21.03.2013 Views

The Bioartificial Pancreas: Progress and Challenges Review

The Bioartificial Pancreas: Progress and Challenges Review

The Bioartificial Pancreas: Progress and Challenges Review

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

DIABETES TECHNOLOGY & THERAPEUTICS<br />

Volume 7, Number 6, 2005<br />

© Mary Ann Liebert, Inc.<br />

<strong>The</strong> <strong>Bioartificial</strong> <strong>Pancreas</strong>: <strong>Progress</strong> <strong>and</strong> <strong>Challenges</strong><br />

SEDA KIZILEL, Ph.D., 1 MARC GARFINKEL, M.D., 1 <strong>and</strong> EMMANUEL OPARA, Ph.D. 2<br />

ABSTRACT<br />

Diabetes remains a devastating disease, with tremendous cost in terms of human suffering<br />

<strong>and</strong> healthcare expenditures. A bioartificial pancreas has the potential as a promising approach<br />

to preventing or reversing complications associated with this disease. <strong>Bioartificial</strong> pancreatic constructs<br />

are based on encapsulation of islet cells with a semipermeable membrane so that cells<br />

can be protected from the host’s immune system. Encapsulation of islet cells eliminates the requirement<br />

of immunosuppressive drugs, <strong>and</strong> offers a possible solution to the shortage of donors<br />

as it may allow the use of animal islets or insulin-producing cells engineered from stem cells.<br />

During the past 2 decades, several major approaches for immunoprotection of islets have been<br />

studied. <strong>The</strong> microencapsulation approach is quite promising because of its improved diffusion<br />

capacity, <strong>and</strong> technical ease of transplantation. It has the potential for providing an effective<br />

long-term treatment or cure of Type 1 diabetes.<br />

INTRODUCTION<br />

DIABETES MELLITUS represents a major public<br />

health problem, as it is the most frequent<br />

endocrine disease in industrialized countries. 1,2<br />

According to the latest estimates, the number<br />

of people with this disease worldwide is 177<br />

million, <strong>and</strong> this figure will double by the year<br />

2025. 3 <strong>The</strong> complications of diabetes are associated<br />

with multiple medical problems related<br />

to ophthalmic, renal, neurological, cerebrovascular,<br />

cardiovascular, <strong>and</strong> peripheral vascular<br />

disease. 4–9 <strong>The</strong> economic burden of diabetes is<br />

related to its management, to the treatment of<br />

its secondary complications, <strong>and</strong> to resultant<br />

productivity loss. 10 While one estimate shows<br />

that the cost of treating diabetes in the United<br />

States in 1997 was $44 billion, 3 another study<br />

<strong>Review</strong><br />

968<br />

has shown that it cost about $100 billion to treat<br />

diabetes <strong>and</strong> its complications in this country<br />

in 1992. 10<br />

Type 1 diabetes, also previously known as<br />

insulin-dependent diabetes mellitus, occurs<br />

when pancreatic islet cells are unable to produce<br />

insulin. Consequently, the blood glucose<br />

concentration becomes high while tissues are<br />

starving for metabolic fuel. In Type 2 diabetes,<br />

previously referred to as non–insulin-dependent<br />

diabetes, the body continues to make at<br />

least some insulin, but is unable to respond<br />

properly to the action of insulin produced by<br />

the pancreas. <strong>The</strong> Diabetes Control <strong>and</strong> Complications<br />

Trial 11 showed that an improved<br />

metabolic control was achieved using intensive<br />

insulin treatment in Type 1 diabetes patients.<br />

However, the aggressive management of dia-<br />

1 Section of Transplantation, Department of Surgery, <strong>The</strong> University of Chicago, Chicago, Illinois.<br />

2 Pritzker Institute of Biomedical Science <strong>and</strong> Engineering, Illinois Institute of Technology, Chicago, Illinois.


THE BIOARTIFICIAL PANCREAS 969<br />

betes with exogenous insulin only delayed the<br />

progression <strong>and</strong> development of complications<br />

of this disease. Also, intensive insulin treatment<br />

could only be applied to fewer than 10%<br />

of patients due an increased risk of severe<br />

episodes of hypoglycemia. Furthermore, it appears<br />

that C-peptide, a cleavage product from<br />

the processing of pro-insulin to insulin, prevents<br />

diabetes <strong>and</strong> hyperglycemia-induced<br />

vascular <strong>and</strong> neural dysfunction in animal <strong>and</strong><br />

clinical models of diabetes <strong>and</strong> should therefore<br />

be used in combination with insulin to<br />

maintain vascular <strong>and</strong> neural functions as well<br />

as blood glucose levels. 12–15<br />

Advances in transplantation <strong>and</strong> in immunosuppression<br />

have made pancreas transplantation<br />

another treatment option for Type 1<br />

diabetes. <strong>The</strong> main objective for pancreas transplantation<br />

is to achieve normal level of glucose<br />

in the blood <strong>and</strong> to free the patient from exogenous<br />

insulin requirements based on multiple<br />

finger stick glucose measurements. 16 Even<br />

though successful pancreas transplantation<br />

provides normal glucose homeostasis, the<br />

requirement of lifelong immunosuppression<br />

makes it unclear whether transplantation is advantageous<br />

over continued insulin treatment. 17<br />

Also, because of the need for immunosuppression,<br />

most pancreas transplantations have been<br />

done simultaneously with a kidney transplant<br />

in patients who have advanced nephropathy. 18<br />

In contrast to pancreas transplantation, islet<br />

transplantation requires no major surgery.<br />

Also, islet transplantation offers an alternative<br />

to exogenous insulin treatment, as it can result<br />

in better glycemic control 19 <strong>and</strong> potentially<br />

avoids surgical complications associated with<br />

whole-organ pancreas transplantation. It is<br />

known that islet cell transplantation can prevent,<br />

<strong>and</strong>, in some cases, reverse existing complications<br />

of diabetes probably because of the<br />

role played by C-peptide. 20 However, major<br />

challenges remain to be addressed before islet<br />

transplantation can be used routinely <strong>and</strong> applied<br />

more widely to patients with diabetes.<br />

One major barrier is the shortage of human<br />

pancreas as a source of islet cells. Also, lifelong<br />

use of immunosuppressive drugs to overcome<br />

the rejection of transplanted tissue poses a significant<br />

risk to patients, as the use of glucocorticoids<br />

<strong>and</strong> cyclosporine have dose-dependent<br />

deleterious effects on glucose homeostasis <strong>and</strong><br />

-cell function, <strong>and</strong> result in increased incidences<br />

of infection <strong>and</strong> cancer. 20–24 <strong>The</strong> introduction<br />

of the steroid-free immunosuppressive<br />

drug regimen has resulted in long-term graft<br />

survival of islets with concomitant control of<br />

blood glucose levels. 25 However, this immunosuppressive<br />

drug regimen is also not without<br />

risks to graft recipients. 26<br />

<strong>The</strong> routine application of islet transplantation<br />

would be tremendously enhanced by an<br />

unlimited supply of donor tissue, a st<strong>and</strong>ard<br />

implantation procedure, <strong>and</strong> the ability to<br />

maintain the transplanted tissue without the requirement<br />

of immunosuppressive drugs. 27 To<br />

achieve these goals, immunoisolation (encapsulation)<br />

of islet cells has been proposed to protect<br />

cells from attack by the host immune system.<br />

Using immunoisolation of the islets,<br />

chronic administration of immunosuppressive<br />

drugs can be theoretically eliminated or minimized,<br />

as transplanted cells are separated from<br />

the host immune system by a biocompatible<br />

<strong>and</strong> semipermeable membrane. 28,29 Moreover,<br />

immunoisolation of pancreatic islet cells for<br />

transplantation into patients with diabetes theoretically<br />

permits grafting of xenogenic islets,<br />

thus exp<strong>and</strong>ing the potential supply of donor<br />

islet tissue. 25 Hence, immunoisolation could<br />

open up the possibility of using islets harvested<br />

from animals, or insulin-producing cells engineered<br />

from stem cells without the requirement<br />

of immunosuppression of transplant recipients.<br />

Porcine <strong>and</strong> bovine insulin amino acid sequences<br />

have considerable sequence homology<br />

with human insulin, 30 <strong>and</strong> therefore pigs <strong>and</strong><br />

cows are considered attractive as options for<br />

xenogeneic donors.<br />

One important consideration in using microencapsulated<br />

islets is defining the optimal<br />

site for transplantation. To date, most research<br />

has considered peritoneal cavity as the optimal<br />

site for implantation of encapsulated islets, as<br />

it is possible to transplant microcapsules into<br />

the peritoneal cavity using a simple procedure<br />

that could be suitable for routine outpatient<br />

use. 29,31–34 Also, this site allows for implantation<br />

of high numbers of islets, which can be retrieved<br />

by peritoneal lavage 35 if necessary. 36<br />

However, this site limits graft viability because<br />

of low blood supply, oxygen tension, <strong>and</strong> con-


970<br />

centration of essential minerals. 34 In addition,<br />

the release of insulin from the peritoneal cavity<br />

to the blood stream is delayed compared<br />

with hormones released to the portal vein. 37<br />

Also, peritoneal macrophages have a high toxicity<br />

against encapsulated islets. 38<br />

Several researchers have also explored the<br />

portal vein of the liver as an alternative site for<br />

transplantation of immunoisolated islets. 39<br />

Based on its double vascularization (hepatic<br />

artery <strong>and</strong> portal vein), the intraportal location<br />

has a high oxygen <strong>and</strong> nutrient supply, which<br />

could be beneficial for the performance <strong>and</strong> the<br />

longevity of microencapsulated cells. 39 Also,<br />

the liver is considered to be an immunologically<br />

privileged site. 40 But, even more clinically<br />

relevant, is the use of percutaneous transhepatic<br />

catheterization, which provides relatively<br />

simple, inexpensive, <strong>and</strong> non-surgical access to<br />

the liver. 41 In addition, <strong>The</strong> International Islet<br />

Registry reports more C-peptide-positive cases<br />

(more than 0.5 ng/mL) 1 year after transplantation<br />

into the portal vein than from any other<br />

site. 42<br />

This article reviews the recent progress <strong>and</strong><br />

challenges remaining for the successful development<br />

of a bioartificial pancreas, <strong>and</strong> discusses<br />

the requirements to bring this technology<br />

closer to clinical application.<br />

HISTORICAL DEVELOPMENT<br />

<strong>The</strong> pancreatic origin of diabetes was discovered<br />

in 1889 by Mering <strong>and</strong> Minkowski,<br />

when surgical removal of the pancreas caused<br />

dogs to develop diabetes. In 1913, Murlin <strong>and</strong><br />

Kramer prepared extracts of bovine pancreas<br />

to lower the blood glucose in a dog with diabetes.<br />

However, the benefit of the extract was<br />

explained by the presence of lactate. Finally, in<br />

1922, Banting <strong>and</strong> Best developed a method for<br />

preparing a pancreatic extract, <strong>and</strong> a few years<br />

later, injection of the active factor in this extract,<br />

insulin, became the main therapy for Type 1 diabetes.<br />

As of today, the treatment options for Type<br />

1 diabetes are exogenous insulin with external<br />

glucose monitoring, whole-organ pancreas<br />

transplantation, islet cell transplantation, the artificial<br />

pancreas, <strong>and</strong> the bioartificial pancreas.<br />

KIZILEL ET AL.<br />

Among these treatments, the bioartificial<br />

pancreas avoids the use of immunosuppressive<br />

drugs while providing moment-to-moment<br />

glucose homeostasis consistent with a wellfunctioning<br />

native pancreas. It is important in<br />

this context to distinguish between the terms<br />

“artificial pancreas” (sometimes called “mechanical<br />

artificial pancreas”) <strong>and</strong> “bioartificial<br />

pancreas.”<br />

<strong>The</strong> mechanical artificial pancreas consists of<br />

a glucose sensor, an insulin pump, <strong>and</strong> a computer,<br />

which determines the rate of insulin delivery.<br />

<strong>The</strong> sensor may be implanted in the<br />

vena cava, <strong>and</strong> may require periodic replacement.<br />

<strong>The</strong> development of a glucose biosensor<br />

has been challenged by the difficulty of creating<br />

a sensitive, stable, <strong>and</strong> accurate sensor, despite<br />

the capability of a mechanical artificial<br />

pancreas to regulate glucose levels. An error in<br />

sensing, computation, or delivery could result<br />

in insulin overdosing, which could be potentially<br />

risky with life-threatening hypoglycemia,<br />

43–46 an unacceptable potential side effect<br />

of an artificial pancreas. It is also very difficult<br />

to produce a mechanical artificial pancreas that<br />

responds quickly enough to changes in blood<br />

sugar (islets respond in less than 10 min). 47<br />

In contrast, a bioartificial pancreas is a device<br />

that substitutes for the endocrine portion<br />

of the pancreas. Devices in this category contain<br />

synthetic materials <strong>and</strong> functional islets<br />

encapsulated within a semipermeable membrane<br />

to protect cells from the host immune response.<br />

<strong>The</strong> semipermeable membrane permits<br />

exchange of nutrients, glucose, <strong>and</strong> insulin<br />

with the host, but excludes the diffusion of immunoglobulins,<br />

complement, <strong>and</strong> white blood<br />

cells. <strong>The</strong> device may be implanted into a vascularized<br />

site 48,49 or into the peritoneal cavity. 36<br />

<strong>The</strong> idea of using ultrathin polymer membrane<br />

microcapsules was proposed by Chang 50<br />

in 1964 for the immunoprotection of transplanted<br />

cells. Nearly 2 decades later, the concept<br />

of bioencapsulation was successfully<br />

shown to maintain glucose homeostasis in rats<br />

with diabetes using encapsulated allogeneic<br />

islets. 32 When alginate microcapsules containing<br />

islets were implanted in rats, normal blood<br />

glucose levels were achieved for 13 weeks. <strong>The</strong><br />

microcapsule used in that study consisted of an<br />

inner alginate core surrounded by a polylysine


THE BIOARTIFICIAL PANCREAS 971<br />

membrane, which was then surrounded by an<br />

outer polyethyleneimine coating. In later studies,<br />

the outer layer was replaced by the more<br />

biocompatible alginate coating because of an<br />

inflammatory reaction induced by polyethyleneimine.<br />

51 Since then, a variety of studies have<br />

been performed to underst<strong>and</strong> the requirements<br />

for successful transplantation of encapsulated<br />

islet cells, but there is still a need for<br />

convincing evidence of success of encapsulated<br />

islet transplantation in either non-human primates<br />

or humans. 49<br />

IMPEDIMENTS TO THE PROGRESS OF<br />

CELL ENCAPSULATION TECHNOLOGY<br />

Fibrotic overgrowth of the implanted microcapsules<br />

is one of the major obstacles to progress<br />

in cell encapsulation technology. Invariably,<br />

the materials used for encapsulation are<br />

not completely inert, <strong>and</strong> can induce foreign<br />

body <strong>and</strong> inflammatory reactions. As a result of<br />

this fibrous tissue overgrowth, the diffusion of<br />

nutrients, oxygen, hormones, <strong>and</strong> waste products<br />

through the capsule is diminished, <strong>and</strong> encapsulated<br />

islets are destroyed because of hypoxia,<br />

starvation, <strong>and</strong> the secretion of nitric<br />

oxide by the stimulated macrophages. 52 This fibrotic<br />

reaction to implanted microcapsules had<br />

previously been associated with commercially<br />

available alginate with high mannuronic acid<br />

content, which activates macrophages in vivo,<br />

resulting in fibroblast proliferation <strong>and</strong> eventual<br />

overgrowth. 53 However, later studies did<br />

not support that notion. 33 It has also been<br />

shown that polylysine may cause a fibrotic response<br />

through the induction of cytokines. 54<br />

Another important component of capsular<br />

design is permselectivity. Most capsule designs<br />

are based on the presumption that the effectiveness<br />

of immunoisolation of a polymer capsule<br />

is closely related to the pore size of the capsule<br />

membrane. 55–57 However, pore sizes of<br />

polymer membranes are not homogeneous by<br />

nature, <strong>and</strong> consist of a broad spectrum of<br />

sizes. 58 Total immunoprotection of encapsulated<br />

cells can only be provided if the polymer<br />

membrane does not have any pores larger than<br />

the antibody complement component. 59 This<br />

fact has been largely ignored by researchers<br />

<strong>and</strong> has resulted in inconsistent findings in the<br />

field. 60–66<br />

Mechanism of immunologic attack of allografts<br />

<strong>and</strong> xenografts<br />

With regard to specific immunologic attack<br />

of encapsulated cell implants, there are differences<br />

in the immune protection requirement of<br />

allografts versus xenografts. An allograft is a<br />

graft between genetically different individuals<br />

within the same species, while a xenograft is a<br />

graft between individuals from different<br />

species. Allograft rejection occurs as result of<br />

activation of cellular immunity by interactions<br />

of host T cells with a graft, while humoral immunity,<br />

including antibodies <strong>and</strong> complement<br />

proteins, is responsible for the rejection of<br />

xenografts. 67,68 In the direct pathway of antigen<br />

presentation, the T cell receptor recognizes<br />

T cells presented by donor-type antigen-presenting<br />

cells along with Class I or II major histocompatibility<br />

complex (MHC) molecules.<br />

<strong>The</strong> idea of immunoisolation using a polymer<br />

membrane is to separate allogeneic or xenogeneic<br />

tissue from the immune system of the<br />

recipient. <strong>The</strong>refore, microencapsulation of<br />

islets within effective permselective membranes<br />

would prevent contact with immunoglobulin,<br />

complement components, <strong>and</strong><br />

inflammatory cells. However, there is also a potential<br />

immune response towards antigens<br />

shed by encapsulated allogeneic or xenogeneic<br />

cells. Such antigens could be therapeutic agent<br />

themselves, cell surface molecules, or cell components<br />

(phospholipids, DNA), including<br />

those released upon cell death. Shedding of<br />

antigens from encapsulated cells would initiate<br />

a molecular tissue response around the implant,<br />

which could affect the viability <strong>and</strong> function<br />

of the encapsulated cells. As a result of this<br />

immunological reaction, these shedded antigens<br />

may be internalized, processed, <strong>and</strong> presented<br />

in association with host Class II MHC<br />

molecules (macrophages <strong>and</strong> dendritic cells) to<br />

host CD4 helper T cells (Fig. 1). <strong>The</strong> recognition<br />

of antigens through this indirect pathway<br />

may lead to the activation of T helper cells,<br />

which then secrete cytokines <strong>and</strong> regulate cellmediated<br />

immune response <strong>and</strong> inflammation.<br />

69,70 Small molecules, such as reactive oxy-


972<br />

FIG. 1. Schematic representation of antigen recognition<br />

via (A) the direct pathway <strong>and</strong> (B) the indirect pathway.<br />

APC, antigen-presenting cells; TCR, T cell receptor.<br />

gen species <strong>and</strong> cytokines, may also pass<br />

through the polymer membrane <strong>and</strong> damage<br />

the transplanted tissue.<br />

Owing to the complexity of the immune response<br />

mechanism, it is important to underst<strong>and</strong><br />

the mechanism of immune protection,<br />

<strong>and</strong> considerable efforts have been made to investigate<br />

this issue. Chen et al. 71 developed cell<br />

lines that have resistance to cytokines <strong>and</strong> oxygen<br />

radical-induced damage. Recently, macrophage<br />

depletion has also been used as a tool to<br />

protect xenografts from immune destruction. It<br />

has been found that depletion of peritoneal<br />

macrophages with clodronate liposomes improved<br />

the survival of macroencapsulated<br />

porcine neonatal pancreatic cell clusters. 35<br />

Safley et al. 72 evaluated the cellular immune response<br />

in non-obese diabetic (NOD) mice after<br />

two different pathways of T cell co-stimulation<br />

were blocked. Alginate–poly-L-lysine-encapsulated<br />

porcine islet xenografts were transplanted<br />

intraperitoneally in NOD mice treated<br />

with CTLA4-immunoglobulin to block CD28/<br />

B7 <strong>and</strong> with anti-CD154 monoclonal antibody<br />

to inhibit CD40/CD40–lig<strong>and</strong> interactions. It<br />

was concluded that blocking two different<br />

pathways of T cell co-stimulation inhibited T<br />

cell-dependent inflammatory responses, <strong>and</strong><br />

KIZILEL ET AL.<br />

significantly prolonged the survival of encapsulated<br />

islet xenografts.<br />

Another approach to improving the survival<br />

of microencapsulated islets can be to use local<br />

immunosuppression. Studies have shown that<br />

preimplantation of an immunoisolating device<br />

improved the survival of an encapsulated islet<br />

graft <strong>and</strong> reduced fibroblast outgrowth. 73 Functional<br />

capsules may also be designed to release<br />

the immunosuppressive agent in a controlled<br />

manner. Local immunosuppression induced using<br />

this approach may prevent the occurrence<br />

of an undesirable reaction around the implant.<br />

Inflammatory reaction in response to the implantation<br />

of cell-free capsules involves a sequence<br />

of events similar to a foreign body reaction,<br />

starting with an acute inflammatory<br />

response <strong>and</strong> leading to a chronic inflammatory<br />

response or granulation tissue development<br />

<strong>and</strong> fibrous capsule development. <strong>The</strong> intensity<br />

<strong>and</strong> duration of each of these responses<br />

are dependent upon several factors, such as the<br />

extent of injury created in the implantation,<br />

biomaterial chemical composition, surface free<br />

energy, surface charge, porosity, surface<br />

roughness, <strong>and</strong> implant size <strong>and</strong> shape. 74,75 <strong>The</strong><br />

biocompatibility of the material is determined<br />

by the extent <strong>and</strong> deviation from the optimal<br />

wound healing conditions. 76<br />

Sources of donor islets<br />

Despite all of the advances in islet transplantation<br />

<strong>and</strong> in encapsulation technology, some<br />

challenges must be still overcome before the<br />

bioartificial pancreas can be broadly applied.<br />

<strong>The</strong> most significant of these challenges is the<br />

shortage of donor islets. This shortage is the<br />

motivation for researchers to find alternative<br />

sources of insulin-producing cells. Promising<br />

approaches for resolving this problem are differentiation<br />

of stem cells into cells with -celllike<br />

characteristics77–80 <strong>and</strong> genetic engineering<br />

of adult cells for secretion of recombinant insulin.<br />

81–86 Based on similarities between mechanisms<br />

that control the development of both the<br />

adult pancreas <strong>and</strong> the central nervous system,<br />

methods promoting neural differentiation of<br />

embryonic stem cells have been adapted by researchers<br />

to derive insulin-producing cells. 87–91<br />

Hori et al. 92 investigated the possibility of directing<br />

neural progenitors to develop into glu-


THE BIOARTIFICIAL PANCREAS 973<br />

cose-responsive insulin-producing cells using<br />

inductive signals involved in normal pathways<br />

of islet development. Compared with other<br />

methods for insulin-producing cell development<br />

from human stem cells, 91,93 the method<br />

developed by this group produced insulin at<br />

the highest levels yet achieved from an exp<strong>and</strong>able,<br />

human stem cell-derived tissue.<br />

However, the method developed in the study<br />

still needs to be improved, perhaps through the<br />

addition of glucagon-like peptide-1, transforming<br />

growth factor- lig<strong>and</strong>s, or other factors<br />

to potentiate -cell maturation, growth,<br />

<strong>and</strong> insulin secretion. 94–96<br />

Gene therapy has also been suggested as a<br />

treatment of insulin-dependent diabetes mellitus.<br />

This strategy can be applied by preventing<br />

the autoimmune destruction of -cells, 97,98 by<br />

regenerating -cells, 99,100 or by engineering insulin-secreting<br />

surrogate -cells. 99,101–103 Sapir<br />

et al. 104 recently suggested the potential use of<br />

adult human liver as an alternate tissue for autologous<br />

-cell-replacement therapy. Using<br />

pancreatic <strong>and</strong> duodenal homeobox gene 1<br />

(PDX-1) <strong>and</strong> soluble factors, they managed to<br />

induce a comprehensive developmental shift of<br />

adult human liver cells into functional insulinproducing<br />

cells. According to the findings of<br />

that study, PDX-1-treated human liver cells expressed<br />

insulin, stored it in defined granules,<br />

<strong>and</strong> secreted the hormone in a glucose-regulated<br />

manner. When these cells were implanted<br />

under the renal capsule of immunodeficient<br />

mice with diabetes, the cells ameliorated hyperglycemia<br />

for prolonged periods of time. <strong>The</strong><br />

technique offers both the potential of a cell-replacement<br />

therapy for patients with diabetes<br />

<strong>and</strong> allows the patient to be the donor of his or<br />

her own insulin-producing tissue.<br />

Intestinal K cells have also been engineered<br />

to express <strong>and</strong> secrete insulin in a glucose dependent<br />

manner, generating functional cells.<br />

105,106 With the development of robust <strong>and</strong><br />

safe gene therapy, this strategy may lead to effective<br />

therapy in the future.<br />

Preservation of encapsulated cell systems<br />

<strong>The</strong> last challenge for a bioartificial pancreas<br />

to become a routine clinical application is the<br />

long-term cryopreservation of encapsulated<br />

cells. Validated technologies are required for<br />

long-term preservation of encapsulated cell systems<br />

to maintain a product inventory, <strong>and</strong> in order<br />

to meet end-user dem<strong>and</strong>s. One of the current<br />

strategies for overcoming the problem of<br />

cryopreservation of tissue is the reduction of cryoprotectant<br />

[dimethyl sulfoxide (DMSO)] concentration<br />

or complete replacement by equally<br />

powerful substances. Recent work in Dr. Opara’s<br />

laboratory has demonstrated that cryopreservation<br />

of islet cells cultured overnight in the presence<br />

of 10% polyvinylpyrrolidone yielded higher<br />

intact islet recovery compared with islets frozen<br />

in the presence of DMSO <strong>and</strong> glycerol. 107 <strong>The</strong><br />

lower islet cell integrity <strong>and</strong> function were explained<br />

on the basis of hypothesis that low-molecular-weight<br />

compounds, such as DMSO <strong>and</strong><br />

glycerol, permeate the cell <strong>and</strong> interact hydrophobically<br />

with intracellular proteins, which<br />

results in perturbed cytoskeletal architecture of<br />

the frozen cells <strong>and</strong> diminished islet cell integrity<br />

<strong>and</strong> function.<br />

<strong>The</strong> other promising technical improvement<br />

of the cryopreservation technology is the reduction<br />

of total sample volume. Miniaturization<br />

of the cryosubstrates from 1 mL to microliters<br />

reduces temperature gradients in cryosubstrates,<br />

which makes it possible to achieve<br />

higher freezing rates <strong>and</strong> more homogeneous<br />

freezing of the sample. 108 Islets cryopreserved<br />

with this strategy were highly functional even<br />

with the lower DMSO concentrations. 109<br />

Misler et al. 110 studied stimulus–secretion<br />

coupling in whole islets as well as single cells<br />

from carefully selected cryopreserved <strong>and</strong><br />

thawed human islets of Langerhans. <strong>The</strong>y found<br />

that without using any of the recent advances in<br />

cryopreservation technique, cryopreserved <strong>and</strong><br />

thawed human islets, which were selected based<br />

on their smooth surface <strong>and</strong> diameter, responded<br />

to glucose in a calcium- <strong>and</strong> metabolicdependent<br />

fashion. In another study by von<br />

Mach et al., 111 viability of pancreatic islets after<br />

cryopreservation was correlated negatively with<br />

their size, <strong>and</strong> suppression of insulin release was<br />

not observed for islets 300 m.<br />

DIFFERENT FORMS OF BIOARTIFICIAL<br />

PANCREAS<br />

Various configurations have been studied for<br />

the purpose of immunoisolation of islets. <strong>The</strong>se


974<br />

include biohybrid vascular devices, extravascular<br />

chambers, <strong>and</strong> encapsulation. 112,113 Encapsulation<br />

is the technique of islet immunoisolation<br />

using biopolymeric spheres of<br />

different sizes. Currently, results in rodents<br />

<strong>and</strong> dogs with diabetes indicate that spherical<br />

micro- <strong>and</strong> macrocapsules appear to have the<br />

highest therapeutic potential. 60,65,114 A microcapsule<br />

typically contains only a few hundred<br />

cells or a single islet, <strong>and</strong> to provide a therapeutic<br />

dosage, tens of hundreds of thous<strong>and</strong>s<br />

of cells are required. In contrast, macrocapsules<br />

employ much larger depots to contain the full<br />

therapeutic dosage in one or a few implants.<br />

Macrocapsules are shaped as cylinders or<br />

planes, <strong>and</strong> typically one dimension is in the<br />

range of 2–6 mm (Fig. 2a). 115,116 One advantage<br />

of the implantation of macrocapsules is the ease<br />

of retrieval in case of complications. However,<br />

despite reports of reduced risks in the implantation<br />

of macrocapsules, it has been shown that<br />

macroencapsulated islets have significantly impaired<br />

insulin secretion because of necrosis in<br />

the center of aggregated islet clumps as a result<br />

of diffusion limitations of nutrients <strong>and</strong><br />

oxygen. 116,117<br />

Encouraging results have been obtained by<br />

immobilization of the islets in a matrix before<br />

final macroencapsulation. 115,118 Limited diffusion<br />

of nutrients, as well as slow exchange of<br />

glucose <strong>and</strong> insulin, occurs as a result of relative<br />

large surface-to-volume ratio of these devices.<br />

To ensure sufficient nutrient <strong>and</strong> oxygen<br />

diffusion to the cells, islet cell density within<br />

the macrocapsules is kept to around 5–10% of<br />

the volume fraction. This approach, however,<br />

requires the implantation of large devices in order<br />

to provide adequate masses of insulin-producing<br />

cells, <strong>and</strong> large grafts are not practical<br />

<strong>and</strong> cannot be transplanted in conventional<br />

sites. 119<br />

In contrast, microencapsulation offers an optimal<br />

volume-to-surface ratio for fast exchange<br />

of hormones <strong>and</strong> nutrients without a large device.<br />

<strong>The</strong> technique involves enclosure of one<br />

or two islets within small spheres, which have<br />

diameters of less than 1 mm (Fig. 2b). Also, because<br />

of reduced volume of the islet transplants,<br />

<strong>and</strong> in contrast to macroencapsulated islets, microcapsules<br />

can theoretically be transplanted in<br />

delicate sites, such as the portal vein. <strong>The</strong> trans-<br />

(a) Macroencapsulation<br />

(b) Microencapsulation<br />

plantation of microcapsules into the peritoneal<br />

cavity, an outpatient-based procedure, 33,120–122<br />

makes it particularly attractive to patients <strong>and</strong><br />

doctors. Interestingly, intraperitoneally implanted<br />

microencapsulated islets have shown<br />

promising results in large animal 122–124 models<br />

as well as in limited clinical trials. 33<br />

TECHNIQUES IN<br />

MICROENCAPSULATION<br />

KIZILEL ET AL.<br />

FIG. 2. Immunoisolated islets within (a) membrane diffusion<br />

chambers (macrocapsules) <strong>and</strong> (b) microcapsules.<br />

Materials used for microencapsulation<br />

Various materials have been used as biopolymeric<br />

coats in islet microencapsulation. <strong>The</strong>se<br />

have included alginate, agarose, tissue-engineered<br />

chondrocytes, polyacrylates, 125,126 <strong>and</strong><br />

poly(ethylene glycol) (PEG).<br />

Alginate–polylysine-based microencapsulation<br />

of islets, first described by Lim <strong>and</strong> Sun, 32<br />

has been found not to interfere with cellular function,<br />

<strong>and</strong> these microcapsules have been shown<br />

to be stable for years in small <strong>and</strong> large animals<br />

as well as in human beings. 127–132 Alginate is a<br />

linear polysaccharide extracted from seaweed. 133<br />

It is composed of three types of 1 4-linked<br />

polyuronic acid blocks containing poly-L-guluronic<br />

acid segments (G blocks), poly-D-manuronic<br />

acid segments (M blocks), <strong>and</strong> segments<br />

of alternating L-guluronic <strong>and</strong> D-mannuronic<br />

acid residues, <strong>and</strong> it has gel-forming properties<br />

in the presence of most polyvalent cations. 133,134<br />

Impurities, such as monomers, catalysts, <strong>and</strong> initiators<br />

present in synthetic polymers, may contribute<br />

to the failure of the encapsulated islet im-


THE BIOARTIFICIAL PANCREAS 975<br />

plants. 135 Other impurities, such as pyrogens <strong>and</strong><br />

mitogens, can be found in polymers derived<br />

from natural substances, such as seaweed. 135 Up<br />

to 90% of the impurities can be removed by electrophoresis<br />

or by the Klock extraction purification<br />

procedure. 130,135 Conflicting reports exist on<br />

whether alginate with high M blocks or high G<br />

blocks results in increased fibrosis. 53 It has been<br />

shown, however, that the inflammatory response<br />

to alginate becomes less with purification,<br />

irrespective of the composition used. 36,136<br />

<strong>The</strong> immunoisolating permselectivity of alginate<br />

microcapsules can be achieved by soaking<br />

the initial alginate beads in 0.05–0.1%<br />

polylysine dissolved in normal saline for 6–20<br />

min. This occurs as a result of the binding of<br />

negatively charged carboxyl groups on the alginate<br />

to positively charged -amino groups on<br />

the amino acid polymer. <strong>The</strong> droplets are again<br />

rinsed with normal saline, followed by the addition<br />

of an outer alginate layer by soaking in<br />

a lower concentration of 0.06–0.25% sodium alginate<br />

for 4 min. It is extremely important that<br />

the outer coating of alginate on top of the<br />

polylysine membrane is complete. In the case<br />

of incomplete cover of the polylysine coating<br />

<strong>and</strong> when an impure alginate is used for the<br />

outer coating, microcapsules may become completely<br />

covered by cell overgrowths within 1<br />

week of transplantation because of the inflammatory<br />

reactions attributable to polylysine <strong>and</strong><br />

the impurities in the alginate. 137 To dissolve the<br />

intracapsular sodium alginate by chelation of<br />

the cross-linking calcium, the resulting microcapsules<br />

are treated with 55 mM sodium citrate<br />

for 7 min. <strong>The</strong> hollow microcapsules containing<br />

a few islets are then washed with saline<br />

(Fig. 3). It appears that liquefaction of the inner<br />

alginate core of microcapsules is required<br />

for optimal function of the enclosed islets. 138<br />

One successful transplantation of encapsulated<br />

islets performed with this technique was reported<br />

by a team of investigators at Duke University.<br />

<strong>The</strong> encapsulated islets were made<br />

with an outer alginate coating <strong>and</strong> inner poly-<br />

L-ornithine layer for permselectivity. To liquefy<br />

the inner alginate core of each bead a salt treatment<br />

was used. After transplantation, encapsulated<br />

islet cells were shown to keep a baboon<br />

with diabetes from requiring exogenous insulin<br />

for more than 9 months. 122<br />

FIG. 3. Microencapsulation of islets with alginate–<br />

polylysine.<br />

For a capsule to be stable in vivo it has to be<br />

water insoluble, as capsule dissolution will<br />

elicit a continuing inflammatory response <strong>and</strong><br />

will ultimately expose the transplanted cells to<br />

the host. Both alginate <strong>and</strong> polylysine are water-soluble<br />

polymers <strong>and</strong> are both expected to<br />

elicit an inflammatory or foreign body response.<br />

However, findings indicate that the<br />

polyelectrolyte complex formed during encapsulation<br />

is biocompatible <strong>and</strong> less soluble in<br />

water than the individual components. 62,139,140<br />

Differences in biocompatibility were also noted<br />

between empty <strong>and</strong> cell-containing capsules in<br />

NOD mice, 141,142 which could be due to the difficulty<br />

of producing reproducible cell-containing<br />

capsules. For better biocompatibility, one<br />

group has suggested the use of high guluronic<br />

acid-containing alginates, 53 while another<br />

group modified alginate–polylysine capsules<br />

with PEG. 132 An alternative approach has been<br />

chosen to obtain stronger <strong>and</strong> more biocompatible<br />

microcapsules using synthetic polyacrylates<br />

such as the water-insoluble synthetic<br />

copolymer hydroxyethyl methacrylate-methyl<br />

methacrylate. 143 This research group has selected<br />

the acrylate monomers because of their<br />

diversity <strong>and</strong> has shown that mammalian cells


976<br />

may be microencapsulated in uncharged <strong>and</strong><br />

polyelectrolyte polymer, in polyelectrolyte<br />

complexes, <strong>and</strong> inside a cohesive precipitate<br />

from a destabilized emulsion, without loss of<br />

viability.<br />

Polymerization of acrylamide monomer on<br />

islet cells encapsulated in agarose microspheres<br />

has also been reported. 144 However, when<br />

polymerization is in direct contact with tissue,<br />

this can generate excessive local heating <strong>and</strong><br />

cytotoxicity. 145 Poly(styrene sulfonic acid), one<br />

of the most potent complement-interacting<br />

polymers, was selected for the purpose of consuming<br />

cytolytic complement activity <strong>and</strong><br />

mixed with agarose for the preparation of microbeads<br />

to protect xenogeneic islets in mice<br />

with diabetes from the humoral immune reaction.<br />

63,146 Antibodies <strong>and</strong> complement proteins<br />

play a major role in the rejection of xenografts.<br />

Binding of antibodies to the antigens on the<br />

xenogeneic cell surface cannot alone damage<br />

the islet cell. For destruction of the xenogeneic<br />

cells to occur, complement activation by antigen–antibody<br />

complexes present on the cell<br />

surface is required. 147 Various polymers bearing<br />

hydroxyl groups such as regenerated cellulose,<br />

cellulose acetate, <strong>and</strong> poly(ethylene-covinyl<br />

alcohol) activate the complement system<br />

through the alternative pathway, 148 whereas<br />

other polymers bearing sulfonic acid or sulfate<br />

groups strongly interact with complement proteins<br />

<strong>and</strong> decrease the cytolytic complement activity.<br />

149<br />

As previously noted, a fibrotic reaction after<br />

implantation would limit the diffusion of essential<br />

nutrients or metabolites, leading to reduced<br />

viability or impaired functional activity<br />

of the encapsulated cells. Several studies have<br />

reported the usefulness of coating membrane<br />

surfaces with poly(ethylene oxide) (PEO) in order<br />

to reduce protein adsorption <strong>and</strong> associated<br />

fibrotic reaction. 150 This was achieved by<br />

exposing the poly(acrylonitrile-vinyl chloride)<br />

membrane surface to amine-terminated PEO<br />

after acid hydrolysis of the nitrile group to a<br />

carboxylic acid. Membrane surfaces were also<br />

exposed to PEO-succinimide after base reduction<br />

to an amine. 151 <strong>The</strong> in vivo response<br />

proved that a smaller number of macrophages<br />

<strong>and</strong> foreign body giant cells were present on<br />

the PEO-grafted membrane surface without<br />

KIZILEL ET AL.<br />

any change in transport properties. <strong>The</strong> other<br />

application to modify the membrane surface<br />

was achieved by synthesizing a graft copolymer<br />

having poly-L-lysine as the backbone <strong>and</strong><br />

the monomethoxy PEG as pendant chains. Microcapsules<br />

with sodium alginate were formed<br />

using this polycationic copolymer, which demonstrated<br />

reduced protein adsorption, complement<br />

binding, <strong>and</strong> cell adhesion in vitro compared<br />

with materials with unmodified poly-<br />

L-lysine. 132<br />

That artificial materials used for immunoisolation<br />

purposes are not completely inert <strong>and</strong><br />

can induce foreign body <strong>and</strong> inflammatory reactions<br />

have led researchers to study tissue-engineered<br />

capsules of rat chondrocytes. Pollok<br />

et al. 126 proposed the encapsulation of islets<br />

with a layer of chondrocytes <strong>and</strong> their matrix<br />

to prevent immunorecognition <strong>and</strong> destruction<br />

of transplanted allogeneic or xenogeneic islets.<br />

This investigation demonstrated the membrane’s<br />

ability as an immunoisolation barrier<br />

by utilizing the immunoprivileged properties<br />

of the chondrocyte matrix. This approach<br />

might offer a therapeutic advantage for patients<br />

with diabetes, using the patient’s own<br />

chondrocytes from a cartilage biopsy specimen<br />

as the encapsulation material.<br />

PEG is another polymer that has been utilized<br />

for the purpose of encapsulating islets. In<br />

order to obtain stable <strong>and</strong> biocompatible gels<br />

without any cytotoxicity generation, Pathak et<br />

al. 152 <strong>and</strong> Cruise et al. 153 reported rapid photopolymerization<br />

of water-soluble PEG-based<br />

macromers in direct contact with islet cells.<br />

PEG is biocompatible, nontoxic, non-immunogenic,<br />

<strong>and</strong> hydrophilic <strong>and</strong> can be chemically<br />

cross-linked into hydrogels for a variety of applications.<br />

154,155 Diffusion profiles of proteins<br />

through PEG diacrylate hydrogels showed that<br />

gels formed with the proper formulation were<br />

capable of being immunoprotective. <strong>The</strong> semipermeable<br />

properties of cross-linked PEG hydrogels<br />

made by interfacial photopolymerization<br />

of PEG diacrylate were also studied by<br />

Cruise et al. 156 by forming hydrogels upon<br />

poly(vinylidiene fluoride) microporous filters.<br />

This approach allowed them to study the effect<br />

of molecular weight <strong>and</strong> concentration of the<br />

PEG diacrylate on the diffusivity of biological<br />

molecules.


THE BIOARTIFICIAL PANCREAS 977<br />

Techniques used for droplet generation<br />

<strong>The</strong> other major consideration in microencapsulation<br />

technology is how to get the cells<br />

into the individual polymer capsules. Techniques<br />

to form such a physical barrier include<br />

air-jet syringe pump droplet generator, interfacial<br />

photopolymerization, <strong>and</strong> selective withdrawal.<br />

One of the main devices used for microencapsulation<br />

of islets with alginate–polylysine is<br />

the air-jet syringe pump droplet generator. 157<br />

<strong>The</strong> device consists of an air jacket surrounding<br />

an alginate nozzle through which alginate<br />

solution is injected. 158,159 Islets are suspended<br />

in a solution of 1.4–3% sodium alginate, <strong>and</strong><br />

spherical droplets of this suspension are<br />

formed by an air-jet syringe pump generator.<br />

As alginate droplets are forced out of the end<br />

of the needle by the syringe pump, the droplets<br />

are pulled off by the shear forces set up by the<br />

flowing air stream. <strong>The</strong> size of the spherical<br />

droplets is controlled by adjusting the flow rate<br />

of the air jacket. <strong>The</strong> spherical droplets are collected<br />

in a funnel containing HEPES acid buffer<br />

supplemented with 1.1% CaCl2 solution, which<br />

transformed alginate droplets into rigid beads<br />

by gelation. 110 Excess fluid is drained with the<br />

aid of a filtering device consisting of a nylon<br />

mesh <strong>and</strong> a stopcock at the exit end of the funnel,<br />

<strong>and</strong> the droplets are washed in normal<br />

saline.<br />

As noted earlier, an air jet-syringe pump extrusion<br />

method generates gel droplets containing<br />

entrapped islets from the suspension of the<br />

islets in aqueous sodium alginate solution.<br />

However, there are various constraints concerning<br />

the use of this procedure to produce<br />

microcapsule diameters of less than 700 m. To<br />

form perfectly spherical capsules, the viscosity<br />

of the gel-forming liquid must be greater than<br />

30 cp. Also, to prevent the blockage of the needle<br />

by the islets, the minimum internal diameter<br />

of the needle must be greater than 300 m.<br />

Finally, the volumetric airflow rate must remain<br />

below 2,000 mL/min in order to produce<br />

capsules of uniform diameter. This procedure<br />

was improved by employing an electrostatic<br />

droplet generator to form uniform, smooth,<br />

<strong>and</strong> perfectly spherical microcapsules having a<br />

diameter about 150–500 m. 160 A droplet,<br />

which is charged with high static voltage, is<br />

suspended from a needle <strong>and</strong> attracted to a collecting<br />

vessel with opposing polarity. Once the<br />

voltage threshold potential is passed, the<br />

droplet moves from the needle to the collecting<br />

vessel. Droplets with predetermined sizes<br />

may be repeatedly generated as the voltage<br />

pulse height, pulse frequency <strong>and</strong> length, <strong>and</strong><br />

extrusion rate of the droplets are adjustable. As<br />

shown in Figure 4, droplets are formed as the<br />

plunger is driven by the syringe pump <strong>and</strong> expelled<br />

towards a collecting vessel containing a<br />

hardening solution, which may be aqueous calcium<br />

chloride in the case of an aqueous droplet<br />

forming liquid containing sodium alginate.<br />

Negative polarity was attached to the needle,<br />

while positive polarity was attached to the<br />

metal ring. As the voltage applied during<br />

droplet formation is a static voltage, the viability<br />

of the encapsulated islets is not compromised.<br />

Biomolecules such as proteins <strong>and</strong> cells can<br />

also be deposited as patterned droplets onto<br />

surfaces for the development of cell-based<br />

biosensing devices for applications such as<br />

high-throughput drug screening. 161 <strong>The</strong>re are<br />

numerous techniques for microfabrication of<br />

patterned polymer surfaces for protein delivery.<br />

Lithographic techniques <strong>and</strong> microcontact<br />

printing have been most widely used to generate<br />

patterned droplets of cells. 162–168 PEG hy-<br />

FIG. 4. Schematic representation of an electrostatic<br />

droplet generator.


978<br />

drogel is the most commonly used material for<br />

cell patterning purposes. In the case of surfaceinitiated<br />

photopolymerization of PEG, the technique<br />

involves immobilization of initiator onto<br />

a substrate surface using a stamp such as<br />

poly(dimethyl siloxane), followed by polymerization<br />

of precursor solution. 169 One recent<br />

study focused on the cellular delivery based on<br />

micro- <strong>and</strong> nanotechnology. 170 To provide an<br />

immunoisolating microenvironment for islet<br />

cells, nanoporous biocapsules were bulk <strong>and</strong><br />

surface micromachined to present uniform <strong>and</strong><br />

well-controlled pore sizes. <strong>The</strong> designs of the<br />

membranes with defined nanopores were fabricated<br />

using a thermally grown silicon oxide<br />

s<strong>and</strong>wiched between two structural layers of<br />

silicon.<br />

Another technique used to generate droplets<br />

of encapsulated islet cells was proposed by<br />

Cruise et al. 153 using interfacial photopolymerization.<br />

<strong>The</strong> technique involves physical adsorption<br />

of the initiator eosin Y on the islet cell<br />

surface, which then allows polymerization to<br />

occur on the islet–prepolymer interface. <strong>The</strong> interfacial<br />

photoinitiation process employed with<br />

this technique results in conformal coating of<br />

cross-linked PEG-based hydrogel on the islet<br />

cell surface.<br />

<strong>The</strong> process of selective withdrawal is another<br />

novel method reported recently for the<br />

purpose of coating cell clusters, such as islet<br />

cells. 171 Briefly, the process involves the insertion<br />

of a tube into a container such that its tip<br />

is suspended at a specific height above an interface<br />

separating two immiscible fluids (Fig.<br />

5). In the case of a low fluid withdrawal rate,<br />

only the upper fluid (oil) is withdrawn through<br />

the tube. Increasing the fluid flow rate or de-<br />

Oil<br />

Water<br />

To pump<br />

FIG. 5. Diagram of the selective withdrawal apparatus.<br />

creasing the height of the tip above the interface<br />

results in a transition where the lower liquid<br />

(water) is incorporated in a thin spout<br />

along with the oil. <strong>The</strong> technique was demonstrated<br />

through encapsulation of a poppy seed<br />

in a poly(styrenesulfonic acid). <strong>The</strong> particles<br />

were coated with the prepolymer solution,<br />

which has styrene sulfonic acid sodium salt<br />

<strong>and</strong> triethylene glycol diacrylate, <strong>and</strong> mixed<br />

with the initiator eosin Y <strong>and</strong> co-initiator triethanolamine<br />

used for the purpose of initiating<br />

polymerization. 171 After the selective withdrawal<br />

step, the coated particles were collected<br />

<strong>and</strong> photopolymerized with a halogen lamp.<br />

<strong>The</strong> technique could also be used to encapsulate<br />

cells through surface initiated photopolymerization<br />

of PEG. <strong>The</strong> photoinitiator eosin Y<br />

could be immobilized on the islet cell surface<br />

through covalent attachment, 169 <strong>and</strong> after the<br />

selective withdrawal process, cells could be collected<br />

<strong>and</strong> illuminated for 2–3 min with an argon<br />

ion laser in order to cross-link the coat<br />

around the cells.<br />

CONCLUSIONS<br />

KIZILEL ET AL.<br />

<strong>The</strong> successful development of a bioartificial<br />

pancreas involves several considerations. Two<br />

major obstacles in islet transplantation are the<br />

limited supply of islet cells <strong>and</strong> the use of immunosuppressive<br />

drugs to prevent transplant<br />

rejection. It is hoped that the use of encapsulated<br />

islets as a form of bioartificial pancreas<br />

would overcome these obstacles. Three potential<br />

sources of islet cell tissue, including human<br />

or allogeneic cells, porcine or xenogenic cells,<br />

<strong>and</strong> engineered cells, are currently under investigation.<br />

Among these sources, human islet<br />

cells would be the least immunogenic; however,<br />

there is a shortage of pancreata retrieved<br />

from human cadaveric donors, <strong>and</strong> these cells<br />

have a limited secretory capacity <strong>and</strong> life span.<br />

<strong>The</strong> use of porcine islet cells could be a better<br />

option, because these are readily available, <strong>and</strong><br />

there is an unlimited supply of donors. However,<br />

one major concern about the use of islets<br />

harvested from pigs has been the possibility of<br />

transmittance of porcine endogenous retroviruses<br />

(PERVs) to humans during transplantation.<br />

This concept arose from studies on the


THE BIOARTIFICIAL PANCREAS 979<br />

infection of humans <strong>and</strong> immunodeficient mice<br />

after pig cell xenotransplantation. 172,173 <strong>The</strong>re<br />

are studies, however, showing no possibility of<br />

transmission of PERVs from pigs to human recipients<br />

of islet xenografts. 120,174,175 Indeed, one<br />

recent study showed that the porcine genome<br />

harbors a limited number of infectious PERV<br />

sequences, which suggests that many breeds of<br />

pig fail to produce PERVs capable of infecting<br />

human cells even in laboratory testing. 176 This<br />

raises the possibility of using special breeds of<br />

pig for an unlimited supply of islet cells for encapsulation<br />

<strong>and</strong> use as a bioartificial pancreas<br />

in clinical xenotransplantation.<br />

<strong>The</strong> other attractive approach to generate unlimited<br />

supply of islets is the use of embryonic<br />

stem cells, which have the ability to differentiate<br />

into a variety of cell lineages in vitro. Recent<br />

studies in small animals showing successful<br />

transplantation of pancreatic stem cells<br />

suggest that this approach could provide an<br />

unlimited source of allogeneic insulin-producing<br />

tissue in the future. 177–179<br />

<strong>The</strong> maintenance of cell viability is another<br />

important concern in the development of a successful<br />

bioartificial pancreas. When cells are encapsulated<br />

<strong>and</strong> implanted in an environment<br />

without a natural circulation, lack of an adequate<br />

supply of oxygen <strong>and</strong> nutrients results in<br />

cell necrosis, making implantation unsuccessful.<br />

To improve long-term islet survival <strong>and</strong><br />

function, vascularization of transplanted islets<br />

has been proposed. 180–182 An ideal architecture<br />

for implantation would be to promote vascularization<br />

around encapsulated cells using a<br />

novel approach. Recently, a technique to form<br />

covalently bonded multilayers of PEG hydrogels<br />

has been developed. This approach may<br />

be used to encapsulate cells with bilayers such<br />

that outer layer would promote vascularization<br />

around the inner layer. 183,184<br />

<strong>The</strong>re are other areas of urgent need for the<br />

successful development of a bioartificial pancreas<br />

through islet cell microencapsulation. All<br />

current techniques for cell microencapsulation<br />

are slow in producing desirable microcapsules,<br />

requiring days to generate enough encapsulated<br />

islets for one transplantation. 185 An ideal<br />

procedure for routine use of encapsulated islets<br />

would require the development of massively<br />

parallel concepts in microencapsulation, which<br />

would generate sufficient quantities of microcapsules<br />

for multiple transplantations in a matter<br />

of minutes. In the absence of a technique<br />

based on such a principle, there is a need for<br />

an adequate procedure for long-term storage of<br />

encapsulated cells to enhance their use for<br />

transplantation. It is also highly desirable to develop<br />

rapid in vitro techniques for determination<br />

of the functional viability of encapsulated<br />

islets prior to transplantation.<br />

In summary, a potential cure for Type 1 diabetes<br />

could be the use of bioartificial pancreatic<br />

constructs based on insulin-secreting cells<br />

(allogeneic or xenogeneic islet cells) that are immunoisolated<br />

with the microencapsulation<br />

technique. Additionally, enhanced survival of<br />

the graft might be supported with a novel approach<br />

to induce neovascularization, which<br />

could make this technology a clinical reality in<br />

the near future. This will in turn serve as an<br />

important progress in cell therapy for the treatment<br />

of diabetes as well as other diseases, such<br />

as cancer, hemophilia, liver failure, <strong>and</strong> Parkinson’s<br />

disease.<br />

REFERENCES<br />

1. Kleinman JC, Donahue RP, Harris MI, Finucane FF,<br />

Madans JH, Brock DB: Mortality among diabetics in<br />

a national sample. Am J Epidemiol 1988;128:389–401.<br />

2. Drury TF: Disability among adult diabetics. In: National<br />

Diabetes Group, eds. Diabetes in America: Diabetes<br />

Data Compiled. Publication 85-1468. Bethesda,<br />

MD: National Diabetes Data Group, 1985.<br />

3. World Health Organization: Definition, Diagnosis<br />

<strong>and</strong> Classification of Diabetes Mellitus <strong>and</strong> Its Complications.<br />

Report of a WHO Consultation. Geneva:<br />

World Health Organization, 1999.<br />

4. Nathan DM: Long term complications of diabetes<br />

mellitus. N Engl J Med 1993;328:1676–1685.<br />

5. Barrett-Connor E, Orchard T: Insulin-dependent diabetes<br />

mellitus <strong>and</strong> ischemic heart disease. Diabetes<br />

Care 1985;8:65–70.<br />

6. Barret-Conor E, Khaw KT: Diabetes mellitus <strong>and</strong> independent<br />

risk factor for stroke. Am J Epidemiol<br />

1988;128:116–123.<br />

7. Bild DE, Selby JV, Sinnock P, Browner WS, Braveman<br />

P, Showstack JA: Lower extremity amputation<br />

in people with diabetes: epidemiology <strong>and</strong> prevention.<br />

Diabetes Care 1989;12:24–29.<br />

8. Geiss LS, Herman WH, Teutsch SM: Diabetes <strong>and</strong><br />

renal mortality in the United States. Am J Public<br />

Health 1985;75:1325–1326.


980<br />

9. Moss SE, Klein R, Klein BE: <strong>The</strong> incidence of vision<br />

loss in a diabetic population. Ophthalmology 1988;<br />

95:1340–1348.<br />

10. Rubin RJ, Altman WM, Mendelson DN: Health care<br />

expenditures for people with diabetes mellitus. J<br />

Clin Endocrinol Metab 1994;78:809A–809F.<br />

11. Diabetes Control <strong>and</strong> Complications Trial Research<br />

Group: <strong>The</strong> effect of intensive treatment of diabetes<br />

on the development <strong>and</strong> progression of long-term<br />

complications in insulin-dependent diabetes mellitus.<br />

N Engl J Med 1993;329:977–986.<br />

12. Ido Y, Vindignia A, Chang K, Stramm L, Chance R,<br />

Heath WF, DiMarchi RD, Di Cera E, Williamson JR:<br />

Prevention of vascular <strong>and</strong> neural dysfunction in diabetic<br />

rats by C-peptide. Science 1997;277:563–566.<br />

13. Johansson BL, Borg K, Fernqvist-Forbes E, Kernell<br />

A, Odergren T, Wahren J: Beneficial effects of C-peptide<br />

on incipient nephropathy <strong>and</strong> neuropathy in patients<br />

with Type 1 diabetes mellitus. Diabet Med<br />

2000;17:181–189.<br />

14. Hansen A, Johansson BL, Wahren J, Bibra HV: Cpeptide<br />

exerts beneficial effects on myocardial blood<br />

flow <strong>and</strong> function in patients with Type 1 diabetes.<br />

Diabetes 2002;51:3077–3082.<br />

15. Ekberg K, Brismar T, Johansson BL, Jonsson B, Lindström<br />

P, Wahren J: Amelioration of sensory nerve<br />

dysfunction by C-peptide in patients with Type 1 diabetes.<br />

Diabetes 2003;52:536–541.<br />

16. Sutherl<strong>and</strong> DE, Gruessner RW, Gruessner AC: <strong>Pancreas</strong><br />

transplantation for treatment of diabetes mellitus.<br />

World J Surg 2001;25:487–496.<br />

17. Venstrom JM, McBride MA, Rother KI, Hirshberg B,<br />

Orchard TJ, Harlan DM: Survival after pancreas<br />

transplantation in patients with diabetes <strong>and</strong> preserved<br />

kidney function. JAMA 2003;290:2817–2823.<br />

18. Sutherl<strong>and</strong> DE, Stratta RJ, Gruessner AC: <strong>Pancreas</strong><br />

transplant outcome by recipient category: single<br />

pancreas versus combined kidney-pancreas. Curr<br />

Opin Organ Transplant 1998;3:231–241.<br />

19. de Groot M, Schuurs TA, van Schilfgaarde R: Causes<br />

of limited survival of microencapsulated pancreatic<br />

islet grafts. J Surg Res 2004;121:141–150.<br />

20. Kendall WFJ, Opara EC: Immunoisolation techniques<br />

for islet cell transplantation. Expert Opin Biol<br />

<strong>The</strong>r 2002;2:503–511.<br />

21. Alej<strong>and</strong>ro R, Feldman EC, Bloom AD, Kenyon NS:<br />

Effects of cyclosporin on insulin <strong>and</strong> C-peptide secretion<br />

in healthy beagles. Diabetes 1989;38:698–703.<br />

22. Gunnarsson R, Klintmalm G, Lundgren G, Wilczek<br />

H, Ostman J, Groth CG: Deterioration in glucose metabolism<br />

in pancreatic transplant recipients given cyclosporine.<br />

Lancet 1983;2:571–572.<br />

23. Schlumpf R, Largiader F, Uhlschmid GK, Baumgartner<br />

D: Is cyclosporine toxic for transplanted pancreatic<br />

islets? Transplant Proc 1986;28:1169–1170.<br />

24. Van Schilfgaarde R, van der Burg MPM, van<br />

Suylichem HG, Goosen HG, Frolich M: Does cyclosporine<br />

influence beta cell function? Transplant<br />

Proc 1986;28:1175–1176.<br />

KIZILEL ET AL.<br />

25. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E,<br />

Warnock GL, Kneteman NM, Rajotte RV: Islet transplantation<br />

in seven patients with Type I diabetes<br />

mellitus using a glucocorticoid-free immunosuppressive<br />

regimen. N Engl J Med 2000;343:230–238.<br />

26. Ryan EA, Lakey JRT, Paty BW, Imes S, Korbutt GS,<br />

Kneteman, NM, Bigam D, Rajotte RV, Shapiro AM:<br />

Successful islet transplantation: continued insulin reverse<br />

provides long-term glycemic control. Diabetes<br />

2002;51:2148–2157.<br />

27. Brunicardi FC, Mullen Y: Issues in clinical islet transplantation.<br />

<strong>Pancreas</strong> 1994;9:281–290.<br />

28. De Vos P, Van Schilfgaarde R: Biocompatibility issues.<br />

In: Chick WL, ed. Cell Encapsulation Technology<br />

<strong>and</strong> <strong>The</strong>rapeutics. Boston: Birkhauser, 1999:<br />

63–79.<br />

29. De Vos P, Van Straaten JF, Nieuwenhuizen AG, de<br />

Groot M, Ploeg RJ, De Haan BJ, Van Schlifgaarde R:<br />

Why do microencapsulated islet grafts fail in the<br />

presence of fibrotic overgrowth? Diabetes 1999;48:<br />

1381–1388.<br />

30. Ortiz C, Zhang D, Xie Y, Davisson VJ, Ben-Amotz<br />

D: Identification of insulin variants using Raman<br />

spectroscopy. Anal Biochem 2004;332:245–252.<br />

31. Lanza RP, Ecker DM, Kuhtreiber WM, Marsh JP,<br />

Ringeling J, Chick WL: Transplantation of islets using<br />

microencapsulation: studies in diabetic rodents<br />

<strong>and</strong> dogs. J Mol Med 1999;77:206–210.<br />

32. Lim F, Sun AM: Microencapsulated islets as bioartificial<br />

endocrine pancreas. Science 1980;210:908–910.<br />

33. Soon-Shiong P, Heintz RE, Merideth N, Yao QX, Yao<br />

ZW, Zheng TL, Murphy M, Moloney MK, Schmehl<br />

M, Harris M, Mendez R, Mendez R, S<strong>and</strong>ford PA:<br />

Insulin independence in a type I diabetic patient after<br />

encapsulated islet transplantation. Lancet<br />

1994;343:950–951.<br />

34. Zekorn TD, Horcher A, Siebers U, Federlin K, Bretzel<br />

RG: Synergistic effect of microencapsulation <strong>and</strong><br />

immunoalteration on islet allograft survival in bioartificial<br />

pancreas. J Mol Med 1999;77:193–199.<br />

35. Omer A, Keegan M, Czismadia E, De Vos P, Van<br />

Rooijen N, Bonner-Weir S, Weir GC: Macrophage depletion<br />

improves survival of porcine neonatal pancreatic<br />

cell clusters contained in alginate macrocapsules<br />

transplanted into rats. Xenotransplantation<br />

2003;10:240–251.<br />

36. de Vos P, Hamel AF, Tatarkiewicz K: Considerations<br />

for successful transplantation of encapsulated pancreatic<br />

islets. Diabetologia 2002;45:159–173.<br />

37. De Vos P, Vegter D, De Haan BJ, Strubbe JH, Bruggink<br />

JE, Van Schilfgaarde R: Kinetics of intraperitoneally<br />

infused insulin in rats. Functional implications<br />

for the bioartificial pancreas. Diabetes 1996;<br />

45:1102–1107.<br />

38. Kessler L, Jesser C, Lombard Y, Karsten V, Belcourt<br />

A, Pinget M, Poindron P: Cytotoxicity of peritoneal<br />

murine macrophages against encapsulated pancreatic<br />

rat islets: in vivo <strong>and</strong> in vitro studies. J Leuk Biol<br />

1996;60:729–736.


THE BIOARTIFICIAL PANCREAS 981<br />

39. Toso C, Oberholzer J, Ceausoglu I, Ris F, Rochat B,<br />

Rehor A, Bucher P, W<strong>and</strong>rey C, Schuldt U, Belenger<br />

J, Bosco D, Morel P, Hunkeler D: Intra-portal injection<br />

of 400 microcapsules in a large animal model.<br />

Transplant Int 2003;16:405–410.<br />

40. Qian JH, Hashimoto T, Fujiwara H, Hamaoka T:<br />

Studies on the induction of tolerance of alloantigens.<br />

I. <strong>The</strong> abrogation of potentials for delayed-type-hypersensitivity<br />

responses to alloantigens by portal venous<br />

inoculations with allogeneic cells. J Immunol<br />

1985;134:3656–3661.<br />

41. Alej<strong>and</strong>ro R, Mintz DH, Noel J, Latif Z, Koh N, Russell<br />

E, Miller J: Islet cell transplantation in Type I<br />

diabetes mellitus. Transplant Proc 1987;19:2359–<br />

2361.<br />

42. Brendel MD, Hering B, Schultz AO, Bretzel RG:<br />

International Islet Transplant Registry. Newsletter<br />

8. Giessen, Germany: Justus-Liebig-University of<br />

Giessen, 1999.<br />

43. Pfeiffer EF: Artificial pancreas, glucose sensors <strong>and</strong><br />

the impact upon diabetology. Int J Artif Organs<br />

1993;16:636–644.<br />

44. Pfeiffer EF, Kerner W, Beischer W: Substitution of<br />

islet cell function by mechanical device: extracorporeal<br />

artificial pancreas. Adv Exp Med Biol 1979;<br />

119:501–508.<br />

45. Kruse-Jarres JD, Braun G, Naegele R, Bresch M,<br />

Lehmann U: Blood glucose monitoring <strong>and</strong> computer<br />

regulation by means of an artificial endocrine<br />

pancreas. Horm Metab Res Suppl 1979;8:42–45.<br />

46. Kerner W, Beischer W, Herfarth C, Pfeiffer EF: Application<br />

of an artificial endocrine pancreas in the<br />

management of the diabetic surgical patient. Horm<br />

Metab Res Suppl 1979;8:159–161.<br />

47. Hunkeler D: Allo transplants xeno: as bioartificial organs<br />

move to the clinic. Ann N Y Acad Sci 2001;944:<br />

1–6.<br />

48. Encapsulation & immunoprotective strategies of<br />

islet cells. Diabetes Technol <strong>The</strong>r 2002;4:361.<br />

49. Prokop A: <strong>Bioartificial</strong> pancreas: materials, devices,<br />

function <strong>and</strong> limitations. Diabetes Technol <strong>The</strong>r<br />

2001;3:431–449.<br />

50. Chang TMS: Semipermeable microcapsules. Science<br />

1964;146:524–525.<br />

51. Sun AM, O’Shea GM, Goosen MF: Injectable microencapsulated<br />

islet cells as bioartificial pancreas.<br />

Appl Biochem Biotechnol 1984;10:87–99.<br />

52. Wieg<strong>and</strong> F, Kroncke KD, Kolb-Bachofen V: Macrophage<br />

generated nitric-oxide as cytptoxic factor in<br />

destruction of alginate-encapsulated islets. Transplant<br />

Proc 1993;56:1206–1212.<br />

53. Soon-Shiong P, Otterlie M, Skjak-Braek G, Smidsrod<br />

O, Heintz R, Lanza RP, Espevik T: An immunologic<br />

basis for the fibrotic reaction to implanted microcapsules.<br />

Transplant Proc 1991;23:758–759.<br />

54. Str<strong>and</strong> BL, Ryan TL, In’t Veld P, Kulseng B, Rokstad<br />

AM, Skjak-Brek G, Espevik T: Poly-L-lysine induces<br />

fibrosis on alginate microcapsules via the induction<br />

of cytokines. Cell Transplant 2001;10:263–275.<br />

55. De Vos P, Wolters GHJ, Fritschy WM, Van Schilfgaarde<br />

R: Obstacles in the application of microencapsulation<br />

in islet transplantation. Int J Artif Organs<br />

1993;16:205–212.<br />

56. Goosen MFA: In: Lanza RR, Chick WL, eds. Immunoisolation<br />

of Pancreatic Islets. Austin, TX: R.G.<br />

L<strong>and</strong>es Co., 1994:21–44.<br />

57. Soon-Shiong P, Feldman E, Nelson R, Heintz R, Yao<br />

Q, Yao Z, Zheng T, Merideth N, Skjak-Braek G, Espevik<br />

T, Smidsrod O, S<strong>and</strong>ford P: Long-term reversal<br />

of diabetes by the injection of immunoprotected<br />

islets. Proc Natl Acad Sci U S A 1993;90:5843–5847.<br />

58. Lanza R, Langer R, Chick WL: Principles of Tissue<br />

Engineering, 2 nd ed. Orl<strong>and</strong>o, FL: Academic Press,<br />

2000.<br />

59. Colton CK, Avgoustiniatos ES: Bioengineering in development<br />

of the hybrid artificial pancreas. J Biomech<br />

Eng 1991;113:152–170.<br />

60. Brissova M, Lacik I, Powers AC, Anilkumar AV,<br />

Wang TG: Control <strong>and</strong> measurement of permeability<br />

for design of microcapsule cell delivery system.<br />

J Biomed Mater Res 1998;39:61–70.<br />

61. Cole DR, Waterfall M, McIntyre M, Baird JD: Microencapsulated<br />

islet grafts in the BB/E rat: a possible<br />

role for cytokines in graft failure. Diabetologia<br />

1992;35:231–237.<br />

62. Fan MY, Lum Z, Levesque L, Tai IT, Sun AM: Reversal<br />

of diabetes in BB rats by transplantation of encapsulated<br />

rat islets. Diabetes 1990;39:519–522.<br />

63. Iwata H, Takagi T, Kobayashi K, Oka T, Tsuki T, Ito<br />

F: Strategy for developing microbeads applicable to<br />

islet xenotransplantation into a spontaneous diabetic<br />

NOD mice. J Biomed Mater Res 1994;28:1201–1207.<br />

64. Lanza RR, Chick WL, eds.: Immunoisolation of Pancreatic<br />

Islets. Austin, TX: R.G. L<strong>and</strong>es Co., 1994.<br />

65. Lacik I, Brissova M, Anilkumar AV, Powers AC,<br />

Wang TG: New capsule with tailored properties for<br />

the encapsulation of living cells. J Biomed Mater Res<br />

1998;39:52–60.<br />

66. Lum ZP, Tai P, Krestow I, Tai T, Sun AM: Xenografts<br />

of microencapsulated rat islets into diabetic mice.<br />

Transplant Proc 1992;53:1180–1183.<br />

67. Iwata H, Murakami Y, Ikada Y: Control of complement<br />

activities for immunoisolation. Ann N Y Acad<br />

Sci 1999;875:7–23.<br />

68. Paul LC: Mechanism of humoral xenograft rejection.<br />

In: Cooper DKC, Kemp E, Reemtsma K, White D,<br />

eds. Xenotransplantation. Berlin: Springer-Verlag,<br />

1991:47–67.<br />

69. Auchincloss HJ, Moses R, Conti D, Sundt T, Smith<br />

C, Sachs DH, Winn HJ: Rejection of transgenic skin<br />

expressing a xeno-class I antigen is CD4-dependent<br />

<strong>and</strong> CD8-independent. Transplant Proc 1990;22:<br />

1059–1060.<br />

70. Sayegh MH, Watschinger B, Carpenter CB: Mechanisms<br />

of T cell recognition of alloantigen. <strong>The</strong> role<br />

of peptides. Transplantation 1994;57:1295–1302.<br />

71. Chen G, Hohmeier HE, Gasa R, Tran VV, Newgard<br />

CB: Selection of insulinoma cell lines with resistance


982<br />

to interleukin-1beta- <strong>and</strong> gamma-interferon-induced<br />

cytotoxicity. Diabetes 2000;49:562–570.<br />

72. Safley SA, Kapp LM, Tucker-Burden C, Hering B,<br />

Kapp JA, Weber CJ: Inhibition of cellular immune<br />

responses to encapsulated porcine islet xenografts<br />

by simultaneous blockade of two different costimulatory<br />

pathways. Transplantation 2005;79:409–418.<br />

73. Rafael E, Wu GS, Hultenby K, Tibell A, Wernerson<br />

A: Improved survival of macroencapsulated islets of<br />

Langerhans by preimplantation of the immunoisolating<br />

device: a morphometric study. Cell Transplant<br />

2003;12:407–412.<br />

74. Anderson JM: Inflammatory response to implants.<br />

ASAIO Trans 1988;34:101–107.<br />

75. Anderson JM: Mechanisms of inflammation <strong>and</strong> infection<br />

with implanted devices. Cardiovasc Pathol<br />

1993;2(Suppl):33S–41S.<br />

76. Anderson JM: Biocompatibility of tissue engineered<br />

implants. In: CW Patrick Jr, AG Mikos, LV McIntire,<br />

eds. Frontiers in Tissue Engineering. Oxford: Elsevier<br />

Science, 1998:152–165.<br />

77. Bonner-Weir S, Sharma A: Pancreatic stem cells. J<br />

Pathol 2002;197:519–526.<br />

78. Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin<br />

R, McKay R: Differentiation of embryonic stem cells<br />

to insulin-secreting structures similar to pancreatic<br />

islets. Science 2001;292:1389–1394.<br />

79. Peck AB, Cornelius JG, Schatz D, Ramiya VK: Generation<br />

of islets of Langerhans from adult pancreatic<br />

stem cells. J Hepatobil Pancreat Surg 2002;9:704–709.<br />

80. Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck<br />

AB, Cornelius JG: Reversal of insulin-dependent diabetes<br />

using islets generated in vitro from pancreatic<br />

stem cells. Nat Med 2000;6:278–282.<br />

81. Chen R, Meseck ML, Woo SL: Auto-regulated hepatic<br />

insulin gene expression in type I diabetic rats.<br />

Mol <strong>The</strong>r 2001;3:584–590.<br />

82. Lee HC, Kim SJ, Kim KS, Shin HC, Yoon JW: Remission<br />

in models of Type I diabetes by gene therapy<br />

using a single-chain insulin analogue. Nature<br />

2000;408:483–488.<br />

83. Tang SC, Sambanis A: Development of genetically<br />

engineered human intestinal cells for regulated insulin<br />

secretion using rAAV-mediated gene transfer.<br />

Biochem Biophys Res Commun 2003;303:645–652.<br />

84. Tang SC, Sambanis A: Preproinsulin mRNA engineering<br />

<strong>and</strong> its application to the regulation of insulin<br />

secretion from human hepatomas. FEBS Lett<br />

2003;537:193–197.<br />

85. Thule PM, Liu JM: Regulated hepatic insulin gene<br />

therapy of STZ-diabetic rats. Gene <strong>The</strong>r 2000;<br />

7:1744–1752.<br />

86. Thule PM, Liu J, Philips LS: Glucose regulated production<br />

of human insulin in rat hepatocytes. Gene<br />

<strong>The</strong>r 2000;7:205–214.<br />

87. Lumelsky N, Blondel O, Laeng P, Velasco I, Ravin<br />

R, McKay R: Differentiation of embryonic stem cells<br />

to insulin secreting structures similar to pancreaticislets.<br />

Science 2001;292:1389–1394.<br />

KIZILEL ET AL.<br />

88. Hori Y, Rulifson IC, Tsai BC, Heit JJ, Cahoy JD, Kim<br />

SK: Growth inhibitors promote differentiation of insulin-producing<br />

tissue from embryonic stem cells.<br />

Proc Natl Acad Sci U S A 2002;99:16105–16110.<br />

89. Blyszczuk P, Czyz J, Kania G, Wagner M, Roll U, St-<br />

Onge L, Wobus AM: Expression of Pax4 in embryonic<br />

stem cells promotes differentiation of nestinpositive<br />

progenitor <strong>and</strong> insulin-producing cells. Proc<br />

Natl Acad Sci U S A 2003;100:998–1003.<br />

90. Moritoh Y, Yamato E, Yasui Y, Miyazaki S, Miyazaki<br />

J: Analysis of insulin-producing cells during in vitro<br />

differentiation from feeder-free embryonic stem<br />

cells. Diabetes 2003;52:1163–1168.<br />

91. Segev H, Fishman B, Ziskind A, Shulman M, Itskovitz-Eldor<br />

J: Differentiation of human embryonic<br />

stem cells into insulin-producing clusters. Stem Cells<br />

2004;22:265–274.<br />

92. Hori Y, Gu X, Xie X, Kim SK: Differentiation of insulin-producing<br />

cells from human neural progenitor<br />

cells. PLoS Med 2005;2:e103.<br />

93. Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki<br />

KL, Tzukerman M: Insulin production by human<br />

embryonic stem cells. Diabetes 2001;50:1691–<br />

1697.<br />

94. Xu G, Stoffers DA, Habener JF, Bonner-Weir S: Exendin-4<br />

stimulates both beta-cell replication <strong>and</strong><br />

neogenesis, resulting in increased beta-cell mass <strong>and</strong><br />

improved glucose tolerance in diabetic rats. Diabetes<br />

1999;48:2270–2276.<br />

95. Gromada J, Brock B, Schmitz O, Rorsman P:<br />

Glucagon-like peptide-1: regulation of insulin secretion<br />

<strong>and</strong> therapeutic potential. Basic Clin Pharmacol<br />

Toxicol 2004;95:252–262.<br />

96. Harmon EB, Apelqvist AA, Smart NG, Gu X, Osborne<br />

DH, Kim SK: GDF11 modulates NGN3 islet<br />

progenitor cell number <strong>and</strong> promotes beta-cell differentiation<br />

in pancreas development. Development<br />

2004;131:6163–6174.<br />

97. Bach JF: Immunotherapy of insulin-dependent diabetes<br />

mellitus. Curr Opin Immunol 2001;13:601–605.<br />

98. Giannoukakis N, Rudert WA, Robbins PD, Trucco<br />

M: Targeting autoimmune diabetes with gene therapy.<br />

Diabetes 1999;48:2107–2121.<br />

99. Docherty K: Gene therapy for diabetes mellitus. Clin<br />

Sci (Lond) 1997;92:321–330.<br />

100. Soria B, Skoudy A, Martin F: From stem cells to beta<br />

cells: new strategies in cell therapy of diabetes mellitus.<br />

Diabetologia 2001;44:407–415.<br />

101. Selden RF, Skoskiewicz MJ, Russell PS, Goodman<br />

HM: Regulation of insulin-gene expression. Implications<br />

for gene therapy. N Engl J Med 1987;<br />

317:1067–1076.<br />

102. Newgard CB: Cellular engineering <strong>and</strong> gene therapy<br />

strategies for insulin replacement in diabetes. Diabetes<br />

1994;43:341–350.<br />

103. Halban PA, Kahn SE, Lernmark A, Rhodes CJ: Gene<br />

<strong>and</strong> cell-replacement therapy in the treatment of<br />

type 1 diabetes: how high must the st<strong>and</strong>ards be set?<br />

Diabetes 2001;50:2181–2191.


THE BIOARTIFICIAL PANCREAS 983<br />

104. Sapir T, Shternhall K, Meivar-Levy I, Blumenfeld T,<br />

Cohen H, Skutelsky E, Eventov-Friedman S, Barshack<br />

I, Goldberg I, Pri-Chen S, Ben-Dor L, Polak-<br />

Charcon S, Karasik A, Shimon I, Mor E, Ferber S:<br />

Cell-replacement therapy for diabetes: generating<br />

functional insulin-producing tissue from adult<br />

human liver cells. Proc Natl Acad Sci U S A<br />

2005;102:7964–7969.<br />

105. Fujita Y, Cheung AT, Kieffer TJ: Harnessing the gut<br />

to treat diabetes. Pediatr Diabetes 2004;5:57–69.<br />

106. Cheung AT, Dayan<strong>and</strong>an B, Lewis JT, Korbutt GS,<br />

Rajotte RV, Bryer-Ash M, Boylan MO, Wolfe MM,<br />

Kieffer TJ: Glucose-dependent insulin release from<br />

genetically engineered K cells. Science 2000;290:<br />

1959–1962.<br />

107. El-Shewy HM, Kendall WFJ, Darrabie M, Collins BH,<br />

Opara EC: Polyvinyl pyrrolidone: a novel cryoprotectant<br />

in islet cell cryopreservation. Cell Transplant<br />

2004;13:237–243.<br />

108. Zimmermann H, Katsen AD, Ihmig FR, Durst CHP,<br />

Shirley SG, Fuhr GR: First steps of an interdisciplinary<br />

approach towards miniaturised cryopreservation<br />

for cellular nanobiotechnology. IEE Proc Nanobiotechnol<br />

2004;151:134–138.<br />

109. Zimmermann H, Zimmermann D, Reuss R, Feilen<br />

PJ, Manz B, Katsen A, Weber M, Ihmig FR, Ehrhart<br />

F, Gessner P, Behringer M, Steinbach A, Wegner LH,<br />

Sukhorukov VL, Vasquez JA, Schneider S, Weber<br />

MM, Volke F, Wolf R, Zimmermann U: Towards a<br />

medically approved technology for alginate-based<br />

microcapsules allowing long-term immunoisolated<br />

transplantation. J Mater Sci Mater Med 2005;16:<br />

491–501.<br />

110. Misler S, Dickey A, Barnett DW: Maintenance of<br />

stimulus-secretion coupling <strong>and</strong> single beta-cell<br />

function in cryopreserved-thawed human islets of<br />

Langerhans. Pflugers Arch 2005;450:395–404.<br />

111. von Mach MA, Schlosser J, Weil<strong>and</strong> M, Feilen PJ,<br />

Ringel M, Hengstler JG, Weilemann LS, Beyer J,<br />

Kann P, Schneider S: Size of pancreatic islets of<br />

Langerhans: a key parameter for viability after cryopreservation.<br />

Acta Diabetol 2003;40:123–129.<br />

112. Lanza RP, Chick WL: Transplantation of encapsulated<br />

cells <strong>and</strong> tissues. Surgery 1997;121:1–9.<br />

113. Lanza RP, Chick WL: Immunoisolation strategies for<br />

the transplantation of pancreatic islets. Ann N Y<br />

Acad Sci 1997;831:323–331.<br />

114. Wang TG: <strong>Progress</strong> in bioartificial pancreas. J Regen<br />

Med 2000;1:93–98.<br />

115. Jain K, Yang H, Cai BR, Haque B, Hurvitz AI, Diehl<br />

C, Miyata T, Smith BH, Stenzel K, Suthanthiran M: Retrievable,<br />

replaceable, macroencapsulated pancreatic<br />

islet xenografts. Long-term engraftment without immunosuppression.<br />

Transplantation 1995;59:319–324.<br />

116. Trivedi N, Keegan M, Steil GM, Hollister-Lock J,<br />

Hasenkamp WM, Colton CK, Bonner-Weir S, Weir<br />

GC: Islets in alginate macrobeads reverse diabetes<br />

despite minimal acute insulin secretory responses.<br />

Transplantation 2001;71:203–211.<br />

117. Lacy PE, Hegre OD: Maintenance of normoglycemia<br />

in diabetic mice by subcutaneous xenografts of encapsulated<br />

islets. Science 1991;254:1782–1784.<br />

118. Scharp DW, Swanson CJ, Olack BJ, Latta PP, Hegre<br />

OD, Doherty EJ, Gentile FT, Flavin KS, Ansara MF,<br />

Lacy P: Protection of encapsulated human islets implanted<br />

without immunosuppression in patients<br />

with type I or type II diabetes <strong>and</strong> in nondiabetic<br />

control subjects. Diabetes 1994;43:1167–1170.<br />

119. van Suylichem PT, Strubbe JH, Houwing H, Wolters<br />

GH, van Schilfgaarde R: Insulin secretion by rat islet<br />

isografts of a defined endocrine volume after transplantation<br />

to three different sites. Diabetologia<br />

1992;35:917–923.<br />

120. Elliott RB, Escobar L, Garkavenko O, Croxson MC,<br />

Schroeder BA, McGregor M, Ferguson G, Beckman<br />

N, Ferguson S: No evidence of infection with porcine<br />

endogenous retrovirus in recipients of encapsulated<br />

porcine islet xenografts. Cell Transplant 2000;9:<br />

895–901.<br />

121. Sun YL, Ma X, Zhou D, Vacek I, Sun AM: Normalization<br />

of diabetes in spontaneously diabetic<br />

cynomologus monkeys by xenografts of microencapsulated<br />

porcine islets without immunosuppression.<br />

J Clin Invest 1996;98:1417–1422.<br />

122. Kendall WF, Collins BH, Hobbs HA, Bollinger RR,<br />

Opara EC: Long-term normoglycemia induced by<br />

microencapsulated porcine islet xenotransplantation<br />

in a diabetic baboon [abstract]. Acta Chir Austriaca<br />

2001;33(Suppl 174):56.<br />

123. Soon-Shiong P, Feldman E, Nelson R, Komtebedde<br />

J, Smidsrod O, Skjak-Braek G, Espevik T, Heintz R,<br />

Lee M: Successful reversal of spontaneous diabetes<br />

in dogs by intraperitoneal microencapsulated islets.<br />

Transplantation 1992;54:769–774.<br />

124. Sun Y, Ma X, Zhou D, Vacek I, Sun AM: Normalization<br />

of diabetes in spontaneously diabetic cynomologous<br />

monkeys by xenografts of microencapsulated<br />

porcine islets without immunosuppression. J Clin Invest<br />

1996;98:1417–1422.<br />

125. Uludag H, De Vos P, Tresco PA: Technology of<br />

mammalian cell encapsulation. Adv Drug Deliv Rev<br />

2000;42:29–64.<br />

126. Pollok JM, Kolin PA, Lorenzen M, Torok E, Kaufmann<br />

PM, Kluth D, Bohuslavizki KH, Gundlach M,<br />

Rogiers X: Islets of Langerhans encapsulated with a<br />

tissue engineered membrane of rat chondrocytes<br />

maintain insulin secretion <strong>and</strong> glucose insulin feedback<br />

for at least 30 days in culture. Transplant Proc<br />

2001;33:1713–1714.<br />

127. S<strong>and</strong>ler S, Andersson A, Eizirik DL, Hellerstrom C,<br />

Espevik T, Kulseng B, Thu B, Pipeleers DG, Skjak-<br />

Braek G: Assessment of insulin secretion in vitro<br />

from microencapsulated fetal porcine islet like cell<br />

clusters <strong>and</strong> rat, mouse, <strong>and</strong> human pancreatic islets.<br />

Transplantation 1997;63:1712–1718.<br />

128. Halle JP, Leblond FA, Pariseau JF, Jutras P, Brabant<br />

MJ, Lepage Y: Studies on small (300 microns) microcapsules:<br />

II—Parameters governing the produc-


984<br />

tion of alginate beads by high voltage electrostatic<br />

pulses. Cell Transplant 1994;3:365–372.<br />

129. Duvivier-Kali VF, Omer A, Parent RJ, O’Neil JJ, Weir<br />

GC: Complete protection of islets against allorejection<br />

<strong>and</strong> autoimmunity by a simple barium alginate<br />

membrane. Diabetes 2001;50:1698–1705.<br />

130. Klock G, Frank H, Houben R, Zekorn T, Horcher A,<br />

Siebers U, Wöhrle M, Federlin K, Zimmermann U:<br />

Production of purified alginates suitable for use in<br />

immunoisolated transplantation. Appl Microbiol<br />

Biotechnol 1994;40:638–643.<br />

131. Kulseng B, Thu T, Espevik T, Skjk-Brk G: Alginate<br />

poly-lysine microcapsules as immune barrier: permeability<br />

of cytokines <strong>and</strong> immunoglobins over the<br />

capsule membrane. Cell Transplant 1997;6:387–394.<br />

132. Sawhney AS, Hubbell JA: Poly(ethylene oxide)graft-poly(L-lysine)<br />

copolymers to enhance the biocompatibility<br />

of poly(L-lysine)-alginate microcapsule<br />

membranes. Biomaterials 1992;13:863–870.<br />

133. Lim F: Microencapsulation of living mammalian<br />

cells. Adv Biotechnol Processes 1988;7:185–197.<br />

134. Smidsrod O, Skjak-Braek G: Alginate as immobilization<br />

matrix for cells. Trends Biotechnol 1990;8:<br />

71–78.<br />

135. Prokop A, Wang TG: Purification of polymers used<br />

for fabrication of an immunoisolation barrier. Ann<br />

N Y Acad Sci 1997;831:223–231.<br />

136. Zekorn T, Klock G, Horcher A, Siebers U, Wohrle M,<br />

Kowalski M, Arnold M, Federlin K, Bretzel R, Zimmermann<br />

U: Lymphoid activation by different crude<br />

alginates <strong>and</strong> the effect of purification. Transplant<br />

Proc 1992;24:2952–2953.<br />

137. Ricker A, Stockberger S: Hyperimmune response to<br />

microencapsulated xenogeneic tissue in NOD mice<br />

[abstract]. Diabetes 1986;35:61A.<br />

138. Garfinkel MR, Harl<strong>and</strong> RC, Opara EC: Optimization<br />

of the microencapsulated islet for transplantation. J<br />

Surg Res 1998;76:7–10.<br />

139. Ar’Rajab A, Bengmark S, Ahren B: Insulin secretion<br />

in streptozotocin-diabetic rats transplanted with immunoisolated<br />

islets. Transplantation 1991;51:571–<br />

574.<br />

140. Calafiore R, Calcinaro F, Basta G, Pietropoalo M,<br />

Falorni A, Piermattei M, Brunetti P: A method for<br />

the massive separation of highly purified, adult<br />

porcine islets of Langerhans. Metabolism 1990;39:<br />

175–181.<br />

141. Carani BS, Edwin G, eds.: Annals of the New York<br />

Academy of Sciences, Vol. 1037: Immunology of Diabetes<br />

III. New York: New York Academy of Sciences,<br />

2005.<br />

142. Mazaheri R, Atkison P, Stiller C, Dupre J, Vose J,<br />

O’Shea G: Transplantation of encapsulated allogeneic<br />

islets into diabetic BB/W rats. Effects of immunosuppression.<br />

Transplantation 1991;51:750–754.<br />

143. Sefton MV, Stevenson WTK: Microencapsulation of<br />

live animal cells using polyacrylates. Adv Polym Sci<br />

1993;107:145–198.<br />

144. Dupuy B, Gin H, Baquey C, Ducassou D: In situ<br />

polymerization of a microencapsulating medium<br />

KIZILEL ET AL.<br />

round living cells. J Biomed Mater Res 1988;22:<br />

1061–1070.<br />

145. Gin H, Dupuy B, Baquey C, Ducassou D, Aubertin<br />

J: Agarose encapsulation of islets of Langerhans: reduced<br />

toxicity in vitro. J Microencapsul 1987;4:<br />

239–242.<br />

146. Takagi T, Iwata H, Tashiro H, Tsuji T, Ito F: Development<br />

of a novel microbead applicable to xenogeneic<br />

islet transplantation. J Control Rel 1994;31:<br />

283–291.<br />

147. Iwata H, Murakami Y, Ikada Y: Control of complement<br />

activities for immunoisolation. Ann N Y Acad<br />

Sci 1999;875:7–23.<br />

148. Deppsich R, Goehl H, Ritz E, Haensch GM: Complement<br />

activation on artificial surfaces in biomedical<br />

therapies. In: Rother K, Till GO, Hänsch GM, eds.<br />

<strong>The</strong> Complement System, 2 nd ed. Berlin: Springer-<br />

Verlag, 1998, pp. 487–504.<br />

149. Pangburn MK: Alternative pathway: activation <strong>and</strong><br />

regulation. In: Rother K, Till GO, Hänsch GM, eds.<br />

<strong>The</strong> Complement System, 2 nd ed. Berlin: Springer-<br />

Verlag, 1998:93–115.<br />

150. Chaikof EL: Engineering <strong>and</strong> material considerations<br />

in islet cell transplantation. Annu Rev Biomed<br />

Eng 1999;1:103–127.<br />

151. Shoichet MS, Winn SR, Gentile FT, Athavale S, Harris<br />

JM: Poly(ethylene oxide) grafted thermoplastic<br />

membranes for use as cellular hybrid bioartificial organs<br />

in the central nervous system. Biotechnol Bioengin<br />

1994;43:563–572.<br />

152. Pathak CP, Sawhney AS, Hubbell JA: Rapid photopolymerization<br />

of immunoprotective gels in contact<br />

with cells <strong>and</strong> tissue. J Am Chem Soc 1992;<br />

114:8311–8312.<br />

153. Cruise GM, Hegre OD, Scharp DS, Hubbell JA: A<br />

sensitivity study of the key parameters in the interfacial<br />

photopolymerization of poly(ethylene glycol)<br />

diacrylate upon porcine islets. Biotechnol Bioengin<br />

1998;57:655–665.<br />

154. Harris JM: Poly(ethylene Glycol) Chemistry: Biotechnical<br />

<strong>and</strong> Biomedical Applications. New York:<br />

Plenum Press, 1992.<br />

155. Sawhney AS, Pathak CP, Hubbell JA: Bioerodible<br />

hydrogels based on photopolymerized poly(ethylene<br />

glycol)-co-poly(-hydroxy acid) diacrylate<br />

macromers. Macromolecules 1993;26:581–587.<br />

156. Cruise GM, Scharp DS, Hubbell JA: Characterization<br />

of permeability <strong>and</strong> network structure of interfacially<br />

photopolymerized poly(ethylene glycol)<br />

diacrylate hydrogels. Biomaterials 1998;19:1287–<br />

1294.<br />

157. Wolters GH, Fritschy WM, Gerrits D, Van Schilfgaarde<br />

R: A versatile alginate droplet generator applicable<br />

for microencapsulation of pancreatic islets.<br />

J Appl Biomater 1992;3:281–286.<br />

158. Opara EC: <strong>The</strong> therapeutic potential of islet cell<br />

transplants in the treatment of diabetes. Expert Opin<br />

Invest Drugs 1998;7:1–11.<br />

159. Hobbs H, Kendall W, Darrabie M, Collins B, Bridges<br />

S, Opara EC: Substitution of polyornithine for


THE BIOARTIFICIAL PANCREAS 985<br />

polylysine in alginate microcapsules [abstract]. Diabetes<br />

2000;49:A111.<br />

160. Hommel M, Sun AM, Goosen MFA, inventors; Connaught<br />

Laboratories Limited, assignee: Droplet generation.<br />

US Patent 4,956,128. September 11, 1990.<br />

161. Sundberg C: High-throughput <strong>and</strong> ultra-highthroughput<br />

screening: solution- <strong>and</strong> cell-based approaches.<br />

Curr Opin Biotechnol 2000;11:47–53.<br />

162. Kane RS, Takayama S, Ostuni E, Ingber DE, Whitesides<br />

GM: Patterning proteins <strong>and</strong> cells using soft<br />

lithography. Biomaterials 1999;20:2363–2376.<br />

163. Singhvi R, Kumer A, Lopez GP, Stephanopoulos GN,<br />

Wang DIC, Whitesides GM, Ingber DE: Engineering<br />

cell shape <strong>and</strong> function. Science 1994;264:696–698.<br />

164. Amirpour ML, Ghosh P, Lackowski WM, Crooks<br />

RM, Pishko MV: Mammalian cell cultures on micropatterned<br />

surfaces of weak-acid, polyelectrolyte<br />

hyperbranched thin films on gold. Anal Chem<br />

2001;73:1560–1566.<br />

165. Chen CS, Mrksich M, Huang S, Whitesides GM, Ingber<br />

DE: Micropatterned surfaces for control of cell<br />

shape, position, <strong>and</strong> function. Biotechnol Prog<br />

1998;14:356–363.<br />

166. Ito Y: Surface micropatterning to regulate cell functions.<br />

Biomaterials 1999;20:2333–2342.<br />

167. Koh W, Revzin A, Simonian A, Reeves T, Pishko M:<br />

Control of mammalian cell <strong>and</strong> bacteria adhesion on<br />

substrates micropatterned with poly(ethylene glycol)<br />

hydrogels. Biomed Microdev 2003;5:11–19.<br />

168. Matsuda T, Sugawara T: Development of surface<br />

photochemical modification method for micropatterning<br />

of cultured cells. J Biomed Mater Res<br />

1995;29:749–756.<br />

169. Kizilel S, Perez-Luna VH, Teymour F: Photopolymerization<br />

of poly(ethylene glycol) diacrylate on<br />

eosin-functionalized surfaces. Langmuir 2004;20:<br />

8652–8658.<br />

170. Desai TA, West T, Cohen M, Boiarski T, Rampersaud<br />

A: Nanoporous microsystems for islet cell replacement.<br />

Adv Drug Deliv Rev 2004;56:1661–1673.<br />

171. Cohen I, Li H, Hougl<strong>and</strong> JL, Mrksich M, Nagel SR:<br />

Using selective withdrawal to coat microparticles.<br />

Science 2001;292:265–267.<br />

172. Patience C, Takeuchi Y, Weiss RA: Infection of human<br />

cells by an endogeneous retrovirus of pigs. Nat<br />

Med 1997;3:282–286.<br />

173. van der Laan LJW, Lockey C, Griffeth BC, Frasier FS,<br />

Wilson CA, Onions DE, Hering BJ, Long Z, Otto E,<br />

Torbett BE, Salomon DR: Infection by porcine endogeneous<br />

retrovirus after islet xenotransplantation in<br />

SCID mice. Nature 2000;407:90–94.<br />

174. Heneine W, Tibell A, Switzer WM, S<strong>and</strong>strom P,<br />

Rosales GV, Mathews A, Korsgren O, Chapman LE,<br />

Folks TM, Groth CG: No evidence of infection with<br />

porcine endogeneous retrovirus in recipients of<br />

porcine islet-cell xenografts. Lancet 1998;352:695–<br />

699.<br />

175. Paradis K, Langford G, Long Z, Heneine W, S<strong>and</strong>strom<br />

P, Switzer WM, Chapman LE, Lockey C,<br />

Onions D, Otto E: Search for cross species transmis-<br />

sion of porcine endogeneous retrovirus in patients<br />

treated with living pig tissue. <strong>The</strong> XEN 111 Study<br />

Group. Science 1999;285:1236–1241.<br />

176. Niebert M, Rogel-Gaillard C, Chardon P, Tonjes RR:<br />

Characterization of chromosomally assigned replication-component<br />

gamma porcine endogenous<br />

retroviruses derived from a large white pig <strong>and</strong> expression<br />

in human cells. J Virol 2002;76:2714–2720.<br />

177. Soria B, Roche E, Berna G, Leon-Quinto T, Reig JA,<br />

Martin F: Insulin secreting cells derived from embryonic<br />

stem cells normalize glycemia in streptozotocin<br />

induced diabetic mice. Diabetes 2000;49:157–162.<br />

178. Soria B, Andreu E, Berna G, Fuentes E, Gil A, Leon-<br />

Quinto T, Martin F, Montanya E, Nadal A, Reig JA,<br />

Ripoll C, Roche E, Sanchez-Andres JV, Segura J: Engineering<br />

pancreatic islets. Pflugers Arch 2000;<br />

440:1–18.<br />

179. Ramiya VK, Maraist M, Arfors KE, Schatz DA, Peck<br />

AB, Cornelius JG: Reversal of insulin dependent diabetes<br />

using islets generated in vitro from pancreatic<br />

stem cells. Nat Med 2000;6:278–282.<br />

180. Zhang N, Richter A, Suriawinata J, Harbaran S, Altomonte<br />

J, Cong L, Zhang H, Song K, Meseck M,<br />

Bromberg J, Dong H: Elevated vascular endothelial<br />

growth factor production in islets improves islet<br />

graft vascularization. Diabetes 2004;53:963–970.<br />

181. Sigrist S, Mechine-Neuville A, M<strong>and</strong>es K, Calenda<br />

V, Braun S, Legeay G, Bellocq JP, Pinget M, Kessler<br />

L: Influence of VEGF on the viability of encapsulated<br />

pancreatic rat islets after transplantation in diabetic<br />

mice. Cell Transplant 2003;12:627–635.<br />

182. Narang AS, Cheng K, Henry J, Zhang C, Sabek O,<br />

Fraga D, Kotb M, Gaber AO, Mahato RI: Vascular<br />

endothelial growth factor gene delivery for revascularization<br />

in transplanted human islets. Pharm Res<br />

2004;21:15–25.<br />

183. Kizilel S: <strong>The</strong>oretical <strong>and</strong> experimental investigation<br />

for interfacial photopolymerization of poly(ethylene<br />

glycol) diacrylate [Ph.D. thesis]. Chicago: Biomedical<br />

Engineering, Illinois Institute of Technology, 2004.<br />

184. Kizilel S, Sawardecker E, Teymour F, Perez-Luna<br />

VH: Sequential formation of covalently bonded hydrogel<br />

multilayers through surface initiated photopolymerization.<br />

Biomaterials 2005 Sept 10 [Epub<br />

ahead of print].<br />

185. De Vos P, De Haan BJ, Van Schilfgaarde R: Upscaling<br />

the production of microencapsulated pancreatic<br />

islets. Biomaterials 1997;18:1085–1090.<br />

Address reprint requests to:<br />

Emmanuel C. Opara, Ph.D.<br />

Pritzker Institute of Biomedical Science &<br />

Engineering<br />

Illinois Institute of Technology<br />

10 West 32 nd Street, E1-116<br />

Chicago, IL 60616<br />

E-mail: opara@iit.edu

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!