19
A Perspective on Bioactive Cell Microencapsulation Argia Acarregui, 1,2 Ainhoa Murua, 1,2 Jose ´ L. Pedraz, 1,2 Gorka Orive 1,2 and Rosa M. Herna ´ndez 1,2 1 NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain 2 Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz, Spain Contents Abstract ........................................................................................................... 283 1. Introduction .................................................................................................... 284 2. Cell Encapsulation Technology .................................................................................... 284 2.1 Biomaterials ................................................................................................ 285 2.1.1 Alginate and Functionalized Alginate Gels ................................................................ 285 2.1.2 Collagen ............................................................................................. 286 2.1.3 Chitosan ............................................................................................. 286 2.1.4 Agarose .............................................................................................. 286 2.1.5 Other Polymers: Poly(N-isopropylacrylamide), Gelatin, Poly(ethylene glycol), Hyaluronic Acid and Cellulose Sulphate 287 2.2 The Requirements of the Technology ........................................................................... 287 2.2.1 Permeability .......................................................................................... 287 2.2.2 Mechanical Integrity, Stability, and Durability .............................................................. 288 2.2.3 The Size, Morphology, and Surface Properties of the Capsules ................................................ 289 2.2.4 Biocompatibility ....................................................................................... 289 2.2.5 Other Issues ........................................................................................... 289 3. Therapeutic Applications and Clinical Trials .......................................................................... 290 3.1 Diabetes ................................................................................................... 290 3.2 Central Nervous System Diseases .............................................................................. 290 3.3 Cardiovascular Diseases...................................................................................... 293 3.4 Liver Diseases ............................................................................................... 293 3.5 Other Diseases .............................................................................................. 294 4. Concluding Remarks and Future Directions and Challenges ........................................................... 295 Abstract Bioactive cell encapsulation has emerged as a promising tool for the treatment of patients with various disorders including diabetes mellitus, central nervous system diseases, and cardiovascular diseases. The implantation of encapsulated cells that secrete a therapeutic product (protein, peptide, or antibody) within a semipermeable membrane provides a physical barrier to mask the implant from immune surveillance at a local level without the need for systemic immunosuppression; this serves to achieve a successful therapeutic function following in vivo implantation. The aim of this review article is to provide an update on the progress in this field. The current state of cell encapsulation technology as a controlled drug delivery system will be covered in detail, and the essential requirements of the technology, the challenges, and the future directions under investigation will be highlighted. The technical and biological advances, together with the increasing experience in the field, may lead to the realization of the full potential of bioactive cell encapsulation in the coming years. REVIEW ARTICLE Biodrugs 2012; 26 (5): 283-301 1173-8804/12/0005-0283/$49.95/0 Adis ª 2012 Springer International Publishing AG. All rights reserved.

A Perspective on Bioactive Cell Microencapsulation

  • Upload
    rosa-m

  • View
    222

  • Download
    1

Embed Size (px)

Citation preview

Page 1: A Perspective on Bioactive Cell Microencapsulation

A Perspective on Bioactive Cell MicroencapsulationArgia Acarregui,1,2 Ainhoa Murua,1,2 Jose L. Pedraz,1,2 Gorka Orive1,2 and Rosa M. Hernandez1,2

1 NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU),

Vitoria-Gasteiz, Spain

2 Networking Biomedical Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz, Spain

Contents

Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284

2. Cell Encapsulation Technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284

2.1 Biomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285

2.1.1 Alginate and Functionalized Alginate Gels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285

2.1.2 Collagen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

2.1.3 Chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

2.1.4 Agarose . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286

2.1.5 Other Polymers: Poly(N-isopropylacrylamide), Gelatin, Poly(ethylene glycol), Hyaluronic Acid and Cellulose Sulphate 287

2.2 The Requirements of the Technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287

2.2.1 Permeability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287

2.2.2 Mechanical Integrity, Stability, and Durability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288

2.2.3 The Size, Morphology, and Surface Properties of the Capsules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

2.2.4 Biocompatibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

2.2.5 Other Issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289

3. Therapeutic Applications and Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

3.1 Diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

3.2 Central Nervous System Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290

3.3 Cardiovascular Diseases. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293

3.4 Liver Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293

3.5 Other Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294

4. Concluding Remarks and Future Directions and Challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295

Abstract Bioactive cell encapsulation has emerged as a promising tool for the treatment of patients with various

disorders including diabetes mellitus, central nervous system diseases, and cardiovascular diseases. The

implantation of encapsulated cells that secrete a therapeutic product (protein, peptide, or antibody) within a

semipermeable membrane provides a physical barrier to mask the implant from immune surveillance at a

local level without the need for systemic immunosuppression; this serves to achieve a successful therapeutic

function following in vivo implantation. The aim of this review article is to provide an update on the progress

in this field. The current state of cell encapsulation technology as a controlled drug delivery system will be

covered in detail, and the essential requirements of the technology, the challenges, and the future directions

under investigation will be highlighted. The technical and biological advances, together with the increasing

experience in the field, may lead to the realization of the full potential of bioactive cell encapsulation in the

coming years.

REVIEWARTICLEBiodrugs 2012; 26 (5): 283-301

1173-8804/12/0005-0283/$49.95/0

Adis ª 2012 Springer International Publishing AG. All rights reserved.

Page 2: A Perspective on Bioactive Cell Microencapsulation

1. Introduction

New molecules with high therapeutic effect are currently

being discovered; in some cases, traditional routes of adminis-

tration may not allow suitable delivery or guarantee maximum

efficacy. Many of these therapeutic molecules are peptides that

degrade easily and proteins that have limited half-lives in vivo;

they are especially difficult to administer at therapeutic con-

centrations and for extended periods of time. The purpose of

drug delivery systems is to maintain sustained therapeutic

concentrations with minimal side effects at specific locations in

the body, modifying the pharmacokinetics of the drug. Nu-

merous technologies using biomaterials in the form of fibers,

capsules, particles, three-dimensional porous scaffolds and in-

jectable gels have been investigated to better control the deliv-

ery of these drugs.[1]

The transplantation of encapsulated cells is a potent strategy

to enable the local and controlled delivery of therapeutic mol-

ecules to specific physiological sites as a means to recover a loss

of function caused by disease or degeneration.[2] Delivery could

be performed by cell encapsulation or by immobilization within

a semipermeable membrane.[1] The semipermeable membrane

may isolate the entrapped cells from the host immune system

and control the outward and inward diffusion of molecules.

Consequently, the long-term therapies of immunosuppressant

drugs could be eliminated or at least reduced. In this applica-

tion, cells may be considered ‘biological factories’ or ‘living cell

medicines’ that can continuously produce and release ther-

apeutic molecules in a biosafe manner.[3]

In the present review, drug delivery system strategies relying

on microcapsules, especially approaches based on cell micro-

encapsulation technology, are highlighted to provide a general

overview of the current research. The review focuses on some of

the most frequently used biomaterials and the most important

requirements of the technology. Special attention is paid to

some of the most encouraging therapeutic applications and

current clinical trials. Unresolved issues in the field and future

directions for investigation are discussed.

2. Cell Encapsulation Technology

Tissue and organ transplantation, as well as drug delivery

technologies, have significantly improved during the last few

years. Specifically, the immobilization of cells from an outside

environment within a permselective (semipermeable) mem-

brane barrier protects the inner cells from both mechanical

stress and from the components of the host immune system

(such as immunoglobulins, complement and immune cells); at

the same time, the membrane allows the inward diffusion of

nutrients and oxygen as well as the outward delivery of waste

products and therapeutic agents (figure 1). Moreover, this

membrane permits the transplantation of cells without the use

of long-term modulating and/or immunosuppressive therapies,

which have potentially severe side effects.[4,5] Cell encapsulation

technology can be used for the treatment of a large variety of

diseases, including hypoparathyroidism,[6] hemophilia B,[7,8]

anemia,[9] dwarfism,[10,11] liver failure,[12] central nervous sys-

tem (CNS) diseases,[13,14] diabetes mellitus,[15] cancer,[16] and

cardiovascular diseases.[17]

Cell encapsulation technology displays important advan-

tages over other delivery systems because it permits a sustained

and controlled release of de novo-produced therapeutic mole-

cules; this ensures the bioactivity of the therapeutic. Additional

advantages include the enhancement of biosafety, because the

toxicity caused by a rapid delivery of high concentrations of the

drug does not occur, and host immune system response might

NutrientsO2

Therapeutic moleculesWaste products

Hostimmune

mediators

Fig. 1. A schematic illustration of the encapsulated cells within the semipermeable membrane (dashed line) that allows the bidirectional diffusion of nutrients,

oxygen, therapeutic products and waste while preventing the entrance of immune cells and antibodies.

284 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 3: A Perspective on Bioactive Cell Microencapsulation

attack the cells (compromising their survival) if they manage to

leave the microcapsule environment. Moreover, the genetically

modified cells can express any protein desired for in vivo ther-

apy without the modification of the host genome.[18] It is

possible to create small capsules (from 100 mm to 500 mm) and

implant them in close contact with the blood stream, improving

oxygen transfer into the cells, whichmay be beneficial in certain

applications. Furthermore, in comparison with other immobi-

lization systems, the high surface/volume ratio of the micro-

capsules increasesmembrane permeability, ensuring an optimal

product exchange to ensure adequate cell viability. Finally,

the microcapsules can be injected directly or transplanted

during minimally invasive surgery into implantation sites

such as the peritoneal cavity[19,20] and subcutaneous tissue,[21]

reducing the frequency of administration and thus improving

patient comfort.

The most widely employed cell microencapsulation method

was originally developed by Lim and Sun[22] and is based

on the polyelectrolyte complexation of alginate with a

polycation, normally poly-L-lysine (PLL) or poly-L-ornithine

(PLO).[20] These types of microcapsules are commonly referred

to as alginate-PLL-alginate (APA) or alginate-PLO-alginate

(APO) microcapsules, respectively. Apart from microcapsules

formed with these polycations, other polymers, such as chito-

san,[23] lactose-modified chitosan,[24,25] oligochitosans,[26,27]

poly(methylene-co-guanidine),[28] sodium silicate,[29] andmulti-

layers,[30-32] have also been employed to cover the alginate

cores. The drawback of these materials is that they are asso-

ciated with cytotoxicity and cell necrosis, which has generated

much controversy among research groups. Comparative stud-

ies using different polycations have alleviated some of these

concerns, but it is still unclear which of these strategies is the

best for cell encapsulation.[33,34] In recent decades, the initial

technique has undergone modifications, leading to several

methods to produce microcapsules, including extrusion meth-

ods,[35,36] emulsion (thermal gelation),[37] microfluidics,[38,39]

and microlithography.[40]

2.1 Biomaterials

Using an appropriate encapsulation biomaterial is one of the

most important requirements in the development of encapsu-

lated cells. These biomaterials play important roles, providing

the three-dimensional and synthetic extracellular environments

that mimic certain beneficial properties of the extracellular

matrix (ECM) and improving immune protection by isolating

entrapped cells from the host tissue.[41] These materials should

have the ability to form a gel in mild and physiological con-

ditions (in terms of temperature, pH, and ionic strength). In

addition, they must be biocompatible and should not interfere

with cellular function. Although the majority of the literature

has employed sodium alginate,[42] other types of natural and

synthetic polymers, including collagen[43] and hyaluronic acid

(HA)[44] or alginate in combination with chitosan,[45] agar-

ose,[46] gelatin,[47] and tyramine,[48] have been explored in the

field of cell encapsulation.

2.1.1 Alginate and Functionalized Alginate Gels

One of the reasons alginate has been so widely employed is

that it allows gel formation in minimally harmful conditions,

which is an attractive attribute for cell encapsulation.[49,50] The

most commonmethod to gel alginate is to combine the aqueous

alginate solution with ionic crosslinking agents such as divalent

cations (i.e. Ca2+). The divalent cations are believed to bind

solely to the G blocks of the alginate chains. Thus, the G blocks

of one polymer form junctions with the G blocks of adjacent

polymer chains in what is termed the egg-box model of cross-

linking[51] (figure 2). However, Donati et al.[52] have reported

similar characteristics of the mixed junctions formed between

alternating M and G blocks (MG blocks). Other studies have

shown that Ba2+ can also bind to M chains.[53] Several func-

tional and mechanical properties of the alginate microcapsules,

including their biocompatibility, biodegradability, mechanical

and chemical stability, and controllable swelling properties,

depend on the alginate composition and concentration, the type

and concentration of the cations added to induce gelling and

polycations.[34,54-56]

Recently, the functionalization and modification of algi-

nates with various peptides and proteins to provide control over

the cell fate is gaining significant attention. These functional

groups activate the intracellular signaling cascades through the

focal contacts that provide tight control over the cell-matrix

interactions.[57] The Arg-Gly-Asp (RGD) peptide sequence

derived from fibronectin is a natural protein sequence present

in the ECM.[58] Alginate itself does not support cell attach-

ment[59] but it is easily modified by the simple coupling of cell

Ca2+ ( )

Fig. 2. Alginate hydrogels prepared by ionic crosslinking (egg-box model).

Only guluronate blocks participate in the formation of a corrugated egg-box-

like structure with interstices in which calcium ions are placed. Reproduced

from Lee and Mooney (ª 2011),[42] with permission from Elsevier.

A Perspective on Bioactive Cell Microencapsulation 285

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 4: A Perspective on Bioactive Cell Microencapsulation

adhesion domains such as the RGD via carbodiimide chem-

istry. As a result of such a coupling, the interaction between

the alginate and the integrin receptors of the enclosed cells

is improved, enhancing cell survival and functionality.[60-64]

Alginate gels have also been functionalized with the Asp-Gly-

Glu-Ala (DGEA) and Tyr-Ile-Gly-Ser-Arg (YIGSR) sequences

derived from other ECM proteins to enhance the gels’ adhesive

interactions with various cell types.[65,66] In a recent study,

Mazzitelli et al.[67] have developed alginate-based micro-

capsules containing the ECM (isolated and purified from the

urinary bladder [UBM]) and Sertoli cells (SCs). The combina-

tion of alginate and the ECM resulted in a synergistic activity of

both materials. The microcapsules offer the mechanical and

material properties of the alginates, which can be controlled

with the consideration of some issues; at the same time, the

ECM increases the bioactivity of the microcapsules to modu-

late and improve the viability and function of the encapsulated

cells. Biomimetic microcapsules using RGD-alginate have also

been employed. These capsules performed the cell adhesion for

the enclosed cells and prolonged their long-term functionality

and drug release for more than 300 days,[68] suggesting that

these capsules promote the in vivo long-term functionality of

the enclosed cells and that the mechanical stability of the cap-

sules is improved.

Another attractive modification is focused on the control of

the biodegradation rate of the alginates under physiological

conditions. An example of such a modification is the partial

oxidation of the alginate chains so that they become sensitive to

hydrolysis.[69,70] The degree of this oxidation as well as the pH

and temperature of themedia influences the degradation rate of

the gels.[71] In a recent study, mesenchymal stem cells (MSC)

were entrapped in oxidized alginate-fibrin microbeads to ach-

ieve a rapid degradation profile and the release of the cells in an

effort to promote bone engineering.[72]

In addition, alginate may also be covalently crosslinked via

the addition of bi-functional molecules such as poly(ethylene

glycol) [PEG] or methacrylate moieties to the alginate chains.[73,74]

The selective modification of the alginate resulted in an en-

hanced covalent stabilization and an increased mechanical

strength of the alginate beads.

2.1.2 Collagen

Collagen is the main component of the ECM of the mam-

malian connective tissues. Some of the advantages it offers in-

clude its biocompatibility, abundance in nature, natural ability

to anchor cells anddegradabilitymediated bymetalloproteases. Its

gelation can be induced by changes in pH, allowing cells to be

encapsulated under mild conditions.[75,76] However, the chal-

lenges of using collagen as a material for cell immobilization

include the high cost of its purification, the natural variability

of the isolated collagen, its immunogenicity, and the variation

in its enzymatic degradation depending on the implant site.

A novel drug delivery system based on collagen-alginate com-

posite structures has been developed to prevent the cell leak-

age, maintain the cell growth, and control the protein secretion

of glial cell-derived neurotrophic factor (GDNF)-secreting

cells.[77] Collagen has been used to engineer a variety of tissues,

including skin,[78] bone,[79,80] heart valves,[81] and ligaments.[82]

2.1.3 Chitosan

Chitosan is a chitin isolated from the exoskeleton of shellfish,

and it has a structure similar to natural glycosaminoglycans. It

can be formed into hydrogels by ionic interaction[83] and

chemical crosslinking with glutaraldehyde.[84] Scaffolds made

of this material can be degraded by lysozymes. Zielinski and

Aebischer[85] encapsulated a genetically modified fibroblast cell

line in chitosan hydrogels to provide the sustained release of

nerve growth factor (NGF) and showed that the amount of

NGF secreted was sufficient to induce the differentiation of co-

cultured neural progenitor PC12 cells. Thermosensitive chito-

san has also been investigated as a material to create an aligned

cell sheet[86] and to encapsulate MSCs and disc cells.[87] In a

recent study, a bi-continuous morphology scaffold for the re-

pair of load-bearing soft tissues was designed and charac-

terized. A crosslinked chitosan hydrogel was used to distribute

the cells and enable cell growth, and an elastomer prepared

from a star-poly (e-caprolactone-co-D,L-lactide) triacrylate

was used to enhance the mechanical properties of the hydrogel.

The chondrocytes within the bi-continuous scaffolds prolif-

erated, exhibited increasedmetabolic activity, and accumulated

an ECM over a 14-day culture period.[88]

2.1.4 Agarose

Agarose is another algae-derived polysaccharide, but unlike

alginate, the formation of its gel microbeads is thermally re-

versible.[45] Its most appealing qualities include the ease of its

processing and a wide range of mechanical (e.g. matrix stiff-

ness) and structural (e.g. pore size) possibilities,[89] which are

influenced by its concentration in the gel and the cooling

rate.[90] There have been many studies involving the enclosure

of various cell types in agarose microparticles.[91-94] In a recent

study, porcine islets were encapsulated in agarose macrobeads

and transplanted into spontaneously diabetic biobreeding (BB)

rats. Normoglycemia was initially restored in all of the islet-

transplanted rats; moderate hyperglycemia developed approx-

imately 30 days after the transplantation and continued

286 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 5: A Perspective on Bioactive Cell Microencapsulation

throughout the study period. All the rats that received encap-

sulated porcine islets continued to gain weight and did not

require exogenous insulin therapy for the entire study.[94] In ad-

dition, agarose canbemodified to include cell-adhesionmoieties to

transform the polymer into a photolabile substrate that can be

controlled with biochemical cues.[95] A possible problem re-

ported in the use of this biomaterial is cellular protrusion.[96]

2.1.5 Other Polymers: Poly(N-isopropylacrylamide), Gelatin,

Poly(ethylene glycol), Hyaluronic Acid and Cellulose Sulphate

Poly(N-isopropylacrylamide) [pNIPAM] is a thermores-

ponsive and biocompatible polymer with a quick phase tran-

sition and a low critical solution temperature (approximately

32�C).[97,98] RGD-conjugated pNIPAM hydrogel microcarriers

produced using calcium alginate as a temporary mold have

been employed as cell culture substrates for enzyme-free chon-

drocyte detachment.[99] In addition to cell adhesion applica-

tions, cell encapsulation using pNIPAM particles has also been

reported. In this case, pancreatic cells were encapsulated into

pNIPAM-based microcapsules that exhibited a reversible sol-

to-gel transition at 30�C.[100] In another approach, pNIPAM

thermoresponsive hydrogels have been used to immobilize a

chondrogenic cell line, revealing that the biomaterial did not

affect the cell viability or proliferation. The results also dem-

onstrated an increase in the synthesis of glycosaminoglycans

during the culture period. This study supported the hypothesis

that thermally reversible pNIPAM hydrogel may be suitable as

an injectable cell carrier and has great potential for cartilage

repair.[101]

Gelatin is obtained by the partial hydrolysis of collagen

derived from the skin, white connective tissue, and bones of

animals. As a natural macromolecule, its major limitation is the

variability caused by its derivation from animal tissue, making

batch-to-batch reproducibility difficult. This problem could be

solved by the use of recombinant gelatins with defined molec-

ular weights.[102] The formation of gelatin cell microparticles is

based on crosslinking reactions.[103,104] Examples of gelatin

microcarriers include Spheramine�[105] and Cultispher�.

Cultispher� gelatin microcarriers (Percell Biolytica AB, Sweden)

are commercially available products and have been widely

evaluated as gelatin cell carriers. They are composed of type-A

porcine gelatin particles crosslinked to form a matrix. These

microcarriers supported the in vitro growth of variousmammalian

cells and have also been used as biodegradable scaffolds for guided

tissue regeneration and cell-based therapies in vivo.[106-108]

Other biomaterials employed for cell encapsulation in the

form of hydrogels include PEG combined with acrylates and

methacrylates, HA, and cellulose sulphate.[109-115] Despite the

fact that none have been characterized and studied as much as

alginate, we could benefit from the advantages that these bio-

materials might offer in the development of alternative cell-

based therapeutic strategies.

2.2 The Requirements of the Technology

During the past few years, important advances have been

achieved in the field of cell microencapsulation. These im-

provements in the technological properties have provided high

levels of permselectivity and structural stability that lasts

through the desired lifetime of the graft. To transition cell en-

capsulation to large-scale clinical applications, several re-

quirements must be considered.[116,117] In this section, we will

discuss the most important requirements of the cell encapsu-

lation technology with regard to alginate microcapsules.

2.2.1 Permeability

The permeability of the membrane is one of the most im-

portant requirements in the development of cell microcapsules.

As previously mentioned, this membrane must be able to con-

trol the influx rate of molecules essential for cell survival

(oxygen and nutrients) and the efflux rate of therapeutic fac-

tors, metabolites, and waste products, while completely ex-

cluding the harmful components of the host immune system

(antibodies and immune cells). Thus, the viability of the im-

mobilized cells will depend on the semipermeablemembrane.[96]

One of the factors that influences the diffusion and perme-

ability of the capsules is the rate of solute diffusion, which is

usually controlled by size-exclusion phenomena and diffusion

rate. The other factor is the membrane exclusion properties

reflected by the exclusion limit or the molecular weight cut-off

(MWCO). In APA microcapsules, using a PLL with a molec-

ular weight of 12–22 kDa, the MWCO (the size of the largest

molecule that is not essentially blocked by the semipermeable

membrane) is approximately 70 kDa. With this MWCO, the

membrane will exclude leukocytes and various immunoglobulins

such as IgM (950 kDa) and IgG (150 kDa) but will allow the

diffusion of the low molecular weight substances necessary for

cell survival (e.g. glucose [180Da], carbon dioxide [44Da] and

the secretory proteins from the cells including albumin

[66 kDa], growth factors [6–50 kDa] and insulin [6 kDa]).[116]

Table I shows the molecular weights of various enzymes, anti-

bodies, complement components, and common metabolites.

A successful method for the optimization of the permeability

of alginate beads is to modify the G and M blocks. The per-

meability of the capsules increases with increasing ratios of G

residues. This effect is directly connected to the character of the

A Perspective on Bioactive Cell Microencapsulation 287

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 6: A Perspective on Bioactive Cell Microencapsulation

long, stiff G blocks and the short, flexible MG blocks in the

high G ratio alginates.[118] In contrast, it was reported that

the reduction of the membranes’ pore size by increasing of the

alginate concentration could ultimately elicit a reduction in

the rate of diffusion.[119] Another approach to modulating

the permeability of themicrocapsule membrane is to change the

contact time with the polycation solution. Increasing the con-

tact time of the alginate beads with PLL solution to more

than 5 minutes significantly decreased the membrane perme-

ability.[120] In this case, the increased number of ionic bonds

between alginate and PLL results in an increased membrane

crosslinking density. Nevertheless, increasing the gelling time

did not change the pore size distribution and, consequently,

permeability was not altered.[15] Multilayered membranes, in

which the properties of each layer can be controlled inde-

pendently, have been developed to achieve the requirements of

permeability, biocompatibility, and suitable mechanical strength

of the capsules. Various approaches have achieved these re-

quirements through the use of alternating layers of alginate and

various combinations of polycations and polyanions[121] or the

use of three-layered agarose microcapsules.[122] The multi-

layered technique has also been used to ensure the complete

immunoisolation of the encapsulated islets through local-

ization of their biological functionalities within the PEG

capsules.[123]

It is essential to include the diffusion rate of the chosen

solute, the pore size, the pore size distribution, the chemistry of

the membrane and solutes, and the thickness of the membrane

in describing the permeability properties of a microcapsule.

However, these features are only rarely documented, making it

difficult to compare the permeability results between different

laboratories. Thus, the authors believe this topic requires fur-

ther investigation.

2.2.2 Mechanical Integrity, Stability, and Durability

The mechanical resistance of the microcapsule is usually

related to the configuration and chemical structure of the

semipermeable membrane. To bear the compression forces and

shear stresses at the implantation site, the semipermeable

membrane should provide an optimum mechanical strength as

for stiffness (resistance to deformation) and toughness (resis-

tance to fracture).[124]

The type of biomaterials employed for the formation of the

matrix and membrane, the type of gelling ion and the encap-

sulation technology are some of the main parameters that

control the mechanical resistance of the microcapsules.

There are various assays to quantify the mechanical resis-

tance, including osmotic pressure tests,[125,126] surface texture

analysis,[127] optical tweezers,[128] micropipette aspiration,[129]

atomic force microscopy (AFM),[130] magnetic bead measure-

ment techniques,[131] and microelectromechanical (MEMS)

microgrippers.[132] Nevertheless, one of the greatest difficulties

is the lack of standardization in the technology to measure the

mechanical integrity, stability, and durability of themicrocapsules.

Table I. The molecular weights of various enzymes, antibodies, complement components and common metabolites presumed to be excluded from or

permeate through alginate poly-L-lysine alginate microcapsules

Molecules presumed to be excluded

from APA microcapsules

Molecular weight (Da) Molecules presumed to permeate

through APA microcapsules

Molecular weight (Da)

Immunoglobulin M 950000 Albumin 66248

Complement C4 210000 Hemoglobin 64000

Complement C5 195000 Tumor necrosis factor 51 000

Immunoglobulin E 190000 Nerve growth factor 13 000

Complement C3 185000 Insulin 9 000

Immunoglobulin A 170000 Complement 3a 5 733

Complement C2 170000 Complement 5a 4 000

Complement C8 163000 Glucose 180

Immunoglobulin D 160000 Tyrosine 163

Immunoglobulin G 150000 Phenylalanine 147

Complement C6 110000 Glutamine 128

Complement C7 100000 Asparagine 114

Complement C9 79000 Creatinine 113

Oxygen (O2) 32

APA= alginate-poly-L-lysine-alginate.

288 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 7: A Perspective on Bioactive Cell Microencapsulation

The challenge is to determine the optimum mechanical resis-

tance for a specific application without modifying other rele-

vant capsule properties.

2.2.3 The Size, Morphology, and Surface Properties of the Capsules

The uniformity of the size and the shape ofmicrocapsules are

far-reaching characteristics because they could affect the in vivo

immune response to the microcapsules. Thus, to warrant a

correct diffusive mass transport, the overall diameter of the

microcapsules should not exceed 300–400 mm.[133] Beneficial

effects, including a better molecular exchange between the en-

closed cells and the surrounding environment as well as a higher

mechanical resistance and improved biocompatibility, could be

amended by reducing the diameter of the cell-enclosing micro-

capsules.[134-137] In fact, when the microcapsule diameter is too

large, it is difficult for the cells in the center of the core to survive

because the diffusion of oxygen and nutrients is limited.[138,139]

Other advantages of these small capsules are the ease of

their implantation into reduced sites such as the CNS[140] and

the eye.[141]

During the last few years, various technologies to produce

small particles (100–200 mm) have arisen, such as coaxial flow

techniques,[134,141,142] microfluidic technologies,[143] and a flow-

focusing system.[141] In a recent study, Santos et al.[141] devel-

oped a promising strategy to form monodisperse 100 mm APA

microcapsules using the flow-focusing system, and implanted

them successfully in the intravitreous space in vivo. After opti-

mizing all of the technical parameters required to produce

the small APA microcapsules, they showed that the cell via-

bility, cell proliferation, and vascular endothelial growth factor

soluble receptor (KDR) secretion were not affected by the

physicochemical changes arising from such a dramatic size re-

duction. Moreover, a strong signal was obtained from the

monitored encapsulated cells during the 3-week in vivo assay,

confirming the viability of the enclosed cells. The challenges

related to these small microcapsules include determining the

lowest cell load required to obtain a 100% encapsulation yield,

evaluating the changes in cell viability, and optimizing the ad-

ministration protocol to achieve the maximal dose within the

smallest administration volume.

2.2.4 Biocompatibility

Another important property that should be considered is the

ability of a biomaterial to result in an appropriate host response

in a specific application. This requirement is related to both the

interaction between the biomaterials and the host system and

the interaction between the biomaterial and the encapsulated

cells.[144] The biocompatibility of microcapsules is a complex

property that involves many characteristics, including material

composition, structure, morphology, degradation, and me-

chanical properties.[50,145,146]

In recent years, various assays have been proposed to

evaluate biocompatibility.[147-150] In a recent study, de Haan

et al.[150] studied the physicochemical changes caused by the

exposure of alginate beads and alginate-PLL capsules prepared

from various types of alginate to human peritoneal fluid by

applying micro-Fourier transform infrared spectroscopy. They

observed the adsorption of components from human peritoneal

fluid and physicochemical changes to the surface that are de-

pendent on the chemical composition of the capsules. Other

studies have reported that the use of intermediate-G alginate in

combination with calcium for the gelation process and PLO as

the polycation resulted in less immune cell adhesion. The period

of membrane formation, during which the polycation diffuses

and binds the alginate gel, governs the biocompatibility of

microcapsules and their physicochemical properties.[34,151] To

prevent and reduce an inflammatory response, some studies

have co-administered anti-inflammatory drugs along with the

encapsulated cells.[20,152-155] In a recent study, Murua et al.[153]

subcutaneously implanted a composite delivery system con-

sisting of erythropoietin (Epo)-secreting myoblasts immobi-

lized in APA microcapsules and dexamethasone-releasing

poly(lactic-co-glycolic acid) [PLGA] microspheres (figure 3).

This composite system enhanced the biocompatibility and

generated an immunoprivileged local environment to limit the

fibrotic layer caused by inflammation. Nevertheless, the major

challenge is to design standardized protocols that facilitate the

identification of alginate properties and the evaluation of bio-

compatibility; this will establish the criteria for alginate se-

lection and purification and thus result in reproducible results

between research groups.

2.2.5 Other Issues

The protection from the immune response that APA mi-

crocapsules provide to the encapsulated cells has allowed the

use of xenogeneic cells from non-human sources. The choice of

cells depends on the intended therapeutic application, such as

the secretion of a naturally bioactive substance (e.g. neuro-

transmitter, cytokine, growth factor, growth factor inhibitor,

or angiogenic factor), the metabolism of a toxic agent, or the

release of an immunizing agent.[3]

Tracking and monitoring the cell-containing microcapsules

in vivo is another requirement that needs to be resolved before

these products can reach large-scale clinical trials. Towards this

aim, various research groups have used non-invasive imaging

techniques, including X-ray,[156] magnetic resonance,[156,157]

A Perspective on Bioactive Cell Microencapsulation 289

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 8: A Perspective on Bioactive Cell Microencapsulation

and bioluminescent imaging.[158] In a recent study, Catena

et al.[158] incorporated the triple reporter gene TGL into

microencapsulated cells to control the location of the cells

and their viability by luminometry. They were able to interrupt

the therapy by the administration of ganciclovir. This system

may represent an appropriate tool to tightly control micro-

encapsulated cells and, moreover, to improve the biosafety of

this type of drug delivery system.

3. Therapeutic Applications and Clinical Trials

Finally, we will describe the most recent therapeutic appli-

cations and clinical trials to provide the readers with a general

overview of the current research area.

3.1 Diabetes

Diabetes mellitus is a metabolic disorder characterized by

hyperglycemia resulting from defects in insulin secretion, in-

sulin action, or both. A possible approach for the safe and

effective cure of these patients is the replacement of the dam-

aged islets of Langerhans.[159] However, the restricted avail-

ability of cadaveric human donor pancreases, coupled with the

need for life-long, general pharmacological immunosuppression,

makes the application of this therapy difficult. The many

contributions of these studies are shedding light on the main

challenges of the encapsulation technology.[160-166] For exam-

ple, an analysis of the global gene (mRNA) and selected

microRNA expression profiles was performed on the islets

derived from a human cadaver after decapsulation of the

cells. The study demonstrated that the tissue culture and

encapsulation process did not affect the viability of the cells.

Although some genes involved in the inflammatory response

were up-regulated, protein expression was not altered by the

encapsulation.[167]

In a recent study, SCs entrapped in alginate-based micro-

capsules induced the reversal of spontaneous diabetes by cre-

ating newly formed functional islets b-cells in the treated

non-obese diabetic mice.[168] Other researchers demonstrated

that the survival of the islet cell aggregates within the micro-

capsules in low-oxygen culture and during transplantation was

superior to that of the intact islets.[169] Opara et al.[170] have

described the new design of a bioartificial pancreas comprising

islets co-encapsulated with angiogenic proteins in permselec-

tive multilayer alginate microcapsules. In this study, the inner

alginate layer encapsulates the islets and the outer layer en-

capsulates the angiogenic proteins, which would otherwise in-

duce neovascularization around the graft within the omentum

pouch. Currently, stem cells from various sources and differ-

entiated into insulin-producing cells (IPCs) are also employed

to treat diabetes.[163,171,172]

3.2 Central Nervous System Diseases

The use of cell-loaded capsules implanted into the damaged

brain area is an interesting approach to the treatment of

CNS diseases requiring chronic administration of therapeutic

products.

Parkinson’s disease is a neurodegenerative disorder charac-

terized by the extensive loss of the dopamine neurons of

the substantia nigra pars compacta and their terminals in the

striatum. Several studies have been carried out using various

types of cells encapsulated in alginate microcapsules in animal

models of Parkinson’s disease; the most widely employed growth

factors in the treatment of this disease are GDNF[173-176] and

vascular endothelial growth factor (VEGF).[177-179] In one of

these studies, GDNF-secreting fibroblasts were immobilized in

Subcutaneous space: immunopriviledged environment

APA microcapsules embedding Epo-secreting C2C12 myoblasts

Dexamethasone-loaded PLGA microspheres

Fat tissue

Fig. 3. Schematic illustration of the immunomodulatory environment

created in the subcutaneous space of implanted mice. Reproduced from

Murua et al. (ª 2011),[153] with permission from Elsevier. APA =alginate-poly-L-lysine-alginate; PLGA =poly(lactic-co-glycolic acid).

290 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 9: A Perspective on Bioactive Cell Microencapsulation

APA microcapsules and implanted into the striatum of

6-hydroxydopamine (6-OHDA) lesioned rats. These micro-

capsules were able to deliver continuous and reliable levels of

GDNF for at least 6months, resulting in substantial behavioral

improvement and good biotolerance.[174] In general, most of

these studies conclude that GDNF and VEGF are more effec-

tive when administered before the induction of the lesion,

thereby exhibiting more of a neuroprotective than a neuro-

rescue effect. In a recent study by Skinner et al.,[13] neonatal

porcine choroid plexus (CP) cells were encapsulated within

alginate-PLO capsules and implanted into the striatum of

6-OHDA lesioned rats. They have shown that the neuro-

restorative proteins delivered by the encapsulated cells improve

the motor behavior and the nigrostriatal dopaminergic activity

of the lesioned rats.

The co-implantation of cells that induced functional recov-

ery and regeneration of the lesioned area combined with en-

capsulated cells that released various types of growth factors

with the aim of improving and increasing the survival of

transplanted, unencapsulated cells after transplantation has

also been studied.[180-184] Shanbhag et al.[184] developed a novel

alginate construct that acts as a multifunctional tissue scaffold

for CNS repair and as a localized growth factor delivery system.

In this alginate construct, they encapsulated genetically modi-

fied brain-derived neurotrophic factor (BDNF) or neuro-

trophin-3 (NT-3)-secreting fibroblasts, and neural progenitor

cell (NPCs) were seeded on the surface of the system. They

showed that the surface of the alginate construct acted as an

optimal growth environment for theNPC attachment, survival,

migration, and differentiation, and the continuous delivery of

BDNF or NT-3 directly influenced the lineage fates of the

NPCs.

Another interesting therapeutic application employing the

immobilization of genetically engineered cells to produce

growth factors is Alzheimer’s disease (AD).[140,185,186] AD is a

progressive neurodegenerative disorder of the elderly, charac-

terized by elevated levels of amyloid b-peptide (Ab) in the brain

and associated with neuronal and vascular toxicity; it is the

most prevalent form of dementia. Recently, Spuch et al.[140]

implanted microencapsulated fibroblast cells releasing VEGF

into a transgenic (APP/PS1) mouse model of AD with degen-

erative changes in the microvasculature. The study reported

an improvement in brain Ab removal and protection of the

cognitive behavior in the APP/PS1 mice after implantation

of themicrocapsules. In addition, they confirmed the promising

therapeutic advantages of VEGF therapy for brain angiogen-

esis, neuroprotection, and the cerebromicrovascular exchange

of substrates and nutrients; the implantation of VEGF-secreting

cells entrapped in microcapsules was presented as a therapeutic

tool for the treatment and prevention of brain amyloidosis.

Other similar approaches include the use of recombinant cells

secreting ciliary neurotrophic factor (CNTF) encapsulated

in alginate microcapsules that were implanted into the brain

of two different AD mouse models: the Ab oligomer-infused

and Tg2576 models. It was reported that the CNTF stimulated

a Janus kinase/signal transducer and activated a transcription-

mediated survival pathway that prevented synaptic and neu-

ronal degeneration; an improvement in both animal models

was observed after the implantation of these alginate micro-

capsules. In the Ab oligomer-infused murine model, a robust

improvement in the cognitive behavior caused by a continu-

ous secretion of the recombinant CNTF was demonstrated.

The results were more successful in the Tg2576 AD mouse

model, where a full recovery of the cognitive functions asso-

ciated with the stabilization of the synaptic protein levels was

observed.[185]

Huntington’s disease (HD) is another important neuro-

degenerative disorder to take into consideration. This disease

is characterized by the preferential loss of the striatal GABA-

ergic medium spiny neurons and includes an array of different

psychiatric manifestations, cognitive decline, and choreiform

movements. The CP epithelial cells secrete several neurotrophic

factors affecting the production of the cerebrospinal fluid

and the maintenance of the extracellular fluid concentra-

tions in the brain.[187] There have been various studies that

employ immobilized CP cells in alginate microcapsules to

achieve an appropriate release of neurotrophic factors into

the brain in rodent[13,14,188,189] and primate[190] models of HD.

These studies reported that the CP cells encapsulated in the

alginate microcapsules prevented the degeneration of the

striatal neurons. Baby hamster kidney (BHK) cells engineered

to produce human CNTF and immobilized into semipermeable

membranes have also been employed in the treatment of

HD with positive results, including protection of the neu-

rons from degeneration and restoration of the neostriatal

functions.[191]

Spinal cord injury (SCI) results in the disruption of the as-

cending and descending axons and subsequently produces a

devastating loss of motor and sensory function. In this regard,

BDNF-producing fibroblasts entrapped in alginate micro-

capsules were implanted in a murine model of SCI.[192,193] En-

capsulated MSCs have also been employed in the treatment of

the secondary inflammatory responses in the SCI animal

models.[194] The in vitro and in vivo results obtained in this study

demonstrated that the MSC-loaded microcapsules may over-

come many of the disadvantages that occur with the administra-

A Perspective on Bioactive Cell Microencapsulation 291

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 10: A Perspective on Bioactive Cell Microencapsulation

tion of the unencapsulated MSC (distribution to non-targeted

tissues after transplantation and retention at a low rate). At the

same time, encapsulation may serve to augment the tissue-

protective behavior of theMSC caused by the elimination of the

proinflammatory macrophages that is stimulated by the cell-

loaded microcapsules at the site of injury.[194]

The clinical trials performed for the previously mentioned

diseases and some others are summarized in table II.

Table II. Clinical trials already carried out or ongoing in the field of cell encapsulation

Therapeutic application Therapeutic agent Type of capsule or material Results References

Type 1 diabetes Encapsulated islets, low

dose immunosuppression

Alginate-PLL 9 months of insulin independence 195

Encapsulated islets, no

immunosuppression

Alginate-PLO-alginate Insulin consumption declined at 6–12 months

(2 patients)

196,197

Diabecell� device (living cell

technologies) encapsulated

neonatal porcine islets

Alginate 9.5 years’ improvement in glycemic control

(1 patient)

198

Little improvement in hypoglycemia

unawareness and a reduction in the exogenous

insulin schedule (12 patients)

199

Encapsulated islets, low

dose immunosuppression

Alginate Neither insulin requirements nor glycemic

control was affected (4 patients)

200

Monolayer islet device Alginate-monolayer

(1–3 cm2)

Recruiting patients, started in 2008

(NCT00790257)

201

Encapsulated beta cells Alginate Recruiting patients, started in 2011

(NCT01379729)

201

Viacyte (Novocell�) Conformal PEG coating Phase I/II study, concluded in 2011

(NCT00260234)

201

Parkinson’s disease Spheramine�. hRPE cells

implants

Gelatin microcarriers Safe and well tolerated implants and

improvement in motor symptoms

202

Ongoing phase II study, started in 2000

(NCT00761436)

201

Ongoing phase II study, started in 2003

(NCT00206687)

201

Alzheimer’s disease NsG0202 implants

(EC delivery/NsGene),

encapsulated NGF-

secreting cells

Hollow fiber Ongoing phase I study, started in 2008

(NCT01163825)

201

Huntington’s disease BHK cells that secrete CNTF Hollow fiber 2-year study (device changed every 6 months);

variability in CNTF levels (6 patients)

203

Amyotrophic lateral sclerosis BHK cells that secrete CNTF Hollow fiber 17 weeks post-implantation CNTF detected

in the CSF (6 patients)

204

Pancreatic carcinoma Encapsulated cells that

secrete the enzyme

cytochrome P450

Cellulose sulphate Tumor regression or stabilization,

no tumor growth

205,206

Hypoparathyroidism Encapsulated parathyroid

tissue

Alginate Calcium and vitamin D replacement reduced

(2 patients)

207

Retinitis pigmentosa Neurotech (NT-501) Hollow fibers Phase II study, no visual benefit 208,209

Intracerebral hemorrhage GLP-1 CellBeads,�.

encapsulated mesenchymal

cells that secrete GLP-1

Alginate Ongoing phase I/II study, started in 2008

(NCT01298830)

201

BHK cells= baby hamster kidney cells; CNTF= ciliary neurotrophic factor;CSF= cerebrospinal fluid; EC = encapsulated cell;GLP-1= glucagon-like peptide 1;

hRPE cells= human retinal pigment epithelial cells; NGF= nerve growth factor; PEG=poly(ethylene glycol); PLL= poly-L-lysine; PLO=poly-L-ornithine.

292 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 11: A Perspective on Bioactive Cell Microencapsulation

3.3 Cardiovascular Diseases

Cardiovascular diseases (CVDs) are the major causes

of morbidity and mortality worldwide. CVDs include myo-

cardial ischemia with the subsequent extended dysfunction

and death of the cardiomyocytes. After a myocardial infarc-

tion (MI), the heart attempts to restore its damaged tissue by

implementing various physiological and compensatory pro-

cesses, including cardiomyocyte hypertrophy, apoptotic myocyte

loss, progressive collagen replacement, and an enlargement of

the left ventricle.[210] Conventional medical management can im-

prove post-MI mortality, relieving symptoms and slowing de-

terioration, but it cannot restore cardiac function. This

treatment merely accommodates the damaged myocardium

rather than effecting direct repair to restore its normal structure

and function.[211] Thus, new therapeutic approaches for MI are

required.

Cell therapy is a novel and promising therapeutic approach

to sustain the endogenous regenerative mechanisms in ischemic

heart disease and heart failure; it is based on the transplantation

of human or animal cells to replace or repair damaged tissues or

cells.[212] Cell transplantation provides an effective and renew-

able source of proliferating and functional cardiomyocytes and

supports the development of a blood vessel network that sus-

tains and nourishes these newly formed cardiomyocytes and

their surrounding ischemic myocardium.[213] Toward this

aim, stem cell transplantation is being investigated as a novel

means to regenerate the heart tissue and enhance cardiac

function.[212,214] Several animal studies have demonstrated that

the administration of various types of adult stem cells using

various delivery routes can have therapeutic benefits, decreas-

ing cardiac infarction size by promoting angiogenesis and

myogenesis and effectively improving the left ventricular and

cardiac function.[215-222] However, there is an important draw-

back to myocardial cell therapy that is related to the low rate of

engraftment and survival of these transplanted cells. The heart

is constantly contracting, and this contributes to the mechan-

ical loss of the injected cells because they are squeezed out of the

myocardium. Large proportions of the injected cells are lost

from the myocardium within the first few minutes after their

injection.[223,224] Not more than 0.1–15% of all the injected cells

are retained within the myocardium.[211,225] To overcome the

massive loss of the cells and improve cell retention and survival,

diverse approaches are being studied.

One outstanding strategy could be the immobilization of the

cells within microcapsules. Microcapsules are larger than the

blood vessel diameter; thus, the heart’s contractile forces are

unable to eliminate the microcapsules into the bloodstream,

significantly increasing the amount of retained microcapsules

and the amount of cells.[225] In a recent study, Al Kindi et al.[17]

reasserted that the size of the microcapsules is an important

factor in the early loss of cells because encapsulated micro-

spheres in APAmicrocapsules (200–400 mm) exhibited a higher

retention rate than small microspheres (100 mm). In addition,

they showed that myocardial function did not improve with

escalating cell doses, showing that the optimal functional im-

provement began at 1.5 · 106 cells.The microencapsulated cells may exert a beneficial influence

on myocardial regeneration by means of a paracrine growth

factor effect. VEGF-secreting xenogeneic Chinese hamster

ovary (CHO) cells were encapsulated in APA microcapsules

and implanted into the infarcted myocardium of rats. This

study reported an improvement in global heart function and

enhanced angiogenesis.[226] In a similar approach, Yu et al.[227]

encapsulatedMSCs in RGD-modified alginate and covered the

droplets with PLL. They reported an improvement in cell at-

tachment, cell growth, and an increase in the expression of

angiogenic growth factor as well as the successful maintenance

of the shape of the left ventricle, the induction of angiogenesis,

and the prevention of the left ventricle remodeling, including

infarct wall thinning and chamber dilation after an MI. These

results suggest that these microencapsulated cells may have a

much greater potential for heart regeneration compared with

the unencapsulated stem cells. Another interesting and prom-

ising approach is the production of heart tissue grafts, in which

alginate and collagen have been combined to encapsulate neo-

natal rat cardiomyocytes. Cardiac cell proliferation and the

formation of interconnected multilayer heart-like tissue, as well

as the spontaneous synchronized contractility of the engineered

heart tissue grafts, were demonstrated.[228]

3.4 Liver Diseases

The liver is a central and vital organ. It performs a wide

range of fundamental metabolic functions, including the de-

toxification and elimination of endogenous wastes (such as

ammonia or bilirubin), drugs, and exogenous toxins, the syn-

thesis of amino acids and proteins, the metabolism and ab-

sorption of carbohydrates and lipids, and the storage and the

delivery of nutrients according to the requirements of the body.

Acute liver failure and chronic liver failure are diseases related

to the inability of the liver to perform its normal synthetic and

metabolic functions. Although liver dialysis can be used in the

short term, the only way to compensate for the absence of the

liver function in the long term is with an organ transplantation.

Several research groups have employed cell microencapsulation

A Perspective on Bioactive Cell Microencapsulation 293

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 12: A Perspective on Bioactive Cell Microencapsulation

technology to create a living cell-based replacement system or a

bioartificial liver to compensate for essential functions. There

have been various in vivo studies of this technology performed

inmany animalmodels, includingGunn rats,[229,230] mice,[12,231-233]

and pigs.[234] These studies generally reported enhanced re-

covery and support and improvement in hepatic metabolic

functions; the technology was presented as a potential alter-

native to transplantation for liver failure. In one of these

studies, immortalized human hepatocytes encapsulated in APA

microcapsules were transplanted intraperitoneally into a mice

model of acute liver failure to evaluate the effects on liver

metabolism and regeneration. This group reported an im-

provement in the survival and biochemical profile, including

lower levels of the cytokines (tumor necrosis factor [TNF]-a,interleukin [IL]-6, hepatocyte growth factor [HGF] and trans-

forming growth factor [TGF]-b1) in the serum of mice trans-

planted with hepatocytes compared with the mice receiving

empty capsules. These lower levels of cytokines could be ex-

plained by an increase in the clearance of these cytokines by

the encapsulated hepatocytes. However, the encapsulated

hepatocytes transplanted into the intraperitoneum did not

stimulate native liver regeneration.[231] Another possible ther-

apy involves the co-encapsulation of hepatocytes with other

types of cells that may maintain the specific function and phe-

notype of the encapsulated hepatocytes.[235,236] Shi et al.[237]

reported that rat hepatocytes and MSCs co-encapsulated in

APA microcapsules improved hepatocyte-specific functions

(albumin secretion and urea synthesis). In vivo studies in a

rat model of acute liver failure demonstrated an increase in

the liver’s survival rate and an improvement in its functions.

In addition, some MSCs transdifferentiated into hepatocyte-

like cells in vivo and expressed albumin, a typical marker for

hepatocytes.

An important issue in the transplantation of cells into the

liver is the evaluation of the mass transfer velocity between the

fluid and the encapsulated hepatocytes as well as the biological

functions of cells. Toward this aim, fluidized bed systems are

being investigated.[234,238-241] These devices provide a suitable

and valuable option to improve nutrient and oxygen transfers,

facilitating the inflow and transformation of lethal toxins as

well as the release of essential proteins from the cell-containing

beads (figure 4).

3.5 Other Diseases

Epo has been widely used in the treatment of the anemia

associated with various chronic conditions, including end-stage

renal disease, malignancy, and human immunodeficiency virus

(HIV) infection.[242] With consideration of that fact, our re-

search group has carried out a careful selection of biomate-

rials and cell lines to produce uniform and biocompatible

microcapsules with long-term functionality. C2C12 myoblasts

genetically engineered to secrete Epo have been successfully en-

trapped in APAmicrocapsules and implanted in allogeneic and

C3A

entrapped into alginate bead

Nutrients

Oxygen

Metabolites

Bioreactor

Red blood cells

Plasma

Plasmadetoxificated

Peristaltic pump

Patientwith

hepaticfailure

Plasmapheresis

a

b

Fig. 4. Principle of the fluidized-bed bioartificial liver for liver support (a). The patient’s blood is withdrawn and separated into plasma and blood cells. Plasma is

perfused through a fluidized-bed bioreactor hosting hepatocytes in order to be detoxified. Plasma and blood cells are mixed again and returned to the patient.

Transfer of nutrients, oxygen and metabolites occurs through the alginate bead structure (b). Reproduced from Gautier et al. (ª 2011),[238] with permission.

294 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 13: A Perspective on Bioactive Cell Microencapsulation

syngeneic mice[35,243,244] as well as in xenogeneic rats using

transient immunosuppression.[9] In a recent in vivo study carried

out in our laboratory, with the aim of generating an im-

munoprivileged local environment to avoid or at least diminish

the fibrotic layer caused by inflammation, we have developed a

composite delivery system that consists of Epo-secreting myo-

blasts immobilized in APAmicrocapsules and dexamethasone -

releasing PLGAmicrospheres.[153] All of these studies included

a single dose of microcapsules, which achieved high and con-

stant hematocrit levels.

The use of microcapsules containing cells that secrete vari-

ous therapeutic products has also been proposed for the treat-

ment of cancer. Some of these therapeutic products are cytokines

that enhance immunity and decrease tumorigenicity.[245] Others

are antiangiogenic proteins (endostatin or hemopexin) that

inhibit the release of VEGF and, thus, the growth and devel-

opment of blood vessels characteristic of and necessary to tu-

mors.[246-250] Others have employed a combined delivery of

the cytokines and antiangiogenic proteins.[251] In a recent

study, Dubrot et al.[252] encapsulated hybridoma cells that se-

crete immunostimulatory antibodies (anti-CD137 and anti-

OX40 mAb) and implanted them in mice with induced tumors.

The immunostimulatory monoclonal antibodies secreted by

the implanted encapsulated hybridoma cells enhance tumor-

specific cellular immunity.

Pelegrin et al.[253] employed cell encapsulation technology

for the treatment of viral diseases. Toward this aim, they im-

mobilized cells secreting an ectopic monoclonal antibody that

neutralizes the FrCasE retrovirus; neutralization prevents the

lethal neurodegeneration caused by the virus in infected new-

born mice. The treated mice showed reduced or undetectable

viremia as well as a lack of the histopathological lesions char-

acteristic of this disease.

Other approaches have recently been proposed by Piller

et al.[254] for the treatment of mucopolysaccharidosis type I

(MPSI). A murine myoblast cell line engineered to overexpress

the enzyme a-L-iduronidase (IDUA) was immobilized in algi-

nate microcapsules and implanted in a MPSI mouse model.

This resulted in a significant induction of enzyme activity,

a reduction in the accumulation of glycosaminoglycan and

a complete normalization of the deposits.

Finally, the treatment of deafness is being studied using

encapsulation technology. The effects of encapsulated BDNF-

expressing Schwann cells on the survival of the auditory neu-

rons in the deaf guinea pig were assessed. The study reported

that the delivery of the neurotrophic factors reduced or pre-

vented the auditory neuron degeneration that causes sensor-

ineural hearing loss.[255]

4. Concluding Remarks and Future Directions

and Challenges

Although several advances have been observed in cell micro-

encapsulation technology for the treatment of many significant

diseases, there are still major challenges to the technology.

These challenges include determining the effects of the encap-

sulation process, determining the appropriate deployment lo-

cation, achieving biocompatibility, increasing immunoprotection,

eliminating hypoxia, and preventing pericapsular fibrotic

overgrowth and post-transplant inflammation. These obstacles

must be resolved for the use of immunoprotective capsules in

clinical applications. Modifications to the microcapsule surface to

prevent fibrotic overgrowth, the application of genetic engineering,

co-encapsulation, an improvement in the oxygen supply or the

establishment of hypoxia resistance as well as other methods are

novel approaches to overcoming the obstacles mentioned above.

A continuing challenge in drug delivery applications is

achieving precise control over the delivery of single or multiple

drugs and sustained or sequential release in response to external

environmental changes, such as mechanical signals. Dynamic

control over the delivery can improve the safety and effective-

ness of the drugs and may provide new therapies.

Thus, the standardization and definition of the protocols for

the assay of the specific requirements of the technology must be

mandated to understand lab-to-lab variations and achieve ad-

equate reproducibility. This ambitious goal requires a multi-

disciplinary approach between scientists and researchers from the

biomedical, physical, and chemical fields as well as other fields.

Acknowledgments

The authors gratefully acknowledge the support to research cell micro-

encapsulation from the ‘‘Ministerio de Ciencia e Innovacion’’, University of

the Basque Country UPV/EHU (UFI11/32) and FEDER funds (SAF2008-

03157). A. Acarregui thanks the ‘‘Gobierno Vasco (Departamento de Edu-

cacion, Universidades e Investigacion)’’ for the Ph.D. fellowship.

In Memoriam

We would like to thank the contribution of Ainhoa Murua for her

extraordinary knowledge and capacity to foster scientific research. The

greatest tribute to her will be our continued promotion of the highest

scientific and educational work. She, who has touched the life of many

colleagues and PhD students, will remain forever alive.

References1. Balmayor ER, Azevedo HS, Reis RL. Controlled delivery systems: from

pharmaceuticals to cells and genes. Pharm Res 2011; 28: 1241-58

2. Nafea EH, Poole-WarrenAM,Martens PJ. Immunoisolating semi-permeable

membranes for cell encapsulation: focus on hydrogels. J Control Release

2011; 154: 110-22

A Perspective on Bioactive Cell Microencapsulation 295

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 14: A Perspective on Bioactive Cell Microencapsulation

3. Hernandez RM, Orive G, Murua A, et al. Microcapsules and microcarriers

for in situ cell delivery. Adv Drug Deliv Rev 2010; 62: 711-30

4. Hunt NC, Grover LM. Cell encapsulation using biopolymer gels for re-

generative medicine. Biotechnol Lett 2010; 32: 733-42

5. Chang TM. Therapeutic applications of polymeric artificial cells. Nat Rev

Drug Discov 2005; 4: 221-35

6. Moskalenko V, Ulrichs K, Kerscher A, et al. Preoperative evaluation of mi-

croencapsulated human parathyroid tissue aids selection of the optimal bio-

artificial graft for human parathyroid allotransplantation. Transpl Int 2007;

20: 688-96

7. Wen J, Vargas AG, Ofosu FA, et al. Sustained and therapeutic levels of

human factor IX in hemophilia B mice implanted with microcapsules: key

role of encapsulated cells. J Gene Med 2006; 8: 362-9

8. Wen J, Xu N, Li A, et al. Encapsulated human primary myoblasts deliver

functional hFIX in hemophilic mice. J Gene Med 2007; 9: 1002-10

9. Murua A, Orive G, Hernandez RM, et al. Xenogeneic transplantation of

erythropoietin-secreting cells immobilized in microcapsules using transient

immunosuppression. J Control Release 2009; 137: 174-8

10. Chang PL, Van Raamsdonk JM, Hortelano G, et al. The in vivo delivery

of heterologous proteins by microencapsulated recombinant cells. Trends

Biotechnol 1999; 17: 78-83

11. Cheng WT, Chen BC, Chiou ST, et al. Use of nonautologous micro-

encapsulated fibroblasts in growth hormone gene therapy to improve growth

of midget swine. Hum Gene Ther 1998; 9: 1995-2003

12. Mei J, Sgroi A, Mai G, et al. Improved survival of fulminant liver failure by

transplantation of microencapsulated cryopreserved porcine hepatocytes in

mice. Cell Transplant 2009; 18: 101-10

13. Skinner SJ, GeaneyMS, LinH, et al. Encapsulated living choroid plexus cells:

potential long-term treatments for central nervous system disease and

trauma. J Neural Eng 2009; 6: 065001

14. Borlongan CV, Thanos CG, Skinner SJ, et al. Transplants of encapsulated rat

choroid plexus cells exert neuroprotection in a rodentmodel of Huntington’s

disease. Cell Transplant 2008; 16: 987-92

15. Vaithilingam V, Kollarikova G, Qi M, et al. Effect of prolonged gelling time

on the intrinsic properties of barium alginate microcapsules and its bio-

compatibility. J Microencapsul 2011; 28: 499-507

16. Salmons B, Brandtner EM, Hettrich K, et al. Encapsulated cells to focus

the metabolic activation of anticancer drugs. Curr Opin Mol Ther 2010; 12:

450-60

17. Al Kindi AH, Asenjo JF, Ge Y, et al. Microencapsulation to reduce me-

chanical loss of microspheres: implications in myocardial cell therapy. Eur J

Cardiothorac Surg 2011; 39: 241-7

18. Murua A, Portero A, Orive G, et al. Cell microencapsulation technology:

towards clinical application. J Control Release 2008; 132: 76-83

19. Qi M, Strand BL, Morch Y, et al. Encapsulation of human islets in novel

inhomogeneous alginate-ca2+/ba2+ microbeads: in vitro and in vivo func-

tion. Artif Cells Blood Substit Immobil Biotechnol 2008; 36: 403-20

20. Giovagnoli S, Blasi P, Luca G, et al. Bioactive long-term release from

biodegradable microspheres preserves implanted ALG-PLO-ALG

microcapsules from in vivo response to purified alginate. Pharm Res 2010;

27: 285-95

21. Murua A, Orive G, Hernandez RM, et al. Cryopreservation based on freezing

protocols for the long-term storage of microencapsulated myoblasts. Bio-

materials 2009; 30: 3495-501

22. Lim F, Sun AM.Microencapsulated islets as bioartificial endocrine pancreas.

Science 1980; 210: 908-10

23. Paul A, Cantor A, Shum-Tim D, et al. Superior cell delivery features of

genipin crosslinked polymeric microcapsules: preparation, in vitro charac-

terization and pro-angiogenic applications using human adipose stem cells.

Mol Biotechnol 2011; 48: 116-27

24. Marsich E, Borgogna M, Donati I, et al. Alginate/lactose-modified chitosan

hydrogels: a bioactive biomaterial for chondrocyte encapsulation. J Biomed

Mater Res A 2008; 84: 364-76

25. Donati I, Haug IJ, Scarpa T, et al. Synergistic effects in semidilute mixed

solutions of alginate and lactose-modified chitosan (chitlac). Biomacro-

molecules 2007; 8: 957-62

26. Orive G, Bartkowiak A, Lisiecki S, et al. Biocompatible oligochitosans as

cationic modifiers of alginate/Camicrocapsules. J BiomedMaterRes BAppl

Biomater 2005; 74: 429-39

27. De Castro M, Orive G, Hernandez RM, et al. Biocompatibility and in vivo

evaluation of oligochitosans as cationic modifiers of alginate/Ca micro-

capsules. J Biomed Mater Res A 2009; 91: 1119-30

28. Rokstad AM, Brekke OL, Steinkjer B, et al. Alginate microbeads are

complement compatible, in contrast to polycation containingmicrocapsules,

as revealed in a human whole blood model. Acta Biomater 2011; 7: 2566-78

29. Dandoy P, Meunier CF, Michiels C, et al. Hybrid shell engineering of animal

cells for immune protections and regulation of drug delivery: towards the

design of ‘‘artificial organs’’. PLoS One 2011; 6: e20983

30. Leung A, Trau M, Nielsen LK. Assembly of multilayer PSS/PAH membrane

on coherent alginate/PLOmicrocapsule for long-term graft transplantation.

J Biomed Mater Res A 2009; 88: 226-37

31. Zhi ZL, Liu B, Jones PM, et al. Polysaccharide multilayer nanoencapsulation

of insulin-producing beta-cells grown as pseudoislets for potential cellular

delivery of insulin. Biomacromolecules 2010; 11: 610-6

32. Kunze A, Giugliano M, Valero A, et al. Micropatterning neural cell cultures

in 3D with a multi-layered scaffold. Biomaterials 2011; 32: 2088-98

33. Ponce S, Orive G, Hernandez R, et al. Chemistry and the biological response

against immunoisolating alginate-polycation capsules of different compo-

sition. Biomaterials 2006; 27: 4831-9

34. TamSK, Bilodeau S,Dusseault J, et al. Biocompatibility and physicochemical

characteristics of alginate-polycation microcapsules. Acta Biomater 2011; 7:

1683-92

35. Murua A, de Castro M, Orive G, et al. In vitro characterization and in vivo

functionality of erythropoietin-secreting cells immobilized in alginate-poly-

L-lysine-alginate microcapsules. Biomacromolecules 2007; 8: 3302-7

36. Calafiore R. Alginate microcapsules for pancreatic islet cell graft immuno-

protection: struggle and progress towards the final cure for type 1 diabetes

mellitus. Expert Opin Biol Ther 2003; 3: 201-5

37. Franco CL, Price J, West JL. Development and optimization of a dual-

photoinitiator, emulsion-based technique for rapid generation of cell-laden

hydrogel microspheres. Acta Biomater 2011; 7: 3267-76

38. Ji XH, ChengW,Guo F, et al. On-demand preparation of quantum dot-encoded

microparticles using a droplet microfluidic system. Lab Chip 2011; 11: 2561-8

39. Domachuk P, Tsioris K, Omenetto FG, et al. Bio-microfluidics: biomaterials

and biomimetic designs. Adv Mater 2010; 22: 249-60

40. Rosellini E, Vozzi G, Barbani N, et al. Three-dimensional microfabricated

scaffolds with cardiac extracellular matrix-like architecture. Int J Artif

Organs 2010; 33: 885-94

41. Ye Z,Mahato RI. Emerging trends in cell-based therapies: preface. AdvDrug

Deliv Rev 2008; 60: 89-90

42. Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog

Polym Sci 2012; 37: 106-26

43. Wu TJ, Huang HH, Hsu YM, et al. A novel method of encapsulating and

cultivating adherent mammalian cells within collagen microcarriers. Bio-

technol Bioeng 2007; 98: 578-85

44. Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical

applications. Adv Mater 2011; 23: H41-56

45. Baruch L, Machluf M. Alginate-chitosan complex coacervation for cell en-

capsulation: effect on mechanical properties and on long-term viability.

Biopolymers 2006; 82: 570-9

296 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 15: A Perspective on Bioactive Cell Microencapsulation

46. Sakai S, Hashimoto I, Tanaka S, et al. Small agarose microcapsules with cell-

enclosing hollow core for cell therapy: transplantation of ifosfamide-

activating cells to the mice with preestablished subcutaneous tumor. Cell

Transplant 2009; 18: 933-9

47. Sakai S, Ito S, Kawakami K. Calcium alginate microcapsules with spherical

liquid cores templated by gelatin microparticles for mass production of

multicellular spheroids. Acta Biomater 2010; 6: 3132-7

48. Sakai S, Kawakami K. Both ionically and enzymatically crosslinkable algi-

nate-tyramine conjugate as materials for cell encapsulation. J BiomedMater

Res A 2008; 85: 345-51

49. Santos E, Zarate J, Orive G, et al. Biomaterials in cell microencapsulation.

Adv Exp Med Biol 2010; 670: 5-21

50. Orive G, Tam SK, Pedraz JL, et al. Biocompatibility of alginate-poly-L-lysine

microcapsules for cell therapy. Biomaterials 2006; 27: 3691-700

51. Braccini I, Perez S.Molecular basis of C(2+)-induced gelation in alginates and

pectins: the egg-box model revisited. Biomacromolecules 2001; 2: 1089-96

52. Donati I, Holtan S, Morch YA, et al. New hypothesis on the role of

alternating sequences in calcium-alginate gels. Biomacromolecules 2005; 6:

1031-40

53. Morch YA, Donati I, Strand BL, et al. Effect of Ca2+, Ba2+, and Sr2+ on

alginate microbeads. Biomacromolecules 2006; 7: 1471-80

54. Orive G, Hernandez RM, Rodriguez Gascon A, et al. History, challenges and

perspectives of cell microencapsulation. Trends Biotechnol 2004; 22: 87-92

55. Drury JL, Dennis RG, Mooney DJ. The tensile properties of alginate

hydrogels. Biomaterials 2004; 25: 3187-99

56. George M, Abraham TE. Polyionic hydrocolloids for the intestinal delivery of

protein drugs: alginate and chitosan – a review. JControlRelease 2006; 114: 1-14

57. Chan G, Mooney DJ. New materials for tissue engineering: towards greater

control over the biological response. Trends Biotechnol 2008; 26: 382-92

58. Augst AD, Kong HJ, Mooney DJ. Alginate hydrogels as biomaterials.

Macromol Biosci 2006; 6: 623-33

59. Smetana Jr K. Cell biology of hydrogels. Biomaterials 1993; 14: 1046-50

60. Rowley JA, Madlambayan G, Mooney DJ. Alginate hydrogels as synthetic

extracellular matrix materials. Biomaterials 1999; 20: 45-53

61. Boontheekul T, Kong HJ, Hsiong SX, et al. Quantifying the relation between

bond number andmyoblast proliferation. Faraday Discuss 2008; 139: 53-70;

discussion 105-28, 419-20

62. Comisar WA, Kazmers NH, Mooney DJ, et al. Engineering RGD nano-

patterned hydrogels to control preosteoblast behavior: a combined com-

putational and experimental approach. Biomaterials 2007; 28: 4409-17

63. Kong HJ, Boontheekul T, Mooney DJ. Quantifying the relation between

adhesion ligand-receptor bond formation and cell phenotype. Proc Natl

Acad Sci U S A 2006; 103: 18534-9

64. Huebsch ND, Mooney DJ. Fluorescent resonance energy transfer: a tool for

probing molecular cell-biomaterial interactions in three dimensions. Bio-

materials 2007; 28: 2424-37

65. Alsberg E, Anderson KW, Albeiruti A, et al. Cell-interactive alginate

hydrogels for bone tissue engineering. J Dent Res 2001; 80: 2025-9

66. DhootNO, Tobias CA,Fischer I, et al. Peptide-modified alginate surfaces as a

growth permissive substrate for neurite outgrowth. J Biomed Mater Res A

2004; 71: 191-200

67. Mazzitelli S, Luca G, Mancuso F, et al. Production and characterization of

engineered alginate-based microparticles containing ECM powder for

cell/tissue engineering applications. Acta Biomater 2011; 7: 1050-62

68. Orive G, De Castro M, Kong HJ, et al. Bioactive cell-hydrogel microcapsules

for cell-based drug delivery. J Control Release 2009; 135: 203-10

69. Boontheekul T, Kong HJ, Mooney DJ. Controlling alginate gel degradation

utilizing partial oxidation and bimodal molecular weight distribution. Bio-

materials 2005; 26: 2455-65

70. Silva EA,MooneyDJ. Spatiotemporal control of vascular endothelial growth

factor delivery from injectable hydrogels enhances angiogenesis. J Thromb

Haemost 2007; 5: 590-8

71. Bouhadir KH, Lee KY, Alsberg E, et al. Degradation of partially oxidized

alginate and its potential application for tissue engineering. Biotechnol Prog

2001; 17: 945-50

72. ZhouH,XuHH. The fast release of stem cells from alginate-fibrinmicrobeads

in injectable scaffolds for bone tissue engineering. Biomaterials 2011; 32:

7503-13

73. Rokstad AM, Donati I, Borgogna M, et al. Cell-compatible covalently re-

inforced beads obtained from a chemoenzymatically engineered alginate.

Biomaterials 2006; 27: 4726-37

74. Gattas-AsfuraKM,Fraker CA, Stabler CL. Covalent stabilization of alginate

hydrogel beads via Staudinger ligation: assessment of poly(ethylene glycol)

and alginate cross-linkers. J Biomed Mater Res A 2011; 99: 47-57

75. Rosenblatt J, Devereux B, Wallace DG. Injectable collagen as a pH-sensitive

hydrogel. Biomaterials 1994; 15: 985-95

76. Senuma Y, Franceschin S, Hilborn JG, et al. Bioresorbable microspheres by

spinning disk atomization as injectable cell carrier: from preparation to in

vitro evaluation. Biomaterials 2000; 21: 1135-44

77. Lee M, Lo AC, Cheung PT, et al. Drug carrier systems based on collagen-

alginate composite structures for improving the performance of GDNF-

secreting HEK293 cells. Biomaterials 2009; 30: 1214-21

78. Ananta M, Brown RA, Mudera V. A rapid fabricated living dermal equiva-

lent for skin tissue engineering: an in vivo evaluation in an acute wound

model. Tissue Eng Part A 2012; 18: 353-61

79. Zhang Y, Wang F, Chen J, et al. Bone marrow-derived mesenchymal stem

cells versus bonemarrow nucleated cells in the treatment of chondral defects.

Int Orthop 2012; 36: 1079-86

80. BitarM, Salih V, Brown RA, et al. Effect of multiple unconfined compression

on cellular dense collagen scaffolds for bone tissue engineering. J Mater Sci

Mater Med 2007; 18: 237-44

81. Flanagan TC, Wilkins B, Black A, et al. A collagen-glycosaminoglycan co-

culture model for heart valve tissue engineering applications. Biomaterials

2006; 27: 2233-46

82. MurrayMM,ForsytheB,ChenF, et al. The effect of thrombin onACL fibroblast

interactions with collagen hydrogels. J Orthop Res 2006; 24: 508-15

83. Chenite A, Chaput C, Wang D, et al. Novel injectable neutral solutions of

chitosan form biodegradable gels in situ. Biomaterials 2000; 21: 2155-61

84. Monteiro Jr OA, Airoldi C. Some studies of crosslinking chitosan-

glutaraldehyde interaction in a homogeneous system. Int J Biol Macromol

1999; 26: 119-28

85. Zielinski BA, Aebischer P. Chitosan as a matrix for mammalian cell encap-

sulation. Biomaterials 1994; 15: 1049-56

86. Dang JM, Leong KW.Myogenic induction of aligned mesenchymal stem cell

sheets by culture on thermally responsive electrospun nanofibers. AdvMater

2007; 19: 2775-9

87. Dang JM, Sun DD, Shin-Ya Y, et al. Temperature-responsive hydroxybutyl

chitosan for the culture of mesenchymal stem cells and intervertebral disk

cells. Biomaterials 2006; 27: 406-18

88. Hayami JW,Waldman SD, Amsden BG. A photocurable hydrogel/elastomer

composite scaffold with bi-continuous morphology for cell encapsulation.

Macromol Biosci 2011; 11: 1672-83

89. Lee KY, Mooney DJ. Hydrogels for tissue engineering. Chem Rev 2001; 101:

1869-79

90. Buckley CT, Thorpe SD, O’Brien FJ, et al. The effect of concentration,

thermal history and cell seeding density on the initial mechanical properties

of agarose hydrogels. J Mech Behav Biomed Mater 2009; 2: 512-21

91. Luo D, Pullela SR, Marquez M, et al. Cell encapsules with tunable transport

and mechanical properties. Biomicrofluidics 2007; 1: 34102

A Perspective on Bioactive Cell Microencapsulation 297

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 16: A Perspective on Bioactive Cell Microencapsulation

92. Sakai S, Hashimoto I, Kawakami K. Agarose-gelatin conjugate membrane

enhances proliferation of adherent cells enclosed in hollow-core micro-

capsules. J Biomater Sci Polym Ed 2008; 19: 937-44

93. Sakai S, Hashimoto I, Kawakami K. Production of cell-enclosing hollow-

core agarose microcapsules via jetting in water-immiscible liquid

paraffin and formation of embryoid body-like spherical tissues from mouse

ES cells enclosed within these microcapsules. Biotechnol Bioeng 2008; 99:

235-43

94. Gazda LS, Vinerean HV, LaramoreMA, et al. Encapsulation of porcine islets

permits extended culture time and insulin independence in spontaneously

diabetic BB rats. Cell Transplant 2007; 16: 609-20

95. Wosnick JH, Shoichet MS. Three-dimensional chemical patterning of trans-

parent hydrogels. Chem Mater 2008; 20: 55-60

96. Uludag H, De Vos P, Tresco PA. Technology of mammalian cell encapsula-

tion. Adv Drug Deliv Rev 2000; 42: 29-64

97. SchmaljohannD. Thermo- and pH-responsive polymers in drug delivery. Adv

Drug Deliv Rev 2006; 58: 1655-70

98. Chaterji S, Kwon IK, Park K. Smart polymeric gels: redefining the limits of

biomedical devices. Prog Polym Sci 2007; 32: 1083-122

99. KimMR, Jeong JH, Park TG. Swelling induced detachment of chondrocytes

using RGD-modified poly(N-isopropylacrylamide) hydrogel beads. Bio-

technol Prog 2002; 18: 495-500

100. Lu HF, Targonsky ED, Wheeler MB, et al. Thermally induced gelable poly-

mer networks for living cell encapsulation. Biotechnol Bioeng 2007; 96:

146-55

101. Sa-Lima H, Tuzlakoglu K, Mano JF, et al. Thermoresponsive poly(N-

isopropylacrylamide)-g-methylcellulose hydrogel as a three-dimensional

extracellular matrix for cartilage-engineered applications. J Biomed Mater

Res A 2011; 98: 596-603

102. Gomez-Guillen MC, Perez-Mateos M, Gomez-Estaca J, et al. Fish gelatin:

a renewablematerial for developing active biodegradable films. Trends Food

Sci Technol 2009; 20: 3-16

103. Payne RG, YaszemskiMJ, Yasko AW, et al. Development of an injectable, in

situ crosslinkable, degradable polymeric carrier for osteogenic cell pop-

ulations. Part 1. Encapsulation of marrow stromal osteoblasts in surface

crosslinked gelatin microparticles. Biomaterials 2002; 23: 4359-71

104. Huang S, Deng T, Wang Y, et al. Multifunctional implantable particles for

skin tissue regeneration: preparation, characterization, in vitro and in vivo

studies. Acta Biomater 2008; 4: 1057-66

105. Stover NP, Watts RL. Spheramine for treatment of Parkinson’s disease.

Neurotherapeutics 2008; 5: 252-9

106. Huss FR, Junker JP, Johnson H, et al. Macroporous gelatine spheres as

culture substrate, transplantation vehicle, and biodegradable scaffold for

guided regeneration of soft tissues: in vivo study in nude mice. J Plast Re-

constr Aesthet Surg 2007; 60: 543-55

107. Gustafson CJ, Birgisson A, Junker J, et al. Employing human keratinocytes

cultured on macroporous gelatin spheres to treat full thickness-wounds:

an in vivo study on athymic rats. Burns 2007; 33: 726-35

108. Sommar P, Pettersson S, Ness C, et al. Engineering three-dimensional

cartilage- and bone-like tissues using human dermal fibroblasts and mac-

roporous gelatine microcarriers. J Plast Reconstr Aesthet Surg 2010; 63:

1036-46

109. Nuttelman CR, Kloxin AM, Anseth KS. Temporal changes in peg hydrogel

structure influence human mesenchymal stem cell proliferation and matrix

mineralization. Adv Exp Med Biol 2006; 585: 135-49

110. AdelowC, Segura T, Hubbell JA, et al. The effect of enzymatically degradable

poly(ethylene glycol) hydrogels on smooth muscle cell phenotype. Bio-

materials 2008; 29: 314-26

111. Hall KK,Gattas-AsfuraKM, Stabler CL.Microencapsulation of islets within

alginate/poly(ethylene glycol) gels cross-linked via Staudinger ligation. Acta

Biomater 2011; 7: 614-24

112. Peroglio M, Grad S, Mortisen D, et al. Injectable thermoreversible hyalur-

onan-based hydrogels for nucleus pulposus cell encapsulation. Eur Spine J.

Epub 2011 Aug 27; DOI 10.1007/s00586-011-1976-2

113. PelegrinM,MarinM,Noel D, et al. Systemic long-term delivery of antibodies

in immunocompetent animals using cellulose sulphate capsules containing

antibody-producing cells. Gene Ther 1998; 5: 828-34

114. Stadlbauer V, Stiegler PB, Schaffellner S, et al. Morphological and functional

characterization of a pancreatic beta-cell line microencapsulated in sodium

cellulose sulfate/poly(diallyldimethylammonium chloride). Xenotransplantation

2006; 13: 337-44

115. Stiegler P, Matzi V, Pierer E, et al. Creation of a prevascularized site for cell

transplantation in rats. Xenotransplantation 2010; 17: 379-90

116. Leung A, Nielsen LK, Trau M, et al. Tissue transplantation by stealth: co-

herent alginate microcapsules for immunoisolation. BiochemEng J 2010; 48:

337-47

117. de Vos P, Bucko M, Gemeiner P, et al. Multiscale requirements for bio-

encapsulation in medicine and biotechnology. Biomaterials 2009; 30: 2559-70

118. Kulseng B, Thu B, Espevik T, et al. Alginate polylysine microcapsules as

immune barrier: permeability of cytokines and immunoglobulins over the

capsule membrane. Cell Transplant 1997; 6: 387-94

119. DembczynskiR, Jankowski T.Determination of pore diameter andmolecular

weight cut-off of hydrogel-membrane liquid-core capsules for immuno-

isolation. J Biomater Sci Polym Ed 2001; 12: 1051-8

120. de Haan BJ, Faas MM, de Vos P. Factors influencing insulin secretion from

encapsulated islets. Cell Transplant 2003; 12: 617-25

121. Schneider S, Feilen PJ, Slotty V, et al. Multilayer capsules: a promising mi-

croencapsulation system for transplantation of pancreatic islets. Bio-

materials 2001; 22: 1961-70

122. Tun T, Inoue K, Hayashi H, et al. A newly developed three-layer agarose

microcapsule for a promising biohybrid artificial pancreas: rat to mouse

xenotransplantation. Cell Transplant 1996; 5: S59-63

123. Weber LM, Cheung CY, Anseth KS. Multifunctional pancreatic islet en-

capsulation barriers achieved via multilayer PEG hydrogels. Cell Transplant

2008; 16: 1049-57

124. Schmidt JJ, Rowley J, Kong HJ. Hydrogels used for cell-based drug delivery.

J Biomed Mater Res A 2008; 87: 1113-22

125. Orive G, Hernandez RM, Gascon AR, et al. Development and optimisation

of alginate-PMCG-alginate microcapsules for cell immobilisation. Int J

Pharm 2003; 259: 57-68

126. Darrabie MD, Kendall Jr WF, Opara EC. Characteristics of Poly-L-Orni-

thine-coated alginate microcapsules. Biomaterials 2005; 26: 6846-52

127. Thanos CG, Bintz BE, Emerich DF. Stability of alginate-polyornithine mi-

crocapsules is profoundly dependent on the site of transplantation. J Biomed

Mater Res A 2007; 81: 1-11

128. Fontes A, Fernandes HP, de Thomaz AA, et al. Measuring electrical and

mechanical properties of red blood cells with double optical tweezers.

J Biomed Opt 2008; 13: 014001

129. Hochmuth RM. Micropipette aspiration of living cells. J Biomech 2000; 33:

15-22

130. Dulinska I, Targosz M, StrojnyW, et al. Stiffness of normal and pathological

erythrocytes studied by means of atomic force microscopy. J Biochem Bio-

phys Methods 2006; 66: 1-11

131. BauschAR,MollerW, Sackmann E.Measurement of local viscoelasticity and

forces in living cells by magnetic tweezers. Biophys J 1999; 76: 573-9

132. Kim K, Liu X, Zhang Y, et al. Elastic and viscoelastic characterization of

microcapsules for drug delivery using a force-feedback MEMS micro-

gripper. Biomed Microdevices 2009; 11: 421-7

133. Sugiura S, Oda T, Izumida Y, et al. Size control of calcium alginate beads

containing living cells usingmicro-nozzle array. Biomaterials 2005; 26: 3327-31

298 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 17: A Perspective on Bioactive Cell Microencapsulation

134. Sugiura S, Oda T, Aoyagi Y, et al. Microfabricated airflow nozzle for mi-

croencapsulation of living cells into 150 micrometer microcapsules. Biomed

Microdevices 2007; 9: 91-9

135. Robitaille R, Leblond FA, Bourgeois Y, et al. Studies on small (<350microm)

alginate-poly-L-lysine microcapsules. V. Determination of carbohydrate

and protein permeation through microcapsules by reverse-size exclusion

chromatography. J Biomed Mater Res 2000; 50: 420-7

136. Canaple L, Rehor A, Hunkeler D. Improving cell encapsulation through size

control. J Biomater Sci Polym Ed 2002; 13: 783-96

137. Chicheportiche D, Reach G. In vitro kinetics of insulin release by micro-

encapsulated rat islets: effect of the size of the microcapsules. Diabetologia

1988; 31: 54-7

138. Beck J, Angus R, Madsen B, et al. Islet encapsulation: strategies to enhance

islet cell functions. Tissue Eng 2007; 13: 589-99

139. Zhang X, Xie Y, Koh CG, et al. A novel 3-D model for cell culture and tissue

engineering. Biomed Microdevices 2009; 11: 795-9

140. Spuch C, Antequera D, Portero A, et al. The effect of encapsulated VEGF-

secreting cells on brain amyloid load and behavioral impairment in a mouse

model of Alzheimer’s disease. Biomaterials 2010; 31: 5608-18

141. Santos E, Orive G, Calvo A, et al. Optimization of 100mum alginate-poly-l-

lysine-alginate capsules for intravitreous administration. J Control Release

2012; 158: 443-50

142. Kontturi LS, Yliperttula M, Toivanen P, et al. A laboratory-scale device for

the straightforward production of uniform, small sized cell microcapsules

with long-term cell viability. J Control Release 2011; 152: 376-81

143. Hong J, deMello AJ, Jayasinghe SN. Bio-electrospraying and droplet-based

microfluidics: control of cell numbers within living residues. Biomed Mater

2010; 5: 21001

144. WangW,LiuX,XieY, et al.Microencapsulation using natural polysaccharides

for drug delivery and cell implantation. J Mater Chem 2006; 16: 3252-67

145. Orive G, Carcaboso AM, Hernandez RM, et al. Biocompatibility evaluation

of different alginates and alginate-based microcapsules. Biomacromolecules

2005; 6: 927-31

146. Williams DF. On the mechanisms of biocompatibility. Biomaterials 2008; 29:

2941-53

147. StarkD,KornmannH,MunchT, et al. Novel type of in situ extraction:Use of

solvent containing microcapsules for the bioconversion of 2-phenylethanol

from L-phenylalanine by Saccharomyces cerevisiae. Biotechnol Bioeng

2003; 83: 376-85

148. JunterGA, Jouenne T. Immobilized viable microbial cells: from the process to the

proteome em leader or the cart before the horse. BiotechnolAdv 2004; 22: 633-58

149. Leinfelder U, Brunnenmeier F, Cramer H, et al. A highly sensitive cell assay

for validation of purification regimes of alginates. Biomaterials 2003; 24:

4161-72

150. de Haan BJ, Rossi A, Faas MM, et al. Structural surface changes and in-

flammatory responses against alginate-based microcapsules after exposure

to human peritoneal fluid. J Biomed Mater Res A 2011; 98: 394-403

151. Tam SK, Dusseault J, Bilodeau S, et al. Factors influencing alginate gel

biocompatibility. J Biomed Mater Res A 2011; 98: 40-52

152. Bunger CM, Tiefenbach B, Jahnke A, et al. Deletion of the tissue response

against alginate-pll capsules by temporary release of co-encapsulated ste-

roids. Biomaterials 2005; 26: 2353-60

153. Murua A, Herran E, Orive G, et al. Design of a composite drug delivery system

to prolong functionality of cell-based scaffolds. Int J Pharm 2011; 407: 142-50

154. Baruch L, Benny O, Gilert A, et al. Alginate-PLL cell encapsulation system

Co-entrapping PLGA-microspheres for the continuous release of anti-

inflammatory drugs. Biomed Microdevices. Epub 2009 Jun 11; DOI

10.1007/s10544-009-9327-3

155. Omer A, Keegan M, Czismadia E, et al. Macrophage depletion improves

survival of porcine neonatal pancreatic cell clusters contained in alginate

macrocapsules transplanted into rats. Xenotransplantation 2003; 10: 240-51

156. Barnett BP, Arepally A, Stuber M, et al. Synthesis of magnetic resonance-,

X-ray- and ultrasound-visible alginate microcapsules for immunoisolation

and noninvasive imaging of cellular therapeutics. Nat Protoc 2011; 6: 1142-51

157. Link TW, Woodrum D, Gilson WD, et al. MR-guided portal vein delivery

andmonitoring ofmagnetocapsules: assessment of physiologic effects on the

liver. J Vasc Interv Radiol 2011; 22: 1335-40

158. Catena R, Santos E, Orive G, et al. Improvement of the monitoring and

biosafety of encapsulated cells using the SFGNESTGL triple reporter sys-

tem. J Control Release 2010; 146: 93-8

159. Shapiro AM, Lakey JR, Ryan EA, et al. Islet transplantation in seven patients

with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive

regimen. N Engl J Med 2000; 343: 230-8

160. de Vos P, Faas MM, Strand B, et al. Alginate-based microcapsules for im-

munoisolation of pancreatic islets. Biomaterials 2006; 27: 5603-17

161. Antosiak-Iwanska M, Sitarek E, Sabat M, et al. Isolation, banking, encap-

sulation and transplantation of different types of Langerhans islets. Pol Arch

Med Wewn 2009; 119: 311-7

162. Schneider S, Klein HH. Preserved insulin secretion capacity and graft func-

tion of cryostored encapsulated rat islets. Regul Pept 2011; 166: 135-8

163. Montanucci P, Pennoni I, Pescara T, et al. The functional performance of

microencapsulated human pancreatic islet-derived precursor cells. Bio-

materials 2011; 32: 9254-62

164. Veriter S, Aouassar N, Adnet PY, et al. The impact of hyperglycemia and the

presence of encapsulated islets on oxygenation within a bioartificial pancreas

in the presence of mesenchymal stem cells in a diabetic Wistar rat model.

Biomaterials 2011; 32: 5945-56

165. Yadav N, Morris G, Harding SE, et al. Various non-injectable delivery sys-

tems for the treatment of diabetes mellitus. Endocr Metab Immune Disord

Drug Targets 2009; 9: 1-13

166. Dufrane D, Goebbels RM, Gianello P. Alginate macroencapsulation of pig

islets allows correction of streptozotocin-induced diabetes in primates up to

6 months without immunosuppression. Transplantation 2010; 90: 1054-62

167. Vaithilingam V, Quayum N, Joglekar MV, et al. Effect of alginate encapsulation

on the cellular transcriptome of human islets. Biomaterials 2011; 32: 8416-25

168. Luca G, Fallarino F, Calvitti M, et al. Xenograft of microencapsulated sertoli

cells reverses T1DM in NOD mice by inducing neogenesis of beta-cells.

Transplantation 2010; 90: 1352-7

169. O’Sullivan ES, JohnsonAS, OmerA, et al. Rat islet cell aggregates are superior to

islets for transplantation in microcapsules. Diabetologia 2010; 53: 937-45

170. Opara EC, Mirmalek-Sani SH, Khanna O, et al. Design of a bioartificial

pancreas(+). J Investig Med 2010; 58: 831-7

171. Ngoc PK, Phuc PV,NhungTH, et al. Improving the efficacy of type 1 diabetes

therapy by transplantation of immunoisolated insulin-producing cells. Hum

Cell 2011; 24: 86-95

172. Shao S, Gao Y, Xie B, et al. Correction of hyperglycemia in type 1 diabetic

models by transplantation of encapsulated insulin-producing cells derived

from mouse embryo progenitor. J Endocrinol 2011; 208: 245-55

173. Kishima H, Poyot T, Bloch J, et al. Encapsulated GDNF-producing C2C12

cells for Parkinson’s disease: a pre-clinical study in chronic MPTP-treated

baboons. Neurobiol Dis 2004; 16: 428-39

174. Grandoso L, Ponce S, Manuel I, et al. Long-term survival of encapsulated

GDNF secreting cells implanted within the striatum of parkinsonized rats.

Int J Pharm 2007; 343: 69-78

175. Shingo T, Date I, Yoshida H, et al. Neuroprotective and restorative effects of

intrastriatal grafting of encapsulated GDNF-producing cells in a rat model

of Parkinson’s disease. J Neurosci Res 2002; 69: 946-54

A Perspective on Bioactive Cell Microencapsulation 299

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 18: A Perspective on Bioactive Cell Microencapsulation

176. Yasuhara T, Shingo T, Muraoka K, et al. Early transplantation of an en-

capsulated glial cell line-derived neurotrophic factor-producing cell dem-

onstrating strong neuroprotective effects in a rat model of Parkinson disease.

J Neurosurg 2005; 102: 80-9

177. Yasuhara T, Shingo T, Muraoka K, et al. The differences between high and

low-dose administration of VEGF to dopaminergic neurons of in vitro and

in vivo Parkinson’s disease model. Brain Res 2005; 1038: 1-10

178. Yasuhara T, Shingo T, Muraoka K, et al. Neurorescue effects of VEGF on a

rat model of Parkinson’s disease. Brain Res 2005; 1053: 10-8

179. Yasuhara T, Shingo T, Kobayashi K, et al. Neuroprotective effects of vas-

cular endothelial growth factor (VEGF) upon dopaminergic neurons in a rat

model of Parkinson’s disease. Eur J Neurosci 2004; 19: 1494-504

180. Date I, ShingoT,OhmotoT, et al. Long-term enhanced chromaffin cell survival

and behavioral recovery in hemiparkinsonian rats with co-grafted polymer-

encapsulated human NGF-secreting cells. Exp Neurol 1997; 147: 10-7

181. Date I, Ohmoto T, Imaoka T, et al. Cografting with polymer-encapsulated

human nerve growth factor-secreting cells and chromaffin cell survival and

behavioral recovery in hemiparkinsonian rats. J Neurosurg 1996; 84: 1006-12

182. Sautter J, Tseng JL, Braguglia D, et al. Implants of polymer-encapsulated

genetically modified cells releasing glial cell line-derived neurotrophic factor

improve survival, growth, and function of fetal dopaminergic grafts. Exp

Neurol 1998; 149: 230-6

183. Ahn YH, Bensadoun JC, Aebischer P, et al. Increased fiber outgrowth from

xeno-transplanted human embryonic dopaminergic neurons with co-

implants of polymer-encapsulated genetically modified cells releasing glial

cell line-derived neurotrophic factor. Brain Res Bull 2005; 66: 135-42

184. Shanbhag MS, Lathia JD, Mughal MR, et al. Neural progenitor cells grown

on hydrogel surfaces respond to the product of the transgene of encapsulated

genetically engineered fibroblasts. Biomacromolecules. Epub 2010 Oct 13;

DOI 10.1021/bm100699q

185. Garcia P, Youssef I, Utvik JK, et al. Ciliary neurotrophic factor cell-based

delivery prevents synaptic impairment and improves memory in mouse

models of Alzheimer’s disease. J Neurosci 2010; 30: 7516-27

186. Klinge PM, Harmening K, Miller MC, et al. Encapsulated native and glu-

cagon-like peptide-1 transfected human mesenchymal stem cells in a trans-

genic mouse model of Alzheimer’s disease. Neurosci Lett 2011; 497: 6-10

187. Emerich DF, Skinner SJ, Borlongan CV, et al. The choroid plexus in the rise,

fall and repair of the brain. Bioessays 2005; 27: 262-74

188. Borlongan CV, Skinner SJ, Geaney M, et al. Neuroprotection by encapsu-

lated choroid plexus in a rodent model of Huntington’s disease. Neuroreport

2004; 15: 2521-5

189. Emerich DF, Schneider P, Bintz B, et al. Aging reduces the neuroprotective

capacity, VEGF secretion, and metabolic activity of rat choroid plexus

epithelial cells. Cell Transplant 2007; 16: 697-705

190. Emerich DF, Thanos CG, Goddard M, et al. Extensive neuroprotection by

choroid plexus transplants in excitotoxin lesioned monkeys. Neurobiol Dis

2006; 23: 471-80

191. Mittoux V, Joseph JM, Conde F, et al. Restoration of cognitive and motor

functions by ciliary neurotrophic factor in a primate model of Huntington’s

disease. Hum Gene Ther 2000; 11: 1177-87

192. Tobias CA, Dhoot NO, Wheatley MA, et al. Grafting of encapsulated

BDNF-producing fibroblasts into the injured spinal cord without immune

suppression in adult rats. J Neurotrauma 2001; 18: 287-301

193. Tobias CA, Han SS, Shumsky JS, et al. Alginate encapsulated BDNF-

producing fibroblast grafts permit recovery of function after spinal cord injury

in the absence of immune suppression. J Neurotrauma 2005; 22: 138-56

194. Barminko J, Kim JH, Otsuka S, et al. Encapsulated mesenchymal stromal

cells for in vivo transplantation. Biotechnol Bioeng 2011; 108: 2747-58

195. Soon-Shiong P,Heintz RE,MeridethN, et al. Insulin independence in a type 1

diabetic patient after encapsulated islet transplantation. Lancet 1994; 343:

950-1

196. Calafiore R, Basta G, Luca G, et al. Microencapsulated pancreatic islet allo-

grafts into nonimmunosuppressed patients with type 1 diabetes: first two

cases. Diabetes Care 2006; 29: 137-8

197. Calafiore R, Basta G, Luca G, et al. Standard technical procedures for mi-

croencapsulation of human islets for graft into nonimmunosuppressed

patients with type 1 diabetes mellitus. Transplant Proc 2006; 38: 1156-7

198. Elliott RB, Escobar L, Tan PL, et al. Live encapsulated porcine islets from a

type 1 diabetic patient 9.5 yr after xenotransplantation. Xenotransplantation

2007; 14: 157-61

199. Wynyard S, Garkavenko O, Elliot R. Multiplex high resolution melting assay

for estimation of Porcine Endogenous Retrovirus (PERV) relative gene

dosage in pigs and detection of PERV infection in xenograft recipients.

J Virol Methods 2011; 175: 95-100

200. Tuch BE, Keogh GW, Williams LJ, et al. Safety and viability of micro-

encapsulated human islets transplanted into diabetic humans. Diabetes Care

2009; 32: 1887-9

201. US National Institutes of Health. ClinicalTrials.gov [online]. Available from

URL: http://www.clinicaltrials.gov [Accessed 2012 May 23]

202. Stover NP, Bakay RA, Subramanian T, et al. Intrastriatal implantation of

human retinal pigment epithelial cells attached to microcarriers in advanced

Parkinson disease. Arch Neurol 2005; 62: 1833-7

203. Bloch J, Bachoud-Levi AC,DeglonN, et al. Neuroprotective gene therapy for

Huntington’s disease, using polymer-encapsulated cells engineered to secrete

human ciliary neurotrophic factor: results of a phase I study. Hum Gene

Ther 2004; 15: 968-75

204. Aebischer P, Schluep M, Deglon N, et al. Intrathecal delivery of CNTF using

encapsulated genetically modified xenogeneic cells in amyotrophic lateral

sclerosis patients. Nat Med 1996; 2: 696-9

205. Lohr M, Hoffmeyer A, Kroger J, et al. Microencapsulated cell-mediated

treatment of inoperable pancreatic carcinoma. Lancet 2001; 357: 1591-2

206. Salmons B, Lohr M, Gunzburg WH. Treatment of inoperable pancreatic

carcinoma using a cell-based local chemotherapy: results of a phase I/IIclinical trial. J Gastroenterol 2003; 38 Suppl. 15: 78-84

207. Hasse C, KlockG, Schlosser A, et al. Parathyroid allotransplantation without

immunosuppression. Lancet 1997; 350: 1296-7

208. Sieving PA, Caruso RC, TaoW, et al. Ciliary neurotrophic factor (CNTF) for

human retinal degeneration: phase I trial of CNTFdelivered by encapsulated

cell intraocular implants. Proc Natl Acad Sci U S A 2006; 103: 3896-901

209. Talcott KE, Ratnam K, Sundquist SM, et al. Longitudinal study of cone

photoreceptors during retinal degeneration and in response to ciliary neu-

rotrophic factor treatment. Invest Ophthalmol Vis Sci 2011; 52: 2219-26

210. Templin C, Kotlarz D, Faulhaber J, et al. Ex vivo expanded hematopoietic

progenitor cells improve cardiac function after myocardial infarction: role of

beta-catenin transduction and cell dose. JMol Cell Cardiol 2008; 45: 394-403

211. Mazo M, Gavira JJ, Pelacho B, et al. Adipose-derived stem cells for myo-

cardial infarction. J Cardiovasc Transl Res 2011; 4: 145-53

212. Choi D, Hwang KC, Lee KY, et al. Ischemic heart diseases: current treat-

ments and future. J Control Release 2009; 140: 194-202

213. Psaltis PJ, Zannettino AC, Worthley SG, et al. Concise review: mesenchymal

stromal cells: potential for cardiovascular repair. Stem Cells 2008; 26:

2201-10

214. Beeres SL, AtsmaDE, vanRamshorst J, et al. Cell therapy for ischaemic heart

disease. Heart 2008; 94: 1214-26

215. Sanz-Ruiz R, Fernandez-Santos E, Dominguez-Munoa M, et al. Early

translation of adipose-derived cell therapy for cardiovascular disease. Cell

Transplant 2009; 18: 245-54

216. Valina C, Pinkernell K, Song YH, et al. Intracoronary administration of

autologous adipose tissue-derived stem cells improves left ventricular func-

tion, perfusion, and remodelling after acute myocardial infarction. Eur

Heart J 2007; 28: 2667-77

300 Acarregui et al.

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)

Page 19: A Perspective on Bioactive Cell Microencapsulation

217. Cai L, Johnstone BH, Cook TG, et al. IFATS collection: human adipose

tissue-derived stem cells induce angiogenesis and nerve sprouting following

myocardial infarction, in conjunction with potent preservation of cardiac

function. Stem Cells 2009; 27: 230-7

218. Miyahara Y, Nagaya N, Kataoka M, et al. Monolayered mesenchymal stem

cells repair scarred myocardium after myocardial infarction. Nat Med 2006;

12: 459-65

219. Rigol M, Solanes N, Farre J, et al. Effects of adipose tissue-derived stem

cell therapy after myocardial infarction: impact of the route of administra-

tion. J Card Fail 2010; 16: 357-66

220. Amado LC, Saliaris AP, Schuleri KH, et al. Cardiac repair with intra-

myocardial injection of allogeneic mesenchymal stem cells after myocardial

infarction. Proc Natl Acad Sci U S A 2005; 102: 11474-9

221. Kim YJ, Huh YM, Choe KO, et al. In vivo magnetic resonance imaging of

injected mesenchymal stem cells in rat myocardial infarction; simultaneous

cell tracking and left ventricular function measurement. Int J Cardiovasc

Imaging 2009; 25 Suppl. 1: 99-109

222. Collins SD, Baffour R, Waksman R. Cell therapy in myocardial infarction.

Cardiovasc Revasc Med 2007; 8: 43-51

223. Yasuda T, Weisel RD, Kiani C, et al. Quantitative analysis of survival of

transplanted smooth muscle cells with real-time polymerase chain reaction.

J Thorac Cardiovasc Surg 2005; 129: 904-11

224. Teng CJ, Luo J, Chiu RC, et al. Massive mechanical loss of microspheres with

direct intramyocardial injection in the beating heart: implications for cellular

cardiomyoplasty. J Thorac Cardiovasc Surg 2006; 132: 628-32

225. Paul A, Ge Y, Prakash S, et al. Microencapsulated stem cells for tissue re-

pairing: implications in cell-based myocardial therapy. Regen Med 2009; 4:

733-45

226. Zhang H, Zhu SJ, Wang W, et al. Transplantation of microencapsulated

genetically modified xenogeneic cells augments angiogenesis and improves

heart function. Gene Ther 2008; 15: 40-8

227. Yu J, Du KT, Fang Q, et al. The use of human mesenchymal stem cells

encapsulated in RGD modified alginate microspheres in the repair of my-

ocardial infarction in the rat. Biomaterials 2010; 31: 7012-20

228. Bai XP, ZhengHX, FangR, et al. Fabrication of engineered heart tissue grafts

from alginate/collagen barium composite microbeads. Biomed Mater 2011;

6: 045002

229. Dixit V,Darvasi R, ArthurM, et al. Restoration of liver function inGunn rats

without immunosuppression using transplanted microencapsulated hep-

atocytes. Hepatology 1990; 12: 1342-9

230. Wong H, Chang TM. Bioartificial liver: implanted artificial cells micro-

encapsulated living hepatocytes increases survival of liver failure rats. Int J

Artif Organs 1986; 9: 335-6

231. Sgroi A, Mai G, Morel P, et al. Transplantation of encapsulated hepatocytes

during acute liver failure improves survival without stimulating native

liver regeneration. Cell Transplant. Epub 2011 Mar 7; DOI 10.3727/096368911X564976

232. Mai G, Nguyen TH, Morel P, et al. Treatment of fulminant liver failure by

transplantation of microencapsulated primary or immortalized xenogeneic

hepatocytes. Xenotransplantation 2005; 12: 457-64

233. Li S, Sun Z, LvG, et al. Microencapsulated UCB cells repair hepatic injure by

intraperitoneal transplantation. Cytotherapy 2009; 11: 1032-40

234. Lv G, Zhao L, Zhang A, et al. Bioartificial liver system based on choanoid

fluidized bed bioreactor improve the survival time of fulminant hepatic

failure pigs. Biotechnol Bioeng 2011; 108: 2229-36

235. Liu ZC, Chang TM. Coencapsulation of hepatocytes and bone marrow stem

cells: in vitro conversion of ammonia and in vivo lowering of bilirubin in

hyperbilirubemia Gunn rats. Int J Artif Organs 2003; 26: 491-7

236. Chang Liu Z, Chang TM. Coencapsulation of hepatocytes and bone marrow

cells: in vitro and in vivo studies. Biotechnol Annu Rev 2006; 12: 137-51

237. Shi XL, Zhang Y, Gu JY, et al. Coencapsulation of hepatocytes with bone

marrow mesenchymal stem cells improves hepatocyte-specific functions.

Transplantation 2009; 88: 1178-85

238. Gautier A, Carpentier B, Dufresne M, et al. Impact of alginate type and bead

diameter on mass transfers and the metabolic activities of encapsulated C3A

cells in bioartificial liver applications. Eur Cell Mater 2011; 21: 94-106

239. Coward SM, Legallais C, David B, et al. Alginate-encapsulated HepG2 cells

in a fluidized bed bioreactormaintain function in human liver failure plasma.

Artif Organs 2009; 33: 1117-26

240. Kinasiewicz A, Gautier A, Lewinska D, et al. Culture of C3A cells in alginate

beads for fluidized bed bioartificial liver. Transplant Proc 2007; 39: 2911-3

241. Miranda JP, Rodrigues A, Tostoes RM, et al. Extending hepatocyte func-

tionality for drug-testing applications using high-viscosity alginate-

encapsulated three-dimensional cultures in bioreactors. Tissue Eng Part C

Methods 2010; 16: 1223-32

242. Murua A, Orive G, Hernandez RM, et al. Emerging technologies in the de-

livery of erythropoietin for therapeutics. Med Res Rev 2011; 31: 284-309

243. Orive G, De CastroM, Ponce S, et al. Long-term expression of erythropoietin

from myoblasts immobilized in biocompatible and neovascularized micro-

capsules. Mol Ther 2005; 12: 283-9

244. Ponce S, Orive G, Hernandez RM, et al. In vivo evaluation of EPO-secreting

cells immobilized in different alginate-PLLmicrocapsules. J Control Release

2006; 116: 28-34

245. CironeP,Bourgeois JM,AustinRC, et al.Anovel approach to tumor suppression

with microencapsulated recombinant cells. Hum Gene Ther 2002; 13: 1157-66

246. Joki T, Machluf M, Atala A, et al. Continuous release of endostatin from micro-

encapsulated engineered cells for tumor therapy. Nat Biotechnol 2001; 19: 35-9

247. Read TA, Sorensen DR, Mahesparan R, et al. Local endostatin treatment of

gliomas administered by microencapsulated producer cells. Nat Biotechnol

2001; 19: 29-34

248. Teng H, Zhang Y, Wang W, et al. Inhibition of tumor growth in mice by

endostatin derived from abdominal transplanted encapsulated cells. Acta

Biochim Biophys Sin (Shanghai) 2007; 39: 278-84

249. Goren A, Dahan N, Goren E, et al. Encapsulated human mesenchymal stem

cells: a unique hypoimmunogenic platform for long-term cellular therapy.

FASEB J 2010; 24: 22-31

250. Kleinschmidt K, Klinge PM, Stopa E, et al. Alginate encapsulated human

mesenchymal stem cells suppress syngeneic glioma growth in the im-

munocompetent rat. J Microencapsul 2011; 28: 621-7

251. Cirone P, Shen F, Chang PL. A multiprong approach to cancer gene therapy

by coencapsulated cells. Cancer Gene Ther 2005; 12: 369-80

252. Dubrot J, Portero A, Orive G, et al. Delivery of immunostimulatory mono-

clonal antibodies by encapsulated hybridoma cells. Cancer Immunol Im-

munother 2010; 59: 1621-31

253. Pelegrin M, Marin M, Oates A, et al. Immunotherapy of a viral disease by in

vivo production of therapeutic monoclonal antibodies. Hum Gene Ther

2000; 11: 1407-15

254. Piller Puicher E, Tomanin R, Salvalaio M, et al. Encapsulated engineered

myoblasts can cure Hurler syndrome: preclinical experiments in the mouse

model. Gene Ther 2012; 19: 355-64

255. Pettingill LN, Wise AK, Geaney MS, et al. Enhanced auditory neuron sur-

vival following cell-based BDNF treatment in the deaf guinea pig. PLoSOne

2011; 6: e18733

Correspondence: Professor Rosa Ma Hernandez, NanoBioCel Group, Labor-

atory of Pharmaceutics, School of Pharmacy, University of the Basque

Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz,

Spain.

E-mail: [email protected]

A Perspective on Bioactive Cell Microencapsulation 301

Adis ª 2012 Springer International Publishing AG. All rights reserved. Biodrugs 2012; 26 (5)