Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Xenotransplantation 2016: 23: 179–201 © 2016 John Wiley & Sons A/S.

doi: 10.1111/xen.12240 Published by John Wiley & Sons Ltd.


XENOTRANSPLANTATION

Review Article

Contribution of polymeric materials to


progress in xenotransplantation of
microencapsulated cells: a review
Mahou R, Passemard S, Carvello M, Petrelli A, Noverraz F, Gerber- Redouan Mahou,1,2,3 Solene
Lemaire S, Wandrey C. Contribution of polymeric materials to Passemard,1,2 Michele Carvello,4
progress in xenotransplantation of microencapsulated cells: a review. Alessandra Petrelli,4 Francßois
Xenotransplantation 2016: 23: 179–201. © 2016 John Wiley & Sons A/S. Noverraz,1,2 Sandrine Gerber-
Published by John Wiley & Sons Ltd. Lemaire1,2 and Christine Wandrey1,2
1
Interfaculty Institute of Bioengineering, Ecole
Abstract: Cell microencapsulation and subsequent transplantation of
Polytechnique Federale de Lausanne, Lausanne,
the microencapsulated cells require multidisciplinary approaches. Physi- 2
Institute of Chemical Sciences and Engineering,
cal, chemical, biological, engineering, and medical expertise has to be Ecole Polytechnique Federale de Lausanne,
combined. Several natural and synthetic polymeric materials and differ- Lausanne, Switzerland, 3Institute for Biomaterials
ent technologies have been reported for the preparation of hydrogels, and Biomedical Engineering, University of Toronto,
which are suitable to protect cells by microencapsulation. However, Toronto, ON, Canada, 4Department of Surgery, San
owing to the frequent lack of adequate characterization of the hydrogels Raffaele Scientific Institute, Milan, Italy
and their components as well as incomplete description of the technol-
ogy, many results of in vitro and in vivo studies appear contradictory or Key words: biocompatibility – cell
cannot reliably be reproduced. This review addresses the state of the art microencapsulation – hydrogels – mechanical
in cell microencapsulation with special focus on microencapsulated cells resistance – permeability – xenotransplantation
intended for xenotransplantation cell therapies. The choice of materials, Abbreviations: Alg-CS, alginate–cellulose sulfate;
the design and fabrication of the microspheres, as well as the conditions APA, alginate–poly(L-lysine)–alginate capsules;
to be met during the cell microencapsulation process, are summarized Ca-alg, calcium alginate; cryo-SEM, cryo-scanning
and discussed prior to presenting research results of in vitro and in vivo electron microscopy; DDA, degree of deacetylation;
studies. Overall, this review will serve to sensitize medically educated ECM, extracellular matrix; FGF-1, heparin-binding
specialists for materials and technological aspects of cell microencapsu- growth factor 1; G, a-L-guluronic acid; HPC,
lation. hydroxypropyl cellulose; IL, interleukin; ISEC, inverse
size-exclusion chromatography; LCST, lower critical
solution temperature; M, D-mannuronic acid;
MWCO, molecular weight cut-off; Na-alg, sodium
alginate; NICCs, neonatal pig islet-like cell clusters;
PEG, poly(ethylene glycol); PLL, poly(L-lysine); PLO,
poly(L-ornithine); PMCG, poly(methylene-co-guani-
dine); PNIPAA, poly(N-isopropylacrylamide); PNVIBA,
poly(N-vinylisobutyramide); RGD, arginylglycylaspar-
tic acid; TNF, tumor necrosis factor; UCST, upper
critical solution temperature; VEGF, vascular
endothelial growth factor
Address reprint requests to Sandrine
Gerber-Lemaire, Institute of Chemical Sciences and
Engineering, Ecole Polytechnique Federale de
Lausanne, EPFL SB ISIC, Batochime, CH-1015
Lausanne, Switzerland
(E-mail: sandrine.gerber@epfl.ch)

Received 29 February 2016;


Accepted 9 May 2016

Drug Administration (FDA) defines cell-based


Introduction
therapy as “the prevention, treatment, cure or miti-
Cell-based therapy is an attractive approach to gation of diseases or injuries in humans by the
treat several end-stage diseases. The Food and administration of autologous, allogeneic or

179
Mahou et al.

xenogeneic cells that have been manipulated or Microencapsulation also includes the entrapment
altered ex vivo” [1]. While the whole-organ trans- of biologically active substances such as cells, tis-
plantation is limited by the shortage of donors and sue, enzymes, bacteria, or DNA. For such applica-
the need of major surgery, cell-based therapy could tions, the term bioencapsulation is frequently used.
overcome both obstacles. Indeed, xenotransplanta- An ambitious challenge in bioencapsulation is cell
tion will offer an inexhaustible source of cells, and microencapsulation, which denotes the entrapment
these cells could be delivered near the target site of cells while maintaining their viability and meta-
using non-invasive procedures. bolic functionality (Fig. 1).
In spite of the enormous potential of such Cell immobilization in polymer-based hydrogels
approach, progress in the field of cell-based ther- was first proposed in 1933 by Bisceglie [4],
apy has been hampered for several reasons, in par- who demonstrated that insulin-producing cells
ticular due to issues of maintaining cell viability remained viable and metabolically active after
and of the identification of “non-self” cells by the immobilization. Three decades later, Chang pro-
immune system causing transplant rejection. posed the use of semipermeable membranes as
Although better patient and transplant survival immune-isolating devices and introduced the term
rates are achievable by the administration of “artificial cells” to define the concept of cell
immunosuppressive treatment, major challenges microencapsulation [5]. As shown in Fig. 1, cell
such as adverse effects associated with these drugs microencapsulation offers protection against
and the risks of long-term immunosuppression are mechanical stress or deteriorating environmental
still to be overcome. effects. The surrounding hydrogel allows for bidi-
The immobilization of cells within a hydrogel rectional diffusion of molecules essential for cell
material has been identified as efficient strategy to metabolism such as oxygen and nutrients, and the
provide mechanical and immune protection to the release of metabolic products. Simultaneously, the
cells, and to maintain their viability and metabolic passage of immune cells and antibodies is
functionality for subsequent therapeutic applica- restricted, giving rise to an immunoprotection for
tions [2,3]. Successful applications undoubtedly the encapsulated cells. Therapies relying on
require a multidisciplinary input from materials sci- microencapsulated cells could therefore result in
entists, chemists, biologists, engineers, and surgeons. the reduction or even avoidance of the administra-
The focus of the present review will be on cell tion of immunosuppressive drugs on the one hand
microencapsulation, even though the immobiliza- and, on the other hand, permit the transplantation
tion and protection of cells is achievable also by of non-human cells, which is a promising alterna-
other techniques such as immobilization in films, tive considering the limited availability of donor
extravascular chambers, or hollow fibers. This organs [6,7]. The therapeutic potential of the trans-
review provides an overview of suitable materials plantation of microencapsulated cells has been
under study for cell microencapsulation and dis- reported for the treatment of a variety of diseases,
cusses the special features of the technologies including liver failure [8–10], renal failure [11–13],
applied so far. The paper is mainly addressed to cancer [14], and diabetes mellitus [15–18].
medically educated readers working on developing
therapies that rely on hydrogels. The selection of
Hydrogels for cell microencapsulation
suitable material, the design and preparation of
spherical hydrogels, as well as the main require- Since the pioneering work of Wichterle on cross-
ments to be fulfilled during the cell immobilization linked poly(hydroxyethyl methacrylate) [19],
process are summarized and discussed. Recent tri-
als toward transplantation of xenogeneic microen-
capsulated cells in order to treat congenital or
acquired hormone/enzyme deficiencies as well as
degenerative/inflammatory diseases complete this
review.

Cell microencapsulation
Microencapsulation denotes the physical entrap-
ment of a gas, liquid, or solid within a surrounding
material with dimensions in the micrometer range.
It has gained interest in domains such as agricul-
ture, food, cosmetics, construction, and analytics. Fig. 1. Schematic representation of cell microencapsulation.

180
Polymeric materials for xenotransplantation

hydrogels have been of great interest to biomate- obtainable in a one-step process while avoiding the
rial scientists [20–24]. Hydrogels consist of a three- use of cross-linking agents. Only physical hydro-
dimensional network of natural [25] or synthetic gels obtained via ionic bonding or induced by tem-
[26] polymer chains. Due to their high water con- perature change are discussed herein.
tent, hydrogels have been recognized to meet the
requirements for bioencapsulation [27–30]. The Physical hydrogels by ionic bonding
cross-linking mechanism determines their classifi- The principle of preparing physical hydrogels via
cation as physical or chemical hydrogels. Hydro- ionic bonding is schematically represented in
gels are called “physical” or “reversible” if the Fig. 2. First, when a polyelectrolyte (polymer bear-
networks are held together by physical forces only. ing many positive or negative charges) interacts
Chemical hydrogels, also referred to as “perma- with multivalent ions of the opposite charge, it
nent,” are obtained when polymers having reactive may form a physical hydrogel known as “ionotro-
groups link together via covalent bonds. Hydrogels pic” hydrogel. Second, when polyelectrolytes of
used for cell microencapsulation are hereinafter opposite charges are mixed, they may gel depend-
designated as microspheres, which is a generic ing on their polymer backbone constitution, con-
name that refers to the size and shape of the mate- centrations, the ionic strength, as well as the pH of
rials. According to other classifications of micro- the solution. The products of such polyanion/poly-
spheres, which take into account the physical cation interactions are known as complex coacer-
structure of the hydrogel, microspheres are called vates, polyelectrolyte complexes, or simplexes. A
microbeads if the hydrogel is radially homoge- polyanion was selected in Fig. 2 as example to
neous. Contrary, the term microcapsule is used if demonstrate the principle. The complexation of
the microsphere is radially heterogeneous, for polycations is achievable similarly.
example, if a microbead was additionally coated A description of polymers that have been
with other polymers or if hydrogel surrounds a liq- explored in terms of their ability to form physical
uid core. hydrogels via ionic bonding is presented below.

Sodium alginate
Physical hydrogels
The designation sodium alginate (Na-alg) does not
Physically cross-linked hydrogels possess physical refer to a unique polymer structure but to a variety
junction domains associated with chain entangle- of polymers, which are composed of the same two
ments, ionic or hydrogen bonding, and hydropho- monomeric units, b-D-mannuronic acid (M) and
bic interactions [31–36]. The interest in these a-L-guluronic acid (G), but arranged in different
physical hydrogels is obvious since they are easily linear sequences [37, 38]. As shown in Fig. 3, the

Fig. 2. Principles to form physical


hydrogels by ionic bonding.

Fig. 3. Na-alg consists of a-L-guluronic


and b-D-mannuronic acid residues.

181
Mahou et al.

Fig. 4. The properties of alg-based


hydrogels depend on the nature of the
applied Na-alg, (solid line) confirmed
influence, (dotted line) controversially
discussed.

monomeric units can be arranged as blocks of dif- The molar mass of the Na-alg and the concen-
ferent lengths or randomly alternated in the poly- tration of its solution are further important param-
mer chains. Due to the advantageous gelling eters. Na-alg is available with molar mass in the
properties in contact with divalent cations, the Na- range of 50 to 3000 kg/mol [48, 49]. The viscosity
alg family is the most frequently used polymeric of a Na-alg solution increases with both the molar
material for cell microencapsulation. However, the mass and the concentration of Na-alg. Therefore, a
properties of alg-based hydrogels are very sensitive compromise between the molar mass of Na-alg
to the nature of the Na-alg and the preparation and its concentration is needed to adapt the viscos-
conditions. Consequently, the knowledge of the ity of the solution to a specific application in terms
molecular and macromolecular characteristics of of cell type and technology. A selection of applica-
Na-alg is crucial for the production of defined tions of Na-alg-based physical hydrogels is listed
hydrogels. Fig. 4 summarizes characteristics of in Table 1.
Na-alg that influence the properties of alg-based In spite of favorable properties, alg-based physi-
hydrogels. cal hydrogels suffer from drawbacks such as lim-
The chemical composition of Na-alg has an ited mechanical stability, insufficient durability,
impact on the stability and permeability of the alg- and too high permeability. They are dissolved
based hydrogels [38–40], attributed to ionic bond- when chelators such as phosphate, lactate, citrate,
ing between G units and divalent cations, referred and non-gelling cations are present above a certain
to as the egg-box model [41]. The importance of concentration [62]. To overcome such problems,
the G units is highlighted by the fact that the
strength of alg-based hydrogels is directly related Table 1. Selected alg-based physical hydrogels applied for cell
to the total content of G units and the average microencapsulation
length of the G blocks in Na-alg [42]. Furthermore,
Divalent
it has been demonstrated that calcium alginate ions Cell type Target Reference
beads (Ca-alg) prepared from Na-alg with high G
2+ a
content are more permeable and exhibit less water Ca BMSC Treatment of stress urinary [50]
incontinence
uptake compared to Ca-alg prepared from Na-alg Hepatocytes Development of bio-artificial liver [51]
with high M content [43]. The composition–bio- Pig islets Impact of implantation sites on the [52]
compatibility relationship, however, is still a mat- biocompatibility
ter of controversy. Some studies reported that Na- ADSCb Study of angiogenic and osteogenic [53,54]
potential of ADSC
alg with a high content of M evokes an inflamma- CSPc Therapeutic approach for cartilage [55]
tory response by stimulating monocytes to produce regeneration
cytokines such as interleukin IL-1, IL-6, and TNF Ba 2+
Rat islets Study of islet function in vitro and [56]
[44]. Moreover, antibodies were found when high- in vivo
Neuroblastoma Cryopreservation of neurospheres by [57]
M alg-based hydrogels were transplanted, but not encapsulation
in the case of high-G alg materials [45]. In contrast, WJMSCd Optimized microencapsulation of MSC [58]
other studies claim high-G alg-based hydrogels to by vibrational nozzle
Ba2+/ Human islets Viability and function after [59,60]
be associated with more severe cell overgrowth
Ca2+ transplantation into diabetic mice.
[46]. In addition to the composition, the purity of ARPE-19e In vitro study of encapsulated human [61]
the Na-alg cannot be neglected [47]. There is a con- retinal pigment epithelial cells
sensus that in vitro and in vivo studies have to be a
Bone marrow mesenchymal stem cells; badipose-derived stem cells; chuman mes-
conducted using highly purified Na-alg, free of enchymal progenitor cells from the subchondral bone marrow; dWharton’s jelly
endotoxin, proteins, and polyphenols. mesenchymal stem cells; ehuman retinal pigment epithelial cells.

182
Polymeric materials for xenotransplantation

subsequent coating of the initially formed hydro-


gels with polycations to form a polyanion–polyca-
tion complex has been proposed. Poly(L-lysine)
(PLL), poly(L-ornithine) (PLO), and chitosan are
the main polycations that have been suggested for
the coating of alg-based hydrogels. A final layer of
Na-alg is needed to neutralize the excess of positive
charges, to turn the surface charge into negative,
and thus to avoid biocompatibility problems
caused primarily by attachment of proteins to a
surface with positive charges.

Poly(L-lysine) and poly(L-ornithine)


Since the first transplantation of islets of Langer-
hans microencapsulated in alg-based hydrogel
coated with PLL was published [63], the applica-
tion of PLL as coating material for alg-based
Fig. 5. Chemical structure of poly(L-lysine) hydrochloride and
hydrogels has been widely reported. Typically, the poly(L-ornithine) hydrochloride.
alginate–poly(L-lysine)–alginate (APA) microcap-
sule comprises three main components: a core of
Ca-alg surrounded by an alginate–PLL complex hydrophilicity, improved mechanical properties,
and an outer coating of Na-alg. The coating rein- and superior resistance to swelling and damage
forces the mechanical resistance of Ca-alg and under osmotic stress [87,88]. In terms of biocom-
allows for controlling the permeability. The sur- patibility, however, the suitability of using PLO
rounding polyanion–polycation complex reduces instead of PLL is still controversial. Likewise PLL,
the osmotic swelling and thus stabilizes the micro- the immunological response to PLO containing
capsule size. Numerous studies using APA to microcapsules was reduced by PEGylation of the
encapsulate cells have been reported [64–70]. outer layer [89]. While some studies claim that
Despite improvement of the physical properties promising results are obtained when replacing PLL
of alg hydrogels upon coating with PLL, the by PLO [90–92], others suggest that PLL remains
immunological response upon transplantation of the best option [93,94].
APA remains a major challenge. A multitude of
studies have demonstrated that Ca-alg is better tol- Chitosan
erated in vivo than polycation-coated microcap- Chitosan is a linear polysaccharide consisting of
sules [71–73]. The proposed mechanism involves randomly distributed b(1?4) linked D-glucosa-
the adhesion of proteins to the surface of the mine and N-acetyl-D-glucosamine units, as illus-
hydrogel when this latter is exposed to blood, trated in Fig. 6. The cationic properties of
plasma, or peritoneal fluid. This adhesion initiates chitosan depend on the degree of deacetylation
the reactive protein cascades and serves as cellular (DDA), which is the fraction of glucosamine
anchor [74–78]. To overcome these biocompatibil- units. The molar mass depends on the source
ity issues, different methods have been proposed to and isolation procedure, and it can reach values
reduce the protein adsorption to the surface of up to 500 kg/mol. Lower molar masses and oli-
APA. For instance, grafting biocompatible poly gomers are obtained by chain degradation [95].
(ethylene glycol) (PEG) pendent chains [79–81], Chitosan is a non-permanently charged cationic
optimizing the APA composition [46] and size [82], polyelectrolyte. Its charge density and solubility
or using epimerized Na-alg [83] was reported. strongly depend on the DDA and pH. With a
Other efforts were directed toward exploring the pKa value of approximately 6.5, chitosan is posi-
suitability of other polycations as coating materi- tively charged and soluble in acidic to neutral
als. It has been suggested that using PLO has media, only if the DDA exceeds 60%. Excep-
advantages compared to the use of PLL [84–86]. tions are oligomers.
PLO is more hydrophilic than PLL due to the dif- Similarly to PLL and PLO, chitosan has been
ference in the chemical structure, as shown in applied to coat Ca-alg or Ba-alg [96]. However,
Fig. 5. PLO binds more efficiently to the surface of microspheres with a Ca-alg core covered by an
Ca-alg, which in turn has a positive impact on the alginate–chitosan complex membrane can also be
physical properties of the microspheres. Micro- made in a one-stage process by dropping Na-alg
spheres coated with PLO exhibited higher into an aqueous solution containing chitosan and

183
Mahou et al.

Fig. 6. The chemical structure of chitosan.

calcium ions [97]. The opposite process, dropping the ratio of Na-alg/CS in the polyanion blend and
chitosan and Ca2+ solutions into Na-alg yields the chemical composition [43,116,117]. A subse-
microspheres with a chitosan core and a chitosan– quent coating with lower molar mass Na-alg
alginate membrane [98]. The stability, permeabil- allowed adjusting the permeability over a wide
ity, and biocompatibility of alg–chitosan micro- range, suitable for cell microencapsulation and
capsules were intensely studied [99–105]. Several immunoprotection, without compromising the
cell types were successfully encapsulated in alg–chi- durability of the microspheres. A number of stud-
tosan microspheres including islets of Langerhans ies using alg-CS/PMCG microcapsules were pub-
[106,107], hepatocytes [108–113], and mesenchymal lished [118–123]. However, the in vivo
stem cells [114,115]. biocompatibility remains a major issue. Indeed,
when a human whole blood model was used to
Sodium cellulose sulfate and poly(methylene-co-guani- assess the inflammatory properties of alg-CS/
dine) PMCG microspheres, they have triggered comple-
Alginate–cellulose sulfate–poly(methylene-co-gua- ment and leukocyte activation over time, although
nidine) microcapsules (alg-CS/PMCG) are pre- they were still less activating than PLL-containing
pared by a two-step process (Fig. 7). First, a microcapsules [71].
polyanion blend of Na-alg and CS is gelled in the
presence of calcium ions. Second, a membrane is Physical hydrogel by temperature response
formed via the addition of PMCG. It was demon- The preparation of temperature-responding
strated that both mechanical resistance and perme- hydrogels is emerging as a promising tool for
ability of alg-CS/PMCG are tunable by changing various biomedical applications [124], including
cell microencapsulation. Such hydrogels are
obtained from polymers that respond to tempera-
ture change and undergo a solgel transition
[125,126]. Derivatives of methylcellulose [127],
chitosan [128], hydroxypropyl cellulose (HPC)
[129], poly(N-vinylisobutyramide) (PNVIBA)
[130], and poly(N-isopropylacrylamide) (PNI-
PAAm) [131–134] have been reported to exhibit
gelation upon temperature change. The latter is a
very attractive temperature-responsive polymer
since it exhibits a sharp solgel transition in water
at 34.3 °C. Thus, injecting a polymer solution
prepared at room temperature (rt) can lead
in situ to the formation of hydrogels at 37 °C.
Moreover, the formation of hydrogels from PNI-
PAAm is tunable by changing the preparation
conditions [135–137].

Chemical hydrogels
Fig. 7. The chemical structures of sodium cellulose sulfate
(CS) and poly(methylene-co-guanidine) hydrochloride Chemical hydrogels are mostly applied in cell
(PMCG). microencapsulation when high mechanical

184
Polymeric materials for xenotransplantation

resistance and long-term durability are required. precursor solution are crucial to maintain cell via-
For this purpose, several combinations of poly- bility and cell–cell adhesion during the encapsula-
mers and preparation conditions have been tested tion process. Mixing cells with highly viscous
in regard to their suitability to encapsulate cells solutions can lead to a significant decrease in cell
within chemical hydrogels. However, only a lim- viability [157].
ited number of combinations have been identified
for this purpose. Table 2 lists some examples that
Hybrid hydrogels
have been used to immobilize cells. Numerous cri-
teria must be considered when designing a chemi- Novel hydrogel types that combine physical and
cal hydrogel for cell microencapsulation. The chemical cross-linking are being emerging as ade-
process of hydrogel formation must not negatively quate candidates for cell microencapsulation. The
influence cell integrity and viability and should not physical interactions allow for fast gelation and
involve harsh conditions, toxic solvents, and reac- spherical shape formation, while biocompatible
tants [156]. Because cells are suspended in a liquid covalent cross-linking ensures the reinforcement of
precursor solution prior to the encapsulation pro- the hydrogel networks, along with tunable perme-
cess, the choice of precursors is limited to water- ability and gel stiffness.
soluble components. The aqueous solution must be For the preparation of APA microspheres with
buffered with appropriate osmolality to prevent covalent cross-linking between adjacent layers,
cell lysis. The rheological properties of the PLL was equipped with phenyl azide residues that
create covalent bonds with Ca-alg when irradiated
with UVA [158]. The photoinitiated cross-linking
Table 2. Examples of chemical hydrogels applied for cell immobiliza- was shown to be cell compatible and yielded stable
tion microspheres up to 3 yr in alkaline buffer (pH 12),
whereas standard APA disappeared within 1 min.
Material Preparation Cell Reference
Another strategy reported the replacement of the
Alginate Click reaction with tetrazine– EGFP- [138] final alginate layer of APA capsules by poly
norbornene modified hydrogel (no expressing (methyl vinyl ether-alt-maleic anhydride) (PMM)
ionic cross-linkings) 3T3
fibroblast
and poly(vinyl dimethyl azlactone-co-methacrylic
PEG Thiol-ene reaction of PEG diacrylate Fibroblasts; [139,140] acid) (PMV) to form stable covalent amide bonds
with thiolated gelatin keratinocytes with PLL, neutralizing the polycation layer [159].
Maleimide, acrylate, and vinyl sulfone- C2C12 [141] Further, the involvement of methacrylate poly-
modified PEG cross-linked with myoblast
peptides mers into physical microspheres, and subsequent
Photopolymerization of fibrinogen-g- BMSC [142] covalent cross-linking, was reported [160,161]. It
PEG-acryloyl and PEG diacrylate was demonstrated that either cross-linked shells or
Photopolymerization of PEG diacrylate Huh-7.5 [143]
cross-linked cores are obtainable by adjusting the
Chitosan Photopolymerization of chitosan grafted Chondrocytes [144]
with lactic acid and methacrylate molar mass of the cross-linker. Approaches based
Chemically cross-linked chitosan [145] on chemical cross-linking through complementary
hydrogel loaded with gelatin reactive groups attached to two oppositely charged
N-succinyl-chitosan gelation with [146]
aldehyde hyaluronic acid
polyelectrolytes were also investigated [162,163].
Dextran Photopolymerization of dextran with Osteoblast– [147] Similarly, microbeads were prepared by ionotropic
benzophenone endothelial gelation of a combination of Ca-alg and sericin as
cell inner core followed by coating with chitosan and
Gelation of methacrylate and lysine Smooth [148]
functionalized dextran muscle cells
further cross-linking with genipin [164,165]. This
Photopolymerization of dextran acrylate Embryonic [149] combination effectively reduced swelling and phys-
stem cells ical disintegration of the microspheres induced by
HA HA cross-linked via disulfide bond Fibroblasts, [150] non-gelling ions and calcium chelating agents.
formation reaction stem cells
Methacrylated HA cross-linked by UV MSC [151]
Higher resistance to mechanical shear force and
exposure improved durability against enzymatic degradation
Peroxidase catalyzed oxidation of Chondrocytes [152] were achieved. The entrapment of vinyl sulfone
tyramine-substituted HA terminated PEG in Ca-alg and subsequent
Conjugate addition of thiol-modified HA Adipocyte [153]
onto PEG diacrylate stem cells Michael-type cross-linking has been reported. The
PVA UV photopolymerization L929 fibroblast [154] mechanical properties of such hybrid microspheres
Click hydrogels formed by hydrazone [155] were adjustable and suitable for cell microencapsu-
bonds
lation [166–168].
PEG: Poly(ethylene glycol); HA: hyaluronic acid; PVA: poly(vinyl alcohol); MSC: mes- Rather than using hydrogels obtained from
enchymal stem cell. oppositely charged polyelectrolytes bearing

185
Mahou et al.

complementary reactive groups for chemical cross- chemokine CXCL12 [186], or the drug dexam-
linking, one-component hybrid hydrogels are also ethasone [187] reduced the inflammatory response
being developed and tested for cell microencapsu- and thus prolonged the graft survival. Also, anti-
lation. A one-component system denotes hybrid cytokine agents [188,189] were covalently
hydrogels prepared from polymers able to form attached on the hydrogel matrix sequestrating the
simultaneously both physical and chemical links. pro-inflammatory cytokines highly expressed in
Such polymers are very often a modified macro- wounded environment.
molecule or biopolymer. For instance, the prepa-
ration of hybrid alg-based microspheres was
Properties of hydrogel microspheres and their assessment
achieved by equipping Na-alg with azide-termi-
nated PEG pendent chains [169]. The azide end In addition to the size and size stability as well as
group forms chemical cross-links via the Staudin- chemical stability, the most important characteris-
ger reaction by incubation in a gelation bath con- tics of microspheres, which determine the applica-
taining phosphine-functionalized agents. Human bility for subsequent transplantation, are their
pancreatic islets were encapsulated using such sys- mechanical resistance, permeability, and biocom-
tems [170–172]. Similarly, Na-alg with thiol end patibility. The selection of suitable methods to
groups was prepared [173,174]. The modified Na- assess these parameters is not always a straightfor-
alg maintained the gelling capacity in the presence ward decision.
of calcium ions, while the thiol end groups
ensured the preparation of a chemically cross- Mechanical resistance
linked network via disulfide bond formation. The mechanical resistance of hydrogel micro-
Being biocompatible, spontaneous, and catalyst spheres is not an absolute parameter. It can be
free, the formation of disulfide bonds yielded evaluated by several methods, each of them pro-
hybrid microspheres in a one-step extrusion pro- viding different information. One method to assess
cess under physiological temperature, pH, and the mechanical resistance is the osmotic pressure
osmolality. Good survival rate and improved pro- test. The microspheres are simply exposed to water
liferation were obtained upon microencapsulation [190]. The sudden influx of water causes the micro-
of liver-derived cells within hybrid microspheres spheres to swell and break. A method to study the
[175]. resistance to mechanical stress is the microsphere
Nonetheless, the strategies described above shearing test. A suspension of microspheres is sub-
mainly focused on the robustness of the biomate- jected to a controlled fluid shear [191]. The number
rials, but other parameters must also be under- of broken microspheres as a function of the
lined. The lifetime and biocompatibility of applied mechanical stress gives an indication of
encapsulated cells should be enhanced to over- their mechanical resistance. Such essays have the
come the problem of graft failure [176]. Actually, advantage of being very simple. However, they
the survival of engrafted cells depends on the cell have limitations:
adhesion to the hydrogel matrix, on the vascular- (i) only qualitative information, restricted quan-
ization improving the delivery of nutrients, oxy- tification; (ii) the osmotic pressure test is reliably
gen, and metabolites but also on the immune applicable only to physical hydrogels.
response of the host body. For this purpose,
Contrary, the evaluation of the resistance and
hybrid hydrogels containing peptides or proteins
deformability of the microspheres under compres-
were developed. Mimicking the ECM and the
sion is considered a quantitative method [192]. A
covalent incorporation of adhesives ligands such
mobile probe compresses the microspheres at a
as galactose [177], RGD [178], and other types of
defined speed. Microspheres having the same size
laminin-derived recognition sequences and colla-
can be characterized and compared by measuring
gen type I sequence [179] have been reported.
the force corresponding to the rupture of the
Besides these molecules, proteins such as fibrin
microsphere and by comparing the mechanical
[180] and collagen [181] but also glycoproteins
resistance at a given degree of compression
[182] were also encapsulated to enhance cell via-
[193,194]. Dependent on the equipment sensitivity
bility. Moreover, the co-encapsulation of vascu-
and the hydrogel stiffness, two methods are com-
larization promoting factors such as VEGF [183]
monly used:
and FGF-1 [184] promoted the neovasculariza-
tion by improving the viability of engrafted 1. Each microsphere is individually com-
encapsulated islets. Last but not least, co-encap- pressed. The mechanical resistance of 20 to
sulation of anti-inflammatory agents such as 30 microspheres needs to be analyzed to
complement receptor 1 sCR1 [185], the obtain statistically meaningful data.

186
Polymeric materials for xenotransplantation

2. A layer of microspheres is compressed. This size is achieved by cryo-scanning electron micro-


method is suitable for capsules that burst at scopy (cryo-SEM), a technique that images the
low force. A narrow size distribution is the internal structure of hydrogel microspheres on a
prerequisite. nanometer scale [202]. However, the manipulation
of the hydrogel during freezing is critical.
Although several methods have been proposed
to study the mechanical properties of hydrogels, Biocompatibility
only few have focused on measuring the mechani- The assessment of the biocompatibility of hydrogel
cal properties of cell-embedding hydrogels. microspheres is a complex task due to the multi-
Ahearne et al. reported a non-destructive, online, tude of interactions between host body and foreign
and real-time method that allows measuring the material and because biocompatibility is not sim-
mechanical properties of hydrogels with incorpo- ply a property of a material but a property of a
rated cells [195]. biomaterial–host system [203]. Biocompatibility
issues of microspheres are often connected with
Permeability their ability to perform with an “appropriate host
The ability to deliver metabolic products and ther- response” in a “specific application” [204]. A bio-
apeutic proteins, but to block the diffusion of compatible hydrogel is considered to be a system
immune cells and antibodies, is the basis of the use that elicits no or not more than a minimal foreign
of cell microencapsulation in biomedical applica- body reaction. The success of cell microencapsula-
tions. A defined and controllable permeability is tion intended for transplantation strongly depends
therefore an essential prerequisite of hydrogel on minimizing this immune response. Upon
microspheres intended for transplantation. implantation of microencapsulated cells, the
The permeability of microspheres can be immune response is activated by the adsorption of
described by the mass transfer or diffusion coeffi- proteins onto the microspheres surface, which will
cient [196,197]. It defines the rate of diffusion of a subsequently stimulate the recruitment of immune
given solute into the hydrogel microsphere. cells [79], and the rejection of the transplant
Another quantification is the minimum size of a through one of the many well-documented path-
solute, which is completely excluded from the ways [205,206]. The immune response also elicits
hydrogel pores. This is usually referred to as the fibrosis around the hydrogels, which subsequently
exclusion limit or molecular weight cut-off starves the encapsulated cells by limiting the diffu-
(MWCO). The determination of solute permeation sion of nutrients, and limits the efflux of bioactive
can be either by diffusion into the microspheres molecules secreted from the cells [45]. Thus, most
(ingress) or from the microspheres (egress). approaches developed to minimize the immune
Several experimental techniques to assess the response to hydrogels are focused on preventing
permeability of microspheres have been described protein adsorption and cellular adhesion to the
relying on different solute types and monitoring surface of hydrogels through the encapsulation of
methods for quantifying the chosen solute. These cells into biologically inert hydrogels, or modifica-
include, for example, dextrans and pullulans as tion of the hydrogel surface with biocompatible
solutes and spectroscopy (fluorescence, UV/vis), materials. Very recently, bead size and shape have
measurement of radioactivity, size-exclusion chro- shown to influence the immune response of the for-
matography with concentration-sensitive detectors, eign body [207]. It was demonstrated that larger
and protein assay kits as monitoring methods beads (1.5 mm) reduced immune reactions and
[198,199]. fibrosis in comparison with smaller beads
An efficient technique that is being used for mea- (0.5 mm) when using different materials, including
suring the permeability is the inverse size-exclusion alginate in rats and non-human primate models
chromatography (ISEC) [200,201]. It has the [208].
advantage that the MWCO and its distribution In addition to the host reaction, the cell compat-
can be determined simultaneously. The drawback ibility of the hydrogel is a general biocompatibility
is that a minimum of 10 ml of microspheres is so prerequisite for a successful therapy using microen-
far needed for each measurement. The optimiza- capsulated cells. Different cell types could require
tion of this method to reduce the volume of hydro- different hydrogel properties in terms of stiffness
gel microspheres would be a great achievement. or hydrophobicity. In this context, also a minimum
The hydrogel mesh size, which is the space avail- of microsphere volume was reported as being
able between the macromolecular chains, has been advantageous to ensure sufficient oxygen supply
also used to describe the permeability of hydrogel and to avoid necrosis [91]. This is in contradiction
microspheres. Direct determination of the mesh with results reported in [208].

187
Mahou et al.

uniformity are obtained. However, the size distri-


Cell microencapsulation techniques
bution significantly increases for diameters below
Extrusion techniques 400 lm [213]. Reasonable production rates are
attainable by optimization [214].
Numerous techniques have been developed for
the production of hydrogel microspheres
Jet breakage by a vibrating nozzle
[209,210]. Most of them are based on forcing the
Small (<1000 lm) and uniform (<3% size devia-
passage of the cell-containing solution through a
tion) microspheres can be obtained by employing
needle or a nozzle, and its extrusion into a gela-
the jet-break vibrating technique [215]. The tech-
tion bath. The simplest method comprises drip-
nology principle is that a laminar liquid jet breaks
ping with only gravitational force as the driving
up into equally sized droplets by superimposed
force. This mode is restricted to low-viscous solu-
vibration of the nozzle. However, given that the
tions and yields hydrogel microspheres with diam-
droplet diameter obtained by this technique is 2 to
eters in the range of 1.5 to 3 mm. However, the
3 times larger than the nozzle diameter, the pro-
challenge in cell microencapsulation is to produce
duction of capsules with diameters <400 lm needs
preferably microspheres with diameters in the
extrusion nozzles of 150 lm. Therefore, this tech-
range of 100 to 400 lm. Indeed, hydrogel micro-
nique is not optimal for the microencapsulation of
spheres with such dimensions have the advantage
large cells or cell clusters. The high mechanical
of a higher surface-to-volume ratio, exhibiting
stress to which the cells are exposed might compro-
good transport of nutrients [211]. Subsequently,
mise their viability.
different processes of droplet formation, which is
the first step of cell microencapsulation, are dis-
Droplet formation by an electrostatic potential
cussed and schematically shown in Fig. 8. A com-
To obtain small uniform droplets, a high electro-
prehensive database of different encapsulation
static potential between the needle and the receiv-
technologies is available on http://www.ge-
ing bath can be used [216,217]. The diameter of the
nialab.de/WG3/.
droplets can be tailored by controlling the electro-
static pulses [218–220].
Droplet formation by coaxial air flow
The principle of the coaxial air-flow droplet gener-
Jet breakage by rotating elements
ator [212] is the use of air flow to separate droplets
This technology is especially suitable for the pro-
from a needle or nozzle tip before they fall due to
duction of large batches of microspheres. Droplets
gravity into a gelling bath. Surface tension gives
are formed by rotating nozzles, a rotating disk, or
the droplets their near-spherical shape. Micro-
a cutting wire [221].
spheres in the range of 200 to 1000 lm with good

Fig. 8. Formation of droplets by: coaxial air flow (1), electrostatic potential (2), rotating disk (jet cutter) (3), and vibrating nozzle
(4). Cells are finally entrapped in hydrogel microspheres after falling down in the gelation bath.

188
Polymeric materials for xenotransplantation

Emulsion techniques drawback to be overcome. There is therefore a


need for scale-up studies to adapt this technology
Emulsion techniques allow the formation of micro-
for cell microencapsulation [250].
spheres with relatively small diameters. The aque-
ous phase (cell-containing solution) is mixed and
dispersed in an organic phase. When the dispersion Conformal polymer coating
reaches equilibrium, gel formation is initiated by
If the average diameter of a cell aggregate is con-
cooling or by the addition of a gelling agent.
sidered as 150 lm, the total volume of a 450-lm-
Although the emulsion process is advantageous for
diameter microcapsule is 27 times larger. This
large-scale production [218–222], its use for cell
“dead” volume is an often-overlooked issue that
microencapsulation is limited due to the broad size
can be related to a delayed metabolic response and
distribution generally obtained. Further, the use of
a slowed diffusion of oxygen and nutrients causing
an organic phase and the significant shear stress
cell necrosis, especially at the center of the micro-
during emulsion may compromise cell survival
sphere. This issue may also represent a limiting fac-
[223].
tor if the transplantation of microencapsulated
cells is aimed, as only limited space is available at
Microfluidic-based cell encapsulation the transplant site. To reduce the microsphere size,
depositing a polymer coating directly on the sur-
The recent advances in microfluidics and
face of a cell was proposed [251]. As shown in
microlithography provided effective tools to
Fig. 9, the total volume equals the size of the cell
control the formation of hydrogel microspheres
aggregate plus the thickness of the coating. The
with desirable shape, size, and size distribution
advantages from a mass transfer perspective are
[224–227]. Microfluidics allow for the preparation
achieved because of the high ratio of surface to
of hydrogels with defined morphology such as
volume. The direct deposit of polymer coatings on
Janus particles, multicompartment microspheres,
the surface of cells has been achieved following dif-
and non-spherical microgels [228–235]. Droplets
ferent processes such as emulsification [208], dis-
are produced when mixing an aqueous precursor
continuous gradient density centrifugation [252],
solution with an oil phase such as mineral oil, sili-
selective withdrawal [253], or interfacial polymer-
cone oil, corn oil, hexadecane oil, or fluorinated oil
ization [254]. Because of the relatively large cell or
[236]. The droplets are further internally cross-
cell cluster diameter, conformal polymer coating
linked by ionic gelation or by photopolymerization
was preferably applied to islets of Langerhans,
to obtain cells entrapped in hydrogel microspheres
using alginate [255], agarose [256], and PEG [257–
[237]. Obviously, the immiscibility of the two
259], and different chemical strategies were
phases is a prerequisite to ensure the formation of
explored to modify covalently the surface of the
spherical droplets [238,239]. However, the viscosity
cells [260]. Polymers such as PLL have been
of the oil has been identified as an important factor
applied for conformal coating after conjugation
controlling the formation of the droplets. The
with biocompatible molecules to attenuate their
results have shown that highly viscous liquids are
toxicity [261–263].
emulsified into larger droplets with lower polydis-
persity [240]. As a proof of concept, several cell
types have been encapsulated within hydrogel
microspheres using microfluidic droplet formation Cell microencapsulation for xenotransplantation
devices [241–247]. Moreover, this technology exhi-
Sites of transplantation
bits ideal solution for single cell encapsulation
[248,249]. The production of a sufficient numbers Different surgical sites have been so far used for
of droplets in a short-time period remains a major the transplantation of encapsulated cells [264,265].

Fig. 9. Comparison of conformal coating


and cell microencapsulation.

189
Mahou et al.

Choosing the surgical site is critical to provide cap- paraphysiological conditions for the cells to work
sules proper intimacy with the outside environment properly are difficult to obtain in these sites; thus,
and graft vascularization. In addition to biological the skin should be used for host biocompatibility
functionality, the ideal site for transplantation and immunoprotection tests. Vascularization could
should be easily accessible for both placement and be improved by using the intramuscular site to
microcapsule retrieval. maintain good surgical accessibility and graft mon-
To accomplish these needs, several studies have itoring.
investigated the peritoneal site. Indeed, the peri- The kidney capsule has been widely used for
toneum is currently considered by several authors murine islet transplantation [280,281]. It provides
a feasible and effective site to implant both encap- good surgical accessibility and retrievability. How-
sulated islets and hepatocytes [266–271]. However, ever, the volume of encapsulated cells required to
it was also shown that the peritoneal cavity has less achieve insulin independence makes this site
chance to provide sufficient oxygen to microencap- unconvincing for clinical translation.
sulated cells compared to the kidney capsule and
muscle [266]. Nonetheless, the peritoneum has
Sources of cells for xenotransplantation
shown good results, when used as implantation
site for encapsulated cells, despite impaired insulin The use of porcine cells for xenotransplantation
secretion to glucose stimuli and progressive loss of has been intensively studied, in particular for islet
function in islet transplantation has been transplantation [282,283]. Shin et al. demonstrated
reported. Furthermore, it has been shown that the that adult porcine islets from pathogen-free minia-
capsules are likely to float into the peritoneal cav- ture pigs transplanted into immunosuppressed dia-
ity with lack of engraftment, clot formation, and betic monkeys induced fast glucose level
subsequent poor nutrition. On the other hand, the normalization. In addition, normoglycemia was
peritoneum allows the transplantation of large maintained for more than 6 months in four cases,
quantity of tissue and further infusions in case of with no serious advert effects resulting from the
loss of graft function with respect to other sites transplantation [284]. Several reports highlighted
[272]. In addition, the biocompatibility can be sig- the advantages to use neonatal pig islet-like cell
nificantly improved by using barium alginate clusters (NICCs) instead of adult pig cells. In par-
microcapsules [273]. To overcome low oxygen ten- ticular, NICCs are easily digested and purified and
sion, some authors proposed the construction of show low level of T-cell response and high resis-
an omental pouch providing a more efficient tance to ischemia and inflammation [285]. Optimal
blood supply by capillary neoangiogenesis functionality was observed after 12 days of culture
[274,275]. Indeed, it was shown that encapsulated [286] and tolerance to xenotransplantation of
islets, transplanted into the omental pouch, are NICCs can be improved by treatment with
able to restore euglycemia in NOD mice without expanded regulatory T cells or molecules targeting
immunocyte infiltration around the capsules innate immunity [287]. The ideal age at which these
[275,276]. cell clusters should be isolated from porcine donors
The portal vein is traditionally used for human was established to be during the first month of life,
islet transplantation and it is now considered the with slight advantages for pigs within the first week
standard site to make islets exert their function. of life [288]. Pig-to-monkey islet xenotransplanta-
Moreover, closer contact to the vessels should pro- tion was also attempted from adult genetically
vide efficient nutrition and blood supply. On the engineered pigs to overcome the metabolic and
other hand, the risk of portal vein occlusion and immunological barriers between species. However,
the difficult retrieval of the capsules (harboring the multitransgenic islet grafts did not show consis-
both islets and hepatocytes) are important issues to tent long-term functionality for several months
be solved to optimize this site of implantation [289]. The use of pig cells was also investigated in
[277]. Nevertheless, portal puncture during acute the context of xenotransplantation for the treat-
liver failure for HT should be balanced as a highly ment of acute liver failure [290]. Alginate-encapsu-
hazardous maneuver due to both coagulopathy lated reaggregated neonatal pig liver cells
and the enhanced risk of occlusion in pathologic demonstrated promising efficacy for the treatment
liver parenchyma. of mice with acute liver failure. In addition, coat-
Skin and subcutaneous tissue represent the most ing of the capsules with chitosan resulted in a
accessible site for microsphere transplantation reduction of the attachment of macrophages [291].
regarding surgical implantation and retrieval [278]. Recently, No et al. reported on the development of
Lack of blood supply could be improved by pre- a method for the triculture of three types of cells
vascularization of the site [279]. However, from rats to produce uniformly sized and shaped

190
Polymeric materials for xenotransplantation

microliver tissues. Encapsulated in collagen–algi- alginate-encapsulated islets, transplanted into the


nate composites, these cells induced a long-term portal vein, in 4 type I diabetic patients obtaining
survival rate of 80% in mice with 90% hepatec- transient insulin independence only in one of them,
tomy [292]. Other sources of transgenic cells have but showing safety, feasibility, and biocompatibil-
been investigated. In particular, wide-type and ity of alginate capsules in humans [299]. More
transgenic tilapia donors were considered for the recently, Matsumoto et al. published the results
harvesting of islet cells [293,294]. Transplantation from a phase I/IIa xenotransplantation study of
of alginate-encapsulated tilapia islets into diabetic APA-encapsulated neonatal porcine islets in
mice resulted in long-term (up to 210 days) meta- 14 patients with unstable type 1 diabetes [300].
bolic control but required immunosuppressive Without immunosuppressive treatment, the trans-
treatment [295]. The significant difference between plantation was safe and resulted in the reduction
tilapia insulin and human insulin is nevertheless a of unaware hypoglycemia events, but did not result
strong limitation to clinical application of tilapia in insulin independence.
islets in cell therapy protocols.
Concluding remarks, future directions, and open questions
Translation of cell microencapsulation into a xenotransplantation
The Holy Grail for transplant investigators is to
cell therapy
avoid the use of immunosuppression. Encapsula-
Since the first reports, in the 1980s, of hydrogel tion of cells and cell clusters could finally lead to this
encapsulation of islet-b-cells for the treatment of goal by setting up the conditions for long-term
type I diabetes mellitus, many studies on animal function cell therapy without the adverse effect of
models addressed the different parameters which immunosuppressive agents. Nonetheless, adequate
can improve biocompatibility, stability, and func- protection of cells from the immune system could
tionality of encapsulated cells. Table 3 lists some expand the field of xenogeneic transplantation.
of the animal experiments and preclinical studies However, despite the recent breakthrough findings,
that evaluated the xenotransplantation of encapsu- crucial issues still remain unsolved. Purity and sta-
lated islets and hepatocytes. bility of encapsulation materials should be
The encapsulation of different islet types in Ca- improved to gain effective biocompatibility [301].
and Ba-alg materials allowed restoring normo- Highly purified alginate-based materials have
glycemia in xenogenic settings, without immuno- brought promising results, but capsular overgrowth
suppression. Similarly, encapsulated xenogenic is still an issue leading to inadequate blood and
hepatocytes were able to revert acute liver failure nutrients supply. Conformal coating of cells and
in rodent models, in the absence of immunosup- cell clusters seems to be one possibility to overcome
pressive treatment. Despite these encouraging problems of cell malnutrition by reducing the dis-
results, the demonstration of effective protocols in tance between cells and capillaries and has shown
patient protocols is still very scarce. In 2011, Cala- better quality in terms of immunogenicity. Despite
fiore et al. reported a phase I clinical trial testing the progress achieved in biotechnology, the balance

Table 3. Xenotransplantation of encapsulated cells in animal models

Transplanted model Cell type Transplantation site Materials for encapsulation Reference

Mice Neonatal porcine islets Peritoneal cavity Ca-alg and Ba-alg [269]
[270]
Rat islets Subcutaneous tissue Agarose/poly (styrene sulfonic acid) [279]
Intraperitoneal space Alg-PLL-PEG [80]
Peritoneal cavity Agarose [185]
Human islets Omental pouch Agarose [275]
Peritoneal cavity Ba-alg [56]
Neonatal pig hepatocytes Abdominal cavity Ba-alg [291]
Rat hepatocytes Peritoneal cavity Alg-PLL-alg [266,267]
Human hepatocytes
Fish islets Abdominal cavity Ba-alg [295]
Rat Pig islets Subcutaneous tissue Ca-alg [278]
Abdominal cavity Ba-alg [296]
Guinea pig hepatocytes Peritoneal cavity acrylonitrile–sodium methallyl sulfonate copolymer [297]
Non-human primates Pig islets Intraportal injection No material [284]
[289]
Intraperitoneal space Ca-alg-PLL [298]

191
Mahou et al.

between immunoprotection and nutrition delivery successfully tested. Cryopreservation and rewarm-
requires further attention. Interestingly, necrosis of ing did not negatively affect the function of
the central region of the encapsulated cells was microencapsulated hepatocytes [306,307]. In con-
identified in several studies as the common charac- clusion, while the potential of cell microencapsula-
teristic for graft failure and thus it was hypothesized tion for the development of cell therapies has been
that poor nutrition and lack of blood supply are the demonstrated in animal models, long-term studies
main causes of loss of function other than failure in are still scarcely reported. Despite promising
immunoprotection [275]. results, translating in particular xenotransplanta-
There is no consensus about the ideal site of tion of microencapsulated cells into a therapy will
transplantation. Many reports gave evidence for require further multidisciplinary efforts. The final
the advantages of intraperitoneal placement of success will not least depend on the development
both encapsulated islets and hepatocytes. Peri- of suitable encapsulation materials and technolo-
toneum is well surgically accessible, but the short- gies.
age of nutrition impairs the engraftment of
encapsulated cells [302,303]. Moreover, the lack of
References
contact with vessels causes a reduced insulin
response to glucose stimuli for the islets. On the 1. Application of current statutory authorities to human
other hand, the omental pouch has shown suffi- somatic cell therapy products and gene therapy prod-
ucts, notice, Oct 14, 1993 (federal register). (Notice
cient angiogenesis potential and could be an ideal from the center for biologics evaluation and research,
site for islet transplantation by virtue of direct por- center for drug evaluation and research, center for
tal delivery. Intraportal infusion has the advantage devices and radiological health, U.S. Food and drug
of providing more accessible blood supply to the administration).
encapsulated cells, but it is affected by the risk of 2. CALAFIORE R, BASTA G, LUCA G et al. Grafts of microen-
capsulated pancreatic islet cells for the therapy of dia-
occlusion. Hence, optimization of the microcapsule betes mellitus in non-immunosuppressed animals.
size is advised in order to avoid unnecessary hydro- Biotechnol Appl Biochem 2004; 39: 159–164.
gel dead volume. Islet-encapsulated therapy has 3. TOMARO-DUCHESNEAU C, SAHA S, MALHOTRA M et al.
been demonstrated to be effective in the treatment Microencapsulation for the therapeutic delivery of
of diabetes in rodent models and large animals. drugs, live mammalian and bacterial cells, and other bio-
pharmaceutics: current status and future directions. J
Feasibility and biocompatibility studies in human Pharm 2013:e103527. doi: 10.1155/2013/103527.
beings have recently highlighted the potential of 4. BISCEGLIE VV. Uber die antineoplastische immunit€ at.
this fascinating technology in translational medi- Krebsforsch 1933; 40: 141–158.
cine, but effective results in patients are still lacking 5. CHANG TMS. Semipermeable microcapsules. Science
[304]. However, enough evidence has been gath- 1964; 146: 524–525.
6. Rapport annuel 2012. Fondation Nationale Suisse pour
ered to justify clinical trials, in particular for por- le don et la transplantation d’organes. Available
cine islet xenotransplantation [305]. The other at: http://www.swisstransplant.org [accessed on 15
therapeutic area, which might largely benefit from February 2016].
xenotransplantation, is the reversal of acute liver 7. AEBISCHER P. Transplantation in humans of encapsulated
failure. In murine models, the engraftment of xenogeneic cells without immunosuppression: a prelimi-
nary report. Transplantation 1994; 58: 1275–1277.
encapsulated xenogenic hepatocytes brought new 8. BONAVITA AG, QUARESMA K, COTTA-DE-ALMEIDA V et al.
advances in cell therapy [266,267]. Xenogeneic Hepatocyte xenotransplantation for treating liver dis-
sources of hepatocytes should be considered in ease. Xenotransplantation 2010; 17: 181–187.
translational protocols given the shortage of liver 9. TENG Y, WANG Y, LI S et al. Treatment of acute hepatic
donors. Further studies in large animal models are failure in mice by transplantation of mixed microencap-
sulation of rat hepatocytes and transgenic human fetal
required to prove the effectiveness of this strategy. liver stromal cells. Tissue Eng Part C -meth 2010; 16:
However, encapsulated hepatocytes have shown, 1125–1134.
by metabolites detossification rate analysis, loss of 10. QIU L, WANG J, WEN X et al. Transplantation of co-
function within 30 days after transplantation. This microencapsulated hepatocytes and HUVECs for treat-
short-term cell functionality could be acceptable ment of fulminant hepatic failure. Int J Artif Organs
2012; 35: 458–465.
for therapies intending to provide a bridge for the 11. COUSSA R, MARTONI C, BHATHENA J et al. Oral microen-
native liver to regenerate upon acute failure, but is capsulated live Saccharomyces cerevisiae cells for use in
objectionable for a long-term function protocol in renal failure uremia: preparation and in vivo analysis.
order to treat chronic liver diseases. In the near J Biomed Biotechnol 2010: Article ID 620827. doi:10.
future, cryopreserved microencapsulated cells 1155/2010/620827.
12. JAIN P, SHAH S, COUSSA R, PRAKASH S. Potentials and
could be stored in a cell therapy bank with the limitations of microorganisms as renal failure biothera-
aim to secure large quantities for patients. Also peutics. Biologics 2009; 3: 233–243.
for this purpose, alginate-based hydrogels were

192
Polymeric materials for xenotransplantation

13. BRODIE JC, HUMES HD. Stem cell approaches for the (methacrylic acid-co-dimethyl acrylamide). J Chem Phys
treatment of renal failure. Pharmacol Rev 2005; 57: 299– 2007; 126: 224501.
313. 35. SCHNEIDER HJ, STRONGIN RM. Supramolecular interac-
14. SALMONS B, GUNZBURG WH. Therapeutic application of tions in chemomechanical polymers. Acc Chem Res
cell microencapsulation in cancer. Adv Exp Med Biol 2009; 42: 1489–1500.
2010; 670: 92–103. 36. DAS P, HEUSER T, WOLF A et al. Tough and catalytically
15. PARETA RA. Bioartificial pancreas: drug delivery active hybrid biofibers wet-spun from nanochitin hydro-
through islet microencapsulation to treat diabetes. Rec gels. Biomacromolecules 2012; 13: 4205–4212.
Patents on Biomed Eng 2012; 5: p41. 37. GOVAN JRW, FYFE JAM, JARMAN TR. Isolation of algi-
16. BASTA G, CALAFIORE R. Immunoisolation of pancreatic nate-producing mutants of pseudomonas fluorescens,
islet grafts with no recipient’s immunosuppression: pseudomonas putida and pseudomonas mendocina.
actual and future perspectives. Curr Diab Rep 2011; 11: J Gen Microbiol 1981; 125: 217–220.
384–391. 38. FISCHER FG, DORFEL H. Die polyuronsauren der brau-
17. MONTANUCCI P, PENNONI I, PESCARA T, BASTA G, nalgen (kohlenhydrate der algen-I). Hoppe-Seylers zeits-
CALAFIORE R. Treatment of diabetes mellitus with chrift fur physiologische chemie 1955; 302: 186–203.
microencapsulated fetal human liver (FH-B-TPN) engi- 39. MØRCH YA, DONATI I, STRAND BL, SKJAK  -BRAEK G.
neered cells. Biomaterials 2013; 34: 4002–4012. Effect of Ca2+, Ba2+, and Sr2+ on alginate microbeads.
18. DUFRANE D, GIANELLO P. Macro- or microencapsulation Biomacromolecules 2006; 7: 1471–1480.
of pig islets to cure type-1 diabetes. World J Gastroen- 40. MOYA ML, MORLEY M, KHANNA O, OPARA EC, BREY
terol 2012; 18: 6885–6893. EM. Stability of alginate microbead properties in vitro.
19. WICHTERLE O, LIM D. Hydrophilic gels for biological J Mater Sci Mater Med 2012; 23: 903–912.
use. Nature 1960; 185: 117–118. 41. GRANT GT, MORRIS ER, REES DA, SMITH PJC, THOM D.
20. HOFFMAN AS. Hydrogels for biomedical applications. Biological interactions between polysaccharides and
Adv Drug Deliver Rev 2002; 54: 3–12. divalent cations: the egg box model. FEBS Lett 1973; 32:
21. CENSI R, Di MARTINO P, VERMONDEN T, HENNINK WE. 195–198.
Hydrogels for protein delivery in tissue engineering. 42. DRAGET KI, SKJAK-BRÆEK G, SMIDSRØD O. Alginate
J Control Release 2012; 161: 680–692. based new materials. Int J Biol Macromol 1997; 21:
22. LIU LS, KOST J, YAN F, SPIRO RC. Hydrogels from 47–55.
biopolymer hybrid for biomedical, food, and functional 43. WANDREY C, ESPINOSA D, REHOR A, HUNKELER D. Influ-
food applications. Polymers 2012; 4: 997–1011. ence of alginate characteristics on the properties of mul-
23. MAWAD D, BOUGHTON EA, BOUGHTON P, LAUTO A. ti-component microcapsules. J. Microencapsulation
Advances in hydrogels applied to degenerative diseases. 2003; 20: 597–611.
Curr Pharm Des 2012; 18: 2558–2575. 44. ESPEVIK T, OTTERLEI M, SKJAK-BRAEK G et al. The
24. SELIKTAR D. Designing cell-compatible hydrogels for involvement of CD14 in stimulation of cytokine produc-
biomedical applications. Science 2012; 336: 1124–1128. tion by uronic acid polymers. Eur J Immunol 1993; 23:
25. GASPERINI L, MANO JF, REIS RL. Natural polymers for 255–261.
the microencapsulation of cells. J R Soc Interface 2014; 45. KULSENG B, SKJAK  -BRAEK G, RYAN L et al. Transplanta-
11: 19 pages. tion of alginate microcapsules: generation of antibodies
26. OLABISI RM. Cell microencapsulation with synthetic against alginates and encapsulated porcine islet-like cell
polymers. J Biomed Mater Res, Part A 2015; 103A: 846– clusters. Transplantation 1999; 67: 978–984.
859. 46. CLAYTON HA, LONDON NJM, COLLOBY PS, BELL PRF,
27. SELIMOVIC S, OH J, BAE H, DOKMECI M, KHADEMHOSSEINI JAMES RFL. The effect of capsule composition on the
A. Microscale strategies for generating cell-encapsulat- biocompatibility of alginate-poly-l-lysine capsules. J.
ing hydrogels. Polymers 2012; 4: 1554–1579. Microencapsulation 1991; 8: 221–233.
28. BILLIET T, VANDENHAUTE M, SCHELFHOUT J, Van VLIER- 47. MONTANUCCI P, TERENZI S, SANTI C et al. Insights in
BERGHE S, DUBRUEL P. A review of trends and limitations behavior of variably formulated alginate-based micro-
in hydrogel-rapid prototyping for tissue engineering. capsules for cell transplantation. Biomed Res Int 2015;
Biomaterials 2012; 33: 6020–6041. 2015: 11. Article ID 965804.
29. GAUVIN R, PARENTEAU-BAREIL R, DOKMECI MR, MERRY- 48. VAUCHEL P, ARHALIASS A, LEGRAND J, KAAS R, BARON
MAN WD, KHADEMHOSSEINI A. Hydrogels and microtech- R. Decrease in dynamic viscosity and average molecular
nologies for engineering the cellular microenvironment. weight of alginate from Laminaria digitata during alka-
WIREs Nanomed Nanobiotechnol 2012; 4: 235–246. line extraction. J Phycol 2008; 44: 515–517.
30. BORG DJ, BONIFACIO E. The use of biomaterials in islet 49. MANCINI M, MORESI M, SAPPINO F. Rheological behav-
transplantation. Curr Diab Rep 2011; 11: 434–444. ior of aqueous dispersions of algal sodium alginates.
31. HABIBI Y, LUCIA LA, ROJAS OJ. Cellulose nanocrystals: J Food Eng 1996; 28: 283–295.
chemistry, self-assembly, and applications. Chem Rev 50. DU XW, WU HL, ZHU YF et al. Experimental study of
2010; 110: 3479–3500. therapy of bone marrow mesenchymal stem cells or mus-
32. KEERL M, SMIRNOVAS V, WINTER R, RICHTERING W. cle-like cells/calcium alginate composite gel for the treat-
Interplay between hydrogen bonding and macromolecu- ment of stress urinary incontinence. Neurourol Urodyn
lar architecture leading to unusual phase behavior in 2013; 32: 281–286.
thermosensitive microgels. Angew Chem Int Ed 2008; 51. LEE JH, LEE DH, PARK JK et al. Potentiality of immobi-
47: 338–341. lized pig hepatocyte spheroids in bioartificial liver sys-
33. WANG H-J, HONG XZ, BA X-W. Sol-gel transition in tem. Transplant Proc 2012; 44: 1012–1014.
nonlinear hydrogen bonding solutions. Macromolecules 52. DUFRANE D, Van STEENBERGHE M, GOEBBELS RM et al.
2007; 40: 5593–5598. The influence of implantation site on the biocompatibil-
34. YILMAZ Y. Transition between collapsed state phases ity and survival of alginate encapsulated pig islets in rats.
and the critical swelling of a hydrogen bonding gel: Poly Biomaterials 2006; 27: 3201–3208.

193
Mahou et al.

53. MOYER HR, KINNEY RC, SINGH KA et al. Alginate 70. BHUJBAL SV, DE HAAN B, NICLOU SP, DE VOS P. A novel
microencapsulation technology for the delivery of adi- multilayer immunoisolating encapsulation system over-
pose-derived stem cells. Ann. Plastic Surg 2010; 65: 497– coming protusion of cells. Sci Rep 2014; 4: 6856: 8 pages.
503. 71. ROKSTAD AM, BREKKE OL, STEINKJER B et al. Alginate
54. MAN Y, WANG P, GUO Y et al. Angiogenic and microbeads are complement compatible, in contrast to
osteogenic potential of platelet-rich plasma and adipose- polycation containing microcapsules, as revealed in a
derived stem cell laden alginate microspheres. Biomateri- human whole blood model. Acta Biomater 2011; 7:
als 2012; 33: 8802–8811. 2566–2578.
55. ENDRES M, WENDA N, WOEHLECKE H et al. Microencap- 72. STRAND BL, RYAN TL, KULSENG B et al. Poly-L-lysine
sulation and chondrogenic differentiation of human mes- induces fibrosis on alginate microcapsules via the induc-
enchymal progenitor cells from subchondral bone tion of cytokines. Cell Transplant 2001; 10: 263–275.
marrow in Ca-alginate for cell injection. Acta Biomater 73. JUSTE S, LESSARD M, HENLEY N et al. Effect of poly-L-
2010; 6: 436–444. lysine coating on macrophage activation by alginate-
56. SCHNEIDER S, FEILEN PJ, BRUNNENMEIER F et al. Long- based microcapsules: assessment using a new in vitro
term graft function of adult rat and human islets encap- method. J Biomed Mater Res, Part A 2005; 72: 389–398.
sulated in novel alginate-based microcapsules after 74. NILSSON B, KORSGREN O, LAMBRIS JD et al. Can cells
transplantation in immunocompetent diabetic mice. Dia- and biomaterials in therapeutic medicine be shielded
betes 2005; 54: 687–693. from innate immune recognition? Trends Immunol 2010;
57. MALPIQUE R, OSORIO  LM, FERREIRA DS et al. Alginate 31: 32–38.
encapsulation as a novel strategy for the cryopreserva- 75. EKDAHL KN, LAMBRIS JD, ELWING H et al. Innate immu-
tion of neurospheres. Tissue Eng Part C Methods 2010; nity activation on biomaterial surfaces: a mechanistic
16: 965–977. model and coping strategies. Adv Drug Deliv Rev 2010;
58. PENOLAZZI L, TAVANTI E, VECCHIATINI R et al. Encapsu- 63: 1042–1050.
lation of Mesenchymal Stem Cells from Wharton’s Jelly 76. BABENSEE JE, CORNELIUS RM, BRASH JL, SEFTON MV.
in Alginate Microbeads. Tissue Eng Part C 2010; 16: Immunoblot analysis of proteins associated with
141–155. HEMA-MMA microcapsules: human serum proteins
59. QI M, MØRCH Y, LACIK I et al. Survival of human islets in vitro and rat proteins following implantation. Bioma-
in microbeads containing high guluronic acid alginate terials 1998; 19: 839–849.
crosslinked with Ca2+ and Ba2+. Xenotransplantation 77. TANG L, UGAROVA TP, PLOW EF, EATON JW. Molecular
2012; 19: 355–364. determinants of acute inflammatory responses to bioma-
60. QI M, STRAND BL, MØRCH Y et al. Encapsulation of terials. J Clin Invest 1996; 97: 1329–1334.
human islets in novel inhomogeneous alginate-Ca2+/ 78. ROKSTAD AM, BREKKE OL, STEINKJER B et al. The
Ba2+ microbeads: In vitro and in vivo function. Artif induction of cytokines by polycation containing micro-
Cells Blood Substit Immobil Biotechnol 2008; 36: 403– spheres by a complement dependent mechanism. Bioma-
420. terials 2013; 34: 621–630.
61. WIKSTROM € J, ELOMAA M, SYVAJ € ARVI
€ H et al. Alginate- 79. SAWHNEY AS, HUBBELL JA. Poly(ethylene oxide)-graft-
based microencapsulation of retinal pigment epithelial poly(L-lysine) copolymers to enhance the biocompatibil-
cell line for cell therapy. Biomaterials 2008; 29: 869–876. ity of poly(L-lysine)-alginate microcapsule membranes.
62. SMIDSRØD O, SKJAK-BRÆK G. Alginate as immobilization Biomaterials 1992; 13: 863–870.
matrix for cells. Trends Biotechnol 1990; 8: 71–78. 80. PARK J-B, JEONG J-H, LEE M, LEE DY, BYUN Y. Xeno-
63. LIM F, SUN AM. Microencapsulated islets as bioartificial transplantation of exendin-4 gene transduced pancreatic
endocrine pancreas. Science 1980; 210: 908–910. islets using multi-component (alginate, poly-L-lysine,
64. ACARREGUI A, PEDRAZ JL, BLANCO FJ, HERNANDEZ  RM, and polyethylene glycol) microcapsules for the treatment
ORIVE G. Hydrogel-based scaffolds for enclosing encap- of type 1 diabetes mellitus. J Biomater Sci Polym Ed
sulated therapeutic cells. Biomacromolecules 2013; 14: 2013; 24: 2045–2057.
322–330. 81. SPSOJEVIC M, PAREDES-JUAREZ GA, VORENKAMP J et al.
65. HUANG X, ZHANG X, WANG X, WANG C, TANG B. Reduction of the inflammatory responses against algi-
Microenvironment of alginate-based microcapsules for nate-poly-L-lysine microcapsules by anti-biofouling sur-
cell culture and tissue engineering. J Biosci Bioeng 2012; faces of PEG-b-PLL diblock copolymers. PLoS ONE
114: 1–8. 2014; 9: 11 pages.
66. DUBROT J, PORTERO A, ORIVE G et al. Delivery of 82. ROBITAILLE R, PARISEAU JF, LEBLOND FA et al. Studies
immunostimulatory monoclonal antibodies by encapsu- on small (<350 lm) alginate-poly-L-lysine microcapsules
lated hybridoma cells. Cancer Immunol Immunother III. Biocompatibility of smaller versus standard micro-
2010; 59: 1621–1631. capsules. J Biomed Mater Res, Part A 1999; 44: 116–
67. SANTOS E, LARZABAL L, CALVO A et al. Inactivation of 120.
encapsulated cells and their therapeutic effects by means 83. KING A, STRAND B, ROKSTAD AM et al. Improvement of
of TGL triple-fusion reporter/biosafety gene. Biomateri- the biocompatibility of alginate/poly-L-lysine/alginate
als 2013; 34: 1442–1451. microcapsules by the use of epimerized alginate as a
68. SANTOS E, ORIVE G, CALVO A et al. Optimization of coating. J Biomed Mater Res, Part A 2003; 64: 533–539.
100 lm alginate-poly-l-lysine-alginate capsules for 84. HOBBS HA, KENDALL WF, DARRABIE M et al. Substitu-
intravitreous administration. J Control Release 2012; tion of polyornithine for polylysine in alginate microcap-
158: 443–450. sules. Diabetes 2000; 49(suppl 1): A111.
69. SHANBHAG MS, LATHIA JD, JUSTIN D et al. Neural pro- 85. BRUNETTI P, BASTA G, FALOERNI A et al. Immunoprotec-
genitor cells grown on hydrogel surfaces respond to the tion of pancreatic islet grafts within artificial microcap-
product of the transgene of encapsulated genetically sules. Int J Artif Organs 1991; 14: 789–791.
engineered fibroblasts. Biomacromolecules 2010; 11: 86. CALAFIORE R, BASTA G, BOSELLI C et al. Effects of
2936–2943. alginate/polyaminoacidic coherent microcapsule

194
Polymeric materials for xenotransplantation

transplantation in adult pigs. Transplant Proc 1997; 29: interaction between alginate and chitosan. Biomaterials
2126–2127. 1998; 19: 1815–1825.
87. TAM SK, BILODEAU S, DUSSEAULT J et al. Biocompati- 104. 
GASERØD O, SANNES A, SKJAK  -BRAEK G. Microcapsules
bility and physicochemical characteristics of alginate- of alginate–chitosan-II. A study of capsule stability and
polycation microcapsules. Acta Biomater 2011; 7: permeability. Biomaterials 1999; 20: 773–783.
1683–1692. 105. ZHENG J, XIE H, YU W et al. Enhancement of surface
88. DARRABIE MD, KENDALL WF, OPARA EC. Characteris- graft density of MPEG on alginate/chitosan hydrogel
tics of poly-L-ornithine-coated alginate microcapsules. microcapsules for protein repellency. Langmuir 2012;
Biomaterials 2005; 26: 6846–6852. 28: 13261–13273.
89. NABAVIMANESH MM, HASHEMI-NAJAFABABI S, VASHE- 106. ROSHANBINFAR K, SALAHSHOUR KORDESTANI S. Encapsu-
GHANI-FARAHANI E. Islets immunoisolation using encap- lating beta islet cells in alginate, alginate-chitosan and
sulation and PEGylation, simultaneously, as a novel alginate-chitosan-PEG microcapsules and investigation
design. J Biosci Bioeng 2015; 119: 486–491. of insulin secretion. J Biomat and Tissue Eng 2013; 3:
90. CHEN AZ, BAI Y, WANG SB, LIU YG, CHEN ZX. Molecu- 185–189.
lar biocompatibility evaluation of poly-L-ornithine- 107. ZHI ZL, KERBY A, KING AJ, JONES PM, PICKUP JC.
coated alginate microcapsules by investigating mRNA Nano-scale encapsulation enhances allograft survival
expression of pro-inflammatory cytokines. J Biomimet- and function of islets transplanted in a mouse model of
ics Biomater Tissue Eng 2012; 14: 53–64. diabetes. Diabetologia 2012; 55: 1081–1090.
91. CALAFIORE R, BASTA G, LUCA G et al. Transplantation 108. KIM SK, LEE JH, LEE DH, PARK JK. Optimization of
of pancreatic islets contained in minimal volume micro- chitosan-alginate encapsulation process using pig hepa-
capsules in diabetic high mammalians. Ann NY Acad tocytes for development of bioartificial liver. J Microbiol
Sci 1999; 875: 219–232. Biotechnol 2005; 15: 7–13.
92. SKINNER SJM, GEANEY MS, LIN H et al. Encapsulated 109. HAQUE T, CHEN H, OUYANG W et al. In vitro study of
living choroid plexus cells: potential long-term treat- alginate-chitosan microcapsules: an alternative to liver
ments for central nervous system disease and trauma. cell transplants for the treatment of liver failure.
J Neural Eng 2009; 6: Article ID 065001. Biotechnol Lett 2005; 27: 317–322.
93. OPARA EC, MIRMALEK-SANI SH, KHANNA O, MOYA ML, 110. SON JH, HA YM, KIM YI et al. Immobilization of cry-
BREY EM. Design of a bioartificial pancreas(+). J Invest opreserved primary rat hepatocytes for the development
Med 2010; 58: 831–837. of a bioartificial liver system. Biotechnol Lett 2006; 28:
94. SKINNER SJM, GEANEY MS, LIN H et al. Encapsulated 51–54.
living choroid plexus cells: potential long-term treat- 111. SEO SJ, KIM IY, CHOI YJ, AKAIKE T, CHO CS. Enhanced
ments for central nervous system disease and trauma. J liver functions of hepatocytes cocultured with NIH 3T3
Neural Eng 2009; 6: 11pp. Article ID 065001. in the alginate/galactosylated chitosan scaffold. Bioma-
95. TERBOJEVICH M, MUZZARELLI RAA. Chitosan. In: terials 2006; 27: 1487–1495.
PHILLIPS GO, WILLIAMS PA, eds. Handbook of Hydro- 112. CHEN F, TIAN M, ZHANG D et al. Preparation and char-
colloids. Cambridge: Woodhead Publishing Ltd, 2000: acterization of oxidized alginate covalently cross-linked
367–378. galactosylated chitosan scaffold for liver tissue engineer-
96. LI S, ZHANG Y, CHEN L et al. The relationship between ing. Mater Sci Eng, C 2012; 32: 310–320.
the inflammatory reponse and cell adhesion on alginate- 113. LV G, ZHAO L, ZHANG A et al. Bioartificial liver system
chitosan-alginate microcapsules after transplantation. based on choanoid fluidized bed bioreactor improve the
J Biomed Mater Res, Part A 2014; 103A: 2333–2343. survival time of fulminant hepatic failure pigs. Biotech-
97. HUGUET ML, DELLACHERIE E. Calcium alginate beads nol Bioeng 2011; 108: 2229–2236.
coated with chitosan: effect of the structure of encapsu- 114. FORSEY RW, TARE R, OREFFO ROC, CHAUDHURI JB.
lated Materials on their release. Process Biochem 1996; Perfusion bioreactor studies of chondrocyte growth in
31: 745–751. alginate–chitosan capsules. Biotechnol Appl Biochem
98. DALY MM, KNORR D. Chitosan–alginate complex coac- 2012; 59: 142–152.
ervate capsules: effects of calcium chloride, plasticizers, 115. KUO YC, WANG CC. Guided differentiation of induced
and polyelectrolytes on mechanical stability. Biotechnol pluripotent stem cells into neuronal lineage in alginate-
Prog 1988; 4: 76–81. chitosan-gelatin hydrogels with surface neuron growth
99. ZHAO W, ZHANG Y, LIU Y et al. Oxygen diffusivity in factor. Colloids Surf B Biointerfaces. 2013; 104: 194–
alginate/chitosan microcapsules. J Chem Technol 199.
Biotechnol 2012; 88: 449–455. 116. RENKEN A, HUNKELER D. Polymethylene-co-guanidine
100. BARTKOWIAK A, LISIECKI S, ORIVE G, PEDRAZ JL. The based capsules: a mechanistic study of the formation
effect of selected parameters of formation on properties using alginate and cellulose sulphate. J. Microencapsula-
of alginate/Ca2+/oligochitosan capsules. J Chem Tech- tion 2007; 24: 20–39.
nol Biotechnol 2006; 81: 511–518. 117. SCHULDT U, HUNKELER D. Alginate-cellulose sulphate-
101. BARUCH L, MACHLUF M. Alginate-chitosan complex oligocation microcapsules: optimization of mass trans-
coacervation for cell encapsulation: effect on mechanical port and mechanical properties. J Microencapsulation
properties and on long-term viability. Biopolymers 2006; 2007; 24: 1–10.
82: 570–579. 118. LACIK I, ANILKUMAR AV, WANG TG. A two-step process
102. ORIVE G, BARTKOWIAK A, LISIECKI S et al. Biocompati- for controlling the surface smoothness of polyelec-
ble oligochitosan as cationic modifiers of alginate/Ca trolyte-based microcapsules. J. Microencapsulation
microcapsules. J Biomed Mater Res B Appl Biomater 2001; 18: 479–490.
2005; 74: 429–439. 119. CANAPLE L, NURDIN N, ANGELOVA N, HUNKELER D, DES-
103. 
GASERØD O, SMIDSRØD O, SKJAK  -BRAEK G. Microcap- VERGNE B. Development of a coculture model of encap-
sules of alginate-chitosan-I: a quantitative study of the sulated cells. Ann NY Acad Sci 2001; 944: 350–361.

195
Mahou et al.

120. ZHANG LY, YAO SJ, GUAN YX. Diffusion characteristics 137. PARK TG, HOFFMAN AS. Estimation of temperature-
of solutes with low molecular weight in sodium alginate/ dependent pore size in poly(N-isopropylacrylamide)
cellulose sulfate-CaCl2/poly(methylene-co-guanidine) hydrogel beads. Biotechnol Prog 1994; 10: 82–86.
capsules. J Chem Eng Data 2003; 48: 864–868. 138. DESAI RM, KOSHY ST, HILDERBAND SA, MOONEY DJ,
121. ZHANG LY, YAO SJ, GUAN YX. Effects of poly(methy- JOSHI NS. Versatile click alginate hydrogels crosslinked
lene-co-guanidine) on microbial growth in an alginate/ via tetrazine-norbornene chemistry. Biomaterials 2015;
cellulose sulphate-CaCl2/poly(methylene-co-guanidine) 50: 30–37.
capsule system. Process Biochem 2005; 40: 189–193. 139. XU K, FU Y, CHUNG W. Thiol-ene-based biological/syn-
122. NAHALKA J, DIB I, NIDETZKY B. Encapsulation of thetic hybrid biomatrix for 3-D living cell culture. Acta
Trigonopsis variabilis D-amino acid oxidase and fast Biomater 2012; 8: 2504–2516.
comparison of the operational stabilities of free and 140. FU Y, XU K, ZHENG X et al. 3D cell entrapment in
immobilized preparations of the enzyme. Biotechnol crosslinked thiolated gelatin-poly(ethylene glycol)
Bioeng 2008; 99: 251–260. diacrylate hydrogels. 2012. Biomaterials 2012; 33:
123. VIKARTOVSKA A, BUCKO  M, MISLOVICOV  A D et al. 48–58.
Improvement of the stability of glucose oxidase via 141. PHELPS EA, ENEMCHUKWU NO, FIORE VF et al. Malei-
encapsulation in sodium alginate-cellulose sulfate-poly mide cross-linked bioactive PEG hydrogel exhibits
(methylene-co-guanidine) capsules. Enzyme Microb improved reaction kinetics and cross-linking for cell
Technol 2007; 41: 748–755. encapsulation and in situ delivery. Adv Mater 2012; 24:
124. KLOUDA L, MIKOS AG. Thermoresponsive hydrogels in 64–70.
biomedical applications. Eur J Pharm Biopharm 2008; 142. ALMANY L, SELIKTAR D. Biosynthetic hydrogel scaffolds
68: 34–45. made from fibrinogen and polyethylene glycol for 3D
125. PEPPAS NA, BURES P, LEOBANDUNG W, ICHIKAWA H. cell cultures. Biomaterials 2005; 26: 2467–2477.
Hydrogels in pharmaceutical formulations. Eur J Pharm 143. CHO NJ, ELAZAR M, XIONG A et al. Viral infection of
Biopharm 2000; 50: 27.46. human progenitor and liver-derived cells encapsulated in
126. PATEL A, MEQUANINT K. Hydrogel biomaterials. In: 3D PEG-based hydrogel. Biomed Mater 2009; 4: 011001.
FAZEL-REZAI R, ed. Biomedical Engineering-Frontiers 144. HONG Y, SONG HQ, GONG YH et al. Covalently cross-
and Challenges. Rijeka, Croatia: InTech, 2011: 275–296. linked chitosan hydrogel: properties of in vitro degrada-
ISBN 979-953-307-028-4, Chapter 14. tion and chondrocyte encapsulation. Acta Biomater
127. STABENFELDT SE, GARCIA AJ, LAPLACA MC. Thermore- 2007; 3: 23–31.
versible laminin-functionalized hydrogel for neural tis- 145. HU X, LI D, GAO C. Chemically cross-linked chitosan
sue engineering. J Biomed Mater Res, Part A 2006; 77A: hydrogel loaded with gelatin for chondrocyte encapsula-
718–725. tion. Biotechnol J 2011; 6: 1388–1396.
128. ZAN J, CHEN H, JIANG G, LIN Y, DING F. Preparation 146. TAN H, CHU CR, PAYNE KA, MARRA KG. Injectable
and properties of crosslinked chitosan thermosensitive in situ forming biodegradable chitosan–hyaluronic acid
hydrogel for injectable drug delivery systems. J Appl based hydrogels for cartilage tissue engineering. Bioma-
Polym Sci 2006; 101: 1892–1898. terials 2009; 30: 2499–2506.
129. LU X, HU Z, GAO J. Synthesis and light scattering study 147. BRUNSEN A, RITZ U, MATEESCU A et al. Photocrosslink-
of hydroxypropyl cellulose microgels. Macromolecules able dextran hydrogel films as substrates for osteoblast
2000; 33: 8698–8702. and endothelial cell growth. J Mater Chem 2012; 22:
130. KIM JH, LEE SB, KIM SJ, LEE YM. Rapid temperature/ 19590–19604.
pH response of porous alginate-g-poly(N-isopropylacry- 148. LIU Y, CHAN-PARK MB. A biomimetic hydrogel based
lamide) hydrogels. Polymer 2002; 43: 7549–7558. on methacrylated dextran-graft-lysine and gelatin for
131. SCHMALJOHANN D. Thermo-responsive polymers and 3D smooth muscle cell culture. Biomaterials 2010; 31:
hydrogels in tissue engineering. E-Polymers 2005; 5: 1158–1170.
221–237. 149. FERREIRA LS, GERECHT S, FULLER J et al. Bioactive
132. TAN H, RAMIREZ CM, MILJKOVIC N et al. Ther- hydrogel scaffolds for controllable vascular differentia-
mosensitive injectable hyaluronic acid hydrogel for tion of human embryonic stem cells. Biomaterials 2007;
adipose tissue engineering. Biomaterials 2009; 30: 28: 2706–2717.
6844–6853. 150. CHOH SY, CROSS D, WANG C. Facile synthesis and char-
133. SUWA K, WADA Y, KISHIDA A, AKASHI M. Synthesis and acterization of disulfide-cross-linked hyaluronic acid
functionalities of poly(N-vinylalkylamide). VI. A novel hydrogels for protein delivery and cell encapsulation.
thermosensitive hydrogel crosslinked poly(N-vinylisobu- Biomacromolecules 2011; 12: 1126–1136.
tyramide). J Polym Sci, Part A: Polym Chem 1997; 35: 151. BIAN L, HOU C, TOUS E. The influence of hyaluronic acid
3377–3384. hydrogel crosslinking density and macromolecular diffu-
134. PEROGLIO M, GRAD S, MORTISEN D et al. Injectable ther- sivity on human MSC chondrogenesis and hypertrophy.
moreversible hyaluronan-based hydrogels for nucleus Biomaterials 2013; 34: 413–421.
pulposus cell encapsulation. Eur Spine J 2012; 21: 839– 152. DARR A, CALABRO A. Synthesis and characterization of
849. tyramine-based hyaluronan hydrogels. J Mater Sci
135. EECKMAN F, AMIGHI K, MOES € AJ. Effect of some physio- Mater Med 2009; 20: 33–44.
logical and non-physiological compounds on the phase 153. FLYNN L, PRESTWICH GD, SEMPLE JL, WOODHOUSE KA.
transition temperature of thermoresponsive polymers Adipose tissue engineering with naturally derived scaf-
intended for oral controlled-drug delivery. Int J Pharm folds and adipose-derived stem cells. Biomaterials 2007;
2001; 222: 259–270. 28: 3834–3842.
136. ROLLASON G, DAVIES JE, SEFTON MV. Preliminary report 154. YOUNG CJ, POOLE-WARREN LA, MARTENS PJ. Combin-
on cell culture on a thermally reversible copolymer. Bio- ing submerged electrospray and UV photopolymeriza-
materials 1993; 14: 153–155. tion for production of synthetic hydrogel microspheres

196
Polymeric materials for xenotransplantation

for cell encapsulation. Biotechnol Bioeng 2012; 109: 171. GATTAS  -ASFURA K, FRAKER CA, STABLER CL. Perfluori-
1561–1570. nated alginate for cellular encapsulation. J Biomed
155. ALVES MH, YOUNG CJ, BOZZETTO K, POOLE-WARREN Mater Res, Part A 2012; 100A: 1963–1971.
LA, MARTENS PJ. Degradable, click poly(vinyl alcohol) 172. GATTAS  -ASFURA KM, VALDES M, CELIK E, STABLER CL.
hydrogels: characterization of degradation and cellular Covalent layer-by-layer assembly of hyperbranched
compatibility. Biomed Mater 2012; 7: 11 pages. Article polymers on alginate microcapsules to impart stability
ID 024106. and permselectivity. J Mater Chem B 2014; 2: 8208–
156. NICODEMUS GD, BRYANT SJ. Cell encapsulation in 8219.
biodegradable hydrogels for tissue engineering applica- 173. MAHOU R, WANDREY C. Versatile route to synthesize
tions. Tissue Eng Part B 2008; 14: 149–165. heterobifunctional poly(ethylene glycol) of variable
157. SCHMIDT JJ, ROWLEY J, KONG HJ. Hydrogels used for functionality for subsequent pegylation. Polymers 2012;
cell-based drug delivery. J Biomed Mater Res A 2008; 4: 561–589.
87: 1113–1122. 174. MAHOU R, BORCARD F, CRIVELLI V et al. Tuning the
158. DUSSEAULT J, LEBLOND FA, ROBITAILLE R et al. properties of hydrogel microspheres by adding chemical
Microencapsulation of living cells in semi-permeable cross-linking functionality to sodium alginate. Chem
membranes with covalently cross-linked layers. Bioma- Mater 2015; 27: 4380–4389.
terials 2005; 26: 1515–1522. 175. MAHOU R, MAI NT, DUFRESNE M, LEGALLAIS C, WAN-
159. GARDNER CM, POTTER MA, STOVER € HDH. Improving DREY C. Encapsulation of Huh-7 cells within alginate-
covalent cell encapsulation with temporarily reactive poly(ethylene glycol) hybrid microspheres. J Mater Sci
polyelectrolytes. J Mater Sci: Mater Med 2012; 23: 181– Mater Med 2012; 23: 171–179.
193. 176. LAU HK, KIICK KL. Opportunities for multicomponent
160. MAZUMDER MAJ, BURKE NAD, SHEN F, POTTER MA, hybrid hydrogels in biomedical applications. Biomacro-

STOVER HDH. Core-cross-linked alginate microcapsules mol 2015; 16: 28–42.
for cell encapsulation. Biomacromolecules 2009; 10: 177. TIAN M, HAN B, TAN H, YOU C. Preparation and char-
1365–1373. acterization of galactosylated alginate-chitosan oligomer
161. HILLBERG AL, OUDSHOORN M, LAM JBB, KATHIRGA- microcapsule for hepatocytes microencapsulation. Car-
MANATHAN K. Encapsulation of porcine pancreatic islets bohydr Polym 2014; 112: 502–511.
within an immunoprotective capsule comprising 178. GRIGORE A, SARKER B, FABRY B, BOCCACCINI AR,
methacrylated glycol chitosan and alginate. J Biomed DETSCH R. Behavior of encapsulated MG-63 cells in
Mater Res, Part B 2015; 103B: 503–518. RGD and gelatine-modified alginate hydrogels. Tissue
162. MAZUMDER MAJ, SHEN F, BURKE NAD, POTTER MA, Eng 2014; 20: 2140–2150.

STOVER HDH. Self-crosslinking polyelectrolyte com- 179. WEBER LM, HAYDA KN, HASKINS K, ANSETH KS. The
plexes for therapeutic cell encapsulation. Biomacro- effects of cell-matrix interactions on encapsulated b-cell
molecules 2008; 9: 2292–2300. function within hydrogels functionalized with matrix-
163. GARDNER CM, STOVER € HDH. Reactive polyanions derived adhesive peptides. Biomaterials 2007; 28: 3004–
based on poly(4,4-dimethyl-2-vinyl-2-oxazoline-5-one- 3011.
co-methacrylic acid). Macromolecules 2011; 44: 7115– 180. KIM J-S, LIM J-H, NAM H-Y et al. In situ application of
7123. hydrogel-type fibrin-islet composite optimized for rapid
164. CHEN H, OUYANG W, MARTONI C. Investigation of geni- glycemic control by subcutaneous xenogeneic porcine
pin cross-linked microcapsule for oral delivery of live islet transplantation. J Control Release 2012; 162: 382–
bacterial cells and other biotherapeutics: preparation 390.
and in vitro analysis in simulated human gastrointestinal 181. CAPONE SH, DUFRESNE M, RECHEL M et al. Impact of
model. Int J Polym Sci 2010; 10 pages. Article ID alginate composition: from bead mechanical properties
985137, doi:10.1155/2010/985137. to encapsulated HepG2/C3A cell activities for in vivo
165. NAYAK S, DEY S, KUNDU SC. Silk sericin-alginate-chito- implantation. PLoS ONE 2013; 8: 10 pages.
san microcapsules: hepatocytes encapsulation for 182. CAMPANHA-RODRIGUES AL, GRAZIOLI G, OLIVEIRA TC
enhanced cellular functions. Int J Biol Macromol 2014; et al. Therapeutic potential of laminin-biodritin micro-
65: 258–266. capsules for type 1 diabetes mellitus. Cell Transplant
166. MAHOU R, WANDREY C. Alginate-poly(ethylene glycol) 2015; 24: 247–261.
hybrid microspheres with adjustable physical properties. 183. SIGRIST S, MECHINE-NEUVILLE A, MANDES K et al. Influ-
Macromolecules 2010; 43: 1371–1378. ence of VEGF on the Viability of Encapsulated Pancre-
167. MAHOU R, KOLLARIKOV A G, GONELLE-GISPERT C et al. atic Rat Islets After Transplantation in Diabetic Mice.
Combined electrostatic and covalent polymer networks Cell Transplant 2002; 12: 627–635.
for cell microencapsulation. Macromol Symp 2013; 329: 184. MOYA ML, GARFINKEL MR, LIU X et al. Fibrobalst
49–57. Growth Factor-1 (FGF-1) loaded microbeads enhance
168. MAHOU R, MEIER RPH, BUHLER € L, WANDREY C. Algi- local capillary neovascularization. J Surgical Res 2009;
nate-poly(ethylene glycol)hybrid microspheres for pri- 160: 208–212.
mary cell microencapsulation. Materials 2014; 7: 275– 185. LUAN NM, IWATA H. Xenotransplantation of islets
286. enclosed in agarose microcapsule carrying soluble com-
169. GATTAS  -ASFURA K, STABLER CL. Chemoselective cross- plement receptor 1. Biomaterials 2012; 33: 8075–8081.
linking and functionalization of alginate via Staudinger 186. CHEN T, YUAN J, DUNCANSON S et al. Alginate encapsu-
ligation. Biomacromolecules 2009; 10: 3122–3129. lant incorporating CXCL12 supports long-term allo and
170. HALL KK, GATTAS  -ASFURA K, STABLER CL. Microen- xenoislet transplantation without systemic immune sup-
capsulation of islets within alginate/poly(ethylene glycol) pression. Am J Tranplant 2015; 15: 618–627.
gels cross-linked via Staudinger ligation. Acta Biomater 187. ACARREGUI A, HERRAN  E, IGARTUA M et al. Multifunc-
2011; 7: 614–624. tional hydrogel-based scaffold for improving the

197
Mahou et al.

functionality of encapsulated therapeutic cells and 206. SAYEGH MH, TURKA LA. The role of T-cell costimula-
reducing inflammatory response. Acta Biomater 2014; tory activation pathways in transplant rejection. N Engl
10: 4206–4216. J Med 1998; 338: 1813–1821.
188. LI C-C, METTERS AT, ANSETH KS. Functional PEG-pep- 207. BHUJBAL SV, PAREDES-JUAREZ GA, NICLOU SP,
tide hydrogels to modulate local inflammation induced De VOS P. Factors influencing the mechanical stability
by the proinflammatory cytokine TNFa. Biomaterials of alginate beads applicable for immunoisolation of
2009; 30: 4907–4914. mammalian cells. J Mech Behav Biomed Mater 2014;
189. SU J, HU B-H, LOWE WL, KAUFMAN DB, MESSERSMITH 37: 196–208.
PB. Anti- imflammatory peptide-functionalized hydro- 208. VEISEH O, DOLOFF JC, MA M et al. Size- and shape-
gels for insulin-secreting cell encapsulation. Biomaterials dependent foreign body immune response to materials
2010; 31: 308–314. implanted in rodents and non-human primates. Nat
190. THU B, BRUHEIM P, ESPEVIK T et al. Alginate polycation Mater 2015; 14: 643–651.
microcapsules: I. Interaction between alginate and poly- 209. DULIEU C, PONCELET D, NEUFELD RJ Ed. Birkh€ auser:
cation. Biomaterials 1996; 17: 1031–1040. Boston 1999: 3–17.
191. SKARJA GA, KINLOUGH-RATHBONE RL, PERRY DW, 210. ANGELOVA N, HUNKELER D. Rationalizing the design of
RUBENS FD, BRASH JL. A coneand- plate device for the polymeric biomaterials. Trends Biotechnol 1999; 17:
investigation of platelet biomaterial interactions. 409–421.
J Biomed Mater Res 1997; 34: 427–438. 211. De VOS P, De HAAN B, PATER J, Van SCHILFGAARDE R.
192. 
ROSINSKI S, GRIGORESCU G, LEWINSKA  D et al. Charac- Association between capsule diameter, adequacy of
terization of microcapsules: recommended methods encapsulation, and survival of microencapsulated rat
based on round robin testing. J. Microencapsulation islet allografts. Transplantation 1996; 62: 893–899.
2002; 19: 641–659. 212. CEAUSOGLU I, HUNKELER D. A new microencapsulation
193. DRURY JL, DENNIS RG, MOONEY DJ. The tensile proper- device for controlled membrane and capsule size distri-
ties of alginate hydrogels. Biomaterials 2004; 25: 3187– butions. J. Microencapsulation 2002; 19: 725–735.
3199. 213. VIDAL-SERP DS. Polyelectrolytes-based immunoprotec-
194. JEFFREY L, HINKLEY A, MORGRET LD, GEHRKE SH. Ten- tion systems: polymer purification and surface analysis.
sile properties of two responsive hydrogels. Polymer Thesis EPFL No 3053, 2004.
2004; 45: 8837–8843. 214. YU J, WU P-C, HUANG C-H, YANG C-Y, CHENG C-M.
195. AHEARNE M, YANG Y, El HAJ AJ, THEN KY, LIU KK. High-throughput physically based approach for mam-
Characterizing the viscoelastic properties of thin hydro- malian cell encapsulation. Appl Phys Lett 2013; 103:
gel-based constructs for tissue engineering applications. 153704.
J R Soc Interface 2005; 2: 455–463. 215. HEINZEN C, MARISON I, BERGER A, VON STOCKAR U. Use
196. 
LEWINSKA 
D, ROSINSKI S, HUNKELER D, PONCELET D, of vibration technology for jet break-up for encapsula-
WERYNSKI A. Mass transfer coefficient in characteriza- tion of cells, microbes and liquids in monodisperse
tion of gel beads and microcapsules. J Membr Sci 2002; microcapsules. In Landbauforschung Voelkenrode, Son-
209: 533–540. derheft, num. 241 (Practical aspects of encapsulation
197. LI RH, ALTREUTER DH, GENTILE FT. Transport charac- technologies), p. 19–25, 2002.
terization of hydrogel matrices for cell encapsulation. 216. HALLE JP, LEBLOND FA, PARISEAU JF et al. Studies on
Biotechnol Bioeng 1996; 50: 365–373. small (<300 lm) microcapsules II: parameters governing
198. SCHULDT U, HUNKELER D. Characterization methods for the production of alginate beads by high voltage electro-
microcapsules. Minerva Biotechnologica 2000; 12: 249– static pulses. Cell Transplant 1994; 3: 365–372.
264. 217. GRYSHKOV O, POGOZHYKH D, HOFMANN N et al. Encap-
199. ULUDAG H, De VOS P, TRESCO PA. Technology of mam- sulating non-human primate multipotent stromal cells in
malian cell encapsulation. Adv Drug Deliv Rev 2000; alginate via high voltage for cell-based therapies and cry-
42: 29–64. opreservation. PLoS ONE 2014; 9: 9: e107911: 12 pages.
200. BRISSOVA M, PETRO M, LACIK I, POWERS AC, WANG T. 218. PONCELET D, BABAK V, DULIEU C, PICOT A. A physico-
Evaluation of microcapsule permeability via inverse size chemical approach to production of alginate beads by
exclusion chromatography. Anal Biochem 1996; 242: emulsification-internal ionotropic gelation. Colloids Surf
104–111. A 1999; 155: 171–176.
201. BRISSOVA M, LACIK I, POWERS AC, ANILKUMAR AV, 219. PONCELET D, NEUFELD R, BUGARSK B et al. A parallel
WANG T. Control and measurement of permeability for plate electrostatic droplet generator: parameters affect-
design of microcapsule cell delivery system. J Biomed ing microbead size. Appl Microbiol Biotechnol 1994; 42:
Mater Res 1997; 39: 61–70. 251–255.
202. BERTZ A, WOHL € -BRUHN S, MIETHE S et al. Encapsula- 220. BRANDENBERGER H, WIDMER F. A new multinozzle
tion of proteins in hydrogel carrier systems for con- encapsulation/immobilization system to produce uni-
trolled drug delivery: influence of network structure form beads of alginate. J Biotechnol 1998; 63: 73–80.
and drug size on release rate. J Biotechnol 2013; 163: 221. PRU€ ßE U, FOX B, KIRCHHOFF M et al. New process (jet
243–249. cutting method) for the production of spherical beads
203. WILLIAMS D. Revisiting the definition of biocompatibil- from highly viscous polymer solutions. Chem Eng Tech-
ity. Med Device Technol 2003; 14: 10–13. nol 1998; 21: 29–33.
204. WILLIAMS DF. Summary and definitions. In: Progress 222. PONCELET D, LENCKI R, BEAULIEU C et al. Production of
in Biomedical Engineering: Definition In Biomaterials alginate beads by emulsification/internal gelation I.
(4). Amsterdam: Elsevier Science Publisher BV, 1987: Methodology. Appl Microbiol Biotechnol 1992; 38: 39–
66–71. 45.
205. AUCHINCLOSS H, SACHS DH. Xenogeneic transplantation. 223. RABANEL JM, BANQUY X, ZOUAOUI H, MOKHTAR M,
Annu Rev Immunol 1998; 16: 433–470. HILDGEN P. Progress technology in microencapsulation

198
Polymeric materials for xenotransplantation

methods for cell therapy. Biotechnol Prog 2009; 25: 946– using a droplet-based microfluidic device. Microfluid
963. Nanofluid 2012; 13: 761–767.
224. MAZZITELLI S, CAPRETTO L, QUINCI F, PIVA R, NASTRUZZI 243. DALLE P, MORALES S, BERTHIER J et al. New microfluidic
C. Preparation of cell-encapsulation devices in confined chip for the production of spherical gelled capsules for
microenvironment. Adv Drug Deliv Rev 2013; 65: 1533– cell encapsulation. Technical Proceedings of the 2012
1555. NSTI Nanotechnology Conference and Expo, NSTI-
225. KANG A, PARK J, JU J, JEONG GS, LEE S-H. Cell encap- Nanotech 2012: 298–301.
sulation via microtechnologies. Biomaterials 2014; 35: 244. MARTINEZ CJ, KIM JW, YE C et al. A microfluidic
2651–2663. approach to encapsulate living cells in uniform alginate
226. KIM C, PARK J, KANG JY. A microfluidic manifold with hydrogel microparticles. Macromol Biosci 2012; 12:
a single pump system to generate highly mono-disperse 946–951.
alginate beads for cell encapsulation. Biomicrofluidics 245. VELASCO D, TUMARKIN E, KUMACHEVA E. Microfluidic
2014; 8: 066504. encapsulation of cells in polymer microgels. Small 2012;
227. CHAU M, ADOLHASANI M, THERIEN-AUBIN H et al. 8: 1633–1642.
Microfluidic generation of composite biopolymer micro- 246. ROSSOW T, HEYMAN JA, EHRLICHER AJ et al. Controlled
gels with tunable compositions and mechanical proper- synthesis of cell-laden microgels by radical-free gelation
ties. Biomacromolecules 2014; 15: 2419–2425. in droplet microfluidics. J Am Chem Soc 2012; 134:
228. WAN J. Microfluidic-based synthesis of hydrogel parti- 4983–4989.
cles for cell microencapsulation and cell-based drug 247. HEADEN DM, AUBRY G, LU H, GARCIA AJ.
delivery. Polymers 2012; 4: 1084–1108. Microfluidic-based generation of size-controlled, bio-
229. SEO M, NIE Z, XU S et al. Continuous microfluidic reac- functionalized synthetic polymer microgels for cell
tors for polymer particles. Langmuir 2005; 21: 11614– encapsulation. Adv Mater 2014; 26: 3003–3008.
11622. 248. AKBARI S, PIRBODAGHI T. Microfluidic encapsulation of
230. DENDUKURI D, PREGIBON DC, COLLINS J, HATTON TA, cells in alginate particles via an improved internal gela-
DOYLE PS. Continuous-flow lithography for high- tion approach. Microfluid Nanofluid 2013; 16: 773–
throughput microparticle synthesis. Nat Mater 2006; 5: 777.
365–369. 249. WU L, CHEN P, DONG Y, FENG X, LIU BF. Encapsula-
231. KINTSES B, Van VLIET LD, DEVENISH SRA, HOLFELDER tion of single cells on a microfluidic device integrating
F. Microfluidic droplets: new integrated work-flows for droplet generation with fluorescence-activated droplet
biological experiments. Curr Opin Chem Biol 2010; 14: sorting. Biomed Microdevices 2013; 15: 553–560.
548–555. 250. TENDULKAR S, MIRMALEK-SANI SH, CHILDERS C et al. A
232. KHAN IU, SERRA CA, ANTON N et al. Microfluidic con- three-dimensional microfluidic approach to scaling up
ceived drug loaded Janus particles in side-by-side capil- microencapsulation of cells. Biomed Microdevices 2012;
laries device. Int J Pharm 2014; 473: 239–249. 14: 461–469.
233. MAZUTIS L, VASILIAUSKAS R, WEITZ DA. Microfluidic 251. WILSON JT, CUI W, CHAIKOF EL. Layer-by-layer assem-
production of alginate hydrogel particles for antibody bly of a conformal nanothin PEG coating for intraportal
encapsulation and release. Macromol Biosci 2015; 15: islet transplantation. Nano Lett 2008; 8: 1940–1948.
1641–1646. 252. SEFTON MV, MAY MH, LAHOOTI S, BABENSEE JE. Mak-
234. UTECH S, PRODANOVIC R, MAO AS et al. Microfluidic ing microencapsulation work: conformal coating, immo-
generation of monodisperse, structurally homogeneous bilization gels and in vivo performance. J Controlled
alginate microgels for cell encapsulation and 3D cell cul- Release 2000; 65: 173–186.
ture. Adv Healthcare Mater 2015; 4: 1628–1633. 253. WYMAN JL, KIZILEL S, SKARBEK R et al. Immunoisolat-
235. ALLAZETTA S, HAUSHERR TC, LUTOLF MP. Microfluidic ing pancreatic islets by encapsulation with selective with-
synthesis of cell-type-specific artificial extracellular drawal. Small 2007; 3: 683–690.
matrix hydrogels. Biomacromolecules 2013; 14: 1122– 254. CRUISE GM, HEGRE OD, SCHARP DS, HUBBELL JA. A
1131. sensitivity study of the key parameters in the interfacial
236. LUO RC, CHEN CH. Structured microgels through photopolymerization of poly(ethylene glycol) diacrylate
microfluidic assembly and their biomedical applications. upon porcine islets. Biotechnol Bioeng 1998; 57: 655–
SciRes Soft 2012; 1: 1–23. 665.
237. SHIM TS, KIM SH, YANG SM. Elaborate design strategies 255. BLASI P, LUCA G, MANCUSO F et al. Conformal polymer
toward novel microcarriers for controlled encapsulation coatings for pancreatic islets transplantation. Int J
and release. Part Part Syst Char 2013; 30: 9–45. Pharm 2013; 440: 141–147.
238. GARSTECKI P, FUERSTMAN MJ, STOEN HA, WHITESIDE 256. TERAMURA Y, IWATA H. Bioartificial pancreas: microen-
GM. Formation of droplets and bubbles in a cicrofluidic capsulation and conformal coating of islet of Langer-
T-junction-scaling and mechanism of break-up. Lab hans. Adv Drug Delivery Rev 2010; 62: 827–840.
Chip 2006; 6: 437–446. 257. KIZILEL S, SCAVONE A, LIU X et al. Encapsulation of
239. BAROUD CN, GALLAIRE F, DANGLA R. Dynamics of pancreatic islets within nano-thin functional polyethy-
microfluidic droplets. Lab Chip 2010; 10: 2032–2045. lene glycol coatings for enhanced insulin secretion. Tis-
240. NIE ZH, SEO MS, XU SQ et al. Emulsification in a sue Eng Part A 2010; 16: 2217–2228.
microfluidic flow-focusing device: effect of the viscosities 258. LUAN NM, TERAMURA Y, IWATA H. Layer-by-layer co-
of the liquids. Microfluid Nanofluidics 2008; 5: 585–594. immobilization of soluble complement receptor 1 and
241. MENDES AC, BARAN ET, LISBOA P et al. Microfluidic heparin on islets. Biomaterials 2011; 32: 6487–6492.
fabrication of self-assembled peptide-polysaccharide 259. TERAMURA Y, OOMMEN OP, OLERUD J, HILBORN J, NILS-
microcapsules as 3D environments for cell culture. SON B. Microencapsulation of cells, including islets,
Biomacromolecules 2012; 13: 4039–4048. within stable ultra-thin membranes of maleimide-conju-
242. LIU K, DENG Y, ZHANG N et al. Generation of disk-like gated PEG-lipid with multifunctional crosslinkers. Bio-
hydrogel beads for cell encapsulation and manipulation materials 2013; 34: 2683–2693.

199
Mahou et al.

260. SORELLE JA, KANAK MA, ITOH T et al. Comparison of in the omental pouch of NOD mice. Cell Transplant
surface modification chemistries in mouse, porcine, and 2006; 15: 359–365.
human islets. J Biomed Mater Res, Part A 2015; 103A: 276. KOBAYASHI T, AOMATSU Y, KANEHIRO H, HISANAGA M,
869–877. NAKAJIMA Y. Protection of NOD islet isograft from
261. KOZLOVSKAYA V, ZAVGORODNYA O, KHARLAMPIEVA E. autoimmune destruction by agarose microencapsulation.
Encapsulation and surface engineering of pancreatic Transplant Proc 2003; 35: 484–485.
islets: advances and challenges. In: LIN C, ed. Biomedi- 277. FORT A, FORT N, RICORDI C, STABLER C. Biohybrid
cine, Ch.1. Rijeka, Croatia: InTech 2012: 1–32, (ISBN devices and encapsulation technologies for engineering a
978-953-51-0352-3). bioartificial pancreas. Cell Transplant 2008; 17: 997–
262. WILSON JT, CUI W, KOZLOVSKAYA V et al. Cell surface 1003.
engineering with polyelectrolyte multilayer thin films. 278. VERITER S, MERGEN J, GOEBBELS RM et al. In vivo selec-
J Am Chem Soc 2011; 133: 7054–7064. tion of biocompatible alginates for islet encapsulation
263. WILSON JT, KRISHNAMURTHY VR, CUI W, QU Z, CHAI- and subcutaneous transplantation. Tissue Eng Part A
KOF EL. Noncovalent cell surface engineering with catio- 2010; 16: 1503–1513.
nic graft copolymers. J Am Chem Soc 2009; 131: 18228– 279. WANG W, GU Y, TABATA Y et al. Reversal of diabetes in
18229. mice by xenotransplantation of a bioartificial pancreas

264. NOTH €
U, GROHN P, JORK A et al. 19F-MRI in vivo deter- in a prevascularized subcutaneous site. Transplantation
mination of the partial oxygen pressure in perfluorocar- 2002; 73: 122–129.
bon-loaded alginate capsules implanted into the 280. PETRELLI A, CARVELLO M, VERGANI A et al. IL-21 is an
peritoneal cavity and different tissues. Magn Reson Med antitolerogenic cytokine of the late-phase alloimmune
1999; 42: 1039–1047. response. Diabetes 2011; 60: 3223–3234.
265. TOSO C, MATHE Z, MOREL P et al. Effect of microcapsule 281. JUANG JH, SHEN CR, WANG JJ et al. Magnetic resonance
composition and short-term immunosuppression on imaging study of mouse islet allotransplantation. Trans-
intraportal biocompatibility. Cell Transplant 2005; 14: plant Proc 2010; 42: 4217–4220.
159–167. 282. COOPER DKC, BOTTINO R, SATYANANDA V, WIJKSTROM
266. MAI G, HUY NT, MOREL P et al. Treatment of fulminant M, TRUCCO M. Toward clinical islet xenotransplantation
liver failure by transplantation of microencapsulated pri- – are revisions to the IXA guidelines warranted? Xeno-
mary or immortalized xenogeneic hepatocytes. Trans- transplantation 2013; 20: 68–74.
plant Proc 2005; 37: 527–529. 283. REICHART B, NIEMANN H, CHAVAKIS T et al. Xenotrans-
267. MAI G, HUY NT, MOREL P et al. Treatment of fulminant plantation of porcine islet cells as a potential option for
liver failure by transplantation of microencapsulated pri- the treatment of type 1 diabetes in the future. Horm
mary or immortalized xenogeneic hepatocytes. Xeno- Metab Res 2015; 47: 31–35.
transplantation 2005; 12: 457–464. 284. SHIN JS, KIM JM, KIM JS et al. Long-term control of
268. KOBAYASHI T, AOMATSU Y, IWATA H et al. Indefinite diabetes in immunosuppressed nonhuman primates
islet protection from autoimmune destruction in non- (NHP) by the transplantation of adult porcine islets. Am
obese diabetic mice by agarose microencapsulation J Transplant 2015; 15: 2837–2850.
without immunosuppression. Transplantation 2003; 75: 285. ZHU H, YU L, HE Y et al. Microencapsulated pig islet
619–625. xenotransplantation as an alternative treatment of dia-
269. KOBAYASHI T, AREFANIAN H, HARB G et al. Pro- betes. Tissue Eng Part B 2015; 21: 474–489.
longed survival of microencapsulated neonatal por- 286. JIMENEZ-VERA E, DAVIES S, PHILLIPS P et al. Long-term
cine islet xenografts in immune-competent mice cultured neonatal islet cell clusters demonstrate better
without antirejection therapy. Cell Transplant 2008; outcomes for reversal of diabetes: in vivo and molecular
17: 1243–1256. profiles. Xenotransplantation 2015; 22: 114–123.
270. KOBAYASHI T, HARB G, RAYAT GR. Prolonged survival 287. Van BURCK€ L, SEISSLER J. The potential of local
of microencapsulated neonatal porcine islets in mice immunomodulation and tolerance induction in porcine
treated with a combination of anti-CD154 and anti- islet xenotransplantation. Xenostransplantation 2013;
LFA-1cmonoclonal antibodies. Transplantation 2005; 20: 51.
80: 821–827. 288. NAGARAJU S, BOTTINO R, WIJKSTROM M et al. Islet xeno-
271. CANTARELLI E, PIEMONTI L. Alternative transplantation transplantation: what is the optimal age of the islet-
sites for pancreatic islet grafts. Curr Diab Rep 2011; 11: source pig? Xenotransplantation 2015; 22: 7–19.
364–374. 289. BOTTINO R, WIJKSTROM M, van der WINDT DJ et al. Pig-
272. TUCH BE, KEOGH GW, WILLIAMS LJ et al. Safety and to-monkey islet xenotransplantation using multi-trans-
viability of microencapsulated human islets transplanted genic pigs. Am J Transplant 2014; 14: 2275–2287.
into diabetic humans. Diabetes Care 2009; 32: 1887– 290. RAMACKERS W, KLOSE J, VONDRAN FWR et al.
1889. Species-specific regulation of fibrinogen synthesis with
273. SAFLEY SA, CUI H, CAUFFIEL S, TUCKER-BURDEN C, implications for porcine hepatocyte xenotransplanta-
WEBER CJ. Biocompatibility and immune acceptance of tion. Xenotransplantation 2014; 21: 444–453.
adult porcine islets transplanted intraperitoneally in dia- 291. HAM DS, SONG MS, PARK HS et al. Successful xeno-
betic NOD mice in calcium alginate poly-L-lysine micro- transplantation with re-aggregated and encapsulated
capsules versus barium alginate microcapsules without neonatal pig liver cells for treatment of mice with acute
poly-L-lysine. J Diabetes Sci Technol 2008; 2: 760–767. liver failure. Xenotransplantation 2015; 22: 249–259.
274. KIN T, KORBUTT GS, RAJOTTE RV. Survival and meta- 292. NO DY, JEONG GS, LEE SH. Immune-protected xeno-
bolic function of syngeneic rat islet grafts transplanted geneic bioartificial livers with liver-specific microarchi-
in the omental pouch. Am J Transplant 2003; 3: 281– tecture and hydrogel-encapsulated cells. Biomaterials
285. 2014; 35: 8983–8991.
275. KOBAYASHI T, AOMATSU Y, IWATA H et al. Survival of 293. WRIGHT JR Jr, YANG H, HYRTSENKO O et al. A review of
microencapsulated islets at 400 days posttransplantation piscine islet xenotransplantation using wild-type tilapia

200
Polymeric materials for xenotransplantation

donors and the production of transgenic tilapia express- 300. MATSUMOTO S, TAN P, BAKER J, DURBIN K, TOMIYA M,
ing a “humanized” tilapia insulin. Xenotransplantation DOI M, AZUMA K, ELIOTT RB. Clinical porcine islet
2014; 21: 485–495. xenotransplantation under comprehensive regulation.
294. WHITE DJ. Fish islet xenografts. Xenotransplantation Transplant Proc 2014; 46: 1992–1995.
2014; 21: 124–126. 301. LUDWIG B, LUDWIG S. Transplantable bioartificial pan-
295. SAFLEY SA, CUI H, CAUFFIEL SMD et al. Encapsulated creas devices: current status and future prospects. Lan-
piscine (tilapia) islets for diabetes therapy: studies in dia- genbecks Arch Surg 2015; 400: 531–540.
betic NOD and NOD-SCID mice. Xenotransplantation 302. De VOS P, Van STRAATEN JF, NIEUWENHUIZEN AG et al.
2014; 21: 127–139. Why do microencapsulated islet grafts fail in the absence
296. OMER A, DUVIVIER-KALI V, FERNANDES J et al. Long- of fibrotic overgrowth? Diabetes 1999; 48: 1381–1388.
term normoglycemia in rats receiving transplants with 303. De VOS P, VEGTER D, STRUBBE JH, De HAAN BJ, Van
encapsulated islets. Transplantation 2005; 79: 52–58. SCHILFGAARDE R. Impaired glucose tolerance in recipi-
297. GOMEZ N, BALLADUR P, CALMUS Y et al. Evidence for ents of an intraperitoneally implanted microencapsu-
survival and metabolic activity of encapsulated lated islet allograft is caused by the slow diffusion of
xenogeneic hepatocytes transplanted without immuno- insulin through the peritoneal membrane. Transplant
suppression in Gunn rats. Transplantation 1997; 63: Proc 1997; 29: 756–757.
1718–1723. 304. COOPER DKC. The case for xenotransplantation. Clin
298. KIN T, KORBUTT GS, RAJOTTE RV. Normalization of Transplant 2015; 29: 288–293.
diabetes in spontaneously diabetic cynomologus mon- 305. ELLIS CE, KORBUTT GS. Justifying clinical trials for por-
keys by xenografts of microencapsulated porcine islets cine islet xenotransplantation. Xenotransplantation
without immunosuppression. J Clin Invest 1996; 98: 2015; 22: 336–344.
1417–1422. 306. AGUDELO CA, TERAMURA Y, IWATA H. Cryopreserved
299. BASTA G, MONTANUCCI P, LUCA G et al. Long-term agarose-encapsulated islets as bioartificial pancreas: a
metabolic and immunological follow-up of nonimmuno- feasibility study. Transplantation 2009; 87: 29–34.
suppressed patients with type 1 diabetes treated with 307. SCHNEIDER S, KLEIN HH. Long-term graft function of
microencapsulated islet allografts: four cases. Diabetes cryostored alginate encapsulated rat islets. Eur J Med
Care 2011; 34: 2406–2409. Res 2011; 16: 396–400.

201

You might also like