilage Tissue Engineering, Materials Today
eration in a beagle implant model, Biomed Mater 11 (2016) 055013. Bio (2020) 10 0 051.
[668] T-H Song, J Jang, Y-J Choi, J-H Shim, D-W Cho, 3D-Printed Drug/Cell Carrier [693] NJ Castro, WN Tan, C Shen, LG Zhang, Simulated Body Fluid Nucleation of
Enabling Effective Release of Cyclosporin a for Xenogeneic Cell-Based Ther- Three-Dimensional Printed Elastomeric Scaffolds for Enhanced Osteogenesis,
apy, Cell Transplant 24 (2015) 2513–2525. Tissue Eng Part A 22 (2016) 940–948.
[669] JD Kim, JS Choi, BS Kim, Y Chan Choi, YW Cho, Piezoelectric inkjet printing [694] A Agrawal, Rahbar N, Calvert PD, Strong fiber-reinforced hydrogel, Acta Bio-
of polymers: Stem cell patterning on polymer substrates, Polymer 51 (2010) mater 9 (2013) 5313–5318.
2147–2154. [695] J Perkins, Y Hong, S-H Ye, WR Wagner, S Desai, Direct writing of bio-func-
[670] Y Yan, Z Xiong, Y Hu, S Wang, R Zhang, C Zhang, Layered manufacturing of tional coatings for cardiovascular applications, J Biomed Mater Res A 102
tissue engineering scaffolds via multi-nozzle deposition, Mater Lett 57 (2003) (2014) 4290–4300.
2623–2628. [696] W Xu, X Wang, Y Yan, R Zhang, Rapid Prototyping of Polyurethane for the
[671] C Liu, Y Li, L Zhang, S Mi, Y Xu, W Sun, Development of a novel Creation of Vascular Systems, J Bioact Compatible Polym 23 (2008) 103–114.
low-temperature deposition machine using screw extrusion to fabricate [697] T Cui, Y Yan, R Zhang, L Liu, W Xu, X Wang, Rapid Prototyping of a Dou-
poly(l-lactide-co-glycolide) acid scaffolds, Proc Inst Mech Eng, Part H 228 ble-Layer Polyurethane–Collagen Conduit for Peripheral Nerve Regeneration,
(2014) 593–606. Tissue Eng Part C 15 (2008) 1–9.
[672] AV Mironov, AM Grigoryev, LI Krotova, NN Skaletsky, VK Popov, VI Sevas- [698] W Xiahong, C Tongkui, Y Yongnian, Renji Z Peroneal Nerve Regeneration Us-
tianov, 3D printing of PLGA scaffolds for tissue engineering, J Biomed Mater ing a Unique Bilayer Polyurethane-collagen Guide Conduit, J Bioact Compati-
Res A 105 (2017) 104–109. ble Polym 24 (2009) 109–127.
[673] WY Zhou, SH Lee, M Wang, WL Cheung, Selective Laser Sintering of Tis- [699] H Kai, Xiaohong W Rapid prototyping of tubular polyurethane and
sue Engineering Scaffolds Using Poly(L-Lactide) Microspheres, Key Eng Mater cell/hydrogel constructs, J Bioact Compatible Polym 26 (2011) 363–374.
334-335 (2007) 1225–1228. [700] Hsu S-h, Hung K-C, Y-Y Lin, C-H Su, H-Y Yeh, US Jeng, et al., Water-based
[674] WY Zhou, SH Lee, M Wang, WL Cheung, WY Ip, Selective laser sintering synthesis and processing of novel biodegradable elastomers for medical ap-
of porous tissue engineering scaffolds from poly(l-lactide)/carbonated hy- plications, J Mater Chem B 2 (2014) 5083–5092.
droxyapatite nanocomposite microspheres, J Mater Sci Mater Med 19 (2008) [701] F-Y Hsieh, H-H Lin, Hsu S-h, 3D bioprinting of neural stem cell-laden ther-
2535–2540. moresponsive biodegradable polyurethane hydrogel and potential in central
[675] Zhou WY, Wang M, Cheung WL, Ip WY, Selective laser sintering of poly(l- nervous system repair, Biomaterials 71 (2015) 48–57.
lactide)/carbonated hydroxyapatite nanocomposite porous scaffolds for bone [702] Y-C Tsai, S Li, S-G Hu, W-C Chang, US Jeng, S-h Hsu, Synthesis of thermore-
tissue engineering, Tissue Eng, ( 2010 ), pp. 179-204. sponsive amphiphilic polyurethane gel as a new cell printing material near
[676] S Hoeges, M Lindner, H Fischer, W Meiners, K Wissenbach, Manufacturing of body temperature, ACS Appl Mater Interfaces 7 (2015) 27613–27623.
bone substitute implants using Selective Laser Melting, in: J Vander Sloten, [703] H-H Lin, F-Y Hsieh, C-S Tseng, Hsu S-h, Preparation and characterization of a
P Verdonck, M Nyssen, J Haueisen (Eds.), 4th European Conference of the In- biodegradable polyurethane hydrogel and the hybrid gel with soy protein for
ternational Federation for Medical and Biological Engineering, Springer Berlin 3D cell-laden bioprinting, J Mater Chem B 4 (2016) 6694–6705.
Heidelberg, Berlin, Heidelberg, 2009, pp. 2230–2234. [704] C Zhang, K Zhao, T Hu, X Cui, N Brown, T Boland, Loading dependent swelling
[677] JM Kanczler, S-H Mirmalek-Sani, NA Hanley, AL Ivanov, JJA Barry, C Upton, and release properties of novel biodegradable, elastic and environmental
et al., Biocompatibility and osteogenic potential of human fetal femur-derived stimuli-sensitive polyurethanes, J Controlled Release 131 (2008) 128–136.
cells on surface selective laser sintered scaffolds, Acta Biomater 5 (2009) [705] C Zhang, X Wen, NR Vyavahare, T Boland, Synthesis and characterization
2063–2071. of biodegradable elastomeric polyurethane scaffolds fabricated by the inkjet
[678] TB Bukharova, EN Antonov, VK Popov, TK Fatkhudinov, AV Popova, AV Volkov, technique, Biomaterials 29 (2008) 3781–3791.
et al., Biocompatibility of Tissue Engineering Constructions from Porous Poly- [706] O Guillaume, MA Geven, DW Grijpma, TT Tang, L Qin, YX Lai, et al.,
lactide Carriers Obtained by the Method of Selective Laser Sintering and Bone Poly(trimethylene carbonate) and nano-hydroxyapatite porous scaffolds man-
Marrow-Derived Multipotent Stromal Cells, Bull Exp Biol Med 149 (2010) ufactured by stereolithography, Polym Adv Technol 28 (2017) 1219–1225.
148–153. [707] KEG Dienel, B van Bochove, JV Seppälä, Additive Manufacturing of Bioactive
[679] WS Koegler, LG Griffith, Osteoblast response to PLGA tissue engineering scaf- Poly(trimethylene carbonate)/β -Tricalcium Phosphate Composites for Bone
folds with PEO modified surface chemistries and demonstration of patterned Regeneration, Biomacromolecules 21 (2020) 366–375.
cell response, Biomaterials 25 (2004) 2819–2830. [708] K Fukushima, Poly(trimethylene carbonate)-based polymers engineered for
[680] W Wu, Q Zheng, X Guo, W Huang, The controlled-releasing drug implant biodegradable functional biomaterials, Biomater Sci 4 (2016) 9–24.
based on the three dimensional printing technology: Fabrication and prop- [709] A Ahlinder, T Fuoco, A Finne-Wistrand, Medical grade polylactide,
erties of drug releasing in vivo, J Wuhan Univ Technol 24 (2009) 977. copolyesters and polydioxanone: Rheological properties and melt stabil-
[681] FPW Melchels, B Tonnarelli, AL Olivares, I Martin, D Lacroix, J Feijen, et al., ity, Polym Test 72 (2018) 214–222.
The influence of the scaffold design on the distribution of adhering cells after [710] I Salaoru, Z Zhou, P Morris, GJ Gibbons, Inkjet printing of polyvinyl alco-
perfusion cell seeding, Biomaterials 32 (2011) 2878–2884. hol multilayers for additive manufacturing applications, J Appl Polym Sci 133
[682] S Tanodekaew, S Channasanon, P Kaewkong, P Uppanan, PLA-HA Scaffolds: (2016) 43572.
Preparation and Bioactivity, Procedia Eng 59 (2013) 144–149. [711] D Rigotti, L Fambri, A Pegoretti, Polyvinyl alcohol reinforced with carbon
[683] A Ronca, L Ambrosio, DW Grijpma, Preparation of designed nanotubes for fused deposition modeling, J Reinf Plast Compos 37 (2018)
poly(d,l-lactide)/nanosized hydroxyapatite composite structures by stere- 716–727.
olithography, Acta Biomater 9 (2013) 5989–5996. [712] A Cataldi, D Rigotti, VDH Nguyen, A Pegoretti, Polyvinyl alcohol reinforced
[684] J-W Choi, R Wicker, S-H Lee, K-H Choi, C-S Ha, I Chung, Fabrication of 3D with crystalline nanocellulose for 3D printing application, Materials Today
biocompatible/biodegradable micro-scaffolds using dynamic mask projection Communications 15 (2018) 236–244.
microstereolithography, J Mater Process Technol 209 (2009) 5494–5503. [713] H Sun, L Mei, C Song, X Cui, P Wang, The in vivo degradation, absorption and
[685] M Dadsetan, T Guda, MB Runge, D Mijares, RZ LeGeros, JP LeGeros, et al., Ef- excretion of PCL-based implant, Biomaterials 27 (2006) 1735–1740.
fect of calcium phosphate coating and rhBMP-2 on bone regeneration in rab- [714] M Bartnikowski, TR Dargaville, S Ivanovski, DW Hutmacher, Degradation
bit calvaria using poly(propylene fumarate) scaffolds, Acta Biomater 18 (2015) mechanisms of polycaprolactone in the context of chemistry, geometry and
9–20. environment, Prog Polym Sci 96 (2019) 1–20.
[686] V Luangphakdy, E Walker, K Shinohara, H Pan, T Hefferan, TW Bauer, et al., [715] WP Ye, FS Du, WH Jin, JY Yang, Y Xu, In vitro degradation of
Evaluation of osteoconductive scaffolds in the canine femoral multi-defect poly(caprolactone), poly(lactide) and their block copolymers: influence of
model, Tissue Eng Part A 19 (2013) 634–648. composition, temperature and morphology, React Funct Polym 32 (1997)
[687] JW Lee, KS Kang, SH Lee, J-Y Kim, B-K Lee, D-W Cho, Bone regener- 161–168.
ation using a microstereolithography-produced customized poly(propylene [716] D Gupta, J Venugopal, S Mitra, VR Giri Dev, S Ramakrishna, Nanostructured
fumarate)/diethyl fumarate photopolymer 3D scaffold incorporating BMP-2 biocomposite substrates by electrospinning and electrospraying for the min-
loaded PLGA microspheres, Biomaterials 32 (2011) 744–752. eralization of osteoblasts, Biomaterials 30 (2009) 2085–2094.
[688] JW Lee, K-J Kim, KS Kang, S Chen, J-W Rhie, D-W Cho, Development of a bone [717] SY Chew, R Mi, A Hoke, KW Leong, Aligned Protein-Polymer Composite Fibers
reconstruction technique using a solid free-form fabrication (SFF)-based drug Enhance Nerve Regeneration: A Potential Tissue-Engineering Platform, Adv
releasing scaffold and adipose-derived stem cells, J Biomed Mater Res A 101A Funct Mater 17 (2007) 1288–1296.
(2013) 1865–1875. [718] A-C Albertsson, IK Varma, in: Aliphatic Polyesters: Synthesis, Properties
[689] T Moore, R Adhikari, P Barton, PA Gunatillake, Biodegradable polyurethane and Applications. Degradable Aliphatic Polyesters, Springer Berlin Heidelberg,
Berlin, Heidelberg, 2002, pp. 1–40.
58 D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700

[719] E Malikmammadov, TE Tanir, A Kiziltay, V Hasirci, N Hasirci, PCL and spinning and Its Technologization in Tissue Engineering, Tissue Eng Part B 21
PCL-based materials in biomedical applications, J Biomater Sci Polym Ed 29 (2014) 187–202.
(2018) 863–893. [747] AS Les, RG Ohye, AG Filbrun, M Ghadimi Mahani, CL Flanagan, RC Daniels,
[720] A Cipitria, A Skelton, TR Dargaville, PD Dalton, DW Hutmacher, Design, fab- et al., 3D-printed, externally-implanted, bioresorbable airway splints for se-
rication and characterization of PCL electrospun scaffolds—a review, J Mater vere tracheobronchomalacia, The Laryngoscope 0 (2019).
Chem 21 (2011) 9419–9453. [748] R Datta, M Henry, Lactic acid: recent advances in products, processes and
[721] D Puppi, AM Piras, F Chiellini, E Chiellini, A Martins, IB Leonor, et al., Opti- technologies — a review, J Chem Technol Biotechnol 81 (2006) 1119–1129.
mized electro- and wet-spinning techniques for the production of polymeric [749] DM Bigg, Polylactide copolymers: Effect of copolymer ratio and end capping
fibrous scaffolds loaded with bisphosphonate and hydroxyapatite, J Tissue on their properties, Adv Polym Tech 24 (2005) 69–82.
Eng Regener Med 5 (2011) 253–263. [750] M Rabnawaz, I Wyman, R Auras, S Cheng, A roadmap towards green pack-
[722] RS Bezwada, DD Jamiolkowski, I-Y Lee, V Agarwal, J Persivale, S Trenka-Ben- aging: the current status and future outlook for polyesters in the packaging
thin, et al., Monocryl® suture, a new ultra-pliable absorbable monofilament industry, Green Chemistry 19 (2017) 4737–4753.
suture, Biomaterials 16 (1995) 1141–1148. [751] LS Nair, CT Laurencin, Polymers as Biomaterials for Tissue Engineering and
[723] PD Darney, SE Monroe, CM Klaisle, Alvarado A Clinical evaluation of the Controlled Drug Delivery, Adv Biochem Eng/Biotechnol 102 (2006) 47–90.
Capronor contraceptive implant: Preliminary report, Am J Obstet Gynecol 160 [752] S Doppalapudi, A Jain, W Khan, AJ Domb, Biodegradable polymers—an
(1989) 1292–1295. overview, Polym Adv Technol 25 (2014) 427–435.
[724] D Rohner, DW Hutmacher, TK Cheng, M Oberholzer, Hammer B In vivo ef- [753] RD Alexy, DS Levi, Materials and Manufacturing Technologies Available for
ficacy of bone-marrow-coated polycaprolactone scaffolds for the reconstruc- Production of a Pediatric Bioabsorbable Stent, Biomed Res Int (2013) (2013)
tion of orbital defects in the pig, J Biomed Mater Res B Appl Biomater 66B 11.
(2003) 574–580. [754] YH An, SK Woolf, RJ Friedman, Pre-clinical in vivo evaluation of orthopaedic
[725] JT Schantz, TC Lim, C Ning, SH Teoh, KC Tan, SC Wang, et al., Cranioplasty bioabsorbable devices, Biomaterials 21 (20 0 0) 2635–2652.
after trephination using a novel biodegradable burr hole cover: technical case [755] FA Barber, WD Dockery, Long-Term Absorption of Poly-L-Lactic Acid Interfer-
report, Neurosurgery 58 (2006) ONS–E176 discussion ONS-E. ence Screws, Arthroscopy 22 (2006) 820–826.
[726] http://www.osteopore.com; 2019 [756] WH Warden, D Chooljian, DW Jackson, Ten-Year Magnetic Resonance Imaging
[727] SW Low, YJ Ng, TT Yeo, N Chou, Use of Osteoplug polycaprolactone implants Follow-Up of Bioabsorbable Poly-L-Lactic Acid Interference Screws After Ante-
as novel burr-hole covers, Singapore Med J 50 (2009) 777–780. rior Cruciate Ligament Reconstruction, Arthroscopy 24 (2008) 370.e1–370.e3.
[728] L Teo, SH Teoh, Y Liu, L Lim, B Tan, J-T Schantz, et al., A Novel Bioresorbable [757] K Rezwan, QZ Chen, JJ Blaker, AR Boccaccini, Biodegradable and bioactive
Implant for Repair of Orbital Floor Fractures, Orbit 34 (2015) 192–200. porous polymer/inorganic composite scaffolds for bone tissue engineering,
[729] T Goh Bee, Y Teh Luan, P Tan Danny Ben, Z Zhang, Teoh Swee H Novel 3D Biomaterials 27 (2006) 3413–3431.
polycaprolactone scaffold for ridge preservation – a pilot randomised con- [758] EJ Bergsma, FR Rozema, RR Bos, WC Debruijn, Foreign body reaction to re-
trolled clinical trial, Clin Oral Implants Res 26 (2015) 271–277. sorbable poly(L-lactic) bone plates and screws used for the fixation of unsta-
[730] PS Wiggenhauser, DF Müller, FPW Melchels, JT Egaña, K Storck, H Mayer, ble zygomatic fractures, J Oral Maxillofac Surg 51 (1993) 666–670.
et al., Engineering of vascularized adipose constructs, Cell Tissue Res (2011) [759] C Martin, H Winet, JY Bao, Acidity near eroding polylactide-polyglycolide
1–11. in vitro and in vivo in rabbit tibial bone chambers, Biomaterials 17 (1996)
[731] I Zein, DW Hutmacher, KC Tan, SH Teoh, Fused deposition modeling of novel 2373–2380.
scaffold architectures for tissue engineering applications, Biomaterials 23 [760] X-L Deng, G Sui, M-L Zhao, G-Q Chen, X-P Yang, Poly(L-lactic
(2002) 1169–1185. acid)/hydroxyapatite hybrid nanofibrous scaffolds prepared by electro-
[732] SA Abbah, CXL Lam, DW Hutmacher, JCH Goh, H-K Wong, Biological perfor- spinning, J Biomater Sci Polym Ed 18 (2007) 117–130.
mance of a polycaprolactone-based scaffold used as fusion cage device in a [761] Y-F Chou, JCY Dunn, BM Wu, In vitro response of MC3T3-E1 preosteoblasts
large animal model of spinal reconstructive surgery, Biomaterials 30 (2009) within three-dimensional apatite-coated PLGA scaffolds, J Biomed Mater Res
5086–5093. B Appl Biomater 75B (2005) 81–90.
[733] S-J Kim, M-R Kim, J-S Oh, I Han, S-W Shin, Effects of Polycaprolactone-Tri- [762] AS Dunn, PG Campbell, KG Marra, The influence of polymer blend composi-
calcium Phosphate, Recombinant Human Bone Morphogenetic Protein-2 and tion on the degradation of polymer/hydroxyapatite biomaterials, J Mater Sci
Dog Mesenchymal Stem Cells on Bone Formation: Pilot Study in Dogs, Yonsei Mater Med 12 (2001) 673–677.
Med J 50 (2009) 825–831. [763] W Heidemann, S Jeschkeit, K Ruffieux, JH Fischer, M Wagner, G Kruger, et al.,
[734] CXF Lam, DW Hutmacher, J-T Schantz, MA Woodruff, SH Teoh, Evaluation of Degradation of poly(d,l)lactide implants with or without addition of calci-
polycaprolactone scaffold degradation for 6 months in vitro and in vivo, J umphosphates in vivo, Biomaterials 22 (2001) 2371–2381.
Biomed Mater Res A 90A (2009) 906–919. [764] MS Rahman, Way S, Polyglycolic acid surgical sutures ingynaecological
[735] AA Sawyer, SJ Song, E Susanto, P Chuan, CXF Lam, MA Woodruff, et al., The surgery, BJOG 79 (1972) 849–851.
stimulation of healing within a rat calvarial defect by mPCL–TCP/collagen [765] AE Burns, Biofix fixation techniques and results in foot surgery, J Foot Ankle
scaffolds loaded with rhBMP-2, Biomaterials 30 (2009) 2479–2488. Surg 34 (1995) 276–282.
[736] B Rai, JL Lin, ZXH Lim, RE Guldberg, DW Hutmacher, SM Cool, Differences be- [766] J Otto, M Binnebösel, S Pietsch, M Anurov, S Titkova, AP Öttinger, et al.,
tween in vitro viability and differentiation and in vivo bone-forming efficacy Large-Pore PDS Mesh Compared to Small-Pore PG Mesh, J Invest Surg 23
of human mesenchymal stem cells cultured on PCL-TCP scaffolds, Biomateri- (2010) 190–196.
als 31 (2010) 7960–7970. [767] S Knecht, C Erggelet, M Endres, M Sittinger, C Kaps, E Stüssi, Mechanical
[737] ST Ho, DW Hutmacher, AK Ekaputra, D Hitendra, JH Hui, The evaluation of a testing of fixation techniques for scaffold-based tissue-engineered grafts, J
biphasic osteochondral implant coupled with an electrospun membrane in a Biomed Mater Res B Appl Biomater 83B (2007) 50–57.
large animal model, Tissue Eng Part A 16 (2010) 1123–1141. [768] L Wang, NH Dormer, LF Bonewald, MS Detamore, Osteogenic Differentiation
[738] A Yeo, WJ Wong, S-H Teoh, Surface modification of PCL-TCP scaffolds in rab- of Human Umbilical Cord Mesenchymal Stromal Cells in Polyglycolic Acid
bit calvaria defects: Evaluation of scaffold degradation profile, biomechani- Scaffolds, Tissue Eng Part A 16 (2010) 1937–1948.
cal properties and bone healing patterns, J Biomed Mater Res A 93A (2010) [769] C Erggelet, K Neumann, M Endres, K Haberstroh, M Sittinger, C Kaps, Re-
1358–1367. generation of ovine articular cartilage defects by cell-free polymer-based im-
[739] FA Probst, DW Hutmacher, DF Muller, HG Machens, JT Schantz, [Calvarial re- plants, Biomaterials 28 (2007) 5570–5580.
construction by customized bioactive implant], Handchir Mikrochir Plast Chir [770] N Mahmoudifar, PM Doran, Chondrogenic differentiation of human adi-
42 (2010) 369–373. pose-derived stem cells in polyglycolic acid mesh scaffolds under dynamic
[740] B Rai, ME Oest, KM Dupont, KH Ho, SH Teoh, RE Guldberg, Combination of culture conditions, Biomaterials 31 (2010) 3858–3867.
platelet-rich plasma with polycaprolactone-tricalcium phosphate scaffolds for [771] L Xu, D Cao, W Liu, G Zhou, WJ Zhang, Y Cao, In vivo engineering of a func-
segmental bone defect repair, J Biomed Mater Res A 81A (2007) 888–899. tional tendon sheath in a hen model, Biomaterials 31 (2010) 3894–3902.
[741] JC Reichert, A Cipitria, DR Epari, S Saifzadeh, P Krishnakanth, A Berner, et al., [772] C Martins, F Sousa, F Araújo, B Sarmento, Functionalizing PLGA and PLGA
A Tissue Engineering Solution for Segmental Defect Regeneration in Load- Derivatives for Drug Delivery and Tissue Regeneration Applications, Adv
-Bearing Long Bones, Sci Transl Med 4 (2012) 141ra93. Healthcare Mater 7 (2018) 1701035.
[742] A Cipitria, W Wagermaier, P Zaslansky, H Schell, JC Reichert, P Fratzl, et al., [773] D Puppi, AM Piras, N Detta, D Dinucci, F Chiellini, Poly(lactic-co-glycolic acid)
BMP delivery complements the guiding effect of scaffold architecture without electrospun fibrous meshes for the controlled release of retinoic acid, Acta
altering bone microstructure in critical-sized long bone defects: A multiscale Biomater 6 (2010) 1258–1268.
analysis, Acta Biomater 23 (2015) 282–294. [774] F Danhier, E Ansorena, JM Silva, R Coco, A Le Breton, V Préat, PL-
[743] J Holländer, N Genina, H Jukarainen, M Khajeheian, A Rosling, E Mäkilä, et al., GA-based nanoparticles, An overview of biomedical applications, J Controlled
Three-Dimensional Printed PCL-Based Implantable Prototypes of Medical De- Release 161 (2012) 505–522.
vices for Controlled Drug Delivery, J Pharm Sci 105 (2016) 2665–2676. [775] P Gentile, V Chiono, I Carmagnola, VP Hatton, An Overview of
[744] PF Costa, C Vaquette, Q Zhang, RL Reis, S Ivanovski, DW Hutmacher, Advanced Poly(lactic-co-glycolic) Acid (PLGA)-Based Biomaterials for Bone Tissue
tissue engineering scaffold design for regeneration of the complex hierarchi- Engineering, Int J Mol Sci 15 (2014).
cal periodontal structure, J Clin Periodontol 41 (2014) 283–294. [776] BD Ulery, LS Nair, CT Laurencin, Biomedical Applications of Biodegradable
[745] E Jeon June, C Vaquette, C Theodoropoulos, J Klein Travis, W Hutmacher Diet- Polymers, J Polym Sci, Part B: Polym Phys 49 (2011) 832–864.
mar, Multiphasic construct studied in an ectopic osteochondral defect model, [777] BM Tymrak, M Kreiger, JM Pearce, Mechanical properties of components fab-
J Royal Soc Interface 11 (2014) 20140184. ricated with open-source 3-D printers under realistic environmental condi-
[746] ML Muerza-Cascante, D Haylock, DW Hutmacher, PD Dalton, Melt Electro- tions, Mater Des 58 (2014) 242–246.
D. Puppi and F. Chiellini / Applied Materials Today 20 (2020) 100700 59

[778] CAP Joziasse, H Veenstra, DW Grijpma, AJ Pennings, On the chain stiffness of marate)/poly(dl-lactic-co-glycolic acid) composite scaffolds, Biomaterials 26
poly(lactide)s, Macromol Chem Phys 197 (1996) 2219–2229. (2005) 3215–3225.
[779] JR Dorgan, JS Williams, DN Lewis, Melt rheology of poly(lactic acid): Entan- [806] RA Dilla, CMM Motta, SR Snyder, JA Wilson, C Wesdemiotis, ML Becker, Syn-
glement and chain architecture effects, J Rheol 43 (1999) 1141–1155. thesis and 3D Printing of PEG–Poly(propylene fumarate) Diblock and Triblock
[780] DN Heo, NJ Castro, S-J Lee, H Noh, W Zhu, LG Zhang, Enhanced bone tissue Copolymer Hydrogels, ACS Macro Lett 7 (2018) 1254–1260.
regeneration using a 3D printed microstructure incorporated with a hybrid [807] J Rychlý, A Lattuati-Derieux, B Lavédrine, L Matisová-Rychlá, M Malíková,
nano hydrogel, Nanoscale 9 (2017) 5055–5062. K Csomorová, et al., Assessing the progress of degradation in polyurethanes
[781] S Yuan, F Shen, CK Chua, K Zhou, Polymeric composites for powder-based by chemiluminescence and thermal analysis. II. Flexible polyether- and
additive manufacturing: Materials and applications, Prog Polym Sci 91 (2019) polyester-type polyurethane foams, Polym Degrad Stab 96 (2011) 462–469.
141–168. [808] DG Thompson, JC Osborn, EM Kober, JR Schoonover, Effects of hydrolysis-in-
[782] KF Leong, CK Chua, WS Gui, Verani Building Porous Biopolymeric Microstruc- duced molecular weight changes on the phase separation of a polyester
tures for Controlled Drug Delivery Devices Using Selective Laser Sintering, Int polyurethane, Polym Degrad Stab 91 (2006) 3360–3370.
J Adv Manuf Technol 31 (2006) 483–489. [809] JM Anderson, A Rodriguez, DT Chang, Foreign body reaction to biomaterials,
[783] SH Lim, H Kathuria, JJY Tan, L Kang, 3D printed drug delivery and testing Semin Immunol 20 (2008) 86–100.
systems — a passing fad or the future? Adv Drug Del Rev 132 (2018) 139–168. [810] P Penczek, KC Frisch, B Szczepaniak, E Rudnik, Synthesis and properties of
[784] G Wu, W Wu, Q Zheng, J Li, J Zhou, Z Hu, Experimental study of PLLA/INH liquid crystalline polyurethanes, J Polym Sci, Part A: Polym Chem 31 (1993)
slow release implant fabricated by three dimensional printing technique and 1211–1220.
drug release characteristics in vitro, BioMedical Engineering OnLine 13 (2014) [811] H Honarkar, Waterborne polyurethanes: A review, J Dispersion Sci Technol 39
97. (2018) 507–516.
[785] FK Kasper, K Tanahashi, JP Fisher, AG Mikos, Synthesis of poly(propylene fu- [812] Z Li, J Li, Control of Hyperbranched Structure of Polycaprolac-
marate), Nat Protoc 4 (2009) 518–525. tone/Poly(ethylene glycol) Polyurethane Block Copolymers by Glycerol
[786] Yaszemski MJ, Payne RG, Mikos AG, Poly(propylene fumarate). US: No. and Their Hydrogels for Potential Cell Delivery, The Journal of Physical
5,733,951.; 31 Mar 1998. Chemistry B 117 (2013) 14763–14774.
[787] Domb AJ, Poly(propylene glycol fumarate) compositions for biomedical appli- [813] SL Cooper, AV Tobolsky, Properties of linear elastomeric polyurethanes, J Appl
cations. US patent 4888413; 1989. Polym Sci 10 (1966) 1837–1844.
[788] Y Luo, CK Dolder, JM Walker, R Mishra, D Dean, ML Becker, Synthesis and Bi- [814] PN Lan, S Corneillie, E Schacht, M Davies, A Shard, Synthesis and charac-
ological Evaluation of Well-Defined Poly(propylene fumarate) Oligomers and terization of segmented polyurethanes based on amphiphilic polyether diols,
Their Use in 3D Printed Scaffolds, Biomacromolecules 17 (2016) 690–697. Biomaterials 17 (1996) 2273–2280.
[789] SJ Peter, P Kim, AW Yasko, MJ Yaszemski, AG Mikos, Crosslinking charac- [815] P Alves, P Ferreira, H Gil, Biomedical Polyurethane-Based Materials,
teristics of an injectable poly(propylene fumarate)/β -tricalcium phosphate Polyurethane: Properties, Structure and Applications, Nova Publishers, New
paste and mechanical properties of the crosslinked composite for use as a York, 2012.
biodegradable bone cement, J Biomed Mater Res 44 (1999) 314–321. [816] J Joseph, RM Patel, A Wenham, JR Smith, Biomedical applications of
[790] EL Hedberg, HC Kroese-Deutman, CK Shih, RS Crowther, DH Carney, polyurethane materials and coatings, Transactions of the IMF 96 (2018)
AG Mikos, et al., In vivo degradation of porous poly(propylene fu- 121–129.
marate)/poly(DL-lactic-co-glycolic acid) composite scaffolds, Biomaterials 26 [817] SA Guelcher, Biodegradable Polyurethanes, Synthesis and Applications in Re-
(2005) 4616–4623. generative Medicine, Tissue Eng Part B 14 (2008) 3–17.
[791] MD Timmer, H Shin, RA Horch, CG Ambrose, AG Mikos, In Vitro Cytotox- [818] L Tatai, TG Moore, R Adhikari, F Malherbe, R Jayasekara, I Griffiths, et al.,
icity of Injectable and Biodegradable Poly(propylene fumarate)-Based Net- Thermoplastic biodegradable polyurethanes: The effect of chain extender
works: Unreacted Macromers, Cross-Linked Networks, and Degradation Prod- structure on properties and in-vitro degradation, Biomaterials 28 (2007)
ucts, Biomacromolecules 4 (2003) 1026–1033. 5407–5417.
[792] DD Frazier, VK Lathi, TN Gerhart, WC Hayes, Ex vivo degradation of a [819] PC Caracciolo, NJ Lores, GA Abraham, Chapter 8 - Polyurethane-based struc-
poly(propylene glycol-fumarate) biodegradable particulate composite bone tures obtained by additive manufacturing technologies, in: A-M Holban,
cement, J Biomed Mater Res 35 (1997) 383–389. AM Grumezescu (Eds.), Materials for Biomedical Engineering, Elsevier, 2019,
[793] JP Fisher, D Dean, AG Mikos, Photocrosslinking characteristics and mechanical pp. 235–258.
properties of diethyl fumarate/poly(propylene fumarate) biomaterials, Bioma- [820] KC Hung, CS Tseng, SH Hsu, 5 - 3D printing of polyurethane biomaterials, in:
terials 23 (2002) 4333–4343. SL Cooper, J Guan (Eds.), Advances in Polyurethane Biomaterials, Woodhead
[794] S Pal, Mechanical Properties of Biological Materials, in: S Pal (Ed.), Design of Publishing, 2016, pp. 149–170.
Artificial Human Joints & Organs, Springer US, Boston, MA, 2014, pp. 23–40. [821] TK Merceron, M Burt, Y-J Seol, H-W Kang, SJ Lee, JJ Yoo, et al., A 3D bio-
[795] X Liu, Ii AL Miller, BE Waletzki, MJ Yaszemski, L Lu, Novel biodegradable printed complex structure for engineering the muscle–tendon unit, Biofabri-
poly(propylene fumarate)-co-poly(l-lactic acid) porous scaffolds fabricated cation 7 (2015) 035003.
by phase separation for tissue engineering applications, RSC Adv 5 (2015) [822] K-C Hung, C-S Tseng, Hsu S-h, Synthesis and 3D Printing of Biodegradable
21301–21309. Polyurethane Elastomer by a Water-Based Process for Cartilage Tissue Engi-
[796] MJ Yaszemski, RG Payne, WC Hayes, RS Langer, TB Aufdemorte, AG Mikos, neering Applications, Adv Healthcare Mater 3 (2014) 1578–1587.
The Ingrowth of New Bone Tissue and Initial Mechanical Properties of a De- [823] K-C Hung, C-S Tseng, L-G Dai, Hsu S-h, Water-based polyurethane 3D printed
grading Polymeric Composite Scaffold, Tissue Eng 1 (1995) 41–52. scaffolds with controlled release function for customized cartilage tissue en-
[797] JWM Vehof, JP Fisher, D Dean, J-PCM van der Waerden, PHM Spauwen, gineering, Biomaterials 83 (2016) 156–168.
AG Mikos, et al., Bone formation in transforming growth factor β -1-coated [824] M Tamura, K Matsuda, Y Nakagawa, K Tomishige, Ring-opening polymeriza-
porous poly(propylene fumarate) scaffolds, J Biomed Mater Res 60 (2002) tion of trimethylene carbonate to poly(trimethylene carbonate) diol over a
241–251. heterogeneous high-temperature calcined CeO2 catalyst, Chem Commun 54
[798] MD Timmer, CG Ambrose, AG Mikos, In vitro degradation of polymeric (2018) 14017–14020.
networks of poly(propylene fumarate) and the crosslinking macromer [825] Z Zhang, R Kuijer, SK Bulstra, DW Grijpma, J Feijen, The in vivo and in vitro
poly(propylene fumarate)-diacrylate, Biomaterials 24 (2003) 571–577. degradation behavior of poly(trimethylene carbonate), Biomaterials 27 (2006)
[799] SJ Peter, JA Nolley, MS Widmer, JE Merwin, MJ Yaszemski, AW Yasko, et al., 1741–1748.
In Vitro Degradation of a Poly(Propylene Fumarate)/ β -Tricalcium Phosphate [826] I Engelberg, J Kohn, Physico-mechanical properties of degradable polymers
Composite Orthopaedic Scaffold, Tissue Eng 3 (1997) 207–215. used in medical applications: A comparative study, Biomaterials 12 (1991)
[800] GB Kharas, M Kamenetsky, J Simantirakis, KC Beinlich, A-MT Rizzo, GA Cay- 292–304.
wood, et al., Synthesis and characterization of fumarate-based polyesters for [827] RP Brannigan, AP Dove, Synthesis, properties and biomedical applications of
use in bioresorbable bone cement composites, J Appl Polym Sci 66 (1997) hydrolytically degradable materials based on aliphatic polyesters and polycar-
1123–1137. bonates, Biomater Sci 5 (2017) 9–21.
[801] Z-Y Cai, D-A Yang, N Zhang, C-G Ji, L Zhu, T Zhang, Poly(propylene fu- [828] SBG Blanquer, AWH Gebraad, S Miettinen, AA Poot, DW Grijpma, SP Haimi,
marate)/(calcium sulphate/[beta]-tricalcium phosphate) composites: Prepa- Differentiation of adipose stem cells seeded towards annulus fibrosus cells on
ration, characterization and in vitro degradation, Acta Biomater 5 (2009) a designed poly(trimethylene carbonate) scaffold prepared by stereolithogra-
628–635. phy, J Tissue Eng Regener Med 11 (2017) 2752–2762.
[802] K-W Lee, S Wang, MJ Yaszemski, L Lu, Physical properties and cellular re- [829] E Chiellini, A Corti, S D’Antone, R Solaro, Biodegradation of poly (vinyl alco-
sponses to crosslinkable poly(propylene fumarate)/hydroxyapatite nanocom- hol) based materials, Prog Polym Sci 28 (2003) 963–1014.
posites, Biomaterials 29 (2008) 2839–2848. [830] S-F Chong, AAA Smith, AN Zelikin, Microstructured, Functional PVA Hydro-
[803] EL Hedberg, A Tang, RS Crowther, DH Carney, AG Mikos, Controlled release of gels through Bioconjugation with Oligopeptides under Physiological Condi-
an osteogenic peptide from injectable biodegradable polymeric composites, J tions, Small 9 (2013) 942–950.
Controlled Release 84 (2002) 137–150. [831] N Ben Halima, Poly(vinyl alcohol): review of its promising applications and
[804] EL Hedberg, HC Kroese-Deutman, CK Shih, RS Crowther, DH Carney, insights into biodegradation, RSC Adv 6 (2016) 39823–39832.
AG Mikos, et al., Effect of varied release kinetics of the osteogenic thrombin [832] B Cerroni, R Cicconi, L Oddo, M Scimeca, R Bonfiglio, R Bernardini, et al.,
peptide TP508 from biodegradable, polymeric scaffolds on bone formation in In vivo biological fate of poly(vinylalcohol) microbubbles in mice, Heliyon 4
vivo, J Biomed Mater Res A 72A (2005) 343–353. (2018) pp. e00770-e.
[805] EL Hedberg, CK Shih, JJ Lemoine, MD Timmer, MA K. Liebschner, [833] A Kumar, Han SS PVA-based hydrogels for tissue engineering: A review, Inter-
JA Jansen, et al., In vitro degradation of porous poly(propylene fu- national Journal of Polymeric Materials and Polymeric Biomaterials 66 (2017)
159–182.

You might also like