You are on page 1of 20

3D Bioprinting for Artificial Pancreas

Organ 21
Seon Jae Lee, Jae Bin Lee, Young-Woo Park, and Dong Yun Lee

at the University of Alberta in Canada carrying


21.1 Introduction out human islet transplants. Seven patients were
transplanted more than 800,000 human islets
Type 1 diabetes mellitus (T1DM) results from isolated from recipients two or three through
autoimmune destruction of insulin-producing intrahepatic injection (Shapiro et al. 2000).
beta cell in the islet of the endocrine pancreas. These patients maintained blood glucose control
The disease constitutes 5–10% of the diagnosed for approximately 1 year when accompanied with
diabetes that corresponds to approximately ‘Edmonton protocol’ that is administration of
23 million worldwide. Currently, the most com- several kinds of immunosuppressive medications.
mon method is injection of insulin directly. How- Then the global and multicenter clinical of the
ever, the intensive insulin therapy cannot tightly Edmonton protocol performed by the immune
control the blood glucose levels in the diabetic Tolerance Network showed that 16 of 44 islet
patients due to fluctuation of blood glucose. transplants recipients (44%) had no insulin inde-
Alternatively insulin pump is also used to control pendence for 1 year and 10 experienced complete
the blood glucose with continuous blood glucose transplanted islet defects (Shapiro et al. 2006).
monitoring (CBGM). Still it is necessary to Finally, the proportion of short-term grafts was
develop insulin delivery system according to the up to approximately 80%, but less than 20% of
fluctuation of blood glucose level. grated recipients remained insulin independent
The promising ideal strategy is transplantation until 5 years (Desai and Shea 2017). Thus,
of isolated insulin-secreting pancreatic islets. although islet transplantation is actually applied
Flame of islet transplantation was initially ignited in clinical practice, still it has not been a perfect
treatment. So, there are many challenges
S. J. Lee · J. B. Lee · Y.-W. Park remained. Main issues are donor islet shortage
Department of Bioengineering, College of Engineering, and immune reaction after islet transplantation.
BK21 PLUS Future Biopharmaceutical Human Resource
Training and Research Team, Hanyang University, Seoul, As mentioned above, two to three donors per
South Korea patient are needed. Hence, the efficiency of islet
D. Y. Lee (*) transplantation is drastically reduced due to the
Department of Bioengineering, College of Engineering, shortage of donors. Therefore, xenotransplanta-
BK21 PLUS Future Biopharmaceutical Human Resource tion of pancreatic islet has been proposed as a
Training and Research Team, Hanyang University, Seoul, solution to settle the shortage of donors. In this
South Korea
case, the xenotransplanted islets are dramatically
Institute of Nano Science & Technology (INST), Hanyang rejected due to xenogeneic immune rejection.
University, Seoul, South Korea
e-mail: dongyunlee@hanyang.ac.kr Therefore, immunosuppressive medications

# Springer Nature Singapore Pte Ltd. 2018 355


I. Noh (ed.), Biomimetic Medical Materials, Advances in Experimental Medicine and Biology 1064,
https://doi.org/10.1007/978-981-13-0445-3_21
356 S. J. Lee et al.

should be administered into the body to immuno- protected beta cells (Scharp and Marchetti 2014).
logically protect allogeneic or xenogeneic islets. The principle of encapsulation is that the cells to
The need for lifelong immunosuppressants be implanted are contained in compartments
greatly limits the broad availability of this islet separated by semi-permeable membranes. The
transplantation therapy. However, when immuno- capsule should protect the islets from damage
suppressive medications are administered for a caused by immune response. Therefore, the cap-
long time, they show severe adverse effect such sule is an “immune-isolation capsule”. In addition
as nephrotoxicity, hepatotoxicity, and other to the protective mechanisms provided by the
abnormalities. Therefore, to attenuate the dose capsules, islets in the capsule can also regulate
of immunosuppressive medications, pancreatic blood glucose levels by releasing insulin, while
islet encapsulation with biocompatible polymers small molecules (glucose, oxygen and nutrients)
is developed (Borg and Bonifacio 2011). The and external (metabolic waste) can pass through
biocompatible polymers are used as an this membrane. The encapsulation system is,
immunoprotective barrier to protect the therefore, also considered a “biological artificial
transplanted pancreatic islets. However, the con- pancreas”. Immune-isolation capsule or
ventional encapsulation strategies have several biological artificial pancreas are generally divided
disadvantages such as hypoxia issue, degradabil- into two categories: macroencapsulation and
ity, reproducibility and retrievability. Recently, microencapsulation (Qi 2014).
3D bioprinting technology is considered as alter- Microcapsules and macrocapsules are classi-
native tissue engineering. It can manufacture fied by their size (Fig. 21.1). Microencapsulation
capsules capable of accommodating cells for a use many microscales (100 μm–1 mm) capsules,
transplantable level and inhibit hypoxia by pro- containing single cells or islets that maximize
moting vascularization through structure and surface-to-volume ratio and promote optimized
releasing molecules. nutrient and oxygen exchange (Elliott et al.
In this chapter, therefore, we discuss about the 2007; Tuch et al. 2009; Desai et al. 2004). How-
islet encapsulation technologies with limitation ever, this technique has limited control over
issues and how the proposed 3D bioprinting membrane thickness and pore size, and limits
technologies to overcome the limitations of the number of capsules required for implantation.
encapsulation strategies can be applied. In addi- On the other hand, macroencapsulation capsules
tion, we address research perspectives for making (3–8 cm) contain many cells or islets (Tarantal
commercially available artificial pancreas com- et al. 2009). These larger capsules provide better
plementary to 3D bioprinting technology. control of membrane parameters such as pore size
and porosity and neovascularization by their
rough surface, but limit nutrient and oxygen dif-
21.2 Necessity for Manufacturing fusion and cell response due to capsule thickness
of Artificial Pancreas and large reservoirs (Cornolti et al. 2009; Desai
and Shea 2017; Lembert et al. 2005).
21.2.1 Encapsulation of Islet

To attenuate the dose of immunosuppressive 21.2.2 Microencapsulation


medications, encapsulation of islets with biocom-
patible polymers has been developed, which As mentioned earlier, microencapsulation is a
provides physical barriers to the transplanted method of encapsulating cells on a micro scale
islets and inhibits the immune response from using biocompatible materials. For successful
their recipients (Desai and Shea 2017). Various microencapsulation, studies have been conducted
microencapsulation and macroencapsulation using a variety of materials and these studies have
methods have been developed over the past sev- now reached the clinical trial stage. Some of the
eral decades with the goal of creating immune- fundamental limitations of microencapsulation
21 3D Bioprinting for Artificial Pancreas Organ 357

Fig. 21.1 Structure and function of microcapsule and size and contains multiple cells. Each capsule forms a
macrocapsule. Microcapsule and macrocapsule are classi- semipermeable membrane through biocompetable mate-
fied according to their sizes. (a) Microcapsule is typically rial to protect the inner cells from the immune cell and
prepared in the size of 100 μm–1 mm and contains one or the antibody, while relatively small glucose and insulin are
several cells. (b) Macrocapsule is usually made in 3–8 cm freely diffused

prevent this from becoming a complete technol- microencapsulation technology because it can be
ogy. From now on, we will discuss the process by produced under physiological conditions and
which microencapsulation has been developed don’t affect to islet function and non-toxic
and the limitations of that strategy. (Menard et al. 2010). In this experiment, the
In 1964, cell microencapsulation technology microencapsulated islets survived for the
was first described Chang (1964), and in 1980, 365 day in 1 of 5 animals. The alginate as material
microcapsules were used to treat diabetes, of microcapsule microcapsules was increased the
demonstrating prolonged survival of isograft strength of microcapsules. And decreasing the
islets using alginate-polylysine-polyethy- impurities and increasing the guluronic acid to
leneimine microcapsules (Lim and Sun 1980). mannuronic acid ratio further improved the bio-
Non-encapsulated islets without post-transplant compatibility of alginate microcapsules (Klock
immunosuppression survived for 8 days whereas et al. 1994; Otterlei et al. 1991). After this
encapsulated islets survived to 3 weeks (Lim and study, studies on microencapsulation using algi-
Sun 1980). Thereafter, studies on the materials nate have been actively conducted, but in addition
were conducted, so microcapsule material was to alginate many other polymers such as chitosan,
improved in 1984. The previously used polyeth- agarose, methacrylic acid, polyethylene glycol
ylene imine component was disappeared and algi- (PEG), methyl methacrylate and 2-hydroxyethyl
nate was designed as the outer layer of the methacrylate (HEMA) have been used success-
microcapsule (O’Shea et al. 1984). The use of fully in islet encapsulation studies with limited
alginate has considerably improved the success (de Vos et al. 2010).
358 S. J. Lee et al.

Over the next few decades, research has been case of microcapsules, the smaller size, the more
conducted to develop microcapsules with suffi- fibrotic overgrowth due to the foreign body
cient durability and biocompatibility. The response, which leads to immune rejection in the
experiments was conducted on small animals transplanted cells, resulting in the failure of trans-
and many were successful (Klock et al. 1994; plantation (Veiseh et al. 2015).
O’Shea et al. 1984; Sawhney et al. 1993). To To overcome this problem, we need to
improve the microcapsule, more than 1000 increase the size of the capsule, but it which can
combinations of polyanions and polycations make the capsule layer thicker, inhibiting oxygen,
have been evaluated for relevance to cell encap- nutrient transport and inducing hypoxia. There-
sulation. A polyelectrolyte complex formation fore, there is a dilemma between improving the
process was identified through the experiments microencapsulation strategy and suppressing
with five different polymers, which enabled inde- fibrotic overgrowth. So, when these problems
pendent control over capsule size, wall thickness, are solved, can microencapsulation be
mechanical strength and permeability (Wang commercialized? One of the biggest challenges
et al. 1997). faced by the microencapsulation technique is the
And since then, microencapsulation technol- scale up of the capsulation process. In the case of
ogy has proven effective in many experiments on conventional microencapsulation, the process is
rodents. Between 2000 and 2010, more than usually carried out on a laboratory scale and we
60 studies of microencapsulation strategies stud- need to scale up these fabrication processes to an
ies were performed on rodents and the viability of industrial level. However, in microencapsulation,
islets encapsulated with alginate without immu- controlling the formed diameter is important for
nosuppression was the best for 100 days (Desai proper operation and perturbation of the capsule
and Shea 2017; Souza et al. 2011). And and in order to form a micro-level bead, the flow
encapsulated islet allotransplantation clinical rate at which the material is injected must be
trial, since its inception in 1994, Several clinical controlled.
trials were conducted (Soon-Shiong et al. 1994). So, there is a significant limitation in scaling
These experiments were performed by allo/xeno- up the microencapsulation process at the labora-
transplantation of the encapsulated islets in tory level to the industrial level.
T1DM patients who did not receive immunosup-
pressants and then controlled blood glucose level
normal in the long term. Despite other numerous 21.2.3 Macroencapsulation
clinical trials were conducted by academia and
biotechnology companies, complete encapsulated As mentioned earlier, islet encapsulation is clas-
islets were not have not been developed (Basta sified into two types, depending on their size:
et al. 2011; Calafiore et al. 2006; Jacobs- macrocapsule and microcapsule. Moreover,
Tulleneers-Thevissen et al. 2013; Valdes- macrocapsule is also divided in two types, extra-
Gonzalez et al. 2005, 2010). Each trials was vascular intravascular types (Fig. 21.2). Although
shown to temporarily inhibit diabetes, but in the extravascular and intravascular macroencap-
long term the therapy failed due to islet apoptosis sulation has been developed with individual
and necrosis (Buder et al. 2013). advantages, these strategies also have some
So, what is the limit of microencapsulation problems that prevent these strategies from
that causes the failure of these clinical trials? becoming complete biological artificial pancreas.
Process systems that are not standardized due to From now on, we will discuss the process by
the diversity of raw materials in the which each extravascular and intravascular
manufacturing process and experiments that macroencapsulation has been developed to the
have been mainly done on the rodent model are preclinical stage and limitations of them.
also one cause, but the major reason is fibrotic The extravascular macrocapsule has a diffu-
overgrowth by micro-scale of capsules. In the sion chamber structure and is not connected to
21 3D Bioprinting for Artificial Pancreas Organ 359

Fig. 21.2 Two types of macroencapsulation; Extravascu- where it is implanted intraperitonally or subcutaneously.
lar (a) and Intravascular (b). (a) The cell-laden (b) Arteries and veins are besieged with the macrocapsule,
macrocapsule is located on an external blood vessel which directly borders the vessels

blood vessels, etc. Therefore, it can be implanted Marchetti 2014). Their macrocapsules had a
intraperitonally or subcutaneously, which has pore size of about 450 nm and could inhibit
advantage in that the process of implantation direct cell-cell immunity, which enabled
and removal is somewhat simple (Sakata et al. immunoprotection at allotransplantation available
2012; Schweicher et al. 2014). level. A series of studies at the time demonstrated
The first macroencapsulation used this extra- that encapsulation through these macrocapsules
vascular macrocapsule developed in the 1950s can improve cell viability (Algire and Legallais
(Algire et al. 1954; Prehn et al. 1954). During 1949; Gates et al. 1972; Strautz 1970).
the 1970s, Millipore Corporation developed and Although many studies at this time were
produced commercially available extravascular performed through syngenic cells, transplantation
macrocapsules for implantation (Scharp and failure occurred because of the fibrotic
360 S. J. Lee et al.

overgrowth caused by the lack of proper control. One study in the 1990s (Brauker et al.
biocompetability of the materials used (Desai 1995, 1996) compared the extent of host
and Shea 2017). And it was also due to the neovascularization with membrane pore size
limitations of extravascular microencapsulation differences and found that about 100 times more
that nutrient and oxygen transport depend only neovascularization was formed in the membrane
on diffusion through thick layers. This restriction with the appropriate pore size (Brauker et al.
induces hypoxia and apoptosis/necrosis in the 1995, 1996). And the vascularization was further
encapsulated cells, leading to graft failure accelerated when large pore layers were
(Schweicher et al. 2014). laminated to capsules carrying immune protection
To overcome these limitations, intravascular through a small pore, confirming that this signifi-
macrocapsulation has been studied since the cantly higher level of vascularization was retained
1970s (Chick et al. 1975). The intravascular for 1 year. However, the subcutaneous tissue is a
macrocapsule has a perfusion chamber structure low blood-flow tissue, and more research was
and is directly connected to the arteries and veins needed to confirm whether accelerated vasculari-
of the host (Monaco et al. 1991). The islets are zation is adequate for islet survival. However, the
supplied nutrients and oxygen through the blood subcutaneous tissue originally had a low blood
flowing through the hollow fibers inside the flow, resulting in a somewhat exaggerated vascu-
chamber and releases the insulin. Because there larization effect, so more research was needed to
is a semi-permeable membrane between the confirm whether vascularization is sufficiently
chamber and the hollow fibers, the islets are accelerated in other tissues and it can improve
immunoprotected. the survival of islets (Clark et al. 2000; Fumimoto
The islet containing intravascular et al. 2009; Ryan et al. 2001).
microcapsules chamber made from diverse Another study showed that allograft transplan-
materials whereas tested on a diabetic rat, mon- tation of macroencapsulated islets into the epidid-
key model, which could be expected to replace ymal fat pad of streptozotocin-induced diabetic
the previously advanced extravascular mice yielded normal glucose tolerance for
capsulation (Chick et al. 1977; Orsetti et al. 12 weeks (Suzuki et al. 1998). However, in this
1978; Sun et al. 1977; Tze et al. 1976, 1980; experiment too, there was a problem caused by
Sun et al. 1980). However, blood coagulation hypoxia caused by the encapsulation of large
and hemorrhage occurred in these experiments mass of islets into one capsule. In addition, failure
to confirm the possibility of intravascular micro- of transplantation due to fibrotic overgrowth
capsule (Sun et al. 1980; Tze et al. 1980). This is around the capsule was also observed. Since
because thrombosis is induced by the blood flow then, the biocompetability of the material has
somewhat suppressed at the interface between the gradually improved and macroencapsulation
blood vessel and the capsule, and blood coagula- capsules have demonstrated some success in
tion is caused by the limited biocompetibility of large animals, although the results have not been
macrocapsule itself. Unfortunately, systemic consistent. In nonhuman primates (NHPs), por-
anticoagulation agents cannot be used for T1DM cine islets placed within an alginate macrocapsule
patients, and therefore a method to replace intra- transplanted subcutaneously were found to pro-
vascular macrocapsules is needed. vide normoglycaemia for up to 6 months
Studies have been conducted to overcome (Dufrane et al. 2006).
these limitations, and several studies have been And other macrocapsule studies using alginate
carried out showing that the induction of host sheet structures showed significant results in pre-
neovascularization can solve the problem of clinical trials, but at the same time showed
extravascular capsules have been carried out. limitations in maintaining sheet planarity (Storrs
These attempts to allow blood vessels to form et al. 2001). The thickness of the sheet was
inside the capsule have allowed the host cell to determined by considering the maximum distance
penetrate into the capsule through pore size of oxygen and nutrient diffusion (250 μm). The
21 3D Bioprinting for Artificial Pancreas Organ 361

prepared islet containing alginate sheet was Therefore, 3D bioprinting is emerging as a new
implanted to the omentum of pancreas resected technology for encapsulation of cell due to posi-
dog. As a result of the experiment, fasting tioning alive cells in a specific location (Billiet
euglycemia was confirmed for 84 days. After that, et al. 2012; Wust et al. 2011). Furthermore,
the islet in macrocapule was collected again and it depending on the applied biochemical materials
was confirmed that it was alive. This confirms that and types of cells being encapsulated, a variety of
fasting blood glucose lowering is due to functional tissue or organs can be created (Mur-
macroencapsulation-induced immunoprotection. phy and Atala 2014; Zadpoor and Malda 2017).
However, despite the success of this large In this respect, organ transplantation is focused on
animal, macroencapsulation capsules have yet to 3D bioprinting as a solution to manufacture an
enter the human clinical trial (Scharp and March- alternative organ. Consequently, the desired
etti 2014). For xenotransplantation in humans, organs are manufactured that what kinds of
about 5000–20,000 IEq/kg is needed (Ekser materials or cells are applied to 3D bioprinting.
et al. 2016), There is not yet a way to overcome Therefore, in the field of 3D bioprinting, it is
the increased threat of core hypoxia as these islet essential to find suitable cells and materials for
masses are encapsulated in one capsule. In addi- finally fabricated organs. Additionally, the
tion, currently used macrocapsule production materials have sufficient mechanical properties
techniques are limited in that there is little way to allow 3D structures to manufacture through
to induce vascularization other than controlling the stacking layer by layer method.
pore size. So, if these hypoxia problems cannot be Generally, the 3D bioprinting can be classified
solved, macroencapsulation cannot be presented into two systems depending on the materials, the
as a treatment for T1DM. first is a scaffolding system. A synthetic polymer
So if macroencapsules are improved to solve is mainly applied to the system. It is liquefied
the core hypoxia problem, is it likely to be suc- through a thermal process and the molten poly-
cessfully commercialized? So, if the problem of mer is extruded. Then, the extruded polymer is
hypoxia after transplantation is solved, can cooled to produce the scaffold. In general,
macroencapsulation capsules be commercialized? polylacitc acid (PLA), poly(Lactide-co-glycolic
In macroencapsulation, the capsule is formed in a acid) (PLGA), and polycaprolactone (PCL)
single mass, so hypoxia can occur in the cells of approved by FDA are mainly used as synthetic
the inner core during manufacturing process too. polymers applied in 3D bioprinting due to a supe-
Also, because it is made of a single mass, it is rior biocompatibility and biodegradability to
difficult to distribute the cells properly in capsule. regenerate tissues and organs (Gunatillake and
These points limit the scale up of the Adhikari 2003; Shim et al. 2011). Herein, the
macrocapsule capsule manufacturing process scaffold composed of the synthetic polymer is
and bring it up to the industrial level. mainly used for scaffoling purpose. In addition,
cells are seeded in these scaffolds. Finally, the 3D
structure containing the cells is completed. Pati
et al. (2016). In fact, the scaffold composed of
21.3 3D Bioprinting
blending PLA and PLGA was made by 3D
bioprinting (Shim et al. 2011). Then, rat primary
21.3.1 Introduction of 3D Bioprinting
hepatocytes and a mouse pre-osteoblast MC3T3-
E1 cell encapsulated hydrogels were injected in
3D bioprinting is a technique for positioning bio-
the printed scaffold. After 7 days of injection, the
chemical materials and alive cells in a stacking
encapsulated cells were observed to be
layer by layer at a desired location. Using this
proliferated felicitously. Furthermore, a viability
techique, a 3D structure can be fabricated by
was also maintained until 10 days. Additionally,
controlling the space of the positioned
dual cell laden 3D structure composed of PLA
components (Murphy and Atala 2014).
362 S. J. Lee et al.

containing both osteoblast and chondrocyte was biological artificial pancreas is urgent. Fortu-
successfully constructed (Shim et al. 2012). This nately, 3D bioprinting is a revolutionary system
dual cell laden system maintained a viability and that can be mass-produced through a rapid
proliferation of cells for 1 week appropriately. In automated system (Bak 2003). However, conven-
addition, the surface of a scaffold made of PCL tional micro/macroencapsulation is difficult for
may be modified to improve the affinity with cells mass production of artificial pancreas. The pro-
(Domingos et al. 2013). The second is a scaffold- cess of conventional encapsulation is usually car-
ing free system. A hydrogel is mostly used for this ried out on a laboratory scale. In fact, in the case
system. The hydrogel contains a large amount of of microencapsulation, controlling the formed
water and can provide the optimal environment diameter is important for proper stability and
for cells (Melchels et al. 2012). Unlike the scaf- durability of the capsule. The flow rate through
folding system previously described, the hydrogel which the material is injected must be adjusted to
containing alive cells is positioned in a specific produce an appropriate size of bead. However,
location directly; this hydrogel is called a bio-ink. that is a significant limitation in scaling up the
The bio-ink is applied not only to cells but also to microencapsulation process at the laboratory
various drugs or biomolecules together (Kaigler level to the industrial level. In macroencap-
et al. 2013; Wust et al. 2014). Additionally, the sulation, the cell enclosed form is a single mass.
hydrogel is solidified by physical or chemical In other words unlike the case of microencapsula-
crosslinking to stack layer by layer to complete tion, hypoxia can occur in the cells of the inner
the 3D structure (Nichol et al. 2010; Pescosolido core. This hypoxia can also occur when
et al. 2011; Wang et al. 2006; Yan et al. 2005). In macrocapsule is implanted (O’Sullivan et al.
fact, gelatin metacrylamide containing alive cells 2010). Also, proper cell distribution in the
was printed and crosslinked by a photo-initator to macroencapsulation becomes difficult. As a
solidify immediately after printing. The viability result, that is also a limit to scale up to the indus-
of printed cells right after printing was maintained trial level. However, 3D bioprinting can over-
at over 97%. Consequently, cell-laden hydrogel come the limitations of these conventional
was successfully manufactured (Billiet et al. encapsulation technology. As mentioned earlier,
2014). In addition, an Extra-cellular matrix 3D bioprinting can position biochemical
(ECM) based hydrogel was used to increase the materials and alive cells where desired. There-
viability and bio-functionality of encapsulated fore, the printed cells are properly distributed in
cells (Yeo et al. 2016). To protect cells the 3D structure. In addition, 3D bioprinting has
encapsulated in the ECM based hydrogel, a shell the potential to improve the hypoxia of cells in the
of alginate was formed on the outside to form a 3D structure by vascularization and locally distri-
core-sheath structure. Later, the core-sheath 3D bution of the printed cells (Novosel et al. 2011).
structure containing human adipose stem cells The 3D structure with a special shape can also be
(hASCs) maintained viability for approximately regarded as a building block. Then, the building
1 weeks. It also effectively differentiated in blocks can be made into a larger structure. And
hepatogenic cultures. Likewise 3D bioprinting is 3D bioprinting can create complex shaped artifi-
an innovative field that can mimic human tissues cial structures. Therefore, the disadvantages of
or organs by selecting appropriate materials and conventional encapsulation could overcome the
cells according to a function of the finally limitation of scaling up (Ozbolat 2015; Ozbolat
fabricated 3D structure. and Yu 2013; Pati et al. 2016). In the next
The development of an artificial pancreas sections, we will look at the development of the
containing pancreatic islets using 3D bioprinting current artificial pancreas through 3D bioprinting,
is still an introductory stage. Nowdays, as the and discuss possibiliy of improvement in scaling
shortage of islet donors, the production of up and hypoxia aspects in the artificial pancreas.
21 3D Bioprinting for Artificial Pancreas Organ 363

21.3.2 Scalling Up of Artificial used as a cell source for islet transplantation to


Pancreas T1DM (Chao et al. 2008). It also induced engraft-
ment and vascularization of transplanted islets
The artificial pancreas developed so far was pro- through co-transplantation with pancreatic islets
duced through the scaffolding system. Various (Figliuzzi et al. 2009; Ito et al. 2010). Thus,
cells from primary cells to beta cell like stem MSCs are applied to islet transplantation directly
cells were planted in a framework based on and also involved in differentiation to islet like
FDA approved biocompatible synthesis cells (Chen et al. 2004). To apply ILCA to 3D
polymers. These approaches have introduced the bioprinting, a PLA scaffold was prepared. The
production of artificial pancreas. Initially devel- scaffold was discoidal shape and had a diameter
oped artificial pancreas was scaffolded with of 13 mm and thickness of 4.5 mm. Additionally,
PLGA (Daoud et al. 2011). The final 3D structure the scaffold was obtained to the surface charge
was constructed by seeding the ECM based and hydrophilicity to improve suitability with the
hydrogel containing human pancreatic islet into encapsulated cells. A poly-L-lysine (PLL) coated
the PLGA scaffold. Generally, ECM components on the surface of the PLA to give a charge. Addi-
are critical factors for participations in cellular tionally, the surface of the PLA was etched
adhesion, matrix for supporting cells, and various through argon (Ar) or oxygen (O2) plasma to
signaling pathways with cells (Hamamoto et al. improve hydrophilicity. In addition, an endothe-
2003; Jiang et al. 2002; Kaido et al. 2004). Fur- lial cell attachment factor (ECAF) was coated on
thermore, pancreatic islets also showed better the surface of the PLA to promote vascularization
survival when cultured in a matrix containing around the scaffold. Indeed, a surface modifica-
ECM components (Thomas et al. 1999). Thus, tion of a material changed a cell behavior and
human pancreatic islets were encapsulated via gene expression of adjacent cells (Mrksich
an ECM supplemented gels composed of 2000; Stevens and George 2005). Therefore, the
collagen I, fibronectin and collagen IV. In addi- surface modification of the PLA scaffold
tion, the porosity and pore size of the scaffold expected a synergism with the seeded cells In
were controlled by varying the shape of the addition, a platelet lysate (PL) gel was used to
PLGA scaffold. Therefore, it had a synergism to encapsulate the ILCA into PLA scaffold. In gen-
seeded cells in the ECM based hydrogel. There- eral, the PL gel was an ECM component obtained
fore, human pancreatic islets in the 3D structure from animal platelet. It was often used to encap-
were 1.8-fold more effective insulin release than sulate an endothelial cell for vascular capillary
islets in normal suspension culture. Additionally, formation (Fortunato et al. 2016). In this way,
an expression of insulin secreting genes in the 3D the ILCA was encapsulated in PL gel. Then, the
structure was increased by 50-fold compared to a PL was seeded into the PLA scaffold which is
normal culture. That is because the PLGA scaf- only modified surface by etching N2 or O2
fold provided a proper environment through an plasma. Interestingly, the duration of insulin
improved oxygen and mass transfer. In conclu- secretion maintained in ILCA in the PLA scaffold
sion, 3D bioprinting has informed a start of the longer than in ILCA that was in a normal suspen-
artificial pancreas development through the sion culture. That resulted in the accumulation of
proper combination of the scaffold and ECM insulin secreted from ILCA in the PL gel. In
based hydrogel. addition, a larger PLA disk (diameter 20 mm,
Mesenchymal stem cells (MSCs) thickness 1 mm) treated with Ar plasma, PLL,
differentiated into islet like cell aggregates and ECAF. A cytotoxicity of human unbilical
(ILCA) applied to the manufacture of the artificial vein endothelial cells (HUVECs) with the PLA
pancreas using 3D bioprinting instead of a pri- disk were observed. Certainly, in the case of the
mary cell (Sabek et al. 2016). Indeed, MSCs were PLA disk treated with Ar and PLL, a toxicity of
364 S. J. Lee et al.

HUVECs was observed. In conclusion, although pancreatic cancer surgery, a patient-specific pan-
the cytotoxicity of PLA scaffold occurred, the creas and peripancreatic region were
approach of modifying a surface of the scaffold manufactured by using 3D printing technology.
may assist on the development of a new artificial Then, a plan of the surgery was made based on the
pancreas. printed anatomical pancreas. Therefore, it has not
With the introduction of this scaffolding sys- reported that the human scale pancreas was not
tem, the size of the scaffold can be adjusted used to treat T1DM. Indeed, for performance of
according to the size and design of the scaffold islet transplantation in the clinical trial, the num-
to be printed. In other word, the scaffold can be ber of transplanted islets was 5000–20,000
made in sizes that are possible to apply to real IEq/kg, which was a very large amount (Ekser
human organs. In fact, there was a case that the et al. 2016). A capsule containing cells must have
scaffolding system using 3D bioprinting. It was a sufficient volume. In conclusion, the new
used to manufacture a size similar to an actual approach of developing the artificial pancreas
human ear (Lee et al. 2014a, b). The main part of indicates that the scaffolding system of 3D
the scaffold was composed of PCL. Then, the bioprinting can be used to make a large size of
cell-laden hydrogel was seeded in the PCL scaf- the main scaffold, and the hydrogel containing
fold. Furthermore, the final human scale ear was various cell types as well as pancreatic islets can
printed by using polyethyleneglycol (PEG) with a be seeded into the scaffold.
sacrificial part. The sacrificial part additionally A case of developing the artificial pancreas
supports the main PCL scaffold with a complex with the scaffolding free system has been reported
shape during the process of 3D bioprinting. It is (Marchioli et al. 2015). To make the artificial
widely applied to print complex shapes in the 3D pancreas composed solely of a hydrogel, compos-
bioprinting (King and Tansey 2003). In the print- ite hydrogels were prepared by alginate hydrogel.
ing process, PEG could support the main part of Additionally, various materials such as gelatin,
PCL, but it was easily removed through aqueous hyaluronic acid or Matrigel were mixed with algi-
solution or culture medium. In order to create an nate. To make the artificial pancreas through
element similar to the human ear, chondrocytes these various hydrogel mixtures, the main mate-
and adipocytes that were differentiated from adi- rial, alginate, was pre-crosslinked by a low con-
pose derived stromal cells (ASCs) were centration of calcium ion. Because of the
encapsulated in alginate hydrogel. Chondrogenic pre-crosslinked alginate, the viscosity of the algi-
and adipogenic expression was higher when the nate hydrogel was increased. Therefore, lamina-
cells were co-encapsulated than enclosed sepa- tion was possible to make the 3D structure
rately. Indeed, because of a synergism associated temporarily. In addition, when the temporary 3D
with an treatment of fat tissue for cartilage forma- structure was printed completely, a post-
tion, an injection of the fat tissue may be used to crosslinking was performed by a high concentra-
regenerate a septal cartilage formation (Nakakita tion of calcium ion to strengthen the mechanical
et al. 1999). Unfortunately, the alginate hydrogel properties. There is a lack of mechanical
containing these cells was applied to the artificial properties to stack layer by layer if a hydrogel
ear shaped scaffold. However, the ability of these precusor is only used to print the 3D structure.
cells to differentiate properly could be Therefore, the mechanical properties must be suf-
demonstrated by using another shape of a scaffold ficiently strengthened through physical or chemi-
composed of PCL. In conclusion, it was cal crosslinking. In fact, using pre-crosslinked
suggested that the ear-shaped structure containing alginate could improve the resolution of the
with multi-cells could be sufficiently possible for printed 3D structure if complex structures are
regeneration of the auricular cartilage. printed (Tabriz et al. 2015). In addition, INS1E-
Unfortunately, there has been no report on the beta cell that derived from rat insulinoma was
production of human scale pancreas as using 3D encapsulated in various hydrogel mixtures to
bioprinting. However, in order to perform make the artificial pancreas. Then, a viability of
21 3D Bioprinting for Artificial Pancreas Organ 365

the cell was monitored depending on the without the scaffold. For introduction the artificial
components of the hydrogel mixtures. As a result, pancreas through introducing the concept of
the viability of the cells in the alginate/gelatin advanced assembly system, it is possible to
mixture was maintained at 95% to 21 days after develop a new human scale pancreas capable of
printing. The structure was also preserved proper vascularization.
soundly. Using the only hydrogel to make the
artificial pancreas has indicated that a new
approach can create an environment similar to 21.3.3 Prevention of Hypoxia through
the mechanical properties of the real pancreas. Vascularization
Indeed, the pancreas has a significantly lower
modulus than other organs, skin, muscles and Hypoxia is the most decisive factor of the effi-
skeletons (Liu et al. 2015). In conclusion, the ciency of islet transplantation (Pedraza et al.
development of the artificial pancreas as using 2012). And this is a big problem in the case of
the scaffolding free system can be an approach the technique of forming immunoprotective layer,
to mimic the proper environment of the actual such as encapsulation. In the 3D bioprinting,
pancreas. mainly macro scale capsules are manufactured
The scaffolding system produces the based and therefore, the problem of extravascular
scaffold composed of synthetic polymers non macroencapsulation mentioned above such as
containing cells to print human scale organs. On core hypoxia is maintained. Macro scale capsules
the other hands, the scaffolding free system are mainly implanted in subcutaneous tissue, and
requires only the use of the cell-laden hydrogel the viability of the transplanted islets in the sub-
to make human scale organs. However, because cutaneous site is drastically reduced because the
of the prolonged printing process in order to cells isolated from the surrounding blood vessels.
produce the large scale structure, a viability of As has been mentioned, islets have highly rate of
the cells encapsulated in the hydrogel may be consumption of oxygen and nutrients relative to
drastically reduced (Ozbolat and Yu 2013). their proportion in the pancreas because insulin
Instead of printing tissues or organs of the secretion in islets requires a lot of mitochondrial
human scale at once, there is a new approach to respiration (Sato et al. 2011). Therefore, the oxy-
assemble building blocks or mini-tissues com- gen supply to the transplanted islets is
posed of only hydrogels to complete the final tremendously critical.
human scale organs (Ozbolat 2015; Ozbolat and So, how do we solve this hypoxia problem?
Yu 2013). In fact, to construct a vascular tissue Currently, 3D bioprinting based technologies use
through the scaffolding free system, agarose rods vascularization to solve these hypoxia problems
were prepared by 3D bioprinting to use as build- (Lee et al. 2014a, b). A blood vessel system is
ing blocks to assemble a desired shape of molding formed to form a homogeneous blood flow inside
template (Norotte et al. 2009). Then, uniform the 3d bioprinting based transplantation capsule.
multicellular spheroids were printed into patterns This technique is also being studied in the above-
designed at the template. Thereafter, the printed mentioned macroencapsulation, so it may ques-
spheroids were self-assembled to a large vascular tion whether 3D bioprinting is necessary to solve
tissue depending on the patterns in the template. hypoxia. As mentioned above, because this tech-
Additionally, the formed large vascular tissues nique is also being studied in the macro encapsu-
could be assembled together to make one new lation, 3D bio-printing can be asked if it is a
larger vascular tissue. Thus, building blocks can necessary technique to solve hypoxia. However,
be used to create a new large-scale structure the development of 3D bioprinting based trans-
consisting entirely of hydrogel without scaffolds. plantation capsules offers a variety of strategies to
In conclusion, in order to create the original envi- promote vascularization and therefore, 3D
ronment of pancreas, it is also necessary to pro- bioprinting has utility value as a tool to solve
duce the artificial pancreas with only hydrogel the problems of existing technique.
366 S. J. Lee et al.

Currently, there are two types of vasculariza- cells can establish a perfusion network of vessels
tion techniques applied in the field of 3D in appropriate artificial capsules (Chen et al.
bioprinting: cell based strategy and hydrogel/ 2009; Laschke et al. 2008; Tremblay et al.
scaffold based strategy (Novosel et al. 2011). In 2005). The main disadvantage of this method is
cell based strategy, activation of endothelial cells that at least three operations are required during
through growth factors, adhesion peptides, and the procedure: the implantation for vasculariza-
co-culture with other cells leads to neovascu- tion, the removal of the prevascularized capsule
larization in the capsule and this method proceeds and secondary implantation (Rouwkema et al.
through prevascularization of the capsule 2008). Vascular networks produced in an
followed by transplantation or by inducing in vivo environment allow oxygen and nutrient
neoangiogenesis after transplantation. delivery to transplanted cells immediately when
the capsule is implanted, but the complex
manufacturing process of this method lowers the
21.3.3.1 Cell Based Prevascularization
commercial applicability of this method.
Prevascularization is a method of forming a vas-
cular network through vasculogenesis or angio-
genesis using endothelial cells in the capsule, and 21.3.3.2 Cell Based Neoangiogenesis
this method proceeds either in vitro prevascu- So, how about the recipient host makes vasculari-
larization or in vivo prevascularization. zation by himself? So, how about letting vascu-
In ‘in vitro prevascularization’, endothelial larization take the capsule to the recipient host? If
cells with other cells such as myoblast or fibro- host-induced vascularization can occurs in the
blast are seeded to capsule and they are. The implanted capsule quickly and appropriately, the
structure is cultured in vitro for the purpose of process of manufacturing the capsule will be
constructing a three dimensional much simpler since the process of vascularization
(3D) prevascularized structure (Rivron et al. in the capsule is omitted. If host-induced vascu-
2008). This method has the advantage that the larization occurs in the implanted capsule, the
manufacturing process of the capsule is simpler, process of manufacturing the capsule will be
but it takes time to connect the vascular network much simpler since the process of vascularization
of the host to the network of which the in the capsule is omitted. However, the vasculari-
manufactured capsule to be implanted to the net- zation naturally occurred by the implant is slow
work of the manufactured. Oxygen and nutrient and transplanted cells in the capsules are exposed
diffusion is possible only from 100 to 200 μm to damage by hypoxia during vascularization. To
from the blood vessel (Rouwkema et al. 2008), prevent it, vascularization should be promoted as
Therefore, to survive transplanted cells, the vas- much as possible. In current researches, various
cular network in the capsule must be activated methods of seeding endothelial cells in the cap-
immediately after transplantation, so whether or sule and promoting vascularization through
not these drawbacks can be overcome will deter- angiogenic growth factors or immobilized
mine the success or failure of transplantation. molecules are used.
In contrast to the method of vascularization Angiogenic growth factors activate endothelial
through a pre – seeded capsule, ‘in vivo prevascu- (progenitor) cells and induce migration by gradi-
larization’ uses a nonvascularized construct. Dur- ent to promote neovascularization. In addition,
ing the preliminary implantation, the de novo they stimulate blood vessel formation and matu-
vascularization of the construct proceeds at the ration by inducing cell assembly (Nomi et al.
recipient’s implant site. First, when a 2006). The major growth factors that promote
macroporous capsule is implanted, blood vessels the angiogenesis process are the basic fibroblast
infiltrate and vascular networks are formed. Since growth factor (bFGF), the vascular endothelial
then, several studies have demonstrated that host growth factor (VEGF) and the hepatocyte growth
21 3D Bioprinting for Artificial Pancreas Organ 367

factor (HGF). In addition, the interaction of using 3D bioprinting has been conducted on a
cytokines such as platelet-derived growth factor capsule that does not induce hypoxia. Therefore,
(PDGF), angiopoietin and transforming growth it is important to solve the hypoxia problem
factor beta (TGF beta) act as an indirect angio- through vascularization, but it suppresses the
genic factor and promote the regeneration of immunoprotection function, which is originally
endothelial tubes. These growth factors and intended for the capsule. In the case of cell-
cytokines promote the vascularization process of based vascularization, a vascular network is
endothelial cells seeded in the capsule, which formed in the capsule, which means that
increases the feasibility of host-induced transplanted islets may become more vulnerable
vascularization. to the host immune response. Then, can
However, this method of using growth factors vascularization-based methods be applied to islet
needs to solve one big problem in order to pro- transplantation? Fortunately, we can do cell dis-
duce successful results. The rapid degradation tribution properly within the capsule via 3D
rate of the growth factor can prevent the vascular bioprinting. Therefore, we can manufacture a cap-
network from being formed sufficiently, which sule suitable for artificial pancreas by controlling
can lead to hypoxia in transplanted cells, resulting the density of the formed vascular network to
in failure of transplantation. To solve this prob- such an extent that immune protection can be
lem, the growth factor must be continuously con- sustained without causing cell hypoxia.
trolled released around the capsule (Santos and
Reis 2010). For controlled release of these growth 21.3.3.3 Hydrogel/Scaffold – Based
factors, studies on capsules degradable or Strategy
containing preencapsulated microspheres have So how can we control the density of these vas-
been made. For controlled release of these growth cular networks? Of course, it can regulate itself
factors, studies have been made on capsules and it is more efficient. The proper 3D design of
degradable or contain preencapsulated the capsule has the effect of promoting proper
microspheres (Borselli et al. 2010; Demirdogen vascularization (Liu and Chen 2005). And this
et al. 2010; Ko et al. 1995). In conclusion, in means that the degree of vascularization can be
order for cell-based neoangiogenesis to be suc- controlled according to how we fabricate the 3D
cessful, an appropriate controlled release of the design of capsule is done. We can manufacture
growth factors and thus rapid vascularization are capsules suitable for islet transplantation by
important. controlling vascularization through capsule mate-
So, we have discussed how to perform cell- rial, porosity, pore size, capsule material and
based vascularization through each in vitro/ channel designing.
in vivo prevascularization and neoangiogenesis. Then, how are these factors considered and
Although these methods have advantages and determined when designing a real capsule? First,
disadvantages, they are efficient methods for vas- the porosity and pore size of the capsule, which is
cularization of capsules based on 3D bioprinting one of the most important factors in determining
technology. vascularization, capsule is determined by the den-
However, it is necessary to think again sity of the material and the length of the polymer
whether the methods discussed above are efficient chain, which is one of the most important factors
for islet transplantation. To manufacture artificial in determining vascularization (Keskar et al.
pancreas, we need a capsule to immunoprotect the 2009). And we can consider a variety of materials
transplanted islets from the immune response of that we will use to make 3D printing based
the host. However, it is difficult to fabricate capsules, using scaffolds and hydrogels made
biological artificial pancreas because of the hyp- primarily from synthetic peeolymers (Novosel
oxia induced by the capsule manufactured for this et al. 2011). Since the polymer chain length of
purpose. To overcome this limitation, research each material is different from biocompatibility
368 S. J. Lee et al.

and stability, it is important to select the appropri- 21.4 Vision of the Artificial Pancreas
ate material for capsule design. Future Research
And In addition, the 3D bioplotting method
using hydrogel is mainly used for manufacturing As previously mentioned, islets require a lot of
artificial pancreas compared to other organs and oxygen, so when islets are isolated, they can get
pancreas because porosity can be controlled by hypoxic damage. Especially, because size of
the density of network formed during bioplotting. islets is large (50~350 μm), so the core region of
Of course, creating a capillary network in the them can get this hypoxia damage easily
capsule more directly through 3D bioprinting (Ramachandran et al. 2015). And vascularization
can also be considered. However, forming a com- takes a few days. To solve this problem, forming
plete capillary network is difficult because there is islet cell spheroid (ICS) can be is another strategy
a problem of maximum resolution and production to prevent hypoxia because of smaller size than
time on current 3D bioprinting technology. intact islet. Islet spheroids can be obtained by
Therefore, in the current 3D bioprinting field, clustering single cells obtained by breaking
researchers we use microchannel in capsule to down intact islet cells or β-cell lines. And it is
induce formation of vascular network (Lee et al. efficient to use the spheroids because they main-
2014a, b). A study conducted until 2014 has tain the function despite the small size compared
shown that 3D bioprinting produces a fluidic vas- to the intact islet. Moreover, smaller size of islets
cular channel, and that the angiogenic channel is more suitable than larger when transplanted
that extends from this vascular channel andis is (MacGregor et al. 2005).
connected to the vascular network that is sepa- There are several spheroid formation methods;
rately formed in the capsule. In this channeled hanging drop, centrifugation, non-adherent
capsule made by 3D bioprinting, the capsule surfaces, etc. (Mehta et al. 2012; Sutherland
entry of endothelial cells and other supporting et al. 1981). For example, hanging drop method
cells and soluble factors is much easier, which is using the cell suspended droplets and allows
promotes the vascularization process. Through cell aggregation at the bottom of the droplets by
this vascular channel, we can induce vasculariza- gravity Foty (2011). Centrifugation can also form
tion at the desired position, thereby inducing oxy- spheroids, centrifugal force improves cell aggre-
gen and nutrient transport efficiently while gation (Handschel et al. 2007). Non-adherent sur-
immunoprotecting the cell (Lee et al. 2014a, b). face is using molecules such as chitosan to avoid
from cells adhering to culture dish, so cells can
21.3.3.4 Conclusion aggregate easily (Huang et al. 2011).
So far, we have discussed cell-based and Since the islet spheroids produced by the
hydrogel/scaffold-based vascularization above methods are highly resistant to hypoxia
techniques that can be used to produce artificial and have high functional efficiency, they can
pancreas via 3D bioprinting. Although each reduce the size of the capsules produced when
method has been described separately, proper used in a 3D bioprinting based transplantation
vascularization through 3D bioprinting is more capsule and as a result, it is possible to scale up
effective when using the above methods together. the manufacture process. Also, they can make the
If we resolve hypoxia, one of the biggest manufacturing process more efficient by reducing
problems of artificial pancreas, through vascular- the possibility of core hypoxia that can occur
ization, we will be able to step closer to commer- during 3D printing. In addition, the small size of
cialization of artificial pancreas. the islet spheroid improves resolution by allowing
smaller nozzles to be used in 3D printing. This
allows the making of more detailed vasculariza-
tion induction structures.
21 3D Bioprinting for Artificial Pancreas Organ 369

These advantages of spheroids increase the Government (MSIP & MOHW). Also, this study was
likelihood that 3D bioprinting based artificial partially supported by a grant of the Korea Health Tech-
nology R&D project through the Korea Health Industry
pancreas can be commercialized. Development Institute (KHIDI), funded by the Ministry of
Health & Welfare, Republic of Korea (grant number:
HI14C2099).
21.5 Conclusion

The ideal strategy for treating T1DM is islet


Reference
transplantation. However, transplanted islets are
rejected from host immune cells. To protect the Algire GH, Legallais FY (1949) Recent developments in
immune response, islets are encapsulated with a the transparent-chamber technique as adapted to the
biocompatible polymer for an immune-barrier. mouse. J Natl Cancer Inst 10(2):225–253
Algire GH, Weaver JM, Prehn RT (1954) Growth of cells
The thickness and pore size of polymer mem-
in vivo in diffusion chambers. I. Survival of
brane surrounding islets can be controlled. There- homografts in immunized mice. J Natl Cancer Inst 15
fore, it protects attack from the immune cells and (3):493–507
provide interchange of small molecules; oxygen, Bak D (2003) Rapid prototyping or rapid production? 3D
printing processes move industry towards the latter.
nutrients, insulin, metabolic wastes, etc.
Assembly Autom 23(4):340–345. https://doi.org/10.
However, the conventional encapsulation 1108/01445150310501190
technology has a significant limitation. First, the Basta G, Montanucci P, Luca G, Boselli C, Noya G,
hypoxia occurs in islets encapsulated at the core Barbaro B, Qi M, Kinzer KP, Oberholzer J, Calafiore
R (2011) Long-term metabolic and immunological
of capsule. That reduces viability of islets. Con-
follow-up of nonimmunosuppressed patients with
sequently, the efficiency of the transplantation is type 1 diabetes treated with microencapsulated
dramatically decreased. Second, it is difficult to islet allografts: four cases. Diabetes Care 34
scale up the capsule. In order to be applied to (11):2406–2409. https://doi.org/10.2337/dc11-0731
Billiet T, Vandenhaute M, Schelfhout J, Van
clinical trial, capsule must be possible to contain
Vlierberghe S, Dubruel P (2012) A review of trends
a large amount of islets. Therefore, scaling up the and limitations in hydrogel-rapid prototyping for tissue
capsule is essential. engineering. Biomaterials 33(26):6020–6041. https://
In this chapter, 3D bioprinting is suggested doi.org/10.1016/j.biomaterials.2012.04.050
Billiet T, Gevaert E, De Schryver T, Cornelissen M,
that a new approach can overcome the conven-
Dubruel P (2014) The 3D printing of gelatin
tional limitation in islet encapsulation. The methacrylamide cell-laden tissue-engineered
printed cells are properly distributed in the 3D constructs with high cell viability. Biomaterials 35
structure by the desired positioning system. Addi- (1):49–62. https://doi.org/10.1016/j.biomaterials.
2013.09.078
tionally, forming the vascularization through 3D
Borg DJ, Bonifacio E (2011) The use of biomaterials in
bioprinting can overcome the hypoxia. In addi- islet transplantation. Curr Diab Rep 11(5):434–444.
tion, scaling up of the 3D structures can be possi- https://doi.org/10.1007/s11892-011-0210-2
ble for two systems depending on the used Borselli C, Ungaro F, Oliviero O, d’Angelo I, Quaglia F,
La Rotonda MI, Netti PA (2010) Bioactivation of
materials; scaffolding system, scaffolding free
collagen matrices through sustained VEGF release
system. However, the development of the artifi- from PLGA microspheres. J Biomed Mater Res A
cial pancreas via 3D bioprinting is actually initial 92a(1):94–102. https://doi.org/10.1002/jbm.a.32332
stage. As donors of islets are shortage, develop- Brauker JH, Carrbrendel VE, Martinson LA, Crudele J,
Johnston WD, Johnson RC (1995) Neovascularization
ment of the artificial pancreas is urgently
of synthetic membranes directed by membrane
required. microarchitecture. J Biomed Mater Res 29
(12):1517–1524. https://doi.org/10.1002/jbm.
Acknowledgements This study was supported by Basic 820291208
Science Research Program Brauker J, Martinson LA, Young SK, Johnson RC (1996)
(NRF-2015R1A2A1A05001832) and partially by the Bio Local inflammatory response around diffusion
& Medical Technology Development Program chambers containing xenografts – nonspecific destruc-
(NRF-2015M3A9E2030125) through the National tion of tissues and decreased local vascularization.
Research Foundation (NRF) funded by the Korean
370 S. J. Lee et al.

Transplantation 61(12):1671–1677. https://doi.org/10. Desai T, Shea LD (2017) Advances in islet encapsulation


1097/00007890-199606270-00002 technologies. Nat Rev Drug Discov 16(5):338–350.
Buder B, Alexander M, Krishnan R, Chapman DW, Lakey https://doi.org/10.1038/nrd.2016.232
JR (2013) Encapsulated islet transplantation: strategies Desai TA, West T, Cohen M, Boiarski T, Rampersaud A
and clinical trials. Immune Netw 13(6):235–239. (2004) Nanoporous microsystems for islet cell replace-
https://doi.org/10.4110/in.2013.13.6.235 ment. Adv Drug Deliv Rev 56(11):1661–1673. https://
Calafiore R, Basta G, Luca G, Lemmi A, Montanucci MP, doi.org/10.1016/j.addr.2003.11.006
Calabrese G, Racanicchi L, Mancuso F, Brunetti P Domingos M, Intranuovo F, Gloria A, Gristina R,
(2006) Microencapsulated pancreatic islet allografts Ambrosio L, Bartolo PJ, Favia P (2013) Improved
into nonimmunosuppressed patients with type 1 diabe- osteoblast cell affinity on plasma-modified 3-D
tes: first two cases. Diabetes Care 29(1):137–138 extruded PCL scaffolds. Acta Biomater 9
Chang TM (1964) Semipermeable Microcapsules. Science (4):5997–6005. https://doi.org/10.1016/j.actbio.2012.
146(3643):524–525 12.031
Chao KC, Chao KF, Fu YS, Liu SH (2008) Islet-like Dufrane D, Goebbels RM, Saliez A, Guiot Y, Gianello P
clusters derived from mesenchymal stem cells in (2006) Six-month survival of microencapsulated pig
Wharton’s jelly of the human umbilical cord for trans- islets and alginate biocompatibility in primates: proof
plantation to control type 1 diabetes. PLoS One 3(1): of concept. Transplantation 81(9):1345–1353. https://
e1451. https://doi.org/10.1371/journal.pone.0001451 doi.org/10.1097/01.tp.0000208610.75997.20
Chen LB, Jiang XB, Yang L (2004) Differentiation of rat Ekser B, Bottino R, Cooper DKC (2016) Clinical islet
marrow mesenchymal stem cells into pancreatic islet xenotransplantation: a step forward. EBioMedicine
beta-cells. World J Gastroenterol 10(20):3016–3020 12:22–23. https://doi.org/10.1016/j.ebiom.2016.09.
Chen XF, Aledia AS, Ghajar CM, Griffith CK, Putnam AJ, 023
Hughes CCW, George SC (2009) Prevascularization of Elliott RB, Escobar L, Tan PL, Muzina M, Zwain S,
a fibrin-based tissue construct accelerates the formation Buchanan C (2007) Live encapsulated porcine islets
of functional anastomosis with host vasculature. Tissue from a type 1 diabetic patient 9.5 yr after xenotrans-
Eng Pt A 15(6):1363–1371. https://doi.org/10.1089/ plantation. Xenotransplantation 14(2):157–161.
ten.tea.2008.0314 https://doi.org/10.1111/j.1399-3089.2007.00384.x
Chick WL, Like AA, Lauris V (1975) Beta cell culture on Figliuzzi M, Cornolti R, Perico N, Rota C, Morigi M,
synthetic capillaries: an artificial endocrine pancreas. Remuzzi G, Remuzzi A, Benigni A (2009) Bone
Science 187(4179):847–849 marrow-derived mesenchymal stem cells improve
Chick WL, Perna JJ, Lauris V, Low D, Galletti PM, islet graft function in diabetic rats. Transplant Proc 41
Panol G, Whittemore AD, Like AA, Colton CK, (5):1797–1800. https://doi.org/10.1016/j.transproceed.
Lysaght MJ (1977) Artificial pancreas using living 2008.11.015
Beta cells – effects on glucose homeostasis in diabetic Fortunato TM, Beltrami C, Emanueli C, De Bank PA, Pula
rats. Science 197(4305):780–782. https://doi.org/10. G (2016) Platelet lysate gel and endothelial progenitors
1126/science.407649 stimulate microvascular network formation in vitro:
Clark H, Barbari TA, Stump K, Rao G (2000) Histologic tissue engineering implications. Sci Rep 6:25326.
evaluation of the inflammatory response around https://doi.org/10.1038/srep25326
implanted hollow fiber membranes. J Biomed Mater Foty R (2011) A simple hanging drop cell culture protocol
Res 52(1):183–192 for generation of 3D spheroids. J Vis Exp 51. https://
Cornolti R, Figliuzzi M, Remuzzi A (2009) Effect of doi.org/10.3791/2720
micro- and macroencapsulation on oxygen consump- Fumimoto Y, Matsuyama A, Komoda H, Okura H, Lee
tion by pancreatic islets. Cell Transplant 18 CM, Nagao A, Nishida T, Ito T, Sawa Y (2009) Crea-
(2):195–201 tion of a rich subcutaneous vascular network with
Daoud JT, Petropavlovskaia MS, Patapas JM, Degrandpre implanted adipose tissue-derived stromal cells and adi-
CE, Diraddo RW, Rosenberg L, Tabrizian M (2011) pose tissue enhances subcutaneous grafting of islets in
Long-term in vitro human pancreatic islet culture using diabetic mice. Tissue Eng Part C Methods 15
three-dimensional microfabricated scaffolds. (3):437–444. https://doi.org/10.1089/ten.tec.2008.
Biomaterials 32(6):1536–1542. https://doi.org/10. 0555
1016/j.biomaterials.2010.10.036 Gates RJ, Smith R, Lazarus NR, Hunt MI (1972) Return to
de Vos P, Spasojevic M, Faas MM (2010) Treatment of normal of blood-glucose, plasma-insulin, and weight-
diabetes with encapsulated islets. Adv Exp Med Biol gain in New Zealand obese mice after implantation of
670:38–53 islets of langerhans. Lancet 2(7777):567–570
Demirdogen B, Elcin AE, Elcin YM (2010) Neovascu- Gunatillake PA, Adhikari R (2003) Biodegradable syn-
larization by bFGF releasing hyaluronic acid-gelatin thetic polymers for tissue engineering. Eur Cell Mater
microspheres: in vitro and in vivo studies. Growth 5:1–16 discussion 16
Factors 28(6):426–436. https://doi.org/10.3109/ Hamamoto Y, Fujimoto S, Inada A, Takehiro M, Nabe K,
08977194.2010.508456 Shimono D, Kajikawa M, Fujita J, Yamada Y, Seino Y
21 3D Bioprinting for Artificial Pancreas Organ 371

(2003) Beneficial effect of pretreatment of islets with Laschke MW, Rucker M, Jensen G, Carvalho C, Mulhaupt R,
fibronectin on glucose tolerance after islet transplanta- Gellrich NC, Menger MD (2008) Improvement of vascu-
tion. Horm Metab Res 35(8):460–465. https://doi.org/ larization of PLGA scaffolds by inosculation of in situ-
10.1055/s-2003-41802 preformed functional blood vessels with the host micro-
Handschel JG, Depprich RA, Kubler NR, Wiesmann HP, vasculature. Ann Surg 248(6):939–948. https://doi.org/10.
Ommerborn M, Meyer U (2007) Prospects of 1097/SLA.0b013e31818fa52f
micromass culture technology in tissue engineering. Lee JS, Hong JM, Jung JW, Shim JH, Oh JH, Cho DW
Head Face Med 3:4. https://doi.org/10.1186/1746- (2014a) 3D printing of composite tissue with complex
160X-3-4 shape applied to ear regeneration. Biofabrication 6
Huang GS, Dai LG, Yen BL, Hsu SH (2011) Spheroid (2):024103. https://doi.org/10.1088/1758-5082/6/2/024103
formation of mesenchymal stem cells on chitosan and Lee VK, Lanzi AM, Ngo H, Yoo SS, Vincent PA, Dai GH
chitosan-hyaluronan membranes. Biomaterials 32 (2014b) Generation of multi-scale vascular network
(29):6929–6945. https://doi.org/10.1016/j.biomaterials. system within 3D hydrogel using 3D bio-printing tech-
2011.05.092 nology. Cell Mol Bioeng 7(3):460–472. https://doi.
Ito T, Itakura S, Todorov I, Rawson J, Asari S, Shintaku J, org/10.1007/s12195-014-0340-0
Nair I, Ferreri K, Kandeel F, Mullen Y (2010) Mesen- Lembert N, Wesche J, Petersen P, Doser M, Zschocke P,
chymal stem cell and islet co-transplantation promotes Becker HD, Ammon HP (2005) Encapsulation of islets
graft revascularization and function. Transplantation in rough surface, hydroxymethylated polysulfone
89(12):1438–1445 capillaries stimulates VEGF release and promotes vas-
Jacobs-Tulleneers-Thevissen D, Chintinne M, Ling Z, cularization after transplantation. Cell Transplant 14
Gillard P, Schoonjans L, Delvaux G, Strand BL, (2–3):97–108
Gorus F, Keymeulen B, Pipeleers D, Beta Cell Therapy Lim F, Sun AM (1980) Microencapsulated islets as
Consortium E-F (2013) Sustained function of alginate- bioartificial endocrine pancreas. Science 210
encapsulated human islet cell implants in the peritoneal (4472):908–910
cavity of mice leading to a pilot study in a type 1 dia- Liu WF, Chen CS (2005) Engineering biomaterials to
betic patient. Diabetologia 56(7):1605–1614. https:// control cell function. Mater Today 8(12):28–35.
doi.org/10.1007/s00125-013-2906-0 https://doi.org/10.1016/S1369-7021(05)71222-0
Jiang FX, Naselli G, Harrison LC (2002) Distinct distribu- Liu J, Zheng HY, Poh PSP, Machens HG, Schilling AF
tion of laminin and its integrin receptors in the pan- (2015) Hydrogels for engineering of Perfusable vascu-
creas. J Histochem Cytochem 50(12):1625–1632. lar networks. Int J Mol Sci 16(7):15997–16016
https://doi.org/10.1177/002215540205001206 MacGregor RR, Williams SJ, Tong PY, Kover K, Moore
Kaido T, Yebra M, Cirulli V, Montgomery AM (2004) WV, Stehno-Bittel L (2005) Small rat islets are supe-
Regulation of human beta-cell adhesion, motility, and rior to large islets in in vitro function and in transplan-
insulin secretion by collagen IV and its receptor tation outcomes. Am J Physiol Endocrinol Metab 290
alpha1beta1. J Biol Chem 279(51):53762–53769. (5):771–779
https://doi.org/10.1074/jbc.M411202200 Marchioli G, van Gurp L, van Krieken PP, Stamatialis D,
Kaigler D, Silva EA, Mooney DJ (2013) Guided bone Engelse M, van Blitterswijk CA, Karperien MBJ, de
regeneration using injectable vascular endothelial Koning E, Alblas J, Moroni L, van Apeldoorn AA
growth factor delivery gel. J Periodontol 84 (2015) Fabrication of three-dimensional bioplotted
(2):230–238. https://doi.org/10.1902/jop.2012.110684 hydrogel scaffolds for islets of Langerhans transplan-
Keskar V, Gandhi M, Gemeinhart EJ, Gemeinhart RA tation. Biofabrication 7(2):025009
(2009) Initial evaluation of vascular ingrowth into Mehta G, Hsiao AY, Ingram M, Luker GD, Takayama S
superporous hydrogels. J Tissue Eng Regen M 3 (2012) Opportunities and challenges for use of tumor
(6):486–490. https://doi.org/10.1002/term.183 spheroids as models to test drug delivery and efficacy.
King D, Tansey T (2003) Rapid tooling: selective laser J Control Release 164:192–204
sintering injection tooling. J Mater Process Tech 132 Melchels FPW, Domingos MAN, Klein TJ, Malda J, Bartolo
(1–3):42–48. Pii S0924-0136(02)00257-1. doi: https:// PJ, Hutmacher DW (2012) Additive manufacturing of
doi.org/10.1016/S0924-0136(02)00257-1 tissues and organs. Prog Polym Sci 37(8):1079–1104.
Klock G, Frank H, Houben R, Zekorn T, Horcher A, https://doi.org/10.1016/j.progpolymsci.2011.11.007
Siebers U, Wohrle M, Federlin K, Zimmermann U Menard M, Dusseault J, Langlois G, Baille WE, Tam SK,
(1994) Production of purified alginates suitable for Yahia L, Zhu XX, Halle JP (2010) Role of protein
use in immunoisolated transplantation. Appl Microbiol contaminants in the immunogenicity of alginates. J
Biotechnol 40(5):638–643 Biomed Mater Res B Appl Biomater 93(2):333–340.
Ko C, Dixit V, Shaw W, Gitnick G (1995) In-vitro slow- https://doi.org/10.1002/jbm.b.31570
release profile of endothelial-cell growth-factor Monaco AP, Maki T, Ozato H, Carretta M, Sullivan SJ,
immobilized within calcium alginate microbeads. Borland KM, Mahoney MD, Chick WL, Muller TE,
Artif Cell Blood Sub 23(2):143–151. https://doi.org/ Wolfrum J et al (1991) Transplantation of
10.3109/10731199509117934 islet allografts and xenografts in totally
372 S. J. Lee et al.

pancreatectomized diabetic dogs using the hybrid arti- Pedraza E, Coronel MM, Fraker CA, Ricordi C, Stabler
ficial pancreas. Ann Surg 214(3):339–360 discussion CL (2012) Preventing hypoxia-induced cell death in
361–332 beta cells and islets via hydrolytically activated,
Mrksich M (2000) A surface chemistry approach to study- oxygen-generating biomaterials. P Natl Acad Sci
ing cell adhesion. Chem Soc Rev 29(4):267–273. USA 109(11):4245–4250
https://doi.org/10.1039/a705397e Pescosolido L, Schuurman W, Malda J, Matricardi P,
Murphy SV, Atala A (2014) 3D bioprinting of tissues and Alhaique F, Coviello T, van Weeren PR, Dhert WJA,
organs. Nat Biotechnol 32(8):773–785. https://doi.org/ Hennink WE, Vermonden T (2011) Hyaluronic acid
10.1038/nbt.2958 and dextran-based semi-IPN hydrogels as. Biomater
Nakakita N, Sezaki K, Yamazaki Y, Uchinuma E (1999) Bioprinting Biomacromolecules 12(5):1831–1838.
Augmentation rhinoplasty using an L-shaped auricular https://doi.org/10.1021/bm200178w
cartilage framework combined with dermal fat graft for Prehn RT, Weaver JM, Algire GH (1954) The diffusion-
cleft lip nose. Aesthet Plast Surg 23(2):107–112. chamber technique applied to a study of the nature of
https://doi.org/10.1007/s002669900251 homograft resistance. J Natl Cancer Inst 15
Nichol JW, Koshy ST, Bae H, Hwang CM, Yamanlar S, (3):509–517
Khademhosseini A (2010) Cell-laden microengineered Qi M (2014) Transplantation of encapsulated pancreatic
gelatin methacrylate hydrogels. Biomaterials 31 islets as a treatment for patients with type 1 diabetes
(21):5536–5544. https://doi.org/10.1016/j. mellitus. Adv Med 2014:429710. https://doi.org/10.
biomaterials.2010.03.064 1155/2014/429710
Nomi M, Miyake H, Sugita Y, Fujisawa M, Soker S Ramachandran K, Huang H-H, Stehno-Bittel L (2015) A
(2006) Role of growth factors and endothelial cells in simple method to replace islet equivalents for volume
therapeutic angiogenesis and tissue engineering. Curr quantification of human islets. Cell Transplant
Stem Cell Res Ther 1(3):333–343 24:1183–1194
Norotte C, Marga FS, Niklason LE, Forgacs G (2009) Rivron NC, Liu J, Rouwkema J, de Boer J, van
Scaffold-free vascular tissue engineering using Blitterswijk CA (2008) Engineering vascularised
bioprinting. Biomaterials 30(30):5910–5917 tissues in vitro. Eur Cells Mater 15:27–40
Novosel EC, Kleinhans C, Kluger PJ (2011) Vasculariza- Rouwkema J, Rivron NC, van Blitterswijk CA (2008)
tion is the key challenge in tissue engineering. Adv Vascularization in tissue engineering. Trends
Drug Deliver Rev 63(4–5):300–311. https://doi.org/10. Biotechnol 26(8):434–441. https://doi.org/10.1016/j.
1016/j.addr.2011.03.004 tibtech.2008.04.009
O’Shea GM, Goosen MF, Sun AM (1984) Prolonged Ryan EA, Lakey JR, Rajotte RV, Korbutt GS, Kin T,
survival of transplanted islets of Langerhans Imes S, Rabinovitch A, Elliott JF, Bigam D, Kneteman
encapsulated in a biocompatible membrane. Biochim NM, Warnock GL, Larsen I, Shapiro AM (2001) Clin-
Biophys Acta 804(1):133–136 ical outcomes and insulin secretion after islet trans-
O’Sullivan ES, Johnson AS, Omer A, Hollister-Lock J, plantation with the Edmonton protocol. Diabetes 50
Bonner-Weir S, Colton CK, Weir GC (2010) Rat islet (4):710–719
cell aggregates are superior to islets for transplantation Sabek OM, Farina M, Fraga DW, Afshar S, Ballerini A,
in microcapsules. Diabetologia 53(5):937–945. https:// Filgueira CS, Thekkedath UR, Grattoni A and Gaber
doi.org/10.1007/s00125-009-1653-8 AO (2016) Three-dimensional printed polymeric sys-
Orsetti A, Guy C, Zouari N, Deffay R (1978) Implantation tem to encapsulate human mesenchymal stem cells
of a bio-artificial insulin distributor in dog, using islets differentiated into islet-like insulin-producing
of Langerhans from different animal species. Cr Soc aggregates for diabetes treatment. J Tissue Eng
Biol 172(1):144–150 7 2041731416638198. https://doi.org/10.1177/
Otterlei M, Ostgaard K, Skjak-Braek G, Smidsrod O, 2041731416638198
Soon-Shiong P, Espevik T (1991) Induction of cyto- Sakata N, Sumi S, Yoshimatsu G, Goto M, Egawa S, Unno
kine production from human monocytes stimulated M (2012) Encapsulated islets transplantation: past,
with alginate. J Immunother 10(4):286–291 present and future. World J Gastrointest Pathophysiol
Ozbolat IT (2015) Bioprinting scale-up tissue and organ 3(1):19–26. https://doi.org/10.4291/wjgp.v3.i1.19
constructs for transplantation. Trends Biotechnol 33 Santos MI, Reis RL (2010) Vascularization in bone tissue
(7):395–400. https://doi.org/10.1016/j.tibtech.2015. engineering: physiology, current strategies, major
04.005 hurdles and future challenges. Macromol Biosci 10
Ozbolat IT, Yu Y (2013) Bioprinting toward organ fabri- (1):12–27. https://doi.org/10.1002/mabi.200900107
cation: challenges and future trends. IEEE Trans Sato Y, Endo H, Okuyama H, Takeda T, Iwahashi H,
Biomed Eng 60(3):691–699. https://doi.org/10.1109/ Imagawa A, Yamagata K, Shimomura I, Inoue M
TBME.2013.2243912 (2011) Cellular hypoxia of pancreatic beta-cells due
Pati F, Gantelius J, Svahn HA (2016) 3D bioprinting of tissue/ to high levels of oxygen consumption for insulin secre-
organ models. Angew Chem Int Ed Engl 55 tion in vitro. J Biol Chem 286(14). https://doi.org/10.
(15):4650–4665. https://doi.org/10.1002/anie.201505062 1074/jbc.M110.194738
21 3D Bioprinting for Artificial Pancreas Organ 373

Sawhney AS, Pathak CP, Hubbell JA (1993) Interfacial Millipore filter capsules. Diabetologia 6(3):306–312.
photopolymerization of poly(ethylene glycol)-based https://doi.org/10.1007/Bf01212243
hydrogels upon alginate-poly(l-lysine) microcapsules Sun AM, Parisius W, Healy GM, Vacek I, Macmorine HG
for enhanced biocompatibility. Biomaterials 14 (1977) Use, in diabetic rats and monkeys, of artificial
(13):1008–1016 capillary units containing cultured islets of Langerhans
Scharp DW, Marchetti P (2014) Encapsulated islets for (artificial endocrine pancreas). Diabetes 26
diabetes therapy: history, current progress, and critical (12):1136–1139. https://doi.org/10.2337/diabetes.26.
issues requiring solution. Adv Drug Deliver Rev 12.1136
67–68:35–73. https://doi.org/10.1016/j.addr.2013.07. Sun A, Parisius W, Macmorine H, Sefton M, Stone R
018 (1980) An artificial endocrine pancreas containing
Schweicher J, Nyitray C, Desai TA (2014) Membranes to cultured islets of Langerhans. Artif Organs 4
achieve immunoprotection of transplanted islets. Front (4):275–278
Biosci (Landmark Ed) 19:49–76 Sutherland RM, Carlsson J, Durand R (1981) Spheroids in
Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, cancer research. Cancer Res 41:2980–2984
Warnock GL, Kneteman NM, Rajotte RV (2000) Islet Suzuki K, Bonner-Weir S, Trivedi N, Yoon KH, Hollister-
transplantation in seven patients with type 1 diabetes Lock J, Colton CK, Weir GC (1998) Function and
mellitus using a glucocorticoid-free immunosuppres- survival of macroencapsulated syngeneic islets
sive regimen. N Engl J Med 343(4):230–238. https:// transplanted into streptozocin-diabetic mice. Trans-
doi.org/10.1056/NEJM200007273430401 plantation 66(1):21–28. https://doi.org/10.1097/
Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, 00007890-199807150-00004
Lindblad R, Robertson RP, Secchi A, Brendel MD, Tabriz AG, Hermida MA, Leslie NR, Shu WM (2015)
Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan Three-dimensional bioprinting of complex cell laden
EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, alginate hydrogel structures. Biofabrication 7(4)
Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Tarantal AF, Lee CC, Itkin-Ansari P (2009) Real-time biolu-
Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, minescence imaging of macroencapsulated fibroblasts
Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, reveals allograft protection in rhesus monkeys (Macaca
Bluestone J, Lakey JR (2006) International trial of the mulatta). Transplantation 88(1):38–41. https://doi.org/10.
Edmonton protocol for islet transplantation. N Engl J 1097/TP.0b013e3181a9ee6c
Med 355(13):1318–1330. https://doi.org/10.1056/ Thomas FT, Contreras JL, Bilbao G, Ricordi C, Curiel D,
NEJMoa061267 Thomas JM (1999) Anoikis, extracellular matrix, and
Shim JH, Kim JY, Park M, Park J, Cho DW (2011) apoptosis factors in isolated cell transplantation. Sur-
Development of a hybrid scaffold with synthetic gery 126(2):299–304
biomaterials and hydrogel using solid freeform fabri- Tremblay PL, Hudon V, Berthod F, Germain L, Auger FA
cation technology. Biofabrication 3(3):034102. https:// (2005) Inosculation of tissue-engineered capillaries
doi.org/10.1088/1758-5082/3/3/034102 with the host's vasculature in a reconstructed skin
Shim JH, Lee JS, Kim JY, Cho DW (2012) Bioprinting of transplanted on mice. Am J Transplant 5
a mechanically enhanced three-dimensional dual cell- (5):1002–1010. https://doi.org/10.1111/j.1600-6143.
laden construct for osteochondral tissue engineering 2005.00790.x
using a multi-head tissue/organ building system. J Tuch BE, Keogh GW, Williams LJ, Wu W, Foster JL,
Micromech Microeng 22(8):085014. https://doi.org/ Vaithilingam V, Philips R (2009) Safety and viability
10.1088/0960-1317/22/8/085014 of microencapsulated human islets transplanted into
Soon-Shiong P, Heintz RE, Merideth N, Yao QX, Yao Z, diabetic humans. Diabetes Care 32(10):1887–1889.
Zheng T, Murphy M, Moloney MK, Schmehl M, https://doi.org/10.2337/dc09-0744
Harris M et al (1994) Insulin independence in a type Tze WJ, Wong FC, Chen LM, Oyoung S (1976) Implant-
1 diabetic patient after encapsulated islet transplanta- able artificial endocrine pancreas unit used to restore
tion. Lancet 343(8903):950–951 Normoglycaemia in diabetic rat. Nature 264
Souza YE, Chaib E, Lacerda PG, Crescenzi A, Bernal- (5585):466–467. https://doi.org/10.1038/264466a0
Filho A, D’Albuquerque LA (2011) Islet transplanta- Tze WJ, Tai J, Wong FC, Davis HR (1980) Studies with
tion in rodents. Do encapsulated islets really work? implantable artificial capillary units containing rat
Arq Gastroenterol 48(2):146–152 islets on diabetic dogs. Diabetologia 19(6):541–545
Stevens MM, George JH (2005) Exploring and engineer- Valdes-Gonzalez RA, Dorantes LM, Garibay GN, Bracho-
ing the cell surface interface. Science 310 Blanchet E, Mendez AJ, Davila-Perez R, Elliott RB,
(5751):1135–1138. https://doi.org/10.1126/science. Teran L, White DJ (2005) Xenotransplantation of por-
1106587 cine neonatal islets of Langerhans and Sertoli cells: a
Storrs R, Dorian R, King SR, Lakey J, Rilo H (2001) 4-year study. Eur J Endocrinol 153(3):419–427.
Preclinical development of the islet sheet. Ann N Y https://doi.org/10.1530/eje.1.01982
Acad Sci 944:252–266 Valdes-Gonzalez R, Rodriguez-Ventura AL, White DJ,
Strautz RL (1970) Studies of hereditary-obese mice Bracho-Blanchet E, Castillo A, Ramirez-Gonzalez B,
(Obob) after implantation of pancreatic islets in Lopez-Santos MG, Leon-Mancilla BH, Dorantes LM
374 S. J. Lee et al.

(2010) Long-term follow-up of patients with type 1 dia- Wust S, Muller R, Hofmann S (2011) Controlled position-
betes transplanted with neonatal pig islets. Clin Exp ing of cells in biomaterials-approaches towards 3D
Immunol 162(3):537–542. https://doi.org/10.1111/j. tissue printing. J Funct Biomater 2(3):119–154.
1365-2249.2010.04273.x https://doi.org/10.3390/jfb2030119
Veiseh O, Doloff JC, Ma M, Vegas AJ, Tam HH, Bader Wust S, Godla ME, Muller R, Hofmann S (2014) Tunable
AR, Li J, Langan E, Wyckoff J, Loo WS, hydrogel composite with two-step processing in com-
Jhunjhunwala S, Chiu A, Siebert S, Tang K, bination with innovative hardware upgrade for cell-
Hollister-Lock J, Aresta-Dasilva S, Bochenek M, based three-dimensional bioprinting. Acta Biomater
Mendoza-Elias J, Wang Y, Qi M, Lavin DM, 10(2):630–640. https://doi.org/10.1016/j.actbio.2013.
Chen M, Dholakia N, Thakrar R, Lacik I, Weir GC, 10.016
Oberholzer J, Greiner DL, Langer R, Anderson DG Yan YN, Wang XH, Xiong Z, Liu HX, Liu F, Lin F, Wu
(2015) Size- and shape-dependent foreign body RD, Zhang RJ, Lu QP (2005) Direct construction of a
immune response to materials implanted in rodents three-dimensional structure with cells and hydrogel. J
and non-human primates. Nat Mater 14(6):643–651. Bioact Compat Pol 20(3):259–269. https://doi.org/10.
https://doi.org/10.1038/nmat4290 1177/0883911505053658
Wang T, Lacik I, Brissova M, Anilkumar AV, Prokop A, Yeo M, Lee JS, Chun W, Kim GH (2016) An innovative
Hunkeler D, Green R, Shahrokhi K, Powers AC (1997) collagen-based cell-printing method for obtaining
An encapsulation system for the immunoisolation of human adipose stem cell-laden structures consisting
pancreatic islets. Nat Biotechnol 15(4):358–362. of Core sheath structures for tissue engineering.
https://doi.org/10.1038/nbt0497-358 Biomacromolecules 17(4):1365–1375. https://doi.org/
Wang XH, Yan YN, Pan YQ, Xiong Z, Liu HX, Cheng B, 10.1021/acs.biomac.5b01764
Liu F, Lin F, Wu RD, Zhang RJ, Lu QP (2006) Gener- Zadpoor AA, Malda J (2017) Additive manufacturing of
ation of three-dimensional hepatocyte/gelatin biomaterials, tissues, and organs. Ann Biomed Eng 45
structures with rapid prototyping system. Tissue Eng (1):1–11. https://doi.org/10.1007/s10439-016-1719-y
12(1):83–90. https://doi.org/10.1089/ten.2006.12.83

You might also like