Sei sulla pagina 1di 16

Biochemical Pharmacology 100 (2016) 1227

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Research update

The bioarticial pancreas (BAP): Biological, chemical and engineering


challenges
Veronica Iacovacci* , Leonardo Ricotti, Arianna Menciassi, Paolo Dario
The BioRobotics Institute, Scuola Superiore SantAnna, Viale R. Piaggio 34, 56025 Pontedera (PI), Italy

A R T I C L E I N F O A B S T R A C T

Article history: The bioarticial pancreas (BAP) represents a viable solution for the treatment of type 1 diabetes (T1D). By
Received 25 May 2015 encapsulating pancreatic cells in a semipermeable membrane to allow nutrient, insulin and glucose
Accepted 26 August 2015 exchange, the side effects produced by islets and whole organ transplantation-related immunosuppres-
Available online 4 September 2015
sive therapy can be circumvented. Several factors, mainly related to materials properties, capsule
morphology and biological environment, play a key role in optimizing BAP systems.
Keywords: The BAP is an extremely complex delivery system for insulin. Despite considerable efforts, in some
Bioarticial pancreas
instances meeting with limited degree of success, a BAP capable of restoring physiological pancreas
Cell encapsulation
Type 1 diabetes
functions without the need for immunosuppressive drugs and of controlling blood glucose levels
Biomaterials especially in large animal models and a few clinical trials, does not exist. The state of the art in terms of
Stem cells materials, fabrication techniques and cell sources, as well as the current status of commercial devices and
clinical trials, are described in this overview from an interdisciplinary viewpoint. In addition, challenges
to the creation of effective BAP systems are highlighted including future perspectives in terms of
component integration from both a biological and an engineering viewpoint.
2015 Elsevier Inc. All rights reserved.

1. Introduction more than 29 million suffer from this disease [7] with the number
of deaths due to diabetes expected to double in the next decade [8].
The pancreas, from the Greek pan (all) and kreas (esh), is a 12 T1D is typically managed by exogenous insulin via the use of
15 cm-long soft, lobulated, retroperitoneal organ with both insulin pens, multiple injections or subcutaneous pumps. The
exocrine and endocrine functions [1]. The islets of Langerhans, amount of insulin is calculated on the basis of multiple daily self-
representing approximately 2 million cells in human adults (about monitoring blood glucose (BG) measurements performed by the
1% of pancreatic tissue), enable endocrine functions [2]. In patient and of rules identied by diabetologists. In all cases,
particular, islets b-cells are responsible for the regulation of blood discrete monitoring and bolus-type insulin administration occur.
glucose levels through the production of insulin. When pancreatic These strategies are unable to reproduce a physiological insulin
b-cells are missing or damaged due to autoimmune mechanisms, prole, thus markedly affecting the daily life of patients and giving
type I diabetes (T1D) mellitus occurs resulting in a disorder of rise to secondary complications, such as hyperglycemia or
glucose homeostasis, insulin deciency and hyperglycemia [3,4]. hypoglycemic events that affect the patients health and wellbeing
The rst description of diabetes symptoms dates back to the in the long run.
16th century BC, but the awareness of diabetes associated with In order to overcome the limitations of currently available
pancreatic functions, and the rst attempts to treat it through therapeutic strategies, several solutions have been evaluated,
transplantation or, later on, through exogenous insulin supply, ranging from whole pancreas or islet transplantation, to articial
dates back to the late 19th century [5]. Diabetes is a highly pancreas (AP) systems [9]. This latter solution represents a holy
widespread disease with approximately 347 million individuals grail of diabetes treatment and has been accomplished, albeit only
affected worldwide (10% of whom have T1D) [6]. In America alone, partially, by developing and then combining together portable and
implantable insulin pumps, continuous glucose monitoring
systems (CGMs) and closed loop control algorithms [1012].
Currently available APs show several limitations mainly related to
the delays in glucose sensing and insulin absorption due to the
* Corresponding author. Tel.: +39 050 883074.
E-mail address: v.iacovacci@sssup.it (V. Iacovacci).
subcutaneous site (most commonly exploited both for sensing and

http://dx.doi.org/10.1016/j.bcp.2015.08.107
0006-2952/ 2015 Elsevier Inc. All rights reserved.
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 13

delivery). Another issue is the relatively short lifetime of glucose A compromise for reproducing a natural insulin release prole
sensors [13]. Current research efforts are aimed at enhancing AP while avoiding some of the drawbacks of whole pancreas
performance in order to develop a totally implantable articial transplantation, is represented by islets of Langerhans or b-cell
organ characterized by a more effective insulin supply route e.g., transplantation. The implantation of whole islets was proposed for
the intraperitoneal route, new glucose sensing technologies the rst time by Lacy and Kostianovsky in 1967 [28]. As in the case
[14,15], higher reliability, longer lifetime and new control of pancreas transplantation, irrespective of the source of the
algorithms that reect real time blood glucose levels [16,17]. implanted islets, immunosuppressive drug administration is
Smart procedures that have been recently proposed include required to avoid rejection of the foreign tissue. Thus, methods
miniaturized tools for articial organ relling, functionalized for cell transplantation which do not require the use of
materials, etc. which may in time enable the development of a immunosuppressive drugs are highly desirable. To this aim, the
totally implantable AP [18,19]. Considerable work is still needed to development of semi-permeable, immunoisolating and biocom-
achieve reliable and safe solutions. patible membranes for b-cells or pancreatic islet encapsulation has
Another intriguing approach is based on insulin-containing and been pursued. The encapsulating membrane, typically polymeric,
glucose-responsive materials, which are able to automatically has to be designed to allow the selective permeation of oxygen,
release insulin when the glucose level in the microenvironment is glucose, nutrients, waste products and insulin, and should be also
above a certain threshold [2025]. Although based on smart able to inhibit immune rejection of the encapsulated cells. When
materials and highly miniaturized technologies, this research line referring to b-cells or pancreatic islets encapsulated within a
can be considered as a part of the fully articial pancreas semipermeable, biocompatible membrane, the term bioarticial
approach, since no living elements are embedded in the system. pancreas (BAP) is used [29].
Despite good promises, the issue of insulin depletion in these small This research overview focuses on some of the issues related to
reservoirs (thus limiting their lifetime) is still unsolved. BAP design, with the aim to give an original viewpoint on the topic,
Whole pancreas transplantation represents an effective based not only on biology and materials science, but also on
solution to restore normoglycemia [26]. The introduction of this engineering and mechatronic approaches. Thus, the description of
type of procedure dates back to 1894 (about 30 years or so before BAP development and pre-clinical/clinical applications has been
the discovery of insulin) when the rst xenogenic pancreas enriched via a highly interdisciplinary approach. Section 2
transplantations were performed [27]. On the one hand, this describes a classication of BAP systems, highlighting the
solution can avoid exogenous insulin supply and physiologically advantages and disadvantages of the different strategies reported
restore patient responsiveness to the increased blood glucose in the literature. Section 3 deals with the materials and with the
levels. On the other hand, the shortage of donors, the complexity fabrication techniques exploited for islet encapsulation, highlight-
of transplantation surgical procedures and the need for ing current and future trends of these research efforts. Section 4
immunosuppressive drug therapy to avoid organ rejection, provides an insight on the most critical aspects of the biology and
which by itself produces severe side effects, make pancreas the challenges that need to be taken into account for BAP
replacement a controversial solution for T1D treatment. It is development. The traditional cell sources used and the promise of
often considered a viable option when kidney replacement is stem cell and tissue engineering-based strategies will be analyzed
necessary as well. as well. Insights on recent successful devices, encapsulation

Fig. 1. The bioarticial paancreas approach. (A) Schematic representation of the diffusion mechanism across a semipermeable membrane. The membrane should favor
glucose and nutrients diffusion inside the capsule and insulin and waste diffusion out of it. At the same time, the entry of immune cells should be avoided. (B) Typical
bioarticial pancreas (BAP) implant sites, ranging from the subcutaneous site and peritoneal cavity (most common ones) to the kidney capsule and omentum pouch. (C)
Classication and average dimensions of extravascular encapsulation devices.
14 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

strategies and clinical trials are described in Section 5 with a view to distinguish between macrocapsules (cm), microcapsules (250
to documenting progress in the clinical scenario. Finally Section 6 1000 mm) and nanocapsules (<100 mm; Fig. 1C).
outlines advances in technological components and engineering
solutions, which could contribute in the future to the production of 2.1. Macroencapsulation
safe and effective BAPs.
A macrocapsule is an implantable device in which a large
2. The bioarticial pancreas approach number of pancreatic cells is entrapped with encapsulation
dimensions in the centimeter range. Depending on the implant
Islet transplantation has been explored as a solution for the site, macroencapsulation systems can be distinguished into:
treatment of T1D since the development of collagenase digestion extravascular, typically placed in the peritoneal cavity or subcuta-
of the pancreas [30], the initial step in islet isolation. The rst case neously or intravascular, which are connected to the patient's
of human insulin-independence after the transplantation of cardiovascular system via a shunt [47]. Due to their relatively large
pancreatic islets through intraportal hepatic infusion occurred dimension, extravascular macrocapsules can be made of different
in 1989 [31]. The most outstanding clinical trial in the eld of islet materials: one for the extracellular matrix (ECM) enabling islet
transplantation with steroid-free immunosuppression is known as viability [48], another for the immunoisolating membrane, and an
the Edmonton protocol and was successfully performed in 2000 external surface coating to promote neovascularization [49]. Their
by Shapiro et al. [32]. Seven recipients were insulin-independent a dimensions also favor the formation of cell clusters, enabling
year after transplantation with an acceptable reduction in insulin intercellular communication and better mimicking organ physiol-
dependency and hypoglycemia episodes, but at year 5 the graft ogy [50]. Furthermore, both the dimensions and implant site allow
survival rate was only 10% [33]. The risks associated with this type the use of minimally invasive surgical procedures and enable
of procedure include: thrombogenesis caused by blood-mediated capsule retrieve in case of failure. Macrocapsules can rely on
inammatory reaction; the loss of graft mass; the inability to diffusion mechanisms or ultraltration. Diffusion chambers have
retrieve the islets from the liver [34]; and the immunosuppressive been adopted both for intravascular and extravascular devices
drug therapy required to allow tissue engraftment and immuno- whereas ultraltration has been explored only in intravascular
logic acceptance. In general, even if a reduction in immunosup- systems.
pressive therapy is feasible [35], it is still required for islet The main drawbacks of extravascular macrocapsules involve
transplants. The complications associated with immunosuppres- low surface-to-volume ratios and constraints related to the eligible
sant administration include an increased incidence of infection implant site that do not promote neovascularization. Due to the
and malignancy, decreased wound healing and renal dysfunction lack of a direct vascular access, oxygen and glucose diffusion time,
[3641]. Islet-transplant immunosuppressive regimes can also and insulin release delay are worsened, increasing the risk of
attenuate the efcacy of the therapy itself, by reducing glucose necrosis of cells far from the semipermeable membrane and thus
responsivity [42]. calling for small devices. At the same time, due to the dimensions, a
A BAP has the potential to provide all the benets of islet relatively low cell density (510% of the macrocapsule volume) is
transplantation without the morbidities associated with immuno- desirable, to ensure adequate nutrition [51]. This is somehow
suppressive drugs making xenografts a reality also. The concept of contradictory: lower cell densities claim for larger devices, which
a BAP derived from the success of islet transplantation in so-called imply more signicant diffusion problems. A compromise between
immunoprivileged sites, e.g., the anterior chamber of the eye these conicting needs must be targeted.
[43]. The idea of encapsulating cells in a semipermeable Intravascular macrocapsules allow implanted pancreatic islets
membrane (Fig. 1A) was developed: the introduction of this to remain in direct contact with blood vessels, allowing more
technology dates back to 1933 with the work of Bisceglie et al. [44]. accurate tracking of blood glucose level, insulin absorption delay
Since then efforts have focused on reducing material immunoge- reduction and optimal oxygenation of the implanted cells [52].
nicity and optimizing the implant site (Fig. 1B), capsule dimen- These devices however, also have limitations. Their implantation
sions, characterization and testing procedures, cell sources, and as shunts in blood vessels is a complex procedure while blood
other issues to make this technology sufciently mature for clinical coagulation and platelet deposition, latter causing thrombosis, are
use [45]. potential complications. Blood coagulation is highly limiting, as
To be efcacious, an encapsulation system should permit patients affected by T1D cannot be treated with systemic
optimal viability and function of the pancreatic cells including: (i) anticoagulation medication. These adverse events have led to a
immunoisolation, to avoid the encapsulated cells coming into decrease in intravascular macrocapsule development, although
contact with immune cells, antibodies, complement, etc.; (ii) advances in materials science and technology have renewed
sensitivity to stimulus and rapidity in reactionencapsulated cells interest in this approach [50].
should be able to rapidly detect an increase in the blood glucose
level and to promptly release insulin, thus preventing delays in 2.2. Microencapsulation
blood glucose regulation and avoiding the hormone from staying in
proximity to the cell; (iii) biocompatibilitythe material infrastruc- An alternative to macrocapsules are microcapsules that consist
ture of the BAP should not stimulate brosis and should not of a semipermeable membrane enclosing a small number of islets
activate inammatory processes; (iv) ease of implantation via the of Langerhans (usually 13), suspended in a polymeric gel matrix
use of minimally invasive surgical procedures; (v) long life; and (vi) (e.g., alginate). The smaller dimensions and the higher surface-to-
retrievability in case of failure or unexpected complications. volume ratio assure better diffusion characteristics and enable
The eld of BAP is an area of intensive research with an entire implantation by means of minimally invasive procedures in
issue of Advanced Drug Delivery Reviews dedicated to the topic in locations such as the peritoneal cavity, the kidney capsule or
2014 [46]. To better characterize BAPs and their design require- subcutaneous sites [5254]. However, the limited vascularization
ments, an accurate classication of these systems is required. One of these sites hampers BAP function. Thus, alternative implant sites
classication criterion describes the amount of islet cells such as the omentum pouches have been investigated [55]. In any
encapsulated within the semipermeable membrane, thus the case, the size of microcapsules makes them difcult to retrieve in
dimensions of the implantable capsules provide a system in order the event of failure [56].
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 15

Fabrication strategies and the choice of materials are critical 2.3. Nanoencapsulation
issues in the development of microencapsulation systems. Optimal
pore size and a proper membrane thickness are required to provide To exploit narrower implant sites and overcome diffusion-
immunoprotection and a correct nutrient diffusion. Furthermore, related issues, nanoencapsulation devices have been investigated.
the constitutive material itself should favor cell viability resem- These are less than 100 mm in diameter and can envelop single
bling as closely as possible the natural environment of the cells pancreatic b-cells in a semipermeable membrane, directly bound
[57]. to the cell membrane with a thickness varying between some
A spherical shape is advantageous since it guarantees a high nanometers to some micrometers. Different polyelectrolytes have
surface-to-volume ratio, a short and uniform diffusion time and been successfully investigated to this aim [59,60] and nano-
allows the implantation via injection. As already mentioned, capsules appear particularly advantageous in terms of nutrients
microcapsules range in size from 250 to 1000 mm in diameter. A diffusion and insulin release thanks to their really small
practical rule in designing a microencapsulation system is to have dimensions. One great challenge, but at the same time a great
cells no more than 200 mm from the membrane [54,56,58] to possibility, offered by nanoencapsulation lies in the possibility to
ensure an effective exchange of insulin and glucose, nutrients, embed other nanocomponents, e.g., nanoparticles, in the nano-
oxygen and waste for all encapsulated cells. layers enhancing BAP acceptability and function [61,62].

Fig. 2. Overview of islet encapsulation materials and classication. They can be grouped in polymers, either natural or synthetic, and inorganic materials. For each material
class, details on specic materials (blue box) are provided including chemical formulation, fabrication techniques employed (in red) and a picture from literature showing an
example of encapsulation system with such specic material (references to picture sources are reported in the gure). Natural polymers include: collagen-alginate spheroids
(adapted from [87], reproduced with permission of Elsevier), agarose-based porcine islets encapsulation (adapted with permission from [84] (copyright: Creative Commons))
and alginate-encapsulated human islets (adapted from [205], reproduced with permission of Springer) are reported. Within synthetic polymers, the most commonly
employed materials and material classes are polyethylene glycol (PEG), thermoplastic polymers such as polyvinyl alcohol (PVA) and polyelectrolytes. Examples from
literature are reported (adapted and reproduced from [156] and [130] with permission of Elsevier, and from [154] with permission of American Chemical Society,
respectively). Inorganic materials include silicon (adapted from [134], reproduced with permission of Elsevier), aluminium oxide (adapted from [131], reproduced with
permission of Elsevier), titanium oxide (adapted from [143], reproduced with permission of American Chemical Society) and SU-8 (adapted from [146], reproduced with
permission of AIP Publishing LCC). Membranes pictures based on these materials are reported as examples. The green box points out the techniques that can be exploited for
nanoencapsulation. (For interpretation of the references to color in this gure legend, the reader is referred to the web version of this article.)
16 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

3. Biomaterials for islet encapsulation allow easy capsule fabrication [82]. Agarose [83,84], cellulose
sulfate [85,86] and collagen [87] are also used for cell encapsula-
While islet cell encapsulation represents a promising solution tion with alginate being the most successfully employed natural
for the treatment of T1D, appropriate functional biomaterials that polymer [88].
are suitable for this application and allow BAPs to function Agarose, was used by Iwata et al. in 1988 for the encapsulation
correctly are required. of pancreatic islets and successfully tested in mice [84,89], and
Biomaterials can be distinguished from any other material for dogs [83]. It can be used both for micro and macroencapsulation
their ability to enter in contact with tissues of the human body systems by using temperature-induced gelation processes, and
without causing unacceptable consequences for the recipient [63]. simple extrusion systems [90] and by varying the agarose
For islet encapsulation, material biocompatibility is a fundamental concentration to rene the mechanical properties of the beads
issue where biocompatibility is dened as the ability of a material [91]. To facilitate immunoprotection and graft survival time,
to perform with an appropriate host response in a specic increasing agarose concentration, altering coatings or combining
application [64]. In the case of encapsulated cells, an inamma- agarose with other polymers such as collagen and alginate, have
tory response can result in cell overgrowth on the capsule surface been evaluated as possible strategies. However, many unexpected
that inhibits the diffusion of nutrients and insulin across the host reactions occurred in animals that reect the need to use toxic
membrane. In the context of BAPs, biotolerability is a better molecules in the fabrication process [92,93], and the capability of
descriptor than biocompatibility and can be dened as the ability some complement factors and IgGs to access agarose pores [94],
of a material to reside in the body for long periods of time with only thus making this natural polymer less advantageous as compared
low degrees of inammatory reactions [65]. to alginate.
Various biomaterials have been exploited for islet encapsulation Collagen is a highly versatile polymer able to form hydrogels
ranging from traditional polymers to inorganic materials and also that can be exploited for the fabrication of beads with different
more innovative ones involving micro/nanofabrication (Fig. 2). geometries, membranes, matrices, etc. In the eld of pancreatic
When selecting the material and fabrication technique for an cell encapsulation, it has been used together with crosslinkers [95],
encapsulation device, whether a macrodevice or a micro/nano- with other polymers such as alginates [87], or with outer shells/
capsule, this should confer specic features in terms of membrane coatings, e.g., tetrapolymers of 2-hydroxyethyl methylacrylate
thickness, surface properties, pore dimensions and distribution (HEMA) [96,97] to overcome and control its weak mechanical
[66,67]. Pores must be sufciently large to facilitate the transport of properties and limited stability.
nutrients, insulin (1.352.75 nm) [68], glucose (0.4 nm) [69] and Alginate is a natural anionic polymer composed of 1,40 -linked
waste products and should be small enough to block the mediators b-D-mannuronic acid (M) and a-L-guluronic acid (G) residues in
(e,g., IgG radius = 5.9 nm) and cells (radius = 10 mm) responsible for different sequences. The ratio of M and G blocks inuences
immune rejection [7072]. This can be dened during membrane hydrogel mechanical properties. Due to its good biotolerability,
design, but it is challenging to ensure the required permeability or potentially high degree of purity and easy fabrication without the
molecular weight cut off value for encapsulation membranes [73,74]. need to use toxic solvents, alginate is the most successfully
Studies have shown that capsules with a pore size of even a few tens investigated polymer for pancreatic cell encapsulation.
of nanometers may immunoprotect transplanted cells even if IgGs
and complement proteins permeate the capsule [75,76], whereas 3.1.2. Fabrication with natural polymers
others have found that inhibiting the cytokine- and chemokine- Different fabrication techniques can be applied to alginate and
mediated cross-talk between grafted and immune cells may be to other natural polymer-based hydrogels to produce micro-
benecial for cell survival [77,78]. Membrane thickness can inuence capsules, as well as membranes and sheets that can be used in
diffusion and capsule dimension, while surface properties affect the macrodevices. Animal studies and clinical trial results obtained
immune response. Hydrophobic surfaces promote protein adsorp- with alginate-based BAPs (described further in Section 5) encour-
tion and denaturation that can potentially activate immune aged further research to enhance alginate immunoprotective and
reactions; so they are usually discarded in favor of hydrophilic ones. mechanical properties. In this sense, conformal coatings [98] or
For the same reason, a negative zeta charge is preferred to a positive coatings based on cationic synthetic polymers (e.g., poly-L-lysine
one [79]. Mechanical stability is also important. The material [99] and poly-L-ornithine [100]) can be employed to overcome
selected should not be biodegradable and should last at least as long inhomogeneous pore distribution and reduce membrane size to
as the cells are alive [73]. ensure better immunoprotection, even in the case of xenotrans-
The different classes of materials exploited so far for islet plants [101,102]. Ultra-purication processes can be used to obtain
encapsulation, are described in the following, together with the more chemically stable and biotolerable capsules [103]; further
corresponding micro/nanofabrication techniques. efforts to enhance alginate-based capsule mechanical stability
involve exploring coatings based on polyethylene glycol (PEG)
3.1. Encapsulation with polymers [104] and covalent crosslinking molecules including photoactive
crosslinkers [105,106], aldehyde or hydroxyl groups [107,108].
Polymers are the most commonly employed materials for the Alginate microcapsules are usually manufactured through
fabrication of encapsulating membranes [67]. Both natural and solgel processes and then by extruding the polymer-cell solution
synthetic polymers can be employed: the former are usually used in a crosslinking bath containing divalent cations such as Ca 2+, Ba2
+
as hydrogels due to their viscoelastic behavior, tissue-resembling , etc. [109]. The capsules can be then transferred to a cationic
properties and reduced ability to evoke an inammatory response polyamino acid solution to form a semipermeable membrane
[80]; the latter enable easier and more standardized fabrication around the alginate and then into an alginate solution to form an
processes and are readily available with higher purity, thus additional alginate layer to reduce the immunogenicity of the
resulting in a good efciency in terms of immunoisolation and structure [102,110]. Extrusion relies on gravitational force and
nutrient diffusion [81]. diameters usually approach 2000 mm with such techniques [111].
To reduce the capsule diameter, surface tension needs to be
3.1.1. Natural polymers reduced by using additional energy sources, e.g., air ow, electric
Among natural polymers, polysaccharides are widely employed elds or vibrations [112]. Electrostatic-mediated extrusion exploits
as they do not interfere with cell viability and functionality and a potential difference between the extrusion needle and the gelling
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 17

cationic bath. The exploitation of mechanical energy sources such Aluminum oxide (Al2O3) and titanium oxide (TiO2) membranes
as vibrations [113] or cutting wires [114116] is an alternative can be produced via anodization techniques that result in
especially to increase the extrusion rate even if viscosity and cell membranes with controlled thickness and porosity. On average,
damage related issues are associated with these techniques. An the pores are larger than those obtained with microfabricated
alternative technique to simple droplet extrusion was proposed by structures, whereas membrane thickness is greater than that of Si
Poncelet et al. [117], and optimized by Hoesli et al. using an membranes, but smaller than their polymeric counterparts.
emulsion-based encapsulation involving internal gelation [118]. However, the cheap fabrication process and the high mechanical
stability, as well as the high pore density, which compensates for
3.1.3. Synthetic polymers the increased thickness from a diffusion view point, are among the
An alternative to natural polymers is represented by synthetic advantages of these systems. In vitro and in vivo tests revealed that
polymers that can be synthetized in large quantities via after coating with PEG, the brotic overgrowth and inammatory
standardized processes. They can be chemically modied to response on Al2O3 membranes were quite limited [131,137140].
enhance biotolerability, mechanical properties and morphological TiO2 membranes based on titanium nanotubes are also produced
features, e.g., membrane thickness and pore size [81,119]. Synthetic using anodization processes. Many research efforts are currently
polymers can lead to biocompatibility issues since many of the employed to produce membranes based on Ti nanotubes with
fabrication processes involve toxic solvents that could affect cell different tube dimensions and arrangements obtained by varying
viability. For this reason, synthetic polymers are mainly used to the anodization parameters [141145]. These systems seem
fabricate semipermeable membranes for macrocapsules [67] in promising especially for their application in macrocapsule devices,
the absence of cells. PEG can be used for nano-, micro- and macro- where it is possible to introduce another matrix favoring cell
encapsulation due to a lack of toxic solvents in the fabrication adhesion and viability
process and is thus one of the most successfully employed
synthetic materials. It is often used as a hydrogel, sometimes in 3.3. Other approaches
conjunction with alginate [120] due to its ability to embed a large
amount of water and has a short diffusion time [121]. PEG can be Other approaches used for pancreatic islet encapsulation
polymerized by using UV [122] or physical/chemical crosslinking include the exploitation of photosensitive polymers, e.g., SU-8,
mechanisms [123]. However, in animals, PEG membranes cannot to fabricate nanoimprinted nanoporous membranes. These are
prevent the passage of cytotoxic molecules that can activate the devised to be used in conjunction with microfabricated cuboids to
immune response [124]. enable the encapsulation of single cells or cell clusters [146,147].
Another class of synthetic polymers is represented by SU-8 microstructures are MRI transparent, have good mechanical
thermoplastic polymers, which are eligible for cell encapsulation stability and good diffusion and immunoprotection. However,
due to their greater mechanical and chemical stability. oxygen diffusion is not efcient and there are biocompatibility
Polyurethane [125,126], polytetrauoroethylene (PTFE) [127], issues still to be faced [148]. 2D micro and nanofabrication
polyvinyl alcohol (PVA) [128130] and their copolymers have techniques have been employed to produce nanoporous containers
been successfully used in islet encapsulation. They are often for cell encapsulation as well. In particular, this concept is inspired
used with other polymers acting as substrates for cell adhesion in by Japanese origami art and involves the fabrication of 2D self-
the inner part of the capsule and have been employed both in folding structures that, relying on the minimization of surface
micro and macroencapsulation systems, due to their ability to energy, enable the development of 3D structures that can
form membranes and hollow bers and to be processed in a potentially be exploited for cell encapsulation [149151]. These
variety of shapes through heat melting followed by cooling microstructures can be fabricated by using nickel, copper, gold and
processes [67]. several polymeric materials.

3.2. Encapsulation with inorganic materials 3.4. Biomaterials and techniques for nanoencapsulation

Inorganic materials, e.g., silicon, titanium and aluminum In addition to improving the immunoisolation, biotolerability,
oxides, have been successfully used in islet encapsulation to pancreatic cell viability and functional properties of existing
fabricate membranes with high stability and tight pore size systems, there is considerable ongoing effort to miniaturize b-cell
(typically below 100 nm except for titanium oxide membranes that encapsulation systems. By using nanotechnology, capsule dimen-
have slightly larger pores [56]). The technologies used for the sions and wall thickness can be signicantly reduced to enhance
fabrication of inorganic material-based membranes are less diffusional processes and glucose sensing. Smaller implant sites
mature as compared with those of polymeric systems, but have can also be targeted [152]. Two approaches are being pursued:
signicant potentials for future applications in both macro- and conformal coatings or molecular camouage [153] and fabricat-
micro-capsules. The risk of biocompatibility-related issues is ing nanocapsules by exploiting polyelectrolytes and layer-by-layer
higher for this class of materials than for polymers [131,132] (LBL) deposition methods [154].
requiring the development of coatings to reduce protein adsorp- Molecular camouage can immunoprotect pancreatic islets
tion and enhance membrane biocompatibility [133]. while keeping overall dimensions as low as possible (coatings have
Silicon (Si) membranes are fabricated using photolithography, thickness below 100 mm). Polymers, sometimes in the form of
etching and deposition processes allowing thin membranes to be hydrogels, can covalently bind certain cell membrane constituents
obtained with highly controllable pore distribution and dimen- e.g., membrane proteins [155]. The most exploited material for this
sions. These membranes were used to encapsulate pancreatic cells application is PEG [156]. The ability of these coatings to provide
in the late 1990s [134]. Their advantages lie in the potential to immunoisolation was demonstrated in vitro and in vivo, also by
create hierarchical structures, in their optical transparency and means of pre-clinical studies. However, open issues are related to
easiness of biofunctionalization [135] and they have been the long term stability of such conformal coatings [100].
successfully tested in rats, thus revealing strict dependence from A second approach to reduce the overall dimensions of
pore dimensions [134,136]. Similarly to hydrogel capsules, surface encapsulated pancreatic islets is the LBL technique that involves
chemistry modication and specic coatings can aid in producing the creation of an immunoisolating coating on a single cell or cell
improved outcomes. aggregate by alternating the deposition of polycation and
18 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

polyanion layers to create protective membranes that are only a that the LBL technique can guarantee good BAP performance for up
few nanometers thick [45]. This can improve diffusion, increase to 100 days [61].
responsivity to glucose level, reduce implant volume and enhance
neovascularization. The polyelectrolytes that can accomplish this 4. Biological issues and tissue engineering
task include polyallylamine hydrochloride (PAH) [154]. and PVA
conjugated to a single layer of PEG-phospholipids [59] or biotin/ The replacement of insulin-producing cells in T1D patients is a
streptavidin [157]. In vitro and in vivo tests in rodents indicated challenge that has been addressed by using different strategies
over the last decades. This section highlights the established

Fig. 3. (A) Simplied scheme representing the key developmental steps occurring for the specication of embrionic stem cells (ESCs) into mature b-cells. Relevant pathways
are reported in red, while lateral ones are reported in blue. For each step, the most important known transcription factors governing the process are specied. (B)
Immunouorescence images and photon emission maps for human ESC-derived pancreatic cells engrafted in mice. (Adapted from [187] and reproduced with permission of
Elsevier). (C) Comparison between primary human islets and human ESCs differentiated in vitro to achieve a mature pancreatic phenotype. Immunouorescence images
demonstrate that ESC-derived cells show complete co-localization of insulin with green-uorescent protein (GFP), thus suggesting a good differentiation level. However, the
graphs show that they do not replicate primary cell functionality, since they do not secrete enough insulin in response to high glucose. LG = low glucose; HG = high glucose;
HG + KCl = high glucose plus direct depolarization. (Adapted from [188] and reproduced with permission of Springer). (D) Differentiation of iPSCs in b cells: differentiation
protocol scheme and immunouorescence images showing the expression of C-peptide, glucagon (GCG), somatostatin (SST) and insulin (INS) proteins in cells at day 22.
(Adapted from [193] and reproduced with permission of Nature Publishing Group). (For interpretation of the references to color in this gure legend, the reader is referred to
the web version of this article.)
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 19

approaches toward the extraction, culture and engineering of key producing b-cells. Adenovirus and other vectors have been used to
cell types, the biological issues to be considered in this framework induce the expression of cardinal islet developmental regulators,
and the potential of recent approaches involving the use of specic both in vitro and in vivo [170172]. Recently, it has been found that
stem cell types. a more efcient reprogramming can be achieved by using a forced
expression of four pancreatic transcription factors, namely Pdx1,
4.1. Cell sources: limitations and opportunities Ngn3, Pax4 and MafA, in combination with growth factors such as
betacellulin and exendin-4, the vitamin nicotinamide, and small
The use of cells from human organ donors to replenish diabetic molecules that facilitate DNA binding of transcription factors [173].
patient b-cell mass dates back to 1966 with the rst allogenic Although the reprogramming techniques still need to be furtherly
transplant attempts [158] and has been enhanced by improved developed, in order to guarantee high efcacy and safety, this
surgical techniques [159]. Pancreatic transplants efciently restore approach appears to be very promising: the exocrine pancreatic
metabolic control in diabetic patients. However, the procedure is tissue that is normally discarded after the islet isolation procedure,
severe and it has several drawbacks, already discussed, leading to a in fact, can be used and, after few weeks a second batch of islets
shift towards the use of single cells or islets. derived from the same donor is available for the recipient.
Human islet isolation from the bulk donor pancreas is
performed by digestion and purication steps [32]. Whole islets 4.3. ESCs and iPSCs: toward allogenic/autogenic islet transplantation
have a greater efciency than single b-cells as they retain the
complex multicellular interactions that play a key role in the Stem cells represent a novel approach for the production of
down-regulation of insulin secretion during hypoglycemic epi- transplantable b-cells. Pluripotent stem cells have the unique
sodes [160]. abilities to: self-renew in an undifferentiated state enabling large-
As a result, allotransplantations using rat and pig islets as scale expansion in vitro, and to differentiate towards different
primary xenogeneic tissues have been studied [39,161]. Porcine phenotypes, allowing the generation of all 200 cell types of the
islets are preferred, due to the close homology between porcine human body [174,175]. Thus, theoretically they can be used to
and human insulin and the islet morphological similarity between generate insulin-producing b-cells, multicellular islets or even the
the two species [74]. However, microencapsulation has had more whole pancreas [176,177]. The development of mature b-cells from
negative effects on porcine islets than on rat and human ones embryonic stem cells (ESCs) is an extremely complex process still
[162]. Porcine islets are obtained by using the same protocols as at an early stage [178180]. A simplied scheme of the main
described for human tissues, with the advantage of a potentially developmental steps of pancreas organogenesis is shown in
unlimited supply. However, issues in using porcine tissue are the Fig. 3A. This complex interplay of signaling events and transcrip-
risk of retroviral disease transmission and the absolute need for tional networks results in endocrine progenitors, which give rise to
effective immunobarriers, since they induce a more aggressive ve different hormone-secreting cells: glucagon-secreting a cells,
immune rejection than human cells [163]. somatostatin-releasing d cells, ghrelin-producing e cells, pancre-
The use of immortalized human pancreatic cell lines is another atic polypeptide-secreting PP cells, and insulin-secreting b-cells.
approach that would allow allogeneic instead of xenogeneic ESCs have been extensively studied over the last decade, with a
transplants thus reducing the need for immunoprotection. The focus on producing mature b-cells. Activin A, a transforming
different cell lines produced for this purpose include: (i) bLox5, an growth factor b (TGFb) family member, induces the emergence of
adult b-cell-derived cell line that has been used to clarify the role denitive endoderm [181,182] with retinoic acid supporting
of mitochondria in immune cell-mediated b-cell death, [164] and foregut induction from which multipotential pancreatic cells arise
expresses autoantigens typical of T1D and is a reliable b-cell [183]. Expression of Pdx1 (Pancreatic and duodenal homeobox
surrogate in cell-mediated lymphocytotoxicity assays; (ii) HP62 is a 1 also known as insulin promoter factor 1), along with other
cell line obtained by transfecting human islets with the SV40 virus factors, is a biomarker for the development of pancreatic
that has been used to study b-cell death mechanisms and to progenitors (Fig 3A). Pdx1 expression can be induced by treating
investigate isolation-specic mechanisms associated with the ESCs with noggin (a bone morphogenetic protein antagonist) or
progression to T1D in vivo [165]; (iii) 1.1B4, a cell line obtained by dorsomorphin (a small molecule analog) [181,182,184]. The nal
electrofusion of the exocrine cell line (PANC-1) and freshly isolated steps needed to convert pancreatic progenitors to fully differenti-
b-cells that have been used to investigate mechanisms of ated b-cells are currently unclear [177]. For this reason, the most
autoimmune destruction of pancreatic b-cells, in particular promising results obtained to date have been obtained by
molecular toxicity mechanisms after exposure to the proinam- implanting ESCs in immunocompromised mice at the pancreatic
matory cytokines IL-1b, IFN-g and TNF-a [166]; (iv) EndoC-bH1, a progenitor stage, allowing the cells to mature in vivo [185187]
cell line developed from fetal pancreatic tissue using the lentiviral (Fig. 3B). Nicotinamide, insulin-like growth factor 1 (IGF1) and
vector SV40T. By using an insulin promoter, only insulin-producing hepatocyte growth factor (HGF) have been used to induce mature
cells were immortalized. In mice, these cells showed rather good phenotypes in vitro but have resulted in immature and non-
results, although not comparable with those obtained with glucose-responsive phenotypes [188,189] (Fig. 3C).
primary b-cells [167]; (v) EndoC-bH2 is a second generation Induced pluripotent stem cells (iPSCs) represent another
version of the EndoC-bH1 cell that more closely resembles paradigm with signicant potential in regenerative medicine
primary cells [168]. While these cell lines are interesting models and stem cell-based therapies. Retroviral expression of a limited
for studying b-cell insulin-secretory mechanisms, immunological set of genes (Oct4, Sox 2, Klf4 and c-Myc) can convert adult cells to
processes etc., they are currently inappropriate for cell-replace- a pluripotent state, with transcriptional and epigenetic features
ment purposes, due to scarce reactivity to glucose and low insulin similar to those of ESCs. The ability to reprogram adult cells
production [169]. provides a means to use patients own cells to build tissues/organs
thus facilitating autogenic implants [190192]. The derivation of
4.2. Reprogramming exocrine cells into insulin-producing ones endoderm from iPSCs has been recently reported [192] where
Pdx1-positive pancreatic progenitor cells were obtained from
An intriguing approach, which is recently attracting interest in iPSCs at high efciency and further matured by treatment with
the scientic community, is based on reprogramming acinar cells, forskolin, dexamethasone, Alk5 inhibitor II and nicotinamide in a
which have an exocrine function in the pancreas, to insulin- 3D culture [193] (Fig. 3D). The signaling events modulating this
20 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

process were also claried [194] with high oxygen conditions [197] providing an initial proof of principle for the clinical
facilitating iPSC differentiation into mature b-cells [195]. Micro- application of reprogrammed somatic cells in the treatment of
RNA (miRNA) insertion can also promote pancreatic b-cell diabetes. The ability of iPSCs to differentiate into fully functional
differentiation [196]. Finally, differentiated iPSCs have been pancreatic b-cells, remains controversial due to a limited ability to
implanted in diabetic mouse models and reversed hyperglycemia retain insulin secretory competence upon glucose stimulation.

Fig. 4. Examples of commercial BAP systems. (A) bAir1 chamber system for islet macroencapsulation in its assembled form with connected access ports and X-ray image of
an implanted recipient (adapted with permission from [212] (copyright: Creative Commons)); (B) TheraCyteTM system schematization showing structure and diffusive
mechanisms across the membrane (Reproduced with Theracyte Inc., permission); (C) VC-01TM system in which vascularization is evident (reproduced with permission from
[216]: Nature Publishing Group); (D) Sernovas Cell Pouch SystemTM and its constitutive components (reproduced with permission from [223]: Elsevier); (E) Islet Sheet
system: electron micrograph image and a picture showing the sheet in its implant site are reported respectively (Photo courtesy Scott King/Islet Sheet Medical).
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 21

Nonetheless, a scalable differentiation protocol able to generate could be achieved without multiple implants due to the relatively
hundreds of millions of glucose-responsive b cells from human low cell density of the device (24,000 cells versus the 400,000 re-
pluripotent stem cells in vitro has been described [198]. As a sign of quired) [209].
incomplete maturation, the iPSC-derived b-cells may co-express Cell Pouch SystemTM (Fig. 4D) is a biocompatible polymeric
multi-hormones, but they lack the expression of specic mature macrocapsule devised for subcutaneous implantation that can
pancreatic b-cell markers such as Nkx6.1 and MafA [199]. Despite reproduce a natural environment in the body for the long-term
some unclear issues that have still to be faced, iPSCs represent the survival and function of therapeutic pancreatic cells. The
most promising cell type for BAP applications [200201]. combination with sertolin, a testicular protein that can protect
insulin producing islets from immune system attack, reduces or
5. Recent encapsulation system development and clinical trials eliminates the need for immunosuppressive agents [220222].
Islet sheet device [223] (Fig. 4E) is another macroencapsulation
In this section, attention is focused on the most recent and system consisting of a thin alginate sheet in which pancreatic islets
successful micro- and macro-encapsulation systems (Fig. 4). are entrapped. The main advantages of this system lie in the easy
Nanocapsulation systems are still at an early stage of development implantation, in the peritoneal cavity or subcutaneous site, and in
and neither clinical trials nor commercial products have been its easy retrieval. Each sheet is expected to host about
identied, yet. 100,000 islets, requiring several islet sheets to achieve exogenous
insulin independence [224]. Acceptable neovascularization and
5.1. Microcapsules insulin secretion have been observed in animals trials, but graft
survival in large animals may be an issue. Both large animal testing
Alginate-poly-L-ornithine-alginate microcapsules (PLO) and clinical trials have been scheduled [225226,45].
[202,203], barium alginate microcapsules [53], alginate-encapsu-
lated human islets implanted into the intraperitoneal cavity, under 5.3. Caveats
immunosuppression [204] and alginate-based capsules to correct
diabetes in immunodecient mice [205] are within the most Due to the complexity of the problem and to a lack of adequate
successful systems developed by research teams all over the world. characterization of materials and encapsulation systems, BAP is
Important advancements have been pursued by companies as still far from being an imminent clinical reality, due to
well; the most outstanding example is represented by a system, unavailability of a fully reliable solution. Several efforts have
namely DIABECELL1 [206], consisting of neonatal pig islets focused, in the last decades, on the synthesis or modication of
encapsulated in an alginate-based matrix and implanted in the advanced materials for islet encapsulation. Interesting insights
peritoneal cavity [207]. Clinical trials revealed a 70% reduction in have been recently highlighted, in this eld. However, alginate still
hypoglycemic events and a 20% reduction in insulin dose. However, remains the most suitable polymer used in BAPs, despite its
the need of exogenous insulin injections was not fully eliminated limitations in terms of fabrication reliability and pore dimensions.
[208] Despite the breadth of research efforts also in miniaturizing
encapsulation systems, macrodevices appear to be the most
5.2. Macrocapsules promising BAPs due to the ability to retrieve them in case of failure,
to perform biopsies and to easily incorporate other materials or
Concerning macrocapsule devices, mainly extravascular ones, additional systems that are able to enhance BAP function (e.g., cell
the number of products under development or already on the viability, revascularization, oxygenation, etc.). However, macro-
market is signicantly higher. Some interesting examples are capsule-based BAPs are affected by critical issues related to the
reported in the following. implant site, to esthetics and to the need for the patient to care for
bAir1 device (Fig. 4A) is a subcutaneous insulin bioreactor the device.
containing pancreatic cells immobilized in two at alginate sheet
supplied with oxygen via a gas chamber placed between the 6. Technological issues and engineering challenges
membranes (relled daily by means of an external port) [209211].
The chamber is housed in a plastic case, providing mechanical In this section the contribution of engineering tools to the
protection to the encapsulation device, wrapped in a porous PTFE development of more advanced and functional BAP systems is
membrane impregnated with hydrophobically modied alginate discussed. By enabling/enhancing stem cell differentiation pro-
to avoid immunologic communication between the recipient and cesses, donor shortage and xenograft-related problems could be
the graft tissue. In animals [212] and patients with long-standing eliminated while micro- and nano-components, such as sensors
T1D [213], graft function and regulated insulin secretion were and actuators, could enable the translation of some of the
observed without the need for immunosuppressive drugs. advantages of macrocapsular systems e.g., supply of oxygen or
TheraCyteTM system (Fig. 4B) [214] is a composite structure that VEGF provided by reservoirs or pumps, to microdevices. Finally, it
includes two PTFE membranes bound to a polymeric structure: the is possible that on board sensors may in time provide self-
exterior membrane has tighter pores promoting neovasculariza- monitoring of BAP function.
tion, whereas the inner one provides for pancreatic cell immu-
noisolation. Animal studies have been encouraging [215,216] with 6.1. Miniaturized technologies for stem cell differentiation
the possibility to enhance system performance by infusing
vascular endothelial growth factor (VEGF) [217], pre-implanting The use of technologies at the milli-, micro- and nano-scale may
the device to induce neovascularization before transplantation provide more efcient methods to differentiate pluripotent stem
[218], or exploiting ESCs [45]. cells, accelerating their clinical translation for islet tissue
VC-01TM is a subcutaneous macrocapsule based on ESCs and a engineering and subsequent allogenic (in the case of ESCs) or
semipermeable membrane made of undisclosed polymers (Fig. 4C) even autogenic (in the case of iPSCs) transplantation. External
[219]. It produced encouraging results in mice in terms of signals controlling stem cell fate (stem cell niche) are both
neovascularization and blood glucose regulation. Clinical trials biochemical and biophysical [227,228], such that an effective niche
for this BAP have been initiated although there have been some engineering process must thus include both signal types, in order
questions as to whether independence from exogenous insulin to reproduce the complex series of stimuli that facilitate
22 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

specialization from totipotent stem cells to partly committed Glucose nanosensors, based on optical signaling have been
progenitors and to terminally differentiated and functional cells developed in the form of smart tattoos or systems based on
[229331]. enzymes and quantum dots [259,260]. NEMS technology has
Cellcell and cell-material interactions based on biochemical enabled the possibility to embed miniaturized pumps and infusion
cues are the main driver for stem cell commitment [232,233]. systems based on smart reservoirs or release mechanisms, in
However, surface topography at the cell/scaffold interface also implantable drug delivery devices [261,262]. This technology has
plays a key role. Micro- and nano-structuring of this interface can been developed for AP systems, but would logically be applied to
mimic ECM organization, triggering or regulating specic cellular BAP systems.
activities [234], as substrate stiffness can [235]. New technologies In addition to their use in the design and validation phases to
for fabricating substrates with different surface topographies and verify BAP operation in terms of oxygen and glucose diffusion,
elastic moduli are useful tools for providing pluripotent stem cells these components may also enable the development of hybrid
with precise instructions, promoting their phenotypic maturation devices, i.e., embedding both biological and technological
before their implantation. Other miniaturized tools that can be components. For example, NEMS-based pumps could be used
used to this purpose include microuidic systems that provide a to provide an encapsulation device with substances that enhance
wide range of uidic cues and shear stresses that are recognized cell viability or close the insulin supply loop in case of implant
as crucial for the production of mature phenotypes [229]. failure. Similarly, glucose and oxygen nanosensors could enhance
Parallel-plate channels can also be used to reproduce physiologi- and support BAP sensing capabilities and monitor the correct
cally relevant shear stress values in vitro. More complex operation of the implant. In this way articial components can be
integrated and multiplexed systems can also be fabricated that devised as tools to compensate eventual BAP problems, making
control medium composition, oxygen concentration, pH, ow systems in some ways redundant but more efcient and reliable.
prole, ow rate and shear rate [236]. Mechanical forces (e.g., Currently available nanosensors and actuators are insufciently
traction and compression) inuence gene expression and cell mature from a technological standpoint to promote cross-
function making technologies for applying cyclic uniaxial fertilization between the different BAP functions but with
stretching, equiaxial strain, auxotonic loads and other mechanical additional research focused on addressing the various defects
inputs useful tools in the stem cell niche [237,238]. Additionally, using a multidisciplinary approach could change this situation in
mechanical forces can be applied to cells via ultrasonic pressure the near future.
waves [239].
Electrical inputs can effectively manipulate cell behavior,
especially when the target is an electrically responsive cell 7. Concluding remarks
[240]. Planar type microelectrode arrays (MEAs), consisting of
electrodes embedded in a cell culture platform enable simulta- Despite considerable research efforts since the discovery of
neous effects on cell growth and differentiation [241]. Indium tin insulin and the rst pancreatic transplantation, no optimal
oxide (ITO) microelectrodes [242] and micro-engineered conduc- solution to treat T1D has emerged. Many approaches, ranging
tive polymers [243] subserve a similar function while optical, from whole organ or islet transplant, to AP or BAP systems, have
magnetic and thermal inputs can also inuence cell behavior, been explored to develop a long-term alternative to daily insulin
although to a limited degree [229]. Pulsed infrared lasers injections. Encapsulation of pancreatic islets, cells and stem cells
integrated in optical bers, optomechanical systems, miniaturized using semipermeable membranes can avoid the immunosuppres-
electromagnets and miniaturized heating systems are additional sive therapy required for transplants, but these membranes also
examples of technologies that can drive stem cell differentiation need to be able to allow exchange of insulin, glucose, nutrients and
[244,245]. Synergies between active nanomaterials, internalized waste products with the surrounding environment to provide a
by cells, and external energy sources represent another strategy for physiological milieu. This requires extensive assessment of the
engineering cell differentiation and regulating cell function materials used to create BAPs. Alginate, especially if provided with
[246248]. proper coatings, emerged as the elective material for pancreatic
cell encapsulation, with encouraging clinical trial outcomes. The
6.2. Models, new technological components and BAP development development of easier encapsulation systems that provide
adequate vascularization and oxygen supply are seen as the key
Insufcient oxygenation due to a lack of neovascularization or to achieve implants that are more tolerable for the patient, able to
to inadequate diffusion is a major cause of BAP failure [130,249]. In avoid graft rejection and have a good life expectancy.
designing a new encapsulation system, microcapsule or macro- The development of a functional BAP will require the
device, typical engineering tools can be used to predict oxygen collation and analysis of the benets and limitations of the
diffusive mechanisms. Both analytical models, exploiting simple prototypes that are currently in the clinical trial arena, with a
geometries and zero or rst order kinetic laws [250253] and nite view to identifying and prioritizing the technological aspects that
elements analysis methods (FEMs) [254257], applied to nonlinear need optimization.
kinetics, and more realistic geometries, have been used to study Recently, exciting preclinical results were achieved, thus
oxygenation, nutrients and glucose diffusion in the capsule to demonstrating the high dynamism of this research eld. Veiseh
avoid cell necrosis. Local oxygen partial pressure, oxygen and colleagues demonstrated that in vivo biocompatibility is
membrane permeability and capsule geometry are only a few of largely governed by material geometry: 1.5-mm alginate capsules
the issues inuencing oxygen diffusion, and cell viability. Modeling were able to restore bloodglucose control in rodents and non-
and diffusion equation solving, could aid in dening BAP systems in human primates for 180 days, a period ve times longer than for
terms of the number of cells to be embedded, overall capsule conventional grafts, based on 0.5-mm alginate capsules [263]. In
dimensions, membrane diffusion coefcient, constitutive materi- parallel, Pepper et al. transplanted islets into a prevascularized,
als properties, etc. [258] that would also optimize in vitro and in subcutaneous site created by the temporary placement of a
vivo testing phases. vascular access catheter, in mice. The authors demonstrated that a
Engineering paradigms can also aid BAP development by controlled foreign-body response can be used to generate a
providing micro and nano components such as sensors, electro- prevascularized subcutaneous site supporting islet engraftment,
mechanical systems and actuators as well as nanomaterials. yet preventing the formation of an avascular brotic granular
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 23

capsule and a chronic inammatory response, which contribute to [20] M.J. Taylor, R. Gregory, H. Mitchell, M. Alblihed, A. Alsabih, P. Tomlins, et al.,
graft failure [264]. Insulin pump users would not rule out using an implantable articial
pancreas, Pract. Diabetes 31 (1) (2014) 1823a.
An increase in system complexity will probably be the key to [21] S. Tanna, M.J. Taylor, T.S. Sahota, K. Sawicka, Glucose-responsive UV
achieving better performances, thanks to the integration of polymerised dextranconcanavalin a acrylic derivatised mixtures for closed-
instruments typical of biology, tissue engineering, nanotechnology loop insulin delivery, Biomaterials 27 (8) (2006) 15861597.
[22] Z. Gu, T.T. Dang, M. Ma, B.C. Tang, H. Cheng, S. Jiang, et al., Glucose-responsive
and micro-mechatronics. An ideal BAP would be able to microgels integrated with enzyme nanocapsules for closed-loop insulin
autonomously modulate body reactions by responding to micro- delivery, ACS Nano 7 (8) (2013) 67586766.
environmental changes, maintaining at maximum levels its [23] T. Yang, R. Ji, X.X. Deng, F.S. Du, Z.C. Li, Glucose-responsive hydrogels based on
dynamic covalent chemistry and inclusion complexation, Soft Matter 10 (15)
stability and functionality overtime, yet resulting unobtrusive (2014) 26712678.
for the patient. Although a lifelong BAP is still far to be envisioned, [24] W. Tai, R. Mo, J. Di, V. Subramanian, X. Gu, J.B. Buse, et al., Bio-inspired
future efforts will focus on extending implant lifetime and making synthetic nanovesicles for glucose-responsive release of insulin,
Biomacromolecules 15 (10) (2014) 34953502.
it long enough (at least 23 years) to justify the surgical procedures
[25] G. Jiang, T. Jiang, H. Chen, L. Li, Y. Liu, H. Zhou, et al., Preparation of multi-
needed to install it. The ongoing development of materials, which responsive micelles for controlled release of insulin, Colloid Polym. Sci. 293
increasingly integrate several functionalities [265], and engineer- (1) (2015) 209215.
ing tools raise promises to address this challenge. [26] S.A. White, J.A. Shaw, D.E. Sutherland, Pancreas transplantation, Lancet 373
(9677) (2009) 18081817.
[27] P.W. Williams, Notes on diabetes treated with extract and by grafts of sheeps
Conict of interest pancreas, Br. Med. J. 1303 (1894) .
[28] P.E. Lacy, M. Kostianovsky, Method for the isolation of intact islets of
Langerhans from the rat pancreas, Diabetes 16 (1) (1967) 3539.
The authors declare no conicts of interest. [29] F. Lim, A. Sun, Microencapsulated islets as bioarticial endocrine pancreas,
Science 210 (4472) (1980) 908910.
Acknowledgments [30] R. Howorka, M.E. Wilinska, L.J. Chassin, D.B. Dunger, Roadmap to the articial
pancreas, Diabetes Res. Clin. Pract. 74 (2006) S178S182.
[31] D.W. Scharp, P.E. Lacy, J.V. Santiago, C.S. McCullough, L.G. Weide, L. Falqui,
The authors would like to thank Professor Igor Lack (Polymer et al., Insulin independence after islet transplantation into type I diabetic
Institute, Slovak Academy of Sciences, Slovak Republic) for his patient, Diabetes 39 (4) (1990) 515518.
[32] A.J. Shapiro, J.R. Lakey, E.A. Ryan, G.S. Korbutt, E. Toth, G.L. Warnock, et al.,
support and precious suggestions.
Islet transplantation in seven patients with type 1 diabetes mellitus using a
This work has been supported by the Smart APP (Smart glucocorticoid-free immunosuppressive regimen, N. Engl. J. Med. 343 (4)
Articial Pancreas relled by mechatronic Pills) and the GeT Small (2000) 230238.
[33] E.A. Ryan, B.W. Paty, P.A. Senior, D. Bigam, E. Alfadhli, N.M. Kneteman, et al.,
(TarGeted therapy at Small scale) projects, both funded by the
Five-year follow-up after clinical islet transplantation, Diabetes 54 (7) (2005)
Scuola Superiore di Studi Universitari e di Perfezionamento 20602069.
SantAnna (Pisa, Italy). [34] T. Eich, O. Eriksson, A. Sundin, S. Estrada, D. Brandhorst, H. Brandhorst, et al.,
Positron emission tomography: a real-time tool to quantify early islet
engraftment in a preclinical large animal model, Transplantation 84 (7)
References (2007) 893898.
[35] A.M. Posselt, M.D. Bellin, M. Tavakol, G.L. Szot, L.A. Frassetto, U. Masharani,
[1] D.J. Cunningham, Cunninghams Textbook of Anatomy, 12th ed., Oxford et al., Islet transplantation in type 1 diabetics using an immunosuppressive
University Press, Cary, North Carolina, U.S.A, 1981. protocol based on the antiLFA1 antibody efalizumab, Am. J. Transplant. 10
[2] F.H. Martini, N.J. Timmons, R.B. Tallistch, Human Anatomy, 6th ed., Pearson (8) (2010) 18701880.
Education Inc., Toronto, Canada, 2008. [36] J.F. Gummert, T. Ikonen, R.E. Morris, Newer immunosuppressive drugs a
[3] J.M. Gregory, D.J. Moore, J.H. Simmons, Type 1 diabetes mellitus, Pediatr. Rev. review, J. Am. Soc. Nephrol. 10 (6) (1999) 13661380.
34 (5) (2013) 203215. [37] I. Penn, Post-transplant malignancy, Drug Saf. 23 (2) (2000) 101113.
[4] T.L. Van Belle, K.T. Coppieters, M.G. Von Herrath, Type 1 diabetes: etiology, [38] M.A. Naftanel, D.M. Harlan, Pancreatic islet transplantation, PLoS Med. 1 (3)
immunology, and therapeutic strategies, Physiol. Rev. 91 (1) (2011) 79118. (2004) e58 e58.
[5] N.S. Papaspyros, The History of Diabetes, George Thieme Verlag, Stuttgart, [39] R.P. Robertson, Islet transplantation as a treatment for diabetesa work in
1964. progress, N. Engl. J. Med. 350 (7) (2004) 694705.
[6] T. Scully, Diabetes in numbers, Nature 485 (7398) (2012) S2S3. [40] P.R. Johnson, K.E. Jones, Pancreatic islet transplantation, Semin. Pediatr. Surg.
[7] American Diabetes Association, National Diabetes Statistics Report, 2014, 21 (3) (2012) 272280.
http://www.diabetes.org/diabetes-basics/statistics/ [41] P.G. Stock, J.A. Bluestone, Beta-cell replacement for type I diabetes, Annu. Rev.
[8] World Health Organization, Ten facts about diabetes, 2006, http://www.who. Med. 55 (2004) 133156.
int/features/factles/diabetes/02_en.html [42] A. Hyder, C. Laue, J. Schrezenmeir, Effect of the immunosuppressive regime of
[9] L. Ricotti, T. Assaf, P. Dario, A. Menciassi, Wearable and implantable pancreas edmonton protocol on the long-term in vitro insulin secretion from islets of
substitutes, J. Artif. Organs 16 (1) (2013) 114. two different species and age categories, Toxicol. In Vitro 19 (4) (2005) 541
[10] D.C. Klonoff, The articial pancreas: how sweet engineering will solve bitter 546.
problems, J. Diabetes Sci. Technol. 1 (1) (2007) 7281. [43] H. Browning, P. Resnik, Homologous and heterologous transplantation of
[11] C. Cobelli, E. Renard, B. Kovatchev, Articial pancreas: past, present and pancreatic tissue in normal and diabetic mice, Yale J. Biol. Med. 24 (2) (1951)
future, Diabetes 60 (11) (2011) 26722682. 141.
[12] G.M. Steil, A.E. Panteleon, K. Rebrin, Closed-loop insulin delivery the path to [44] V. Bisceglie, ber die antineoplastische Immunitt, J. Cancer Res. Clin. Oncol.
physiological glucose control, Adv. Drug Deliv. Rev. 56 (2) (2004) 125144. 40 (1) (1934) 122140.
[13] E. Renard, A. Farret, J. Place, A. Wojtusciszyn, J. Bringer, Towards an articial [45] D.W. Scharp, P. Marchetti, Encapsulated islets for diabetes therapy: history,
pancreas at home, Diabetes Metabolism 37 (2011) S94S98. current progress, and critical issues requiring solution, Adv. Drug Deliv. Rev.
[14] Y. Zhang, Y. Wang, J. Jia, J. Wang, Nonenzymatic glucose sensor based on 67 (2014) 3573.
graphene oxide and electrospun NiO nanobers, Sens. Actuat. B Chem. 171 [46] P. de Vos, Cell encapsulation: ready for the next step, Adv. Drug Deliv. Rev. 67
(2012) 580587. (2014) 1.
[15] S. Park, S. Park, S. Park, R.A. Jeong, H. Boo, J. Park, et al., Nonenzymatic [47] H. Uludag, P. De Vos, P.A. Tresco, Technology of mammalian cell
continuous glucose monitoring in human whole blood using electried encapsulation, Adv. Drug Deliv. Rev. 42 (1) (2000) 2964.
nanoporous Pt, Biosens. Bioelectron. 31 (1) (2012) 284291. [48] R.N. Wang, L. Rosenberg, Maintenance of beta-cell function and survival
[16] S.A. Brown, B.P. Kovatchev, M.D. Breton, S.M. Anderson, P. Keith-Hynes, S.D. following islet isolation requires re-establishment of the islet-matrix
Patek, et al., Multinight bedside closed-loop control for patients with type relationship, J. Endocrinol. 163 (2) (1999) 181190.
1 diabetes, Diabetes Technol. Ther. 17 (3) (2015) 203209. [49] N. Lembert, J. Wesche, P. Petersen, M. Doser, P. Zschocke, H.D. Becker, et al.,
[17] S. Del Favero, D. Bruttomesso, F. Di Palma, G. Lanzola, R. Visentin, A. Filippi, Encapsulation of islets in rough surface, hydroxymethylated polysulfone
et al., First use of model predictive control in outpatient wearable articial capillaries stimulates VEGF release and promotes vascularization after
pancreas, Diabetes Care 37 (5) (2014) 12121215. transplantation, Cell Transplant. 14 (23) (2005) 97108.
[18] V. Iacovacci, L. Ricotti, P. Dario, A. Menciassi, Design and development of a [50] A. Prochorov, S. Tretjak, V. Goranov, A. Glinnik, M. Goltsev, Treatment of
mechatronic system for noninvasive relling of implantable articial insulin dependent diabetes mellitus with intravascular transplantation of
pancreas, Mechatron. IEEE/ASME Trans. 20 (3) (2015) 11601169. pancreatic islet cells without immunosuppressive therapy, Adv. Med. Sci. 53
[19] L. Ricotti, T. Assaf, A. Menciassi, P. Dario, A novel strategy for long-term (2) (2008) 240244.
implantable articial pancreas, EMBC, 2011 Annual International Conference [51] P. de Vos, P. Marchetti, Encapsulation of pancreatic islets for transplantation
of the IEEE, August, 2011, pp. 28492853. in diabetes: the untouchable islets, Trends Mol. Med. 8 (8) (2002) 363366.
24 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

[52] J.T. Wilson, E.L. Chaikof, Challenges and emerging technologies in the [82] L. Gasperini, J.F. Mano, R.L. Reis, Natural polymers for the microencapsulation
immunoisolation of cells and tissues, Adv. Drug Deliv. Rev. 60 (2) (2008) 124 of cells, J. R. Soc. Interface 11 (100) (2014) 20140817.
145. [83] L.S. Gazda, H.V. Vinerean, M.A. Laramore, R.D. Hall, J.W. Carraway, B.H. Smith,
[53] B.E. Tuch, G.W. Keogh, L.J. Williams, W. Wu, J.L. Foster, V. Vaithilingam, R. No evidence of viral transmission following long-term implantation of
Philips, Safety and viability of microencapsulated human islets transplanted agarose encapsulated porcine islets in diabetic dogs, J. Diabetes Res. (2014)
into diabetic humans, Diabetes Care 32 (10) (2009) 18871889. 111.
[54] C.K. Colton, Implantable biohybrid articial organs, Cell Transplant. 4 (4) [84] R.W. Holdcraft, L.S. Gazda, H. Adkins, S.G. Harbeck, E.D. Meyer, M.A. Bautista,
(1995) 415436. et al., Enhancement of in vitro and in vivo function of agarose-encapsulated
[55] R. Pareta, J.P. McQuilling, S. Sittadjody, R. Jenkins, S. Bowden, G. Orlando, et al., porcine islets by changes in the islet microenvironment, Cell Transplant. 23
Long-term function of islets encapsulated in a redesigned alginate (8) (2014) 929944.
microcapsule construct in omentum pouches of immune-competent diabetic [85] S. Schaffellner, V. Stadlbauer, P. Stiegler, O. Hauser, G. Halwachs, C. Lackner,
rats, Pancreas 43 (4) (2014) 605613. et al., Porcine islet cells microencapsulated in sodium cellulose sulfate,
[56] J. Schweicher, C. Nyitray, T.A. Desai, Membranes to achieve Transplant. Proc. 37 (1) (2005) 248252.
immunoprotection of transplanted islets, Front. Biosci. (Landmark ed.) 19 [86] V. Stadlbauer, P.B. Stiegler, S. Schaffellner, O. Hauser, G. Halwachs, F. Iberer,
(2014) 49. et al., Morphological and functional characterization of a pancreatic b-cell
[57] J.A. Steele, A.E. Barron, E. Carmona, J.P. Hall, R.J. Neufeld, Encapsulation of line microencapsulated in sodium cellulose sulfate/poly
protein microber networks supporting pancreatic islets, J. Biomed. Mater. (diallyldimethylammonium chloride), Xenotransplantation 13 (4) (2006)
Res. A 100 (12) (2012) 33843391. 337344.
[58] W.F. Mueller-Klieser, R.M. Sutherland, Inuence of convection in the growth [87] B.R. Lee, J.W. Hwang, Y.Y. Choi, S.F. Wong, Y.H. Hwang, D.Y. Lee, et al., In situ
medium on oxygen tensions in multicellular tumor spheroids, Cancer Res. 42 formation and collagen-alginate composite encapsulation of pancreatic islet
(1) (1982) 237242. spheroids, Biomaterials 33 (3) (2012) 837845.
[59] Y. Teramura, Y. Kaneda, H. Iwata, Islet-encapsulation in ultra-thin layer-by- [88] K.Y. Lee, D.J. Mooney, Alginate properties and biomedical applications, Prog.
layer membranes of poly (vinyl alcohol) anchored to poly (ethylene glycol) Polym. Sci. 37 (1) (2012) 106126.
lipids in the cell membrane, Biomaterials 28 (32) (2007) 48184825. [89] H. Iwata, H. Amemiya, T. Matsuda, H. Takano, T. Akutsu, Microencapsulation
[60] Z.I. Zhi, A. Kerby, A.J.F. King, P.M. Jones, J.C. Pickup, Nano-scale encapsulation of Langerhans islets in agarose microbeads and their application for a
enhances allograft survival and function of islets transplanted in a mouse bioarticial pancreas, J. Bioact. Compat. Polym. 3 (4) (1988) 356369.
model of diabetes, Diabetologia 55 (2012) 10811090. [90] S. Fernndez-Cosso, A. Len-Mateos, F.G. Sampedro, M.T.C. Oreja,
[61] H. Dong, T.M. Fahmy, S.M. Metcalfe, S.L. Morton, X. Dong, L. Inverardi, H. Biocompatibility of agarose gel as a dermal ller: histologic evaluation of
Wang, Immuno-isolation of pancreatic islet allografts using pegylated subcutaneous implants, Plast. Reconstr. Surg. 120 (5) (2007) 11611169.
nanotherapy leads to long-term normoglycemia in full MHC mismatch [91] T. Kobayashi, Y. Aomatsu, H. Iwata, T. Kin, H. Kanehiro, Hisanaga, et al.,
recipient mice, PLoS One 7 (12) (2012) 50265. Indenite islet protection from autoimmune destruction in nonobese
[62] V. Kozlovskaya, O. Zavgorodnya, E. Kharlampieva, Encapsulation and surface diabetic mice by agarose microencapsulation without immunosuppression1,
engineering of pancreatic islets: advances and challenges, in: C. Lin (Ed.), Transplantation 75 (5) (2003) 619625.
Biomedicine, In Tech Europe, Croatia, 2012, pp. 134. [92] H. Gin, B. Dupuy, A. Baquey, C.H. Baquey, D. Ducassou, Lack of responsiveness
[63] D.F. Williams, On the mechanisms of biocompatibility, Biomaterials 29 (20) to glucose of microencapsulated islets of Langerhans after three weeks
(2008) 29412953. implantation in the rat-inuence of the complement, J. Microencapsul. 7 (3)
[64] J. Hilborn, L.M. Bjursten, A new and evolving paradigm for biocompatibility, J. (1990) 341346.
Tissue Eng. Regen. Med. 1 (2) (2007) 110119. [93] T. Tun, K. Inoue, H. Hayashi, T. Aung, Y.J. Gu, R. Doi, et al., A newly developed
[65] B.D. Ratner, The biocompatibility manifesto: biocompatibility for the twenty- three-layer agarose microcapsule for a promising biohybrid articial
rst century, J. Cardiovasc. Transl. Res. 4 (5) (2011) 523527. pancreas: rat to mouse xenotransplantation, Cell Transplant. 5 (5) (1996)
[66] J.A.M. Steele, J.P. Hall, D. Poncelet, R.J. Neufeld, Therapeutic cell S59S63.
encapsulation techniques and applications in diabetes, Adv. Drug Deliv. Rev. [94] H. Iwata, T. Takagi, H. Amemiya, Agarose microcapsule applied in islet
67 (2014) 7483. xenografts (hamster to mouse), Transplant. Proc. 24 (3) (1992) 952.
[67] P. de Vos, H.A. Lazarjani, D. Poncelet, M.M. Faas, Polymers in cell [95] L. Marinucci, C. Lilli, M. Guerra, S. Belcastro, E. Becchetti, G. Stabellini, et al.,
encapsulation from an enveloped perspective, Adv. Drug Deliv. Rev. 67 (2014) Biocompatibility of collagen membranes crosslinked with glutaraldehyde or
1534. diphenylphosphoryl azide: an in vitro study, J. Biomed. Mater. Res. A 67 (2)
[68] A. Oliva, J. Faria, M. Llabrs, Development of two high-performance liquid (2003) 504509.
chromatographic methods for the analysis and characterization of insulin [96] C. Yin, S.M. Chia, C.H. Quek, H. Yu, R.X. Zhuo, K.W. Leong, et al., Microcapsules
and its degradation products in pharmaceutical preparations, J. Chromatogr. with improved mechanical stability for hepatocyte culture, Biomaterials 24
B: Biomed. Sci. Appl. 749 (1) (2000) 2534. (10) (2003) 17711780.
[69] J.R. Pappenheimer, E.M. Renkin, L.M. Borrero, Filtration, diffusion and [97] S.M. Chia, A.C.A. Wan, C.H. Quek, H.Q. Mao, X. Xu, L. Shen, et al., Multi-layered
molecular sieving through peripheral capillary membranes, Am. J. Physiol. microcapsules for cell encapsulation, Biomaterials 23 (3) (2002) 849856.
167 (13) (1951) 2578. [98] M.M. Nabavimanesh, S. Hashemi-Najafabadi, E. Vasheghani-Farahani, Islets
[70] L. Leoni, T.A. Desai, Micromachined biocapsules for cell-based sensing and immunoisolation using encapsulation and PEGylation, simultaneously, as a
delivery, Adv. Drug Deliv. Rev. 56 (2) (2004) 211229. novel design, J. Biosci. Bioeng. 119 (4) (2015) 486491.
[71] J. Kang, G. Erdodi, J.P. Kennedy, Third-generation amphiphilic conetworks. III. [99] P. de Vos, B. De Hann, R. Van Schilfgaarde, Effect of the alginate composition
Permeabilities and mechanical properties, J. Polym. Sci. Part A. Polym. Chem. on the biocompatibility of alginate-polylysine microcapsules, Biomaterials
45 (18) (2007) 42764283. 18 (3) (1997) 273278.
[72] R. Lanza, R. Langer, J.P. Vacanti, Principles of Tissue Engineering, Acadamic [100] G. Basta, P. Sarchielli, G. Luca, L. Racanicchi, C. Nastruzzi, L. Guido, et al.,
Press, 2011. Optimized parameters for microencapsulation of pancreatic islet cells: an in
[73] A.M.A. Rokstad, I. Lack, P. de Vos, B.L. Strand, Advances in biocompatibility vitro study clueing on islet graft immunoprotection in type 1 diabetes
and physico-chemical characterization of microspheres for cell mellitus, Transplant. Immunol. 13 (4) (2004) 289296.
encapsulation, Adv. Drug Deliv. Rev. 67 (2014) 111130. [101] R.P. Lanza, D. Ecker, W.M. Khtreiber, J.E. Staruk, J. Marsh, W.L. Chick, A simple
[74] E.S. OSullivan, A. Vegas, D.G. Anderson, G.C. Weir, Islets transplanted in method for transplanting discordant islets into rats using alginate gel
immunoisolation devices: a review of the progress and the challenges that spheres, Transplantation 59 (10) (1995) 14851487.
remain, Endocr. Rev. 32 (6) (2011) 827844. [102] (a) A. Horcher, T. Zekorn, U. Siebers, G. Klck, H. Frank, R. Houben, et al.,
[75] T. Kobayashi, Y. Aomatsu, H. Iwata, T. Kin, H. Kanehiro, M. Hisanaga, et al., Transplantation of microencapsulated islets in rats: evidence for induction of
Indenite islet protection from autoimmune destruction in nonobese brotic overgrowth by islet alloantigens released from microcapsules,
diabetic mice by agarose microencapsulation without immunosuppression1, Transplant. Proc. 26 (2) (1994) 784;
Transplantation 75 (5) (2003) 619625. (b) P. de Vos, M.M. Faas, B. Strand, R. Calaore, Alginate-based microcapsules
[76] R.P. Lanza, W.M. Khtreiber, D. Ecker, J.E. Staruk, W.L. Chick, for immunoisolation of pancreatic islets, Biomaterials 27 (32) (2006) 5603
Xenotransplantatton of porcine and bovine islets without 5617.
immunosuppression using uncoated alginate microspheres, Transplantation [103] G.P.P. Basta, R. Calaore, U.S. Patent No. 8765,937. Washington, DC: U.S. Patent
59 (10) (1995) 13771384. and Trademark Ofce 2014.
[77] C.C. Lin, P.D. Boyer, A.A. Aimetti, K.S. Anseth, Regulating MCP-1 diffusion in [104] J.P. Chen, I.M. Chu, M.Y. Shiao, B.R.S. Hsu, S.H. Fu, Microencapsulation of islets
afnity hydrogels for enhancing immuno-isolation, J. Control. Release 142 (3) in PEG-amine modied alginate-poly (L-lysine)-alginate microcapsules for
(2010) 384391. constructing bioarticial pancreas, J. Ferment. Bioeng. 86 (2) (1998) 185190.
[78] J. Su, B.H. Hu, W.L. Lowe, D.B. Kaufman, P.B. Messersmith, Anti-inammatory [105] V. Breguet, R. Gugerli, M. Pernetti, U. Von Stockar, I.W. Marison, Formation of
peptide-functionalized hydrogels for insulin-secreting cell encapsulation, microcapsules from polyelectrolyte and covalent interactions, Langmuir 21
Biomaterials 31 (2) (2010) 308314. (21) (2005) 97649772.
[79] E.A. Vogler, Protein adsorption in three dimensions, Biomaterials 33 (5) [106] J. Dusseault, F.A. Leblond, R. Robitaille, G. Jourdan, J. Tessier, M. Mnard, et al.,
(2012) 12011237. Microencapsulation of living cells in semi-permeable membranes with
[80] A.S. Hoffman, Hydrogels for biomedical applications, Adv. Drug Deliv. Rev. 64 covalently cross-linked layers, Biomaterials 26 (13) (2005) 15151522.
(2012) 1823. [107] S. Sakai, I. Hashimoto, Y. Ogushi, K. Kawakami, Peroxidase-catalyzed cell
[81] R.M. Olabisi, Cell microencapsulation with synthetic polymers, J. Biomed. encapsulation in subsieve-size capsules of alginate with phenol moieties in
Mater. Res. A 103 (2) (2015) 846859.
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 25

water-immiscible uid dissolving H2O2, Biomacromolecules 8 (8) (2007) [135] D.B. Jaroch, J. Lu, R. Madangopal, N.D. Stull, M. Stensberg, J. Shi, et al., Mouse
26222626. and human islets survive and function after coating by biosilicication, Am. J.
[108] S. Sakai, K. Kawakami, Synthesis and characterization of both ionically and Physiol. Endocrinol. Metab. 305 (10) (2013) E1230E1240.
enzymatically cross-linkable alginate, Acta Biomater. 3 (4) (2007) 495501. [136] T.A. Desai, W.H. Chu, G. Rasi, P. Sinibaldi-Vallebona, E. Guarino, M. Ferrari,
[109] B.T. Stokke, O. Smidsroed, P. Bruheim, G. Skjaak-Braek, Distribution of Microfabricated biocapsules provide short-term immunoisolation of
uronate residues in alginate chains in relation to alginate gelling properties, insulinoma xenografts, Biomed. Microdevices 1 (2) (1999) 131138.
Macromolecules 24 (16) (1991) 46374645. [137] D. Gong, V. Yadavalli, M. Paulose, M. Pishko, C.A. Grimes, Controlled
[110] J.P. Hall, P. de Vos, The need for new therapeutic approaches and the molecular release using nanoporous alumina capsules, Biomed.
bioarticial endocrine pancreas, The Bioarticial Pancreas and Other Microdevices 5 (1) (2003) 7580.
Biohybrid Techniques, Transworld Research Network, Kerala, India, 20091 [138] K.C. Popat, G. Mor, C.A. Grimes, T.A. Desai, Surface modication of
26. nanoporous alumina surfaces with poly (ethylene glycol), Langmuir 20 (19)
[111] D. Poncelet, S.K. Tam, Microencapsulation technologies for a bioartical (2004) 80358041.
endocrine pancreas, The Bioarticial Pancreas and Other Biohybrid [139] E.E.L. Swan, K.C. Popat, C.A. Grimes, T.A. Desai, Fabrication and evaluation of
Techniques, Transworld Research Network, Kerala, India, 20093750. nanoporous alumina membranes for osteoblast culture, J. Biomed. Mater.
[112] U. Prsse, L. Bilancetti, M. Bu9 cko, B. Bugarski, J. Bukowski, P. Gemeiner, et al., Res. A 72 (3) (2005) 288295.
Comparison of different technologies for alginate beads production, Chem. [140] K.E.L. Flamme, G. Mor, D. Gong, T.L. Tempa, V.A. Fusaro, C.A. Grimes, et al.,
Papers 62 (4) (2008) 364374. Nanoporous alumina capsules for cellular macroencapsulation: transport
[113] V. Nemethova, I. Lacik, F. Razga, Vibration technology for and biocompatibility, Diabetes Technol. Ther. 7 (5) (2005) 684694.
microencapsulation: the restrictive role of viscosity, J. Bioprocess. Biotechnol. [141] M. Paulose, H.E. Prakasam, O.K. Varghese, L. Peng, K.C. Popat, G.K. Mor, et al.,
5 (199) (2015) 2. TiO2 nanotube arrays of 1000 mm length by anodization of titanium foil:
[114] D. Serp, E. Cantana, C. Heinzen, U. Von Stockar, I.W. Marison, Characterization phenol red diffusion, J. Phys. Chem. C 111 (41) (2007) 1499214997.
of an encapsulation device for the production of monodisperse alginate [142] M. Paulose, L. Peng, K.C. Popat, O.K. Varghese, T.J. LaTempa, N. Bao, et al.,
beads for cell immobilization, Biotechnol. Bioeng. 70 (1) (2000) 4153. Fabrication of mechanically robust: large area, polycrystalline nanotubular/
[115] U. Pre, U. Jahnz, P. Wittlich, J. Breford, K.D. Vorlop, Bead production with porous TiO2 membranes, J. Membr. Sci. 319 (1) (2008) 199205.
JetCutting and rotating disc/nozzle technologies, Landbauforschung [143] S.P. Albu, A. Ghicov, J.M. Macak, R. Hahn, P. Schmuki, Self-organized, free-
Vlkenrode SH 241 (2002) 110. standing TiO2 nanotube membrane for ow-through photocatalytic
[116] M. Whelehan, I.W. Marison, Microencapsulation using vibrating technology, applications, Nano Lett. 7 (5) (2007) 12861289.
J. Microencapsul. 28 (8) (2011) 669688. [144] S.P. Albu, A. Ghicov, S. Berger, H. Jha, P. Schmuki, TiO2 nanotube layers:
[117] D. Poncelet, R. Lencki, C. Beaulieu, J.P. Halle, R.J. Neufeld, A. Fournier, exible and electrically active ow-through membranes, Electrochem.
Production of alginate beads by emulsication/internal gelation. I. Commun. 12 (10) (2010) 13521355.
Methodology, Appl. Microbiol. Biotechnol. 38 (1) (1992) 3945. [145] J. Schweicher, T.A. Desai, Facile synthesis of robust free-standing TiO2
[118] C.A. Hoesli, K. Raghuram, R.L. Kiang, D. Mocinecov, X. Hu, J.D. Johnson, et al., nanotubular membranes for bioltration applications, J. Appl. Electrochem.
Pancreatic cell immobilization in alginate beads produced by emulsion and 44 (3) (2014) 411418.
internal gelation, Biotechnol. Bioeng. 108 (2) (2011) 424434. [146] J. Kwon, K. Trivedi, N.V. Krishnamurthy, W. Hu, J.B. Lee, B. Gimi, SU-8-based
[119] E. Piskin, Biodegradable polymers as biomaterials, J. Biomater. Sci. Polym. Ed. immunoisolative microcontainer with nanoslots dened by nanoimprint
6 (9) (1995) 775795. lithography, J. Vac. Sci. Technol. B 27 (6) (2009) 27952800.
[120] K.K. Hall, K.M. Gatts-Asfura, C.L. Stabler, Microencapsulation of islets within [147] B. Gimi, Making a case: nanofabrication techniques in encapsulated cell
alginate/poly (ethylene glycol) gels cross-linked via Staudinger ligation, Acta therapy for people with diabetes, J. Diabetes Sci. Technol. 4 (4) (2010) 1016
Biomater. 7 (2) (2011) 614624. 1021.
[121] Y. Teramura, O.P. Oommen, J. Olerud, J. Hilborn, B. Nilsson, [148] B. Gimi, J. Kwon, L. Liu, Y. Su, K. Nemani, K. Trivedi, et al., Cell encapsulation
Microencapsulation of cells, including islets, within stable ultra-thin and oxygenation in nanoporous microcontainers, Biomed. Microdevices 11
membranes of maleimide-conjugated PEG-lipid with multifunctional (6) (2009) 12051212.
crosslinkers, Biomaterials 34 (11) (2013) 26832693. [149] C.L. Randall, E. Gultepe, D.H. Gracias, Self-folding devices and materials for
[122] C.J. Young, L.A. Poole-Warren, P.J. Martens, Combining submerged biomedical applications, Trends Biotechnol. 30 (3) (2012) 138146.
electrospray and UV photopolymerization for production of synthetic [150] B. Gimi, T. Leong, Z. Gu, M. Yang, D. Artemov, Z.M. Bhujwalla, et al., Self-
hydrogel microspheres for cell encapsulation, Biotechnol. Bioeng. 109 (6) assembled three dimensional radio frequency (RF) shielded containers for
(2012) 15611570. cell encapsulation, Biomed. Microdevices 7 (4) (2005) 341345.
[123] F. Cellesi, N. Tirelli, A new process for cell microencapsulation and other [151] T.G. Leong, P.A. Lester, T.L. Koh, E.K. Call, D.H. Gracias, Surface tension-driven
biomaterial applications: thermal gelation and chemical cross-linking in self-folding polyhedra, Langmuir 23 (17) (2007) 87478751.
tandem, J. Mater. Sci. Mater. Med. 16 (6) (2005) 559565. [152] R. Calaore, G. Basta, Clinical application of microencapsulated islets: actual
[124] J.Y. Jang, D.Y. Lee, S.J. Park, Y. Byun, Immune reactions of lymphocytes and prospectives on progress and challenges, Adv. Drug Deliv. Rev. 67 (2014) 84
macrophages against PEG-grafted pancreatic islets, Biomaterials 25 (17) 92.
(2004) 36633669. [153] J.L. Panza, W.R. Wagner, H.L. Rilo, R.H. Rao, E.J. Beckman, A.J. Russell,
[125] S.S. Kadam, M. Sudhakar, P.D. Nair, R.R. Bhonde, Reversal of experimental Treatment of rat pancreatic islets with reactive PEG, Biomaterials 21 (11)
diabetes in mice by transplantation of neo-islets generated from human (2000) 11551164.
amnion-derived mesenchymal stromal cells using immuno-isolatory [154] S. Krol, S. Del Guerra, M. Grupillo, A. Diaspro, A. Gliozzi, P. Marchetti,
macrocapsules, Cytotherapy 12 (8) (2010) 982991. Multilayer nanoencapsulation. New approach for immune protection of
[126] S. George, P.D. Nair, M.V. Risbud, R.R. Bhonde, Nonporous polyurethane human pancreatic islets, Nano Lett. 6 (9) (2006) 19331939.
membranes as islet immunoisolation matricesbiocompatibility studies, J. [155] J.H. Jeong, S.W. Hong, S. Hong, S. Yook, Y. Jung, J.B. Park, et al., Surface
Biomater. Appl. 16 (4) (2002) 327340. camouage of pancreatic islets using 6-arm-PEG-catechol in combined
[127] K. Suzuki, S. Bonner-Weir, J. Hollister, G.C. Weir, A method for estimating therapy with tacrolimus and anti-CD154 monoclonal antibody for
number and mass of islets transplanted within a membrane device, Cell xenotransplantation, Biomaterials 32 (31) (2011) 79617970.
Transplant. 5 (6) (1996) 613625. [156] Y. Teramura, H. Iwata, Bioarticial pancreas: microencapsulation and
[128] S.R. Winn, M.D. Lindner, A. Lee, G. Haggett, J.M. Francis, D.F. Emerich, conformal coating of islet of Langerhans, Adv. Drug Deliv. Rev. 62 (7) (2010)
Polymer-encapsulated genetically modied cells continue to secrete human 827840.
nerve growth factor for over one year in rat ventricles: behavioral and [157] J.T. Wilson, W. Cui, E.L. Chaikof, Layer-by-layer assembly of a conformal
anatomical consequences, Exp. Neurol. 140 (2) (1996) 126138. nanothin PEG coating for intraportal islet transplantation, Nano Lett. 8 (7)
[129] M. Qi, Y. Gu, N. Sakata, D. Kim, Y. Shirouzu, C. Yamamoto, et al., PVA hydrogel (2008) 19401948.
sheet macroencapsulation for the bioarticial pancreas, Biomaterials 25 (27) [158] W.D. Kelly, R.C. Lillehei, F.K. Merkel, Y. Idezuki, F.G. Goetz, T. Maki, et al.,
(2004) 58855892. Allotransplantation of the pancreas and duodenum along with the kidney in
[130] Z. Qi, Y. Shen, G. Yanai, K. Yang, Y. Shirouzu, A. Hiura, et al., The in vivo diabetic nephropathy, Studies 843 (1967) 845.
performance of polyvinyl alcohol macro-encapsulated islets, Biomaterials 31 [159] U. Boggi, S. Signori, F. Vistoli, S. D'Imporzano, G. Amorese, G. Consani, et al.,
(14) (2010) 40264031. Laparoscopic robot-assisted pancreas transplantation: rst world
[131] F. La, K.E. lamme, K.C. Popat, L. Leoni, E. Markiewicz, T.J. La Tempa, B.B. Roman, experience, Transplantation 93 (2) (2012) 201206.
et al., Biocompatibility of nanoporous alumina membranes for [160] D. Bosco, P. Meda, Reconstructing Islet Function in Vitro, Springer, US,
immunoisolation, Biomaterials 28 (16) (2007) 26382645. 1997285298.
[132] T.A. Desai, T. West, M. Cohen, T. Boiarski, A. Rampersaud, Nanoporous [161] A.M. Davalli, Y. Ogawa, L. Scaglia, Y.J. Wu, J. Hollister, S. Bonner-Weir, et al.,
microsystems for islet cell replacement, Adv. Drug Deliv. Rev. 56 (11) (2004) Function, mass, and replication of porcine and rat islets transplanted into
16611673. diabetic nude mice, Diabetes 44 (1) (1995) 104111.
[133] S. Sharma, R.W. Johnson, T.A. Desai, Ultrathin poly (ethylene glycol) lms for [162] S. Sandler, A. Andersson, D.L. Eizirik, C. Hellerstrm, T. Espevik, B. Kulseng,
silicon-based microdevices, Appl. Surf. Sci. 206 (1) (2003) 218229. et al., Assessment of insulin secretion in vitro from microencapsulated fetal
[134] T.A. Desai, D. Hansford, M. Ferrari, Characterization of micromachined silicon porcine islet-like cell clusters and rat, mouse, and human pancreatic islets,
membranes for immunoisolation and bioseparation applications, J. Membr. Transplantation 63 (12) (1997) 17121718.
Sci. 159 (1) (1999) 221231. [163] C.K. Colton, Oxygen supply to encapsulated therapeutic cells, Adv. Drug Deliv.
Rev. 67 (2014) 93110.
26 V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227

[164] Y.L. Lightfoot, J. Chen, C.E. Mathews, Role of the mitochondria in immune- [193] H. Takeuchi, N. Nakatsuji, H. Suemori, Endodermal differentiation of human
mediated apoptotic death of the human pancreatic b cell line bLox5, PLoS pluripotent stem cells to insulin-producing cells in 3D culture, Sci. Rep. 4
One 6 (6) (2011) e20617. (2014) .
[165] Y.L. Lightfoot, J. Chen, C.E. Mathews, Immune-mediated b-cell death in type [194] K.M. Loh, L.T. Ang, J. Zhang, V. Kumar, J. Ang, J.Q. Auyeong, et al., Efcient
1 diabetes: lessons from human b-cell lines, Eur. J. Clin. Invest. 42 (11) (2012) endoderm induction from human pluripotent stem cells by logically
12441251. directing signals controlling lineage bifurcations, Cell Stem Cell 14 (2) (2014)
[166] S. Vasu, N.H. McClenaghan, J.T. McCluskey, P.R. Flatt, Mechanisms of toxicity 237252.
by proinammatory cytokines in a novel human pancreatic beta cell line, [195] F. Hakim, T. Kaitsuka, J.M. Raeed, F.Y. Wei, N. Shiraki, T. Akagi, et al., High
1.1 B4, Biochim. Biophys. Acta 1840 (1) (2014) 136145. oxygen condition facilitates the differentiation of mouse and human
[167] P. Ravassard, Y. Hazhouz, S. Pechberty, E. Bricout-Neveu, M. Armanet, P. pluripotent stem cells into pancreatic progenitors and insulin-producing
Czernichow, et al., A genetically engineered human pancreatic b cell line cells, J. Biol. Chem. 289 (14) (2014) 96239638.
exhibiting glucose-inducible insulin secretion, J. Clin. Invest. 121 (9) (2011) [196] R. Lahmy, M. Soleimani, M.H. Sanati, M. Behmanesh, F. Kouhkan, N. Mobarra,
3589. miRNA-375 promotes beta pancreatic differentiation in human induced
[168] R. Scharfmann, S. Pechberty, Y. Hazhouz, M. von Blow, E. Bricout-Neveu, M. pluripotent stem (hiPS) cells, Mol. Biol. Rep. 41 (4) (2014) 20552066.
Grenier-Godard, et al., Development of a conditionally immortalized human [197] Z. Alipio, W. Liao, E.J. Roemer, M. Waner, L.M. Fink, D.C. Ward, et al., Reversal
pancreatic b cell line, J. Clin. Invest. 124 (5) (2014) 2087. of hyperglycemia in diabetic mouse models using induced-pluripotent stem
[169] B.N. Newby, N. Terada, C.E. Mathews, In search of a surrogate: engineering (iPS)-derived pancreatic b-like cells, Proc. Natl. Acad. Sci. U. S. A. 107 (30)
human beta cell lines for therapy, Trends Endocrinol. Metab. 25 (8) (2014) (2010) 1342613431.
378380. [198] F.W. Pagliuca, J.R. Millman, M. Grtler, M. Segel, A. Van Dervort, J.H. Ryu, et al.,
[170] J. Lee, T. Sugiyama, Y. Liu, J. Wang, X. Gu, J. Lei, et al., Expansion and conversion Generation of functional human pancreatic b cells in vitro, Cell 159 (2) (2014)
of human pancreatic ductal cells into insulin-secreting endocrine cells, Elife 2 428439.
(2013) e00940. [199] R. Calaore, G. Basta, Stem cells for the cell and molecular therapy of type
[171] F. Furuya, H. Shimura, K. Asami, S. Ichijo, K. Takahashi, M. Kaneshige, et al., 1 diabetes mellitus (T1D): the gap between dream and reality, Am. J. Stem
Ligand-bound thyroid hormone receptor contributes to reprogramming of Cells 4 (1) (2015) 2231.
pancreatic acinar cells into insulin-producing cells, J. Biol. Chem. 288 (22) [200] R. Calaore, P. Montanucci, G. Basta, Stem cells for pancreatic b-cell
(2013) 1615516166. replacement in diabetes mellitus: actual perspectives, Curr. Opin. Organ
[172] Q. Zhou, J. Brown, A. Karanek, J. Rajagopal, D.A. Melton, In vivo Transplant. 19 (2) (2014) 162168.
reprogramming of adult pancreatic exocrine cells to b-cells, Nature 455 [201] S. Pellegrini, F. Ungaro, A. Mercalli, R. Melzi, G. Sebastiani, F. Dotta, et al.,
(2008) 627632. Human induced pluripotent stem cells differentiate into insulin-producing
[173] M.J. Lima, K.R. Muir, H.M. Docherty, R. Drummond, N.W. McGowan, S. Forbes, cells able to engraft in vivo, Acta Diabetol. (2015) 111.
et al., Suppression of epithelial-to-mesenchymal transitioning enhances ex [202] R. Calaore, G. Basta, G. Luca, A. Lemmi, M.P. Montanucci, G. Calabrese, et al.,
vivo reprogramming of human exocrine pancreatic tissue toward functional Microencapsulated pancreatic islet allografts into nonimmunosuppressed
insulin-producing b-like cells, Diabetes 62 (8) (2013) 28212833. patients with type 1 diabetes, Diabetes Care 29 (1) (2006) 137138.
[174] G.Q. Daley, The promise and perils of stem cell therapeutics, Cell Stem Cell 10 [203] G. Basta, P. Montanucci, G. Luca, C. Boselli, G. Noya, B. Barbaro, et al., Long-
(6) (2012) 740749. term metabolic and immunological follow-up of nonimmunosuppressed
[175] M. Li, G.H. Liu, J.C.I. Belmonte, Navigating the epigenetic landscape of patients with type 1 diabetes treated with microencapsulated islet allografts
pluripotent stem cells, Nat. Rev. Mol. Cell Biol. 13 (8) (2012) 524535. four cases, Diabetes Care 34 (11) (2011) 24062409.
[176] G. Lanzoni, T. Oikawa, Y. Wang, C.B. Cui, G. Carpino, V. Cardinale, et al., Concise [204] https.//clinicaltrials.gov/ct2/show/record/NCT01379729.
review: clinical programs of stem cell therapies for liver and pancreas, Stem [205] D. Jacobs-Tulleneers-Thevissen, M. Chintinne, Z. Ling, P. Gillard, L.
Cells 31 (10) (2013) 20472060. Schoonjans, G. Delvaux, et al., Sustained function of alginate-encapsulated
[177] G. Tan, A.G. Elefanty, E.G. Stanley, b-cell regeneration and differentiation: human islet cell implants in the peritoneal cavity of mice leading to a pilot
how close are we to the holy grail? J. Mol. Endocrinol. 53 (3) (2014) R119 study in a type 1 diabetic patient, Diabetologia 56 (7) (2013) 16051614.
R129. [206] https//clinicaltrials.gov/ct2/show/results/NCT00940173.
[178] H. Edlund, Pancreatic organogenesisdevelopmental mechanisms and [207] R.B. Elliott, L. Escobar, P.L. Tan, M. Muzina, S. Zwain, C. Buchanan, Live
implications for therapy, Nat. Rev. Genet. 3 (7) (2002) 524532. encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after
[179] H.P. Shih, A. Wang, M. Sander, Pancreas organogenesis: from lineage xenotransplantation, Xenotransplantation 14 (2) (2007) 157161.
determination to morphogenesis, Annu. Rev. Cell Dev. Biol. 29 (2013) 81105. [208] http://www.lctglobal.com/Products-and-Services/Diabecell.
[180] F.C. Pan, C. Wright, Pancreas organogenesis: from bud to plexus to gland, Dev. [209] E. Dolgin, Encapsulate this, Nat. Med. 20 (1) (2014) 911.
Dyn. 240 (3) (2011) 530565. [210] http://beta-o2.com/the-insulin-bioreactor-sair/.
[181] J. Jiang, M. Au, K. Lu, A. Eshpeter, G. Korbutt, G. Fisk, et al., Generation of [211] U. Barkai, G.C. Weir, C.K. Colton, B. Ludwig, S.R. Bornstein, M.D. Brendel, et al.,
insulin-producing islet-like clusters from human embryonic stem cells, Stem Enhanced oxygen supply improves islet viability in a new bioarticial
Cells 25 (8) (2007) 19401953. pancreas, Cell Transplant. 22 (8) (2013) 14631476.
[182] E. Kroon, L.A. Martinson, K. Kadoya, A.G. Bang, O.G. Kelly, S. Eliazer, et al., [212] T. Neufeld, B. Ludwig, U. Barkai, G.C. Weir, C.K. Colton, Y. Evron, et al., The
Pancreatic endoderm derived from human embryonic stem cells generates efcacy of an immunoisolating membrane system for islet
glucose-responsive insulin-secreting cells in vivo, Nat. Biotechnol. 26 (4) xenotransplantation in minipigs, PLoS One 8 (8) (2013) e70150.
(2008) 443452. [213] B. Ludwig, A. Reichel, A. Steffen, B. Zimerman, A.V. Schally, N.L. Block, et al.,
[183] K.A. DAmour, A.G. Bang, S. Eliazer, O.G. Kelly, A.D. Agulnick, N.G. Smart, et al., Transplantation of human islets without immunosuppression, Proc. Natl.
Production of pancreatic hormoneexpressing endocrine cells from human Acad. Sci. U. S. A. 110 (47) (2013) 1905419058.
embryonic stem cells, Nat. Biotechnol. 24 (11) (2006) 13921401. [214] http://www.theracyte.com/TheTechnology.htm.
[184] B. Bose, S.P. Shenoy, S. Konda, P. Wangikar, Human embryonic stem cell [215] I.R. Sweet, O. Yanay, L. Waldron, M. Gilbert, J.M. Fuller, T. Tupling, et al.,
differentiation into insulin secreting b-cells for diabetes, Cell Biol. Int. 36 (11) Treatment of diabetic rats with encapsulated islets, J. Cell Mol. Med. 12 (6b)
(2012) 10131020. (2008) 26442650.
[185] T.C. Schulz, H.Y. Young, A.D. Agulnick, M.J. Babin, E.E. Baetge, A.G. Bang, et al., [216] M. Kumagai-Braesch, S. Jacobson, H. Mori, X. Jia, T. Takahashi, A. Wernerson,
A scalable system for production of functional pancreatic progenitors from et al., The TheraCyteTM device protects against islet allograft rejection in
human embryonic stem cells, PLoS One 7 (5) (2012) e37004. immunized hosts, Cell Transplant. 22 (7) (2013) 11371146.
[186] A. Rezania, J.E. Bruin, J. Xu, K. Narayan, J.K. Fox, J.J. ONeil, et al., Enrichment of [217] N. Trivedi, G.M. Steil, C.K. Colton, S. Bonner-Weir, G.C. Weir, Improved
human embryonic stem cell-derived NKX6. 1-expressing pancreatic vascularization of planar membrane diffusion devices following continuous
progenitor cells accelerates the maturation of insulin-secreting cells in vivo, infusion of vascular endothelial growth factor, Cell Transplant. 9 (1) (1999)
Stem Cells 31 (11) (2013) 24322442. 115124.
[187] K. Kirk, E. Hao, R. Lahmy, P. Itkin-Ansari, Human embryonic stem cell derived [218] A.K. Srenby, M. Kumagai-Braesch, A. Sharma, K.R. Hultenby, A.M.
islet progenitors mature inside an encapsulation device without evidence of Wernerson, A.B. Tibell, Preimplantation of an immunoprotective device can
increased biomass or cell escape, Stem Cell Res. 12 (3) (2014) 807814. lower the curative dose of islets to that of free islet transplantationstudies
[188] C.L. Basford, K.J. Prentice, A.B. Hardy, F. Sarangi, S.J. Micallef, X. Li, et al., The in a rodent model, Transplantation 86 (2) (2008) 364366.
functional and molecular characterisation of human embryonic stem cell- [219] http://viacyte.com/products/vc-01-diabetes-therapy/#.
derived insulin-positive cells compared with adult pancreatic beta cells, [220] http://www.sernova.com/technology/.
Diabetologia 55 (2) (2012) 358371. [221] https.//www.clinicaltrials.gov/ct2/show/NCT01652911?
[189] S.J. Micallef, X. Li, J.V. Schiesser, C.E. Hirst, Q.C. Yu, S.M. Lim, et al., INS GFP/w term=NCT01652911&rank=1.
human embryonic stem cells facilitate isolation of in vitro derived insulin- [222] J. Kriz, G. Vilk, D.M. Mazzuca, P.M. Toleikis, P.J. Foster, D.J. White, A novel
producing cells, Diabetologia 55 (3) (2012) 694706. technique for the transplantation of pancreatic islets within a vascularized
[190] S.M. Wu, K. Hochedlinger, Harnessing the potential of induced pluripotent device into the greater omentum to achieve insulin independence, Am. J.
stem cells for regenerative medicine, Nat. Cell Biol. 13 (5) (2011) 497505. Surg. 203 (6) (2012) 793797.
[191] S.I. Nishikawa, R.A. Goldstein, C.R. Nierras, The promise of human induced [223] http://www.hanumanmedicalfoundation.org/islet-sheet-project.html.
pluripotent stem cells for research and therapy, Nat. Rev. Mol. Cell Biol. 9 (9) [224] S.R. King, R. Dorian, R.W. Storrs, Requirements for encapsulation technology
(2008) 725729. and the challenges for transplantation of islets of Langerhans, Graft 4 (7)
[192] V. Tabar, L. Studer, Pluripotent stem cells in regenerative medicine: (2001) 491.
challenges and recent progress, Nat. Rev. Genet. 15 (2) (2014) 8292.
V. Iacovacci et al. / Biochemical Pharmacology 100 (2016) 1227 27

[225] R. Storrs, R. Dorian, S.R. King, J. Lakey, H. Rilo, Preclinical development of the [246] L. Ricotti, T. Fujie, H. Vazo, G. Ciofani, R. Marotta, R. Brescia, et al., Boron
islet sheet, Ann. N. Y. Acad. Sci. 944 (1) (2001) 252266. nitride nanotube-mediated stimulation of cell co-culture on micro-
[226] J. Schweicher, C. Nyitray, T.A. Desai, Membranes to achieve engineered hydrogels, PLoS One 8 (8) (2013) e71707.
immunoprotection of transplanted islets, Front. Biosci. (Landmark ed.) 19 [247] L. Ricotti, R.P. das Neves, G. Ciofani, C. Canale, S. Nitti, V. Mattoli, et al., Boron
(2014) 49. nitride nanotube-mediated stimulation modulates F/G-actin ratio and
[227] Y. Sun, C.S. Chen, J. Fu, Forcing stem cells to behave: a biophysical perspective mechanical properties of human dermal broblasts, J. Nanopart. Res. 16 (2)
of the cellular microenvironment, Annu. Rev. Biophys. 41 (2012) 519. (2014) 114.
[228] P. Costa, F.V. Almeida, J.T. Connelly, Biophysical signals controlling cell fate [248] L. Ricotti, A. Menciassi, Nanotechnology in biorobotics: opportunities and
decisions: how do stem cells really feel? Int. J. Biochem. Cell Biol. 44 (2012) challenges, J. Nanopart. Res. 17 (2) (2015) 110.
22332237. [249] G.C. Weir, Islet encapsulation: advances and obstacles, Diabetologia 56 (7)
[229] E. Ghafar-Zadeh, J.R. Waldeisen, L.P. Lee, Engineered approaches to the stem (2013) 14581461.
cell microenvironment for cardiac tissue regeneration, Lab Chip 11 (18) [250] G. Reach, M.Y. Jaffrin, J.F. Desjeux, A u-shaped bioarticial pancreas with
(2011) 30313048. rapid glucose-insulin kinetics: in vitro evaluation and kinetic modelling,
[230] L. Ricotti, A. Menciassi, Engineering stem cells for future medicine, Biomed. Diabetes 33 (8) (1984) 752761.
Eng. IEEE Trans. 60 (3) (2013) 727734. [251] E.S. Augoustiniatos, C.K. Colton, Effect of external oxygen mass transfer
[231] Y.L. Han, S. Wang, X. Zhang, Y. Li, G. Huang, H. Qi, et al., Engineering physical resistances on viability of immunoisolated tissue, Ann. N. Y. Acad. Sci. 831 (1)
microenvironment for stem cell based regenerative medicine, Drug Discov. (1997) 145166.
Today 19 (6) (2014) 763773. [252] A.S. Johnson, R.J. Fisher, G.C. Weir, C.K. Colton, Oxygen consumption and
[232] C.M. Metallo, J.C. Mohr, C.J. Detzel, J.J. de Pablo, B.J. Van Wie, S.P. Palecek, diffusion in assemblages of respiring spheres: performance enhancement of
Engineering the stem cell microenvironment, Biotechnol. Progress 23 (2007) a bioarticial pancreas, Chem. Eng. Sci. 64 (22) (2009) 44704487.
1823. [253] J.L. Dulong, C. Legallais, What are the relevant parameters for the geometrical
[233] J.A. Burdick, G. Vunjak-Novakovic, Engineered microenvironments for optimization of an implantable bioarticial pancreas? J. Biomech. Eng. 127
controlled stem cell differentiation, Tissue Eng. Part A 15 (2) (2008) 205219. (7) (2005) 10541061.
[234] C.J. Bettinger, R. Langer, J.T. Borenstein, Engineering substrate topography at [254] P. Buchwald, Exploratory FEM-based multiphysics oxygen transport and cell
the micro-and nanoscale to control cell function, Angew. Chem. Int. Ed. 48 viability models for isolated pancreatic islets, Proceedings of the COMSOL
(30) (2009) 54065415. Conference, Boston 2008; Comsol, Inc., Boston, 2008.
[235] A.J. Engler, S. Sen, H.L. Sweeney, D.E. Discher, Matrix elasticity directs stem [255] J.L. Dulong, C. Legallais, Contributions of a nite element model for the
cell lineage specication, Cell 126 (4) (2006) 677689. geometric optimization of an implantable bioarticial pancreas, Artif. Organs
[236] J. Lii, W.J. Hsu, H. Parsa, A. Das, R. Rouse, S.K. Sia, Real-time microuidic 26 (7) (2002) 583589.
system for studying mammalian cells in 3D microenvironments, Anal. Chem. [256] J.L. Dulong, C. Legallais, S. Darquy, G. Reach, A novel model of solute transport
80 (10) (2008) 36403647. in a hollow-ber bioarticial pancreas based on a nite element method,
[237] V.F. Shimko, W.C. Claycomb, Effect of mechanical loading on three- Biotechnol. Bioeng. 78 (5) (2002) 576582.
dimensional cultures of embryonic stem cell-derived cardiomyocytes, Tissue [257] P. Buchwald, S.R. Cechin, Glucose-stimulated insulin secretion in isolated
Eng. Part A 14 (1) (2008) 4958. pancreatic islets: multiphysics FEM model calculations compared to results
[238] W.Y. Sim, S.W. Park, S.H. Park, B.H. Min, S.R. Park, S.S. Yang, A pneumatic micro of perifusion experiments with human islets, J. Biomed. Sci. Eng. 6 (5A)
cell chip for the differentiation of human mesenchymal stem cells under (2013) .
mechanical stimulation, Lab Chip 7 (12) (2007) 17751782. [258] J.L. Dulong, C. Legallais, A theoretical study of oxygen transfer including cell
[239] J.W. Choi, B.H. Choi, S.H. Park, K.S. Pai, T.Z. Li, B.H. Min, et al., Mechanical necrosis for the design of a bioarticial pancreas, Biotechnol. Bioeng. 96 (5)
stimulation by ultrasound enhances chondrogenic differentiation of (2007) 990998.
mesenchymal stem cells in a brin-hyaluronic acid hydrogel, Artif. Organs 37 [259] K.J. Cash, H.A. Clark, Nanosensors and nanomaterials for monitoring glucose
(7) (2013) 648655. in diabetes, Trends Mol. Med. 16 (12) (2010) 584593.
[240] M. Yamada, K. Tanemura, S. Okada, A. Iwanami, M. Nakamura, H. Mizuno, [260] S.A. John, M. Ottolia, J.N. Weiss, B. Ribalet, Dynamic modulation of intraular
et al., Electrical stimulation modulates fate determination of differentiating glucose imaged in single cells using a FRET-based glucose nanosensor,
embryonic stem cells, Stem Cells 25 (3) (2007) 562570. Pgers Archiv.-Eur. J. Physiol. 456 (2) (2008) 307322.
[241] B.C. Wheeler, G.J. Brewer, Designing neural networks in culture, Proc. IEEE 98 [261] M. Staples, K. Daniel, M.J. Cima, R. Langer, Application of micro-and nano-
(3) (2010) 398406. electromechanical devices to drug delivery, Pharm. Res. 23 (5) (2006) 847
[242] N. Tandon, A. Marsano, R. Maidhof, K. Numata, C. Montouri-Sorrentino, C. 863.
Cannizzaro, et al., Surface-patterned electrode bioreactor for electrical [262] S. Rahiman, B.A. Tantry, Nanomedicine current trends in diabetes
stimulation, Lab Chip 10 (6) (2010) 692700. management, J. Nanomed. Nanotechol. 3 (2012) 5.
[243] F. Greco, T. Fujie, L. Ricotti, S. Taccola, B. Mazzolai, V. Mattoli, Microwrinkled [263] O. Veiseh, J.C. Doloff, M. Ma, A.J. Vegas, H.H. Tam, A.R. Bader, et al., Size-and
conducting polymer interface for anisotropic multicellular alignment, ACS shape-dependent foreign body immune response to materials implanted in
Appl. Mater. Interfaces 5 (3) (2013) 573584. rodents and non-human primates, Nat. Mater. 4 (2015) 643652.
[244] M.K. Kreysing, T. Kieling, A. Fritsch, C. Dietrich, J.R. Guck, J.A. Ks, The optical [264] A.R. Pepper, B. Gala-Lopez, R. Pawlick, S. Merani, T. Kin, A.J. Shapiro, A
cell rotator, Opt. Express 16 (21) (2008) 1698416992. prevascularized subcutaneous device-less site for islet and cellular
[245] M.T. Tsai, W.J. Li, R.S. Tuan, W.H. Chang, Modulation of osteogenesis in human transplantation, Nat. Biotechnol. 33 (5) (2015) 518523.
mesenchymal stem cells by specic pulsed electromagnetic eld stimulation, [265] M.A. McEvoy, N. Correll, Materials that couple sensing, actuation,
J. Orthop. Res. 27 (9) (2009) 11691174. computation, and communication, Science 347 (6228) (2015) 1261689.

Potrebbero piacerti anche