“What we think, or what we know, or what we believe is, in the end, of little consequence. The only consequence is what we do.” John Ruskin (1819 - 1900)

For my family Till min familj Für meine Familie A családomnak

List of Papers

This thesis is based on the following papers, which are referred to in the text by their Roman numerals.

I Brandhorst, H., Friberg, A., Andersson, H.H., Felldin, M., Foss, A., Salmela, K., Lundgren, T., Tibell, A., Tufveson, G., Korsgren, O., Brandhorst, D. (2009) The importance of tryptic- like activity in purified enzyme blends for efficient islet isola- tion. Transplantation, 87:370-375

II Friberg, A.S., Ståhle, M., Brandhorst, H., Korsgren, O., Brand- horst, D. (2008) Human islet separation utilizing a closed auto- mated purification system. Cell Transplantation, 17:1305-1313

III Friberg, A.S., Brandhorst, H., Buchwald, P., Goto, M., Ricordi, C., Brandhorst, D., Korsgren, O. (2011) Quantification of the islet product: presentation of a standardized current good manu- facturing practices compliant system with minimal variability. Transplantation, 91(6):677-683

IV Friberg, A.S., Lundgren, T., Malm, H., Nilsson, B., Felldin, M., Jenssen, T., Kyllönen, L., Tufveson, G., Tibell, A., Kor- sgren, O. Transplantable functional islet mass – predictive bio- markers of graft function in islet after kidney transplanted pa- tients. Manuscript

Reprints were made with permission from the respective publishers.

Contents

Introduction ...... 11 Aims ...... 12 General aims ...... 12 Specific aims ...... 12 Background ...... 13 History of mellitus ...... 13 From antiquity to ...... 13 The big discovery! ...... 14 Forms of diabetes and epidemiology ...... 15 Transplantation as therapy for diabetes ...... 16 Islet transplantation ...... 18 Short history of clinical islet transplantation ...... 18 Islet isolation procedure ...... 19 Regulatory considerations...... 19 Islet donors and organ transport ...... 21 Enzymes for islet isolation ...... 21 Islet purification ...... 23 Islet culture...... 24 Islet isolation quality control ...... 26 Measuring clinical success ...... 28 Materials and methods ...... 31 Results and discussion ...... 34 Identification of a previously unrecognized enzyme activity (Paper I) ..... 34 Evaluation of enzymatic digestion of rat ...... 34 Evaluation of enzymatic digestion of human pancreas ...... 35 Progress in the field since Paper I was published ...... 36 Automated gradient making system (Paper II) ...... 36 Functionality of pump-made gradients ...... 37 Islet separation with pump-made gradients ...... 37 Progress in the field since Paper II was published (2008) ...... 38 Digital imaging analysis (DIA) and presentation of a GMP-friendly islet quantification technique (Paper III) ...... 38 DIA validation ...... 39

Computer-assisted DIA versus manual methods ...... 39 Evaluator versus sample variation ...... 39 Closed system islet evaluation ...... 40 Progress in the field since Paper III was published (2011) ...... 40 Prediction of islet graft potency (Paper IV) ...... 41 Correlations to short term outcome ...... 42 Transplanted functional islet mass (TFIM) model ...... 43 Conclusions ...... 45 Specific conclusions ...... 45 General conclusion...... 45 Critical considerations regarding research design and methods ...... 46 Paper I ...... 46 Paper II ...... 46 Paper III ...... 46 Paper IV ...... 47 Future perspectives ...... 48 Popular science summary ...... 51 Svensksammanfattning ...... 53 Acknowledgments ...... 55 References ...... 61

Abbreviations

AIRarg Acute insulin response to argentine APS Automated purification system BAEE N-benzoyl-L-arginine-ethyl ester BD Brain death BMI Body mass index C-peptide Connecting-peptide CI Collagenase class I CII Collagenase class II CIT Cold ischemia time CV% Coefficient of variation × 100% CP/GCr Change in C-peptide × glucose-1 × creatinine-1 ratio DM Diabetes mellitus DMC Dimethylcasein DIA Digital imaging analysis DTZ Dithiocarbazone, dithizone ELISA Enzyme-linked immunosorbent assay EP European parliament GMP Good manufacturing practices HbA1c Hemoglobin A1c HTK Histidine-tryptophan-ketogluterate IAK Islet after kidney IE Islet equivalent IL Interleukin IVGTT Intravenous glucose tolerance test MCP-1 Macrophage chemotractant protein-1 MMTT Mixed meal tolerance test NP Neutral protease PP-cells Pancreatic polypeptide cells PZ 4-phenylazobenzyloxycarbonyl-L-prolyl-L- leucylglycyl-L-prolyl-D-arginine SEM Standard error of the mean SGM Standard gradient maker SUIT Secretory units of islet in transplantation T1DM Type 1 diabetes mellitus T2DM Type 2 diabetes mellitus TF Tissue factor

TLM Two-layer method UW University of Wisconsin solution

Introduction

Diabetes is a widespread and increasingly prevalent disease. Currently, - cell replacement therapy via transplantation of islets of Langerhans is a via- ble means to maintain control of blood sugar levels and reduce the risk of hypoglycemia in defined populations of patients with brittle type I diabetes mellitus or those requiring partial or whole pancreatectomy. The process of islet isolation across centers suffers from variability and, despite important advances, remains to be standardized. Standardization of the isolation process, quality control parameters, quantification before transplant, and even variables associated with transplant outcome are needed for meaningful comparisons between labs. In this thesis some variables affecting islet isolation success, evaluation of end product, and short-term islet engraftment are uncovered, addressed and evaluated. A previously disregarded enzyme activity, tryptic-like activity, has been identified to influence pancreas digestion efficiency and islet isola- tion success in both the preclinical and clinical situations. An effectively closed, automated pump system for the consistent and flexible creation of density gradients for separation of islet from non-islet tissue was developed. Islet quantification as evaluated with computer-assisted digital imaging analysis and introduction of a closed system that allows for the evaluation of the entire preparation at once is presented. Establishment of a hereto rela- tively unused transplant evaluation parameter, CP/GCr, was used to identi- fy donor, islet isolation, and quality control variables associated with early graft function in an islet after kidney patient population. Through standardization, isletologists will be better equipped to advance the field more cheaply and efficiently with greater security and certainty. This thesis addresses aspects critical to the standardization of human pan- creas processing for islet transplantation as well as some of those essential for evaluation of transplant function in the clinic.

11

Aims

General aims The work contained in this thesis was carried out to improve reproducibility, safety, and reliability of islet isolation parameters and identify variables af- fecting transplantation outcome through standardization and adherence to good manufacturing practices where relevant.

Specific aims

Paper I To characterize efficient enzyme blends by evaluating the effect of a tryptic-like activity on rat and human islet isolation outcome To evaluate the effect of tryptic-like activity on islet viability, mor- phology and function in vitro and in vivo

Paper II To standardize the generation of continuous density gradients for use in human islet purification using an automated, closed system gra- dient making procedure

Paper III To identify sources of evaluation error in the quantification of islet isolation products To improve upon the widely used standard manual counting proce- dure with a GMP-friendly islet evaluation method

Paper IV To identify factors predictive of early islet engraftment in islet after kidney transplant patients as measured by the change in pre- to post- transplant C-peptide/(glucose × creatinine) ratios To create a best-fit model for prediction of early islet engraftment

12

Background

History of diabetes mellitus From antiquity to insulin “Diabetes is a dreadful disease, not very frequent among men, being a melting down of the flesh and limbs into urine. The patients never stop making water and the flow is incessant, like the opening of aquaducts.” Aretaeus of Cappadocia, ca. 100 AD

Descriptions of diabetes can be traced back to antiquity. In Thebes, Egypt, a document that came to be known as the Ebers Papyrus from circa 1535 BC, found between legs of a mummy, contains the first known description of a condition to what today could be called diabetes. It describes a patient where the body has “shrunken with disease” and suggests remedies “for the sup- pression of his thirst (and) for curing his mortal illness” (1). Sushruta an Indian surgeon from around 600 BC described a condition that produced “madhumeha” or honey-like urine. He also advised for the sedentary to be- come more active in the pursuit of curing the disease (2). In China about 400 BC in the Yellow Emperor’s Canon on the Traditional Chinese Medicine, the oldest Chinese medical book, a condition called “XiaoKe symptom”, meaning weight loss due to thirst is described. The first authoritative de- scription of diabetes is from the 2nd century AD when the physician Aretaeus gave the condition the name “diabetes” meaning “siphon”, used to describe the excessive flow of urine associated with the disease (see quote above). The Latin term mellitus, which means “sweet honey”, was added to the name diabetes as introduced by Thomas Willis in 1674 (3). In 1776, Mat- thew Dobson noted that serum was sweet in taste as was the urine and con- cluded that the sweetness was due to sugar after boiling down 2 quarts of urine (4). The organ responsible for diabetes mellitus (DM) was long thought to be the kidney due to the excessive urination. Thomas Cawley, upon autopsy of a patient with diabetes, noted a shrunken pancreas with stones, however, no particular connection to diabetes was raised at the time of the report in 1788 (5). In 1797, John Rollo described different sugar levels in the urine of a patient with diabetes based on the type of foods ingested. An “animal” diet (i.e. meat) was recommended to reduce sugar excretion (6). Paul Langerhans, in his 1869 doctoral thesis, described for the first time round groups of cells spread out at regular intervals throughout the pancrea-

13

tic parenchyma. He himself had no knowledge of the function of the cell clusters he described (7). The name “islets of Langerhans” was given by Frenchman Edouard Laguesse in 1893 in recognition of Langerhans’ thesis work (8). Unfortunately, Langerhans passed away in 1888, the year before the coupling of the pancreas to DM was realized. Joseph von Mering and Oskar Minkowski removed the of two dogs and, with the organs re- moved, the animals displayed the characteristics of human DM almost im- mediately. When placing a small portion of pancreas in a newly pancreatec- tomized dog, hyperglycemia (elevated blood sugar levels, a defining charac- teristic of DM) was avoided until removal or degeneration of the organ (9). In 1894, P. Watson Williams transplanted 3 small sheep’s pancreas segments under the skin of a young boy who died 3 days later. The boy’s pancreas noted as “small, shriveled in appearance” (10). After analyzing the cadaver pancreases of those who had diabetes or pan- creatic disease the pathologist Eugene Opie in 1901 made the connection of the damaged islets of Langerhans to the metabolic disorder (11). The “inter- nal secretion” of the pancreas leading to diabetes was linked to the islets of Langerhans and the race to isolate the secretion was on. The name for the substance providing the internal secretion was given by Meyer in 1909 as “insuline” in reference to the Greek word insula meaning “island” (12).

The big discovery! There were many failed attempts to isolate the internal secretion of the pan- creas, some more successful than others, but it wasn’t until 1921 that the work leading to widespread therapy was initiated (13). In the hot summer of 1921, Fredrick G. Banting convinced James R. Macleod to give him some lab space and help from his technicians. In the end it was Charles H. Best who assisted Banting, both initially unpaid, who isolated the internal secre- tion of the pancreas. From May to the end of July they produced the initial results of injecting “internal secretions” of the pancreas into dogs (14). Eventually, they convinced Macleod to involve Joseph B. Collip, a biochem- ist, to assist in the purification of what was initially named “isletin”, later named “insulin” upon the recommendation of Macleod. On January 11, 1922, a 14 year old patient was the first patient to receive insulin injections, one in each buttocks, and went on to live to 27 years of age (15). In 1923, Banting and Macleod shared the Nobel Prize for their efforts. Banting im- mediately shared half of his prize with Best and Macleod half of his with Collip. Eli Lilly and Company was approached to scale up, refine production and make insulin available to the general public (15). Banting and Best sold the rights to insulin for $1 to the University of Toronto (which Best does not recall ever receiving) (16).

14

Forms of diabetes and epidemiology Standardization of treating diabetes came already in 600 BC when Sushruta delineated between two major forms of the disease. In the1880s Etienne Lancereaux dubbed them diabetes maigre and diabetes gras which mean diabetes of the “thin” and “fat”, respectively (17). It was not until 1979 that the classifications of type 1 (T1DM), previously called insulin-dependent DM or juvenile diabetes, and type 2 (T2DM), previously called non-insulin- dependent DM or adult-onset diabetes, came into being (18). Of all patients with DM globally, roughly 10% have T1DM and the other 90% are considered to have T2DM. For the purposes of this thesis the focus of T1DM and its complications are considered, however effects of hypergly- cemia as discussed in the next sections affect those with T2DM as well.

Type 1 diabetes mellitus T1DM is typically attributed to destruction of the -cells (19) the only cells in the body that produce insulin. The absence of -cells and/or the inability to produce enough insulin requires the exogenous use of the for alleviation of hyperglycemia and to confer long-term survival. T1DM can occur at any age but typically develops in children or young adults. The causes of T1DM remain largely unknown, and despite a number of theories (20), it remains unclear as to what exactly may trigger its development and, correspondingly, there is no known cure. The incidence of T1DM is also increasing worldwide (21-25). Geograph- ic and seasonal changes in incidence of T1DM have been observed (26). Ethnicity also associates with differences in rates of diagnoses within the same country (27). Depending on the population considered, T1DM inci- dence is anywhere from 0.1 (China and Venezuela) to 37 cases per 100,000 (Finland and Sardinia) (28). In another study considering the Finnish popula- tion, in 1980 there were 31.4 cases per 100,000 per year (age-adjusted inci- dence) whereas in 2005 this rose to 64.2 per 100,000 per year (29) and the rate of the increase of incidence is about 4% annually (29, 30). Contrary to the Finnish reports is a recent Swedish report wherein the population demo- graphics point to a tapering of T1DM incidence in the last decade (31).

Type 2 diabetes mellitus T2DM is characterized by both insulin resistance and dysregulation of glu- cose metabolism. It is associated with age, obesity and physical inactivity among others (27). The latest data from the United States Center for Disease Control concerning prevalence of diabetes estimates 25.8 million or 8.3% of the US population has diabetes, both diagnosed and undiagnosed (27). T2DM incidence is also increasing at an alarming rate not only among adults but also in cases involving children (30). Increased physical activity and diet restrictions are typically prescribed, as is insulin in some cases.

15

Complications of diabetes Living with chronic hyperglycemia can lead to many serious and life threat- ening complications and morbidity (32). Some complications include de- struction of microvasculature leading to retinopathy, nephropathy, slow wound healing, and in some severe cases blindness, dialysis and/or amputa- tion. Since morality has been related to the progression of renal dysfunction (33) treating diabetes may prolong lifespan. Diabetes is a major risk factor for cardiovascular disease as well (34). Neuropathy and loss of feeling over time, reduced life-expectancy (32) and quality of life (35) are unfortunate consequences of diabetes for many. Intensive insulin treatment with the goal of normoglycemia can reduce the risk for some of these complications but can lead to increased episodes of hypoglycemia in some cases (36). Hypog- lycemic episodes, seen most often in long-standing T1DM patients with glycemic lability, which if serious enough and left acutely untreated, can be life threatening. In cases where frequently low blood sugars are encountered a condition known as hypoglycemia unawareness can develop. In hypogly- cemia unawareness the patient does not recognize dangerously low blood sugars and the body’s own counterregulatory mechanisms are insufficient to mount a sufficient response (37) which can lead to diabetic coma and/or death (32).

Costs of diabetes As diabetes incidence increases so will the economic costs, which are al- ready staggering. In 2007 in the US alone, diabetes accounted for $174 bil- lion in healthcare costs (27). That equates to $580/person/year in the US, less than the cost of a full gym membership. Compared to non-diabetic indi- viduals a person with T2DM has three-fold higher healthcare costs, a person with T1DM six- to seven-fold higher (38). There is also a personal cost as- sociated with diabetes, possibly life-threatening hypoglycemia, and is the source of worry in this patient group (39).

Transplantation as therapy for diabetes Pancreas versus islet transplantation Since the first pancreas transplantations in humans were performed 1966 (40), over 30,000 vascularized pancreas transplantations have been per- formed (41). Pancreas transplantation is usually offered to patients who have total loss of -cell function and even in some cases of T2DM (42). Pancreas transplantation is associated with a high level of -cell functional capacity and typically the patients have immediate benefits of normalized glycemia (43). Risks of pancreas transplantation mainly include morbidity and mortal- ity associated with the surgical complications (44). These patients require lifelong immunosuppression.

16

A less invasive alternative to pancreas transplantation is islet transplanta- tion. Recipients of islet allotransplants can enjoy reduction of glycemic labil- ity, including reduction of insulin requirements and reduced episodes of hypoglycemia even five years after transplant (45, 46). Measurements of hemoglobin (Hb)A1c, which is an indication of long-term (~3 month aver- age) glycemia, typically increase with diabetes onset (47). Indeed, reduced HbA1c levels that are observed in the majority of islet recipients (45, 46, 48). Subjects with diabetes undergoing dialysis may have beneficial effects in terms of reduced macro- and microangiopathy as well as increased long-term survival if a kidney transplant is combined with a simultaneous islet trans- plant (48). Islet graft recipients currently require immunosuppression. The negative side effects of immunosuppression must be weighed against what is gained by the transplant. Current immunosuppressive regimens can cause side effects such as mouth ulcers, anemia, diarrhea, hypertension, de- terioration of renal function, and more (45, 49, 50). Despite these side ef- fects, overall patients’ quality of life measurements indicate a positive out- look towards islet transplantation as diabetes therapy (51). A necessity of islet transplantation is the proper use of scarce organs for transplantation purposes (52). In whole pancreas transplantation, organs from larger donors are associated with increased risk of graft failure (53) whereas they are preferred for islet cell processing (54). Allocation of organs deemed non-ideal for whole organ transplantation yet acceptable for islet processing can yield optimal distribution of available tissues (52).

Cost analysis of islet versus pancreas transplantation Cost analysis of islet and whole pancreas transplantation has been performed (55, 56). In a study of a Swiss-French islet transplant consortium, cost for islet transplantation was slightly higher than pancreas transplantation (56). A major conclusion of that analysis was that “a better reproducibility of islet processing will be essential for the diffusion of this technology” (56). Less than 20% of the pancreases they accepted resulted in transplantation (56), a number lower than current transplantation rates at other centers (57-59).

Indications for islet transplant Patients qualifying for islet transplants are usually over 18 years of age and present with labile diabetes despite intensive insulin management, they have had diabetes a minimum of 5 years, have documented hypoglycemia un- awareness or related autonomic non-responsiveness, absence of stimulated C-peptide (<0.2 ng/ml) to arginine stimulation test, and progressive second- ary complications). Patients receiving islet transplants should also have sta- ble mental health, no history of non-adherence to prescribed medications, have a body mass index (kg/m2) <30 and be free of a host of other confound- ing conditions (e.g. pregnancy, hepatitis B or C positive, other infections or malignancies) (60).

17

Islet autotransplantation Islet autotransplantation for intractable pancreatitis or other cases requiring pancreatectomy can prevent surgically induced severe diabetes without the need of immunosuppressive drugs (61-67). Auto- and alloimmunity or im- munosuppression may play critical roles in the observed potency of trans- planted islets (68, 69). The gradual loss of islet allotransplant survival over time could be due to the immune insults experienced by the islets (70-73) and cases of islet autotransplantation allow a special insight into this pheno- menon (68). Cellular immunity is not the only explanation of differences between auto- and allotransplantation outcomes. There could be islet viabili- ty differences from living donors compared to brain dead donors (74) not to mention logistical differences between the two methods.

Goals of islet transplantation Initial results of islet transplantation focused on insulin independence as the primary endpoint. Freedom from exogenous insulin is the ultimate goal for transplant recipients. However, for the subset of patients qualifying for islet transplantation, labile, autonomic unresponsive T1DM is life-threatening (75). Glycemic control yields increased confidence and freedom from psy- chological stresses associated with progressive diabetes (76). Indeed, despite continuing with exogenous insulin after receiving an islet transplant, HbA1c levels can be maintained at acceptable levels without episodes of hypogly- cemia (46, 77). Suggested goals of achieving metabolic control with islet transplantation (78), not just insulin independence, are gaining momentum in the quest to provide for as many patients as possible (79).

Islet transplantation Short history of clinical islet transplantation The introduction of insulin therapy revolutionized the treatment for diabetes patients and prolonged the lives of millions of people. However, in certain cases of uncontrollable, so called brittle diabetes, intensive insulin therapy is recommended to be used with caution (48). For these patients, uncontrolla- ble blood sugars and hypoglycemia unawareness remain a persistent, poten- tially lethal threat. These patients are ideal candidates for islet transplanta- tion as a therapy for their disease (33). The first human islet transplant took place at the University of Minnesota in 1974 (80). Initial success was low, and between 1990-1998 only 12% reached insulin independence for more than one week (81). Despite protocol improvements no real breakthroughs came until 2000 when the group in Edmonton reported insulin independence in 7 consecutive patients with an average follow-up of almost one year (82). The so-called Edmonton protocol

18

involved transplanting a marginal mass of purified islets from two to four pancreases to patients on an immunosuppression regime free of glucocorti- coids and reduced dosage of calcineuren inhibitor (82). After the release of the Edmonton protocol an explosion of research fa- cilities and transplantation activity ensued (83). Funding increased and an international trial of the Edmonton protocol ensued involving 10 centers from the US, Canada and Europe (45). Developments in islet isolation (dis- cussed in detail later), transplantation and immunosuppression protocols continue to improve the clinical procedure.

Islet isolation procedure The complex islet isolation process is notoriously inconsistent, partly due to the empirical nature of progress in the field and, partly due to lack of stan- dardization of the process itself. Identifying variables critical to islet isola- tion success and improving process standardization is made even more chal- lenging considering the regulations associated with production of biologicals for clinical use.

Regulatory considerations Regulatory bodies such as the United States’ Food and Drug Administration (FDA) and the European Parliament (EP) were established with the goal, among others, to prevent the spread of diseases to the general population by controlling the production, traceability, quality and safety of food, cosmetics and therapeutics as well as, where appropriate, potency testing of prospec- tive drugs, organs, tissues or cells used for therapeutic applications (84). For instance, in the US the regulations governing islet cell processing for trans- plantation are extensive as islets fall under different regulatory categories; a) biologic b) drug c) somatic cell therapy (85). EP rules also require the main- tenance of good manufacturing practices (GMP) conditions for the manufac- ture of islets as outlined in directive 2006/17/EC Annex IV and detailed in the COM-directive from 2006. Both the FDA and EP state that the exposure of product at any time to the atmosphere, a so-called “open system”, must be in a controlled environment of the highest grade possible. The exposed product should preferably be in a Class 100 (Class A for EP) environment. This corresponds to a maximum load of 100 particles larger than 0.5 m in a cubic foot (ft3) of air or for EP regulations at maximum 3,500 particles no larger than 0.5 m in a cubic meter (equivalent to 99 particles/ft3). To reach such high levels of safety and cleanliness GMP facilities are designed according to strict standards (60).

19

Implications of GMP regulations for islet production Costs associated with building a GMP cell production facility can prevent the establishment of new islet transplant centers. Even low cost approaches such as the modification of existing lab space to achieve GMP level processing can be prohibitive for centers looking to offer islet transplant therapy (86). To avoid the costs and maintenance of clean rooms necessary for open system processing, some clinically available therapies use closed system technologies. In contrast to open systems, which include any open bottle, spike ports, luer ports or any other exposure to the environment, closed sys- tem technology avoids any opening to the surrounding environment and meets GMP regulations for exposure of product. Such technology is com- monly used in blood banks with tubing welders to make validated connec- tions between bags. Other branches of cell therapy incorporate closed system technology with the intention of facilitating the transition from research to the clinic (87-89). Despite stringent precautions of GMP facilities to maintain sterility, con- tamination of human islet preparations is occasionally observed (90). A ma- jority of contaminations are limited generally to organ transport, prior to GMP facility processing, or the early steps of the isolation process (91-93). Even when contamination is present, some patients expressed no signs of infection, perhaps due to prophylactic antibiotic administration (93, 94). In contrast, some preparations with negative cultures have led to infection as well as those with positive cultures (94). Such de novo contamination during islet processing is rare (90, 92) but can even occur under GMP conditions (93). It should be kept in mind that major surgeries and transplantation pro- cedures are performed on a regular basis in operating theaters with exposure to a much less regulated environment.

Closed system islet isolation In the pursuit of islet isolation standardization and development one should consider closed system technology to improve safety and compliance with GMP. As mentioned above, the enormous costs that are associated with rea- lizing a GMP lab from scratch favor the introduction of closed systems when possible. Due to the complex nature of large-scale islet processing, technol- ogies that fulfill functional requirements for the use in blood banks, such as apheresis, elutriation or customized systems, have not been implemented in clinical islet transplantation so far (95, 96). Despite some movement of the islet field toward closed system processing (59, 95-97), a fully closed system has yet to be presented for all steps at one center. Major efforts were made in the presented studies to create standardized systems compliant with closed system processing that were flexible enough to allow for the most demand- ing requirements in the field (98, 99).

20

Islet donors and organ transport Islet donor criteria Presently, the main source of islets for transplantation to patients with T1DM comes from brain-dead, cadaveric donors (83). Living donor distal pancreatectomy and islet allotransplantation has been performed once in the post-Edmonton protocol era with initial insulin independence in both the patient and the recipient (100). Significant variability of organ donors (101, 102) and islet isolation cost makes selection of optimal pancreases for islet isolation a critical challenge (58). O’Gorman et al. (58) sought to meet that challenge and have reported on the standardization of cadaveric, brain-dead pancreas donors for islet isolation. The literature is full of support (103) for their summary of donor criteria which includes age (104-112), cause of death (110, 112, 113), cold ischemia time (CIT, time of organ harvest to organ processing) (59, 108, 110, 112), body mass index (BMI, kg/m2) (54, 59, 107, 112), procurement team (59, 107, 111, 113, 114), length of hospital stay (111) (reference 115 cites reduced islet function with longer hospitalization) (115), serum amy- lase (59, 112, 116), use of vasopressors (59), donor blood glucose (110, 111), and donor medical and social history (111). More recently, Hubert et al. reported preprocurement acute insulin response to argentine (AIRarg), an in vivo functional test of -cell mass, to be predictive of isolation success in humans (117).

Organ transport University of Wisconsin solution (UW) or the two-layer method (TLM, a layer of UW on top of high-oxygen concentration perflurocarbon) have been the dominant methods of pancreas preservation in recent years. The debate of which organ preservation solution is better with regard to islet isolation success remains inconclusive (112). Smaller studies of UW versus TLM preservation methods identified the TLM as optimal (113, 118-120) whereas no beneficial effect of TLM storage was observed in two larger studies (121, 122). The largest analysis of pancreas preservation methods (n=802) found similar islet isolation success rates of UW or TLM transported pancreases but identified a third transport solution, histidine-tryptophan-ketogluterate (HTK), as negatively associated with islet isolation success (112). A large scale analysis with Custradiol pancreas preservation has yet to be performed.

Enzymes for islet isolation Successful preparation of isolated islets of Langerhans was first achieved by Claes Hellerström using microdissection on rodent pancreases (123). Islets isolated in this fashion typically express high viability, however, the tedious technique is impractical for large-scale islet isolation.

21

The initial use of collogenolytic enzymes for islet isolation was per- formed by Moskalewski using a crude collagenase product from Clostridium histolyticum for the dissociation of guinea pig pancreata (124). In the years that followed, Moskalewski’s procedure was continuously refined for the isolation of islets from the human pancreas (125-128) but a significant ad- vance in terms of isolated islet yield was the production and use of highly purified and defined enzyme blends (128-130). Experiments in rats indicated that only collagenase class I (CI) and class II (CII), representing seven different isoforms, and an unspecific protease, such as neutral protease (NP), must be present to obtain a complete libera- tion of islets from within the pancreatic acinar tissue of the pancreas (131- 133). For that reason, commercial enzyme production has aimed in the re- cent past on purifying crude enzyme products, consisting of twelve or more different enzymes and other bacterial products (134), to the highest possible extent. Impurities that have to be removed include bacterial endotoxin, which is related to increased cytokine production in human peripheral blood mononuclear cells and correlates negatively with engraftment in a rat islet transplantation model (135, 136). Another component that has to be careful- ly titrated is NP, which can reduce islet morphological and functional integr- ity, in contrast to purified collagenase that does not seem to have a detrimen- tal effect on islet viability if overdosed (137, 138). In spite of the efforts to provide highly purified enzyme blends, lot-to-lot variability in terms of isolation efficiency remains a problem that has per- sisted from the very beginning of enzymatic islet isolation until now (124, 139, 140). This variability was partially explained by a lot-dependent degra- dation of CI observed in retrospective high-pressure liquid chromotography analysis resulting in different ratios between CI and CII (139, 140). Prospec- tive studies in rats confirmed that the ratio between CI and CII not only in- fluences the efficiency of islet release from the pancreas but determines also the amount of NP that is required for efficient pancreas dissociation (141, 142). In humans the CI/CII ratio also was observed as an important parame- ter in effective enzyme batches (143). However, all these findings do not explain the drop in islet isolation out- come and transplant activities observed worldwide subsequent to the re- placement of one product (Liberase HI) by another one (Serva NB 1) that fulfills the guidelines for quality assurance in clinical islet transplantation (57, 144). Since assays used by individual labs provide analytical data dif- ferent from those of the manufacturers (137, 145) it was hypothesized that besides collagenase and neutral protease other additional enzymes that are not listed on the certificate of analysis may play a significant role in pancreas digestion. In fact, other enzymatic activities have been reported to act as a key component in the isolation of porcine islets (146). Recently, there have been advances in enzyme products from a number of manufacturers (147-150) as well as novel assays proposed to measure more

22

accurately non-degraded, active forms of CI (151). The importance of intact or degraded/truncated CI (100 kDa) for efficient pancreas dissociation is debatable as conflicting reports have emerged (148, 152). The importance of NP and the role of endogenous pancreatic enzymes involved in dissociation of the pancreas remain somewhat ambiguous yet undoubtedly significant (133).

Islet purification The native pancreas is mainly composed of acinar, ductal, and vascular tis- sue, representing the exocrine pancreas, whereas islets, as the endocrine component, represent on average 1.3% of the pancreas which equates to 0.5 to 1.3 ml of tissue (153). After enzymatic dissociation of the pancreas the vast majority of the resulting digest is still composed of exocrine tissue. Studies in rodents and humans have shown that pure preparations are de- sirable when graft function and patient well-being are considered. In rodents crude digest or increased exocrine contamination leads to decreased graft function or engraftment compared to pure islets (154). In humans, large packed tissue volumes are thought to be at least partially responsible for a host of severe complications encountered during intraportal infusion early in the history of islet transplantation (99, 154, 155). Transplanting purified islet tissue is currently the standard procedure in the field of human islet trans- plantation (82, 105, 156). Current methods of islet purification include the use of a COBE 2991 cell separator and that of continuous density gradients (99, 155, 157, 158). This method allows for large tissue volumes, such as those attained from pancreas digestion, to be loaded and separated in a single procedure (155, 157). The technology to produce a large-scale continuous density gradient for islet separation on a COBE 2991 was established about 20 years ago (99, 155) and despite some changes to islet separation equipment and materials, such as COBE refrigeration (159) and osmolarity modifications (160-162), it still represents the current standard for human islet purification (45). The equip- ment for generation of density gradients is composed of an open system standard gradient maker (SGM). The SGM consisting of two beakers in which media of different densities are poured (see Figure 4 in the Materials and methods section). The density solutions are then mixed using a manually controlled magnetic stir plate with the magnet in the heavy solution chamber (155). The importance of proper mixing was also stressed in the first paper using continuous density separation for islet purification (Figure 1) (155). Since the technique is now used worldwide, there are a variety gradient maker models, magnets and, of course, different personnel responsible for magnet stirring speeds to generate the gradients. The open chambers, often reused, are not conducive to closed system processing.

23

Figure 1. Experiment showing the importance of proper mixing of solutions from the original publication employing continuous density gradients for large-scale islet isolation [ref. (156)]. Open circles ( ) shows proper mixing and closed circles ( ) represent poorly mixed solutions. The figure is repro- duced with permission.

Various density solutions are used for continuous density production in- cluding Ficoll (59, 158), a University of Wisconsin solution (UW)/Ficoll mix (162, 163) and iodixanol-based medias (156, 164). Optimal densities for isopycnic human islet separation vary. The Minneapolis group has shown that separation densities for islets prepared for autotransplantation were found to be higher than that of islets for allotransplantation by about 0.015 g/ml (98). Although there are exceptions (57), functional separation densities used to isolate clinical islets have a range of about 0.025 g/ml and can be anywhere from 1.060 to 1.116 g/ml (98, 162, 165). A major limitation of the standard SGM is that it must use the density range corresponding to that of the heavy and light solutions mixed. Once the islets have separated on the density gradient the preparation needs to be collected. To dilute the density solutions used for the separation collection is done using a wash solution. The preparation is typically col- lected in 12 to 20 fractions of about 20-30 ml each (59, 162). With progress- ing steps in the collection process, density solution gets heavier, and since the inherent density of exocrine is heavier than endocrine tissue, it becomes progressively impure. Similar purities are combined, washed, and either transplanted immediately or cultured. The islet purification procedure poses a major challenge to achieving a closed system for islet isolation.

Islet culture Isolated islets were for a long time transplanted immediately post-isolation (45, 80). However, increasing evidence that islet culture is beneficial for islet

24

function, despite the possible loss in number, has changed standard practice (166-168). The introduction of a period of culture is advantageous in many respects as it allows time for transport of patients to transplantation centers, allows time for administration of prophylactic immunosuppression in islet recipients, provides a substantial opportunity for islet preparation evaluation, and may reduce immunogenicity of the preparation (169).

Flasks vs. bags The cultured tissue concentration is strictly controlled to avoid overseeding which may lead to hypoxic damage due to competition for oxygen. Even large (>100 m diameter) islets by themselves have been reported to have necrotic cores (170, 171). Culturing islets in bags may be beneficial for islets in many respects. Since islets do not remain suspended in culture media they sink to the bot- tom of the culture vessel. Compared to petri dishes or culture flasks, gas- permeable bags can improve oxygen transfer directly through the islet/vessel interface while receiving nutrients from the media above (59), a theory also put in practice with silicon rubber membranes (172). Clinically established bags produced in high volume yield advantages of low cost, convenient tran- sition of islet culture to bags used clinically and GMP-friendly, semi-closed culture conditions (59). Culture in bags is a natural precursor to final prepa- ration of islets for transplantation which routinely occurs through gravity infusion with islets in bags (173).

Islet preparation quantification The establishment of the islet equivalent (IE) as a unit of volume measure- ment initiated standardization of islet quantification (174). Since then, the large majority of centers pool pure and impure fractions into culture flasks, retrieve samples from each, and then trained counters manually evaluate the sizes of up to (and sometimes beyond) 200 irregularly-shaped individual islets microscopically. Evaluation of islet purity, number, size distribution, islet fragmentation, entrapment in exocrine tissue and day-to-day changes in these values is considered important for some clinical islet programs (105, 175, 176).

Sampling validation A common problem in the evaluation of an islet preparation is the inconsis- tency of sampling. Intra- and inter-technician variability seen in sample evaluation is often high (177) even when a large sample number is taken. Unlike single cell suspensions, an even distribution of pancreatic tissue par- ticles is difficult to obtain due to their relatively large size and size distribu- tion. Large dilution factors compound error in the inherently variable as- sessment of islet number, size distribution and purity. Defining a method for sampling that produces consistent results between samples and technicians,

25

as well as between centers, would tremendously benefit islet quantification comparability. Despite standardization efforts variability in counting is large (178). To minimize manual influences automated or computerized techniques have been developed to assess islet preparations (179-187), some technically ad- vanced and costly (188, 189). A major obstacle to achieve accurate and completely automated islet evaluation is variability in the staining of islet tissue, which can make it difficult to identify tissues appropriately. One major advantage of more readily available imaging equipment is the adherence to documentation requirements according to GMP regulations (178). A simple, intuitive, affordable program to aid in evaluation of islet sample purity, counting and size distribution has been developed at our lab in Uppsala (190) and elsewhere (189). These programs could prove a valua- ble tool to standardize islet evaluation and to aid intra and inter-lab compari- sons of islet counts and purity.

Islet isolation quality control Regulatory bodies require tissue identity, purity, potency and safety to be carried out on clinically bound tissues. Meeting these requirements for islets can be met with relatively simple assays.

Islets of Langerhans – what do they do and what should we test? In healthy individuals, islets of Langerhans are popularly known as the or- gans primarily responsible for maintaining levels of physiologically optimal blood glucose levels. The islets themselves are composed of a number of cell types which respond to various environmental stimuli, primarily blood glu- cose levels. Islet cell types include -cells, -cells, -cells, -cells, PP-cells and more (endothelial cells, pericytes, grehlin). -cells produce insulin (C- peptide as a byproduct) and compose between 28-75% of the cells in an islet (191). The glucagon producing -cells make up about 10-65% of the islet, - cells which manufacture somatostatin constitute from 1.2-22% (191) and remaining cells <2%. The kinetics of -cell insulin secretion in response to glucose is characte- rized by a biphasic release (Figure 2). This biphasic pattern is seen in both native pancreas and in isolated islets, implying that mechanisms involved are inherent properties of islets themselves (192).

26

Figure 2. Normal physiological insulin release in response to glucose sti- mulation. Reprinted with permission from ref. (192)

By measuring the biphasic pattern with the glucose-stimulated perifusion assay, it is possible to visualize insulin release over time. The perifusion assay has an advantage over static glucose stimulation methods since it pro- vides information about intracellular events that would otherwise be missed. Important markers of efficient packaging of ready-release insulin at the cell periphery, mobilization of insulin granules as well as transport of these gra- nules to the cell surface are among the events visualized. Appropriate insulin release means all physiological machinery is in place and functioning well. Dysfunction of insulin secretion can depend on a variety of mechanistic va- riables, everything from the first step of efficient glucose sensing (193) to pathways involved in insulin exocytosis (194). Islet insulin content is another parameter thought to be associated with is- let quality. Brandhorst et al. have shown that insulin content depends on the donor pancreas but does not change drastically despite the insults adminis- tered during islet isolation (195). By measuring the insulin content a func- tional reserve of insulin may be relevant considering the metabolic demands of the recipients. Other assays measuring metabolic activity or viability including oxygen consumption rate (OCR) (196), glucose-stimulated increased increment of OCR (197-199), ATP/ADP (200, 201), variations of glucose-stimulated in- sulin secretion (45, 156, 201, 202), cytometric -cell viability (203) and combinations (201) of these methods have yet to be validated as predictive of graft function in humans, however, some are promising.

Cytokines Cytokine expression can alert islet scientists of the inflammatory status of the tissue to be transplanted. For instance, macrophage chemotractant pro- tein-1 (MCP-1) and tissue factor (TF) expressed by islets are related to post- transplant cross-linked fibrin levels, a marker that coagulation has occurred (204, 205). Islet TF expression as part of the instant blood mediated inflam- matory reaction is associated with the unfavorable outcome of short-term

27

post-transplant clinical C-peptide release (206). Donor islet interleukin (IL)- 10 is associated with islet graft survival and function (73). The role of IL-6 is ambiguous as it has immunomodulatory effects protecting islets (207) and at the same time it may contribute to diabetes development (208). IL-8 is an inflammatory cytokine associated with neutrophil migration and can be in- duced by hypoxia, steroids and IL-1 its expression an indication of islet damage (209). The above mentioned cytokines are those pertaining to this thesis but it should be known that there are a number of others (167).

Measuring clinical success Measures of clinical success initially focused on insulin independence. However, the life-threatening aspects of brittle diabetes (those aspects quali- fying patients for transplant) include diabetic coma and severe glycemic lability, conditions greatly solved to a large extent even with partially func- tioning islet grafts (46, 77). A shift in the field from obtaining insulin inde- pendence to a more therapeutic approach to obtaining normoglycemia has occurred recently (79). To best use available tissue, determination of islet engraftment and prediction of transplantation success (however measured) are hindered by a number of variables not yet standardized or validated.

Engraftment endpoints Effective prediction of how an individual islet preparation will function after transplantation has remained an elusive goal (210), in part due to the wide array of available measures for graft function. Endpoints of graft success have been related to insulin independence rate (118, 156, 211), -score (212), secretory units of islet in transplantation (SUIT) index (213), C- peptide × glucose-1 and C-peptide × glucose-1 × creatinine-1 ratios (CP/GCr) (214), HbA1c (77), fasting and stimulated C-peptide (215), oral glucose to- lerance test, mixed meal tolerance test (MMTT), intravenous tolerance tests with acute insulin response to glucose or AIRarg (166, 216, 217) and more (218, 219). Recently, the Pittsburgh group, in addition to a number of the above mentioned tests, measured IE per unit insulin reduction as reported by Deng et al. to be about 24,000 IE/U. The same study (166) and another (48) measured plasma C-peptide-to-creatinine ratio to account for possible renal dysfunction in their islet after kidney (IAK) populations. C-peptide is perhaps the best candidate to measure functional islet capaci- ty. C-peptide, which is released in conjunction with insulin in a 1:1 ratio (Figure 3), is recognized as the best measure for functional insulin secretory capacity according to the American Diabetes Association (220). C-peptide positivity in islet recipients has been associated with relief from hypoglyce- mia and reduction of glycemic lability despite the continuation of exogenous insulin therapy (77). Assays for both insulin and C-peptide are widely avail- able but C-peptide benefits from a longer half-life in circulation making it a

28

more stable measurement parameter (221). Furthermore, fasting (>8 hr with- out food intake) C-peptide levels are highly correlated to the 90-minute MMTT peak C-peptide values (222). There could however be cross- reactivity with circulating split proinsulin leading to higher than actual C- peptide readings (66) but with modern assays this phenomenon has largely been dealt with (220).

Figure 3. The processing of proinsulin to insulin and C-peptide which are released in a one-to-one molar ratio. Image reproduced from ref. (192) with permission.

Since C-peptide is cleared via the kidney, renal function should also be accounted for, especially in patients suspected of possible kidney dysfunc- tion at the time of transplant (166, 214) or even long afterwards. Measure- ments of creatinine or albumin clearance are two options for evaluating renal function (223). Plasma creatinine is easily attained from the same blood samples as those taken for C-peptide and glucose. By combining C-peptide levels in relation to both glucose and creatinine concentrations as proposed by Faradji et al. (214), the CP/GCr provides a relatively simple, standardized measure of islet graft function.

Reported predictors of transplant success The ability to predict transplantation outcome evades islet scientists (210). There are however some hints as to the most important factors. Numerous reports have correlated the number of IE transplanted (83, 105, 214, 217, 224) or the -cell mass (225), donor age (109, 224), and CIT (226) to trans- plant function. Most of those studies however did not account for renal func- tion. In light of the nephrotoxic effects some immunosuppressive agents

29

have on transplant recipients (227) it may be prudent to introduce such a measure to account for renal clearance dysfunction.

30

Materials and methods

Please refer to the appropriate section in the respective papers for descrip- tions of materials and methods. More detailed descriptions for some methods are described below.

Pump-made gradients Instead of using the SGM with its two-chamber, magnetic stirring system (Figure 4) (155) a pair of computer-controlled pumps were used to create density gradients. Bags containing either light or heavy density solutions were sterilely welded to a pumpable tube, which was again sterilely welded to the COBE bag set. Each pumpable tube passed through a pump and by controlling the pump speed, the volume of each solution could be controlled to yield desired densities and volumes of each density. Mixing of high and low density solutions in this system occurs when the solutions reach the spinning COBE bag. Observations with dyed solutions led to this finding. A schematic of the pump-made density gradient system and appropriate separation of islet and non-islet tissue is shown in Figure 5.

Figure 4. The open standard gradient maker (SGM) system with two chambers for holding heavy and light density solutions. The tube between the chambers allows light density solution to flow into the heavy density solution chamber while being mixed by a magnet.

31

Figure 5. The effectively closed, pump-made gradient system. From L to R; computer program controlling the two pumps for heavy and light density so- lutions, pumps and bags for light and heavy solutions, COBE 2991 with spinning bag, visualization of densities in bag and islet/exocrine separation in the light and heavy portions of the density gradient.

DIA evaluation The DIA program was used in papers I, II, and III and for some data in paper IV. The DIA macro used in conjunction with Leica Qwin software was de- veloped according to standards in the field (174) and utilized capabilities for quantification that are otherwise impractical without computer-assisted eval- uation. The macro logs information relevant to operator and sample information, fulfilling some aspects of traceability as advised according to GMP. Howev- er, the entered data is not secured by password, electronic signature or con- tra-signing, a requirement of data used for clinical data document security. Pancreatic tissue when stained with dissolved dithizone (DTZ), a zinc- chelator, preferentially dyes the endocrine portions red (228). The program works by first allowing the user to select red-stained tissue for islet quantifi- cation, then, other colors (white or lighter colors) for purity quantification, allows some editing to remove bubbles or other unwanted objects and auto- mates analysis. Values reported by the program include date analyzed, sample ID, opera- tor ID, purity based on area of islet to total tissue, purity based on individual particle volume contributions to islet and total tissue, number of islet par- ticles, number of non-islet particles, size index (IE/number islets), fragmen- tation index (accounting for perimeter/area measurements for each particle), separation into the classical size range categories established in 1990 (174) (plus a 20-50 m range) and color selection data values for the islet and non- islet color selections.

32

The fact that the program is not completely automated depends on the fact that there is a differential staining of islets. This confounding factor limits the extent of automation of the software used to analyze images of islets stained with DTZ using light microscopy.

33

Results and discussion

The goal of the islet isolation process aims to yield the maximum number of islets with optimal viability for transplantation to patients for alleviation of the serious complications associated with loss of glycemic control. Variabili- ty in all steps of the isolation process, from donor care to islet isolation to recipient care, can affect this result (see Background). The work presented in each paper/manuscript aimed to improve aspects of the standardization of islet isolation and transplantation variables to better optimize relevant proto- cols and improve standardization.

Identification of a previously unrecognized enzyme activity (Paper I) One of the most critical steps in obtaining enough islets for transplantation is the identification of efficient pancreatic digestive enzymes (105, 111, 229). Due to inconsistent results and lot-to-lot (and intra-lot) variations from commercially available enzyme products (230), further characterization of enzymatic factors influencing human islet isolation success are urgently required. A number of enzymatic activities are measurable even in purified enzyme blends including digestion of the substrate BAEE which reflects tryptic or tryptic-like activity (TLA) [depending on protocol used (146)]. The TLA, a contemporarily ignored enzyme activity, was evaluated in a rat model and subsequently for human islet isolation. The standardization of the evaluation of TLA required activities to be re- lated to the context of collagenase parameters (BAEE-U/PZ-U activities ratio and CII/CI ratios). The BAEE-U/PZ-U ratio is defined as the TLA- ratio. Neutral protease (NP) levels were also adjusted according to guidance from previous experiments. For the rat experiments this was accomplished with a fixed ratio of CII/CI and constant and appropriate levels of NP.

Evaluation of enzymatic digestion of rat pancreas A Lewis rat model was first used, employing sequentially increasing TLA- ratios from 1.3 to 10%. In the rat model, increasing TLA-ratios correlated

34

with significantly decreased digestion times with no difference in islet via- bility, morphology or yield. Purity for 1.3% TLA-ratio isolated islets was lower than that of 5.0 or 10% TLA-ratio islets. In vitro testing for insulin content was lower for the 10% TLA-ratio islets compared to 1.3% islets yet functionality testing as measured by stimulation indices were similar for all groups. No difference in in vivo functionality was seen in the different TLA- ratio isolated rat islets as all mice from each group that were transplanted under the kidney capsule were cured from streptozotocin-induced diabetes until nephrectomy. These experiments warranted testing human pancreas digestion since functional viability remained intact yet regarding islet yields there were TLA-ratio dependent differences.

Evaluation of enzymatic digestion of human pancreas Controlling the ratios of collagenase classes and NP for human islet isolation was important to ensure that comparisons were valid when considering dif- ferent TLA-ratios. As such, collagenase CII/CI ratios were restricted to a narrow range and NP levels were adjusted to appropriate levels. TLA-ratios were increased stepwise from 1.3% to a maximum of 12.6%. Increasing TLA-ratios resulted in correlation to increased isolation suc- cess rates as measured by total islet yield and adherence to quality testing parameters. A low TLA-ratio of 1.3% was associated with long average di- gestion times and isolation failure. The highest islet equivalent (IE) count per gram pancreas weight was seen with the 9.1% TLA-ratio at almost 4000 IE/g (n=16). The dose response of the TLA-ratio did not cause differences in packed tissue volume nor percent digested pancreas. This indicates a more efficient release of islets from the surrounding exocrine tissue, not in the digestion of the macrostructure of the pancreas. The quality parameters of post culture IE recoveries and purities were similar in all TLA-ratio categories. This indicated no effect of differences in this range of TLA-ratios on islet loss after culture. Islet size distribution was also similar in all TLA-ratio groups, a distribution matching closely that of initial studies evaluating the Liberase enzyme (129). Preliminary clinical data from pancreases isolated with TLA-ratio enzymes ranging from 5.7% to 12.6% in nine islet-after-kidney transplant patients showed no influence of different TLA-ratios on basal or stimulated C-peptide values, 0.57±0.10 and 1.87±0.54 ng/ml respectively. The characterization of TLA in commercially available enzyme products for clinical islet isolation appears to play a critical role in terms of isolation efficiency in meeting criteria for transplantable islet preparations without sacrificing islet morphology, viability or function.

35

Progress in the field since Paper I was published The effect of TLA on islet isolation is used by the lab in Uppsala as an im- portant enzyme batch selection criterion. However, “test vials” of enzyme are still used in order to evaluate the pancreatic digestion quality before committing to large scale purchase. Excluding the pancreas acquisition fees, the enzyme is the single most expensive reagent in the islet isolation proto- col and arguably the most critical component for success. Our lab and other researchers have also focused on evaluation of other re- cently commercially available collagenase and neutral protease products (57, 147-150, 231). It is apparent that highly purified enzyme products meeting general safety, regulatory, purity and dosing guidelines can yield acceptable digestion kinetics and sufficient islet quality to warrant clinical use. The emergence of new enzyme products is good for islet isolation advancement. As a side note, the introduction of TLA as presented awakens the need for evaluation of unrecognized enzyme activities. Assays measuring enzyme activities for use in human pancreas digestion are being developed (151) and will continue to help better characterize enzymes for controlled cell disper- sion. Regarding TLA, no formal articles on the effects or activities of TLA have been reported since the publication of Paper I (to date of submission of this thesis, April 2011). The specific mechanism of action of TLA remains elucidation.

Automated gradient making system (Paper II) Density gradient creation is a technically complex procedure essential for large-scale islet purification with the intention of clinical transplant. The procedure has remained largely unchanged since its inception (99, 155), a testament to the durability of the methodology. However, the standard pro- cedure of gradient making for islet purification is somewhat unsuitable for high-quality, high-reproducibility, good manufacturing practices (GMP)- level methodology due to manual variability and as a possible source of con- tamination. As made evident in the first article describing the density gradient manu- facturing procedure, which is still used today, mixing density solutions prop- erly can dramatically affect the quality of the gradient (Figure 1) (155). This variability can come from a number of different parameters such as the chamber size, tube size connecting light and heavy density solution cham- bers, magnet model and spinning speed. As an open procedure, this manually controlled method provides many opportunities for improvement when considering standardization and manu- facture according to GMP procedures. We theorized that by controlling flow

36

rates of the individual density solutions that we could create reproducible and standardized gradients in a closed-system. A closed, computer-controlled, automatic purification system (APS) was compared to standard gradient maker (SGM) methods for the generation of density gradients for human islet purification.

Functionality of pump-made gradients Reliability of the technology needed to be established prior to use for human islet purification. To measure the gradient making efficacy of the APS, the expected volume of density gradients were compared to that of the SGM. For 400 ml gradients, the APS achieved a more complete recovery of ex- pected gradient volume (98.2±2.0% vs. 90.0±1.1%, p<0.05) indicating that computerized dosing for density gradient manufacture is reliable. To test the versatility of the APS we attempted to produce non-linear, continuous density gradients. The system readily constructed sigmoidal gra- dients, increasing volumes of either the heavy or light portions of the gra- dients. Unpublished observations include the creation of the UW/Ficoll gra- dient protocol from the University of Chicago (162) which creates linear densities between (1.063-1.074 g/ml) without prior mixing of the two densi- ties of the UW (1.045 g/ml) and heavy Ficoll (1.100 g/ml).

Islet separation with pump-made gradients To test the ability of pump-made gradients to separate islets of Langerhans from contaminating exocrine tissue, both the APS and SGM were used to create linear gradients. Both methods recovered similar overall islet equiva- lents (IE) and purities. There was no difference in the number of purified fractions used for tissue culture between SGM and APS. Any change to the isolation procedure requires testing for functionality, viability and inflammatory status. Quality control parameters were similar in regard to perifusion glucose-stimulated insulin secretion stimulation indices and ADP/ATP ratio indicating viable, functional islets from both groups. Cytokine expression was not different for TF expression. Differences arose in the APS separated islets which expressed lower IL-6, IL-8 and MCP-1 versus SGM. Since the purest fractions were used for quality control purpos- es it cannot be ruled out that this affected in vitro test results due to higher islet purity in APS islets (72.2±4.5 vs. 80.9±3.9%, p<0.05). This observation is in-line with the SGM and APS insulin content per DNA measurements of 4.1±0.7 vs. 5.0±0.8 ng/ng (p<0.05), respectively. This small difference in purity could have implications for the cytokine expression levels (232, 233), however, the influence of minor changes in purity and the repercussions for islet quality control and clinical transplantation remain outside the scope of this investigation.

37

The closed, automated gradient making system proved a feasible and flex- ible system to solve problems associated with standardization of islet processing. The APS also resolves one of the major obstacles to realizing a closed system for the entire islet isolation process and in meeting GMP pro- duction requirements.

Progress in the field since Paper II was published (2008) Adoption of this method has been initiated at two centers. Technicalities have slowed progress at one center and the other center in Leiden, Nether- lands will likely implement the pump-made gradient system in their clinical islet production unit soon (Marten Engelse, personal communication, March 29, 2011). Plans to implement the procedure at the City of Hope in Duarte, CA, USA are underway.

Digital imaging analysis (DIA) and presentation of a GMP-friendly islet quantification technique (Paper III) The standard used for quantification of islets after isolation and prior to transplantation involves manually sampled and counted aliquots, a procedure known for high variation, both subjective and technical in nature. Most cen- ters have dedicated “counters” to ensure some level of reproducibility. Var- iation between counters at different centers remains high despite standardiza- tion efforts (178). Sampling variation can also contribute to uncertainty of counts (177). Methods of automated analysis of islet samples (179, 183, 188) have unfortunately not been widely applied. This could be due to cost of some apparatuses used (>$100,000 USD in one case) or the complexity of software used to analyze images. The challenge of adapting the islet quantification procedure to closed sys- tem technology is daunting. Consequently, in order to quantify islets in com- pliance with GMP practices, state-of-the-art clean rooms, expensive facilities and experienced personnel are required (234). Flask-based culture methods ubiquitously utilized by islet labs throughout the world pose a serious chal- lenge to reaching a closed system islet isolation method. Using customized, computer-assisted digital image analysis (DIA) ma- cros, a purity and volume based (PV) system to evaluate islets in a GMP- friendly manner was evaluated with respect to counting variation and com- pared to that of standard manual methods.

38

DIA validation The DIA system required calibration according to a standard suitable for islet evaluation. Our coauthors in Miami sent blinded pools of calibrated microspheres of known size distribution and quantity for assessment (177). The DIA program recorded aspects of microsphere size distribution, with values almost identical to expected, establishing the validity of the program to reliably record sizes accurately. The total islet equivalents (IE) of the pre- pared microsphere pools matched with expected values. Despite the accura- cy of the system to evaluate microsphere sizes, the average percent coeffi- cients of variation (CV%, CV% = standard deviation × average-1 × 100%) of total IE values for the pools (n=15) was 21±10%, indicating large evaluation variation.

Computer-assisted DIA versus manual methods DIA was compared to manual methods to determine the variation between evaluators. Variation in islet samples (n=14) as evaluated between 3 individ- uals reduced by almost half when using DIA for both IE (31% vs. 17%, p=0.002) and islet purity (20% vs. 13%, p=0.033). Although, the absolute values of IE and purity were not significantly different between the two me- thods. This data established that DIA can reduce variation between individu- als compared to manual evaluation. Accuracy in size distribution, performed better by computers than hu- mans, allowed individual sizing of islets. The DIA used for this study ac- counted for individual islet particle area when calculating IE, avoiding grouping of particles into size categories as is customary with the standard, manual method. To assess the difference between the standard method to that of DIA, DIA analyzed IE (calculated based on individual sizes) were compared to IE calculated based on the size ranges (standard method). An observed 15% reduction of total IE using DIA corresponded well with a reduction of 16% seen in another similar DIA system (189). This means that current thresholds for transplantation of, for instance, 5,000 IE/kg should be revised to 4,250 IE/kg or 300,000 IE to 255,000 IE when using DIA from sampled pools for total IE estimation.

Evaluator versus sample variation The identification of the source of the variation in islet evaluation can be important for methodologies aiming to increase quantification reliability. To examine contributions of counter versus sample errors, 14 pools of islets were sampled 5 times by one individual with extensive islet isolation and sampling experience. Each pool yielded a CV% value for IE and purity as analyzed by DIA. The average CV%s were, for IE counts 20.5%±7.5%, and

39

for purity 14.3%±5.7%. Then, 3 individuals analyzed 20 images (four pools worth of samples) to identify if evaluator differences were the cause of the high CV%s. However, with CV%s of 6.6±3.2% for IE and 5.8±3.2% for purity it was apparent that variation came from sampling.

Closed system islet evaluation The measurement of an islet equivalent is a volume (174). Correspondingly, the total volume of the islet preparation should be able to be used for the quantification of transplantable endocrine mass. By knowing the purity and the pellet volume, a theoretical estimation of total IE should be possible. We set out to use the purity and volume (PV) of microsphere/tissue pellets in a GMP-friendly bag system to evaluate total IE. By performing analysis on the entire cell preparation we hoped to avoid large variations in outcome. Standard curves of the microsphere pools were generated to establish vo- lumes for known amounts of IE. The standard curves allowed translation of known, measured volumes to surface areas of pellets in bags. A known amount of microspheres was mixed with exocrine tissue and evaluated with both the standard manual and PV methods. The values of expected IE did not differ with either method. However, manual IE resulted in a total CV% of 44.3% and a range spanning 258 k IE, whereas PV evalua- tion resulted in a CV% of 10.7% and range of 60 k IE, a remarkable differ- ence. In this analysis, again, we saw major contributions of variation that appeared to come from sampling, not evaluators. Purity CV% values for each method were similar approximating 10.5% and differed from expected by +7% for the manual method and +3% for PV. Again, like the islet analy- sis, purity CV%s were nominally lower than those for IE. A semi-closed islet evaluation system is presented which is able to reduce variation compared to standard manual methods. The major contribution of variation in islet evaluation appears to be sample related since evaluator errors were consistently lower in comparison. Variation in IE and purity values reduced by using DIA compared to standard manual methods. Islet quantification according to GMP by utilization of the PV method also re- duced variation compared to manual methods and is the first such model allowing adherence to strict standards for clinical islet products.

Progress in the field since Paper III was published (2011) This paper was recently published (less than two months ago). However, since publication an imaging modality for visualizing 3 dimensional islet volume and cellular content from histological whole pancreas sections has been reported (235). Although not used for islet isolation quantification it represents the fast-paced development of computer-assisted methodology for quantification and identification of tissues of interest.

40

Prediction of islet graft potency (Paper IV) Predictive evaluation of islet transplant engraftment would benefit islet re- search in many respects. By knowing what variables affect individual islet transplantation function, the decision to transplant a particular batch can be made and thereby possibly avoid procedural complications, possible immu- nization and costs associated with unnecessary transplantation procedures. As mentioned earlier, a number of reports correlate to transplant success to various parameters, including the number of IE transplanted (83, 105, 214, 217, 224) or the -cell mass (225), donor age (109, 224), or CIT (226). Strict criteria are in place for clinical islet transplantation trials, such as that of used for the International Trial of the Edmonton Protocol, which re- quired at least 5,000 IE/kg recipient weight (45) for a transplant to take place. The Edmonton trial allowed numerous transplantations to the same patient in the hopes of reaching insulin independence (45). Many other cen- ters have adopted that same multi-transplant ideology. Upon subsequent transplantations this creates a problem in evaluation to determine which graft may function and which may not. Currently, such information can only reli- ably be taken from solitary islet transplants, a limitation in the quest to iden- tify important predictive markers. The islet isolation center in Uppsala is one of the most active in the world as part of the Nordic Network for Islet Transplantation (236). As such, a considerable number of transplantations have been performed. To maximize the amount of information to be gained, the short term islet engraftment, as measured by the change in pre- and 28-day posttransplant islet graft function using the established C-peptide (× 100) × glucose-1 × creatinine-1 ratio ( CP/GCr) was used. Usable data from 110 islet after kidney (IAK) trans- plantations were retrospectively analyzed using univariate regression of var- ious donor, islet isolation, quality control and recipient variables. Similarly, using multiple, backwards-selection regression an optimized transplantable functional islet mass (TFIM) predictive model was generated to identify variables important for engraftment.

41

Variables analyzed include the following:

Donor variables Age Body mass index (BMI) Cold ischemia time (CIT) Number of other organs harvested Days in intensive care for transplantation Cause of death Local or distant donor hospital (all Sex have individual organ harvest teams) Weight

Islet processing variables Preservation solution Islet purity Enzyme used Total tissue volume Total islet equivalents (IE) Days cultured

Quality control measures Dynamic perifusion glucose-stimulated insulin secretion stimulation index Insulin content

Recipient parameters Age IE/kg body weight Sex Pretransplant insulin Body weight Pretransplant HbA1c

Correlations to short term outcome No correlation of CP/GCr for either the univariate or multiple regression analysis was identified for the donor or recipient age, sex and body weights, days in intensive care, cause of death, body mass index, number of other organs harvested for transplantation, local or distant harvest team, preserva- tion solution, enzyme used, islet purity, days cultured, islet insulin content, and pretransplant insulin and HbA1c. Already in this “non-significant” list we can see a difference from the literature in that donor age did not associate to function as it has in smaller studies [n=55 for ref (109) and n=8 for reference (224)]. The variables associated with engraftment for both univariate regression (Spearman r2 and p-values shown) and the TFIM model were cold ischemia time (CIT) (r2=0.091, p=0.0015), islet equivalents (IE) (r2=0.130, p<0.001), total tissue volume (r2=0.050, p=0.025), and dynamic perifusion glucose- stimulated insulin secretion stimulation index (SI) (r2=0.090, p=0.0033). Each of the above mentioned parameters are easily attainable prior to trans- plantation. Individually, the amount of endocrine tissue transplanted had the highest correlation to short-term engraftment, a rather unsurprising (83, 105, 214, 217, 224) but welcome observation. The IE, although significant, ac- counted for only 13% of the transplant outcome variability.

42

Transplanted functional islet mass (TFIM) model All variables were used to calculate and create the predictive TFIM model. The TFIM had an adjusted r2 of 0.387 (p<0.001) meaning it explains 38.7% of the variation in CP/GCr. This is a substantial proportion considering the few parameters taken into account. The TFIM model considers the 4 variables CIT, IE, total tissue volume, and SI and each alter the expected CP/GCr to varying degrees. Each can affect the others and, consequently, for the model to function properly all four variables are required. The influence of each factor on CP/GCr gives us a better understanding of what they mean in reality, not just how significant something is. For in- stance, for every 100,000 IE transplanted or increase of 2 units SI, the TFIM increased by 0.13. It decreased by the same amount with 1250 l tissue vo- lume or about 20 minutes CIT between 4-8 hrs storage. The confounding nature of the model is seen in the influences of IE and total tissue volume. IE is positively associated with outcome and total tissue volume (which inherently has an IE component) is negatively associated. Since such low volumes of tissue were transplanted the effect of the non- islet portion of the tissue volume likely plays a role in the negative contribu- tion of total tissue. Indeed, the exocrine tissue can contain cytokine secreting cells and may harm the graft by enhancing proinflammatory pathways (232). The exact influence of the different exocrine components is not fully known but the TFIM model definitely suggests that whatever the influence, it most likely is not beneficial for early transplant function. Cold ischemia time is a perpetual factor influencing islet isolation (59, 108, 110, 112) and transplantation success (226). Here, in regards to the TFIM model we can get an idea of how important it is. The CP/GCr reduc- es 0.13 for every 20 minutes in hours 4-8 after procurement, the same incre- ment increase for 100,000 IE. The highest single transplant achieved a CP/GCr of 1.5, the equivalent of a little less than 4 hrs CIT. The TFIM is reduced even more with increasing cold storage time. The SI, a measure of -cell insulin secretory capacity, was higher in better functioning islet preparations. The capacity of isolated islets to secrete insu- lin efficiently and strongly in the presence of very high glucose levels re- flects, essentially, expected islet functionality in the transplant situation. In this IAK population the CP/GCr as a measure of engraftment worked well. The main differences of this report to others measuring C- peptide/glucose (214) or C-peptide/creatinine (48, 166) is the larger number of transplants and the consideration of both glucose and creatinine to nor- malize serum C-peptide levels. CP/GC was reported initially by Faradji et al. (214) but not investigated further in their study due to its relative weakness to the simple C-peptide/glucose ratio. Their study of 22 patients included 4 with previous kidney transplants, all of which had “normal and stable renal

43

function” (214). Indeed, in our population with variable renal function no correlations were observed without adjusting for creatinine. Despite the simplicity of the TFIM model a considerable portion of early islet graft outcome variability is explained by parameters easily attainable pretransplant. The TFIM provides a straightforward and potent tool to guide the decision to utilize a specific islet preparation for clinical transplantation.

44

Conclusions

Specific conclusions

Paper I Measurement of TLA ratio can supplement already reported enzyme activities for the identification of effective batches of enzyme for human pancreas digestion In vitro and in vivo function of islets were seemingly unaffected by dif- ferent TLA-ratios

Paper II Removal of manual variation is achievable using a GMP-level, closed system, computer-controlled pump system for the generation of density gradients for the large-scale separation of islets from exocrine tissue

Paper III A GMP-friendly islet quantification system using DIA reduced variabili- ty compared to standard manual methods Sampling variation remains the main source of islet quantification error

Paper IV The simple CP/GCr correlated early IAK transplant engraftment to measures easily attainable pretransplant An optimized TFIM model using 4 variables (1 donor-, 2 isolation-, and 1 quality control-related) explained almost 40% of the variation in en- graftment

General conclusion Reproducibility, safety, reliability and adherence to good manufacturing practices in islet isolation methods was achieved through work in Papers I- III and variables affecting engraftment were identified through standardiza- tion of outcome measures and a predictive model in Paper IV.

45

Critical considerations regarding research design and methods

Paper I There are a number of confounding factors present in islet isolation not im- mediately translatable to other labs and, as such, standardization of all va- riables is not possible. For instance, the human pancreas is perhaps the best assay for enzyme evaluation yet at the same time maybe the worst. Variation comes from a number of sources, e.g. age, islet capsule variability (101), genetics, endogenous pancreatic enzyme secretions [possibly affected by CIT (133) and/or brain death (74)] islet processing variables, patient care prior to islet isolation, etc. The fact that the ratio of females to males increased with increasing TLA- ratios cannot exclude the possibility that gender may have played a role in the results of the isolation success rates. Also, the highest TLA ratio group had the heaviest pancreases which may have influenced results.

Paper II The volumes of APS made gradients for human islet separation were 400 ml and those of the SGM were 300 ml. As such it may be expected that a better separation may occur although no difference was observed. There was about a 15 minute delay in density gradient manufacture between the SGM and APS-made gradients, and which was produced first was not recorded al- though they did alternate. Comparisons of expected gradient used the same volume.

Paper III The ability of each individual to measure the islet or exocrine portions of the images is subjective. However, when using the DIA software the differences in selection were minimized compared to manual methods as mentioned in Paper III. The standard curve for the pellets in bags with a known amount of stan- dardized microspheres is not ideal (Paper III, Fig. 3). There is a large spread of analyzed area in 150 k and 200 k IE populations. Also, the method of imaging the pellets in bags is not as standardized as it should be. Develop-

46

ment to further standardize this process and even automate digital pellet image evaluation is a top priority. As mentioned in the discussion of Paper III, confirmation of these results using even higher numbers of IE needs to be performed before acceptance of this method into the clinic.

Paper IV Not all values of possible interest were taken into account in this analysis. Being a retrospective study this is one of the defining limitations of the pre- sented work. Also, only pancreases taken for clinical transplant were consi- dered. Negative corollaries of graft function, such as recipient autoantibody status (70, 71, 156) were not assessed.

47

Future perspectives

The ultimate goal for islet isolation technologies from cadaver pancreas do- nors is maximal yield and functional capacity of isolated and transplanted islets according to regulatory requirements. Restricting future perspectives to cadaver donor pancreases as a source for -cell replacement therapy, some of the following developments may come to fruition.

Optimization of donor variables As presented by Hubert et al. predictive markers for islet isolation success (117) are still being discovered. Other factors possibly influencing islet iso- lation or transplantation outcome might include brain death, as evidenced by islet potency reported in whole pancreatectomy, autotransplanted patients (68). Patient care immediately after brain death could be a major factor in improving organ quality and viability of tissue. Continuing development in transport solutions, improving oxygenation of hypoxic tissue will likely in- crease tissue recovery and potency (237).

Enzymatic islet isolation Continued characterization of the actions of enzyme components on human pancreas dissociation will help us understand even better the critical processes involved in efficient islet release from exocrine tissue. Assays characterizing these activities and even quick histological analysis may be employed to determine the chances of isolation success.

Islet harvest The work presented in this thesis will help realize a more standardized islet isolation process. Other procedural improvements to reduce variation be- tween labs as well as further GMP processing include closing the system immediately post pancreas digestion. Lakey et al. have reported results with canine islet harvesting using an angled sluice device with seemingly good results. They effectively reduced media volume and concentrated digested tissue without centrifugation (95). However, this system was not adopted by the islet community and its production was subsequently dropped. Other methods to concentrate crude pancreas dilutions from greater than 5 L in- clude the system presented by Klaffschenkel et al. that used the COBE 2991 cell processor (97). This method utilizes repeated centrifugations on packed

48

tissue, a stressful process for living cells. Manufacturing another device sim- ilar in principle to the sluice relying on the passive nature of large cell clus- ters to sediment rapidly out of solution would be optimal. Minimizing me- chanical trauma most likely will improve cell viability and recovery as well as allow for online concentration of cell product.

Loading pancreatic digest to the COBE 2991/tissue purification/islet collection The presented APS provides essential technology to completely close the entire islet isolation procedure. Unfortunately, the COBE bag set itself is not a validated closed system. Loading tissue into the COBE 2991 cell processor and subsequent collection of purified tissue is still performed with open sys- tems. During this stage, unsterile equipment is in close proximity to surfaces in direct contact with product. A proposed bag system to load incubated tissue and subsequently collect the various separated fractions would allow for collection in a closed system, eliminating the need for open flasks or conical tubes. In its current form the automation of density gradients could confer sub- stantial benefits for centers that tailor density gradients to the specific densi- ty of individual pancreases (98, 163, 165, 238). Also, studies easily per- formed with the APS, such as preferentially heavy or light gradients have not been investigated.

Islet quantification and culture The closed system quantification of islet tissue is presented in this thesis but there may be other methods that could do the same job. The use of optical coherence tomography (OCT), a technique that can create three dimensional reconstruction of objects up to 500 m, could provide online yields from purification collection to during infusion on a per particle basis or, if OCT scanners become available, they could scan bags with the tissue sediment. Frequent media change has been examined by the Lille group (239) with promising results. Online media change as employed in other cell expansion technologies (240) could also be employed with islets, reducing workload and, if the evidence from the islet center Lille is any indication, increase tissue viability and maybe even potency.

Engraftment assessment The nature of multiple transplantations makes evaluation of engraftment and contributions of individual islet grafts to functional -cell capacity difficult. Identifying the extent each islet preparation contributes to long-term function is still virtually impossible.

49

Fully automated islet isolation The process of islet isolation and culture could one day be fully automated and optimized on a per pancreas basis. For this to occur, advances in all as- pects of the presented isolation procedure will need to be made. It will re- quire substantial human and material capital, dedicated doctors, technicians and engineers.

50

Popular science summary

Diabetes mellitus is an increasingly prevalent and costly disease. Insulin injections are required for those with type 1 diabetes mellitus to survive long term. This is due to the destruction of the pancreatic -cells, the only cells in the body that produce insulin. The -cells are located in small cell clusters called islets of Langerhans found in the pancreas, and these islets can be transplanted from cadaver donors to type 1 diabetes patients. The use of donor pancreas organs for islet isolation and transplantation is one way to replace insulin producing islet cells. However, the process of islet isolation is highly variable and not all isolations result in transplantable islet numbers and quality. Additionally, the impact of individual islet preparations to their function in transplanted patients is not standardized to the extent possible. This thesis aimed to identify critical variables associated with islet isolation success and the evaluation of posttransplant function. A critical step in the islet isolation process is the enzymatic breakdown of pancreatic tissue to release islets from surrounding tissues. Outcome varia- bility is in large part due to enzyme variations. We introduce an as yet un- identified critical enzyme activity for the efficient digestion of donor pan- creases for islet isolation. Changes in this enzyme activity led to marked differences in islet isolation success rates without sacrificing islet quality. These results should help standardize evaluation of enzymes for clinical islet isolation. Another critical isolation step is the purification of the digested pancreas. The crude pancreatic digest is mainly composed of tissue that does not con- tain insulin-producing islets. Therefore, we use a separation technique based on density gradients to separate islets from the other tissue. To improve on the widely used manual method for gradient making we developed a com- puter-controlled pump system to create the desired density gradients using approved technologies. The pump-made gradients worked as well as the man-made gradients and did so with minimal effort. Evaluating the number of islets after isolation and before transplantation is another source of variation. Using small samples, manual methods that are currently used to microscopically size each islet to obtain total islet yields are not reliable. We developed a digital imaging analysis system to measure islet yield and purity. The principle is that instead of counting each islet we can obtain similar numbers but with greater accuracy by measuring the total tissue pellet volume and islet purity. Indeed, by simply using digital counting

51

methods instead of manual methods, variation in counting reduced by almost half. By using the pellet volume/purity method, variation was also reduced compared to manual methods. Another benefit of the new method is that it is an easy and clean method. Once we have an islet preparation we would like to know how it works in the transplanted patient. C-peptide is released with insulin so it works as a measure of insulin release. Both blood sugar levels and creatinine affect C- peptide levels and therefore we measure all three in order to obtain a proper measure of the transplant’s function. By using this measurement we identi- fied four pretransplant factors that affect outcome. One factor was related to the organ transport time, one to function of the islets, and two to the trans- planted tissue volume. When these four easily attainable factors were put into a predictive model, about 40% of the variation in the islet transplant outcome was explained. The work contained in this thesis identifies and optimizes a number of elements critical to consistent islet isolation and transplantation success.

52

Svensksammanfattning

Diabetes mellitus är en allt vanligare och kostsam sjukdom. Insulininjektio- ner krävs för att personer med diabetes mellitus typ 1 ska överleva långsik- tigt. Detta beror på att -celler, de enda cellerna i kroppen som bildar insulin, förstörs. -cellerna finns i de Langerhanska öarna i bukspottkörteln och kan transplanteras från avlidna donatorer till typ 1-diabetiker. Användningen av bukspottkörtlar för ö-isolering och transplantation är ett sätt att ersätta de insulin-producerande cellöarna. Processen att isolera öar är dock mycket varierande och alla isoleringar leder inte till transplanterbara öar av tillräck- ligt antal och kvalitet. Dessutom saknas standardiserade metoder för att mäta fuktionen hos öarna i transplanterade patienter. Denna avhandling syftar till att identifiera de kritiska variablerna i samband med ö-isoleringsprocessen samt att utvärdera funktionen hos transplantatet. Ett av de viktigaste momenten i ö-isoleringsprocessen är den enzymatiska nedbrytningen av bukspottskörteln. Utfall i variationen är till stor del på grund av enzymvariationer. Vi har introducerat en ännu oidentifierad kritisk enzymaktivitet för effektiv nedbrytning av bukspottkörtlar för ö-isolering. Förändringar i den här nya enzymaktiviteten ledde till betydande skillnader i framgången av ö-isoleringen, utan att ö-kvaliteten försämrades. Mätningar av den här enzymaktiviteten kommer att ha betydelse för ö- isoleringsstandisering. Ett annat kritiskt isoleringssteg är reningen av den nedbrutna bukspottkör- teln. Bukspottkörteln består till 98% av vävnad som inte innehåller öar med insulinproducerande celler. Därför används en separationsteknik baserad på densitetsgradienter för att separera öarna från övrig vävnad. För att förbättra den utbredda manuella metoden för att göra densistetsgradienter har vi ut- vecklat ett datorstyrt pumpsystem för att skapa önskade densitetsgradienter. De automatiserade datorstyrda gradienterna fungerade lika bra som de tidi- gare manuella i vår studie. En annan källa till variation är bedömningen av antalet öar efter isolering och före transplantation. Med små prover är manuella metoder som för när- varande används för att uppskatta storlek på varje ö för att få total ö-mängd inte tillförlitliga. Vi har utvecklat ett digitalt bildanalyssystem för att mäta mängden öar samt renheten. Principen är att istället för att räkna varje ö kan vi få liknande värden men med större noggrannhet genom att mäta den totala volymen vävnad och renhet. Genom att använda den digitala metoden i stäl-

53

let för manuella metoder, minskade variation nästan till hälften. Den största nyttan med den nya metoden är att den också är mycket användarvänlig. När vi har en ö-preparation, vill vi veta hur den kommer att fungerar i den transplanterade patienten. Man kan göra det genom att mäta ett antal olika parametrar. C-peptid frisätts på samma sätt som insulin och fungerar därför som ett mått på insulinfrisättningen. Blodsocker och kreatinin kan påverka C-peptidnivån och därför mätte vi alla tre för att få ett ordentligt mått på transplantatets funktion. Med hjälp av den här mätningen har vi identifierat fyra pretransplantfaktorer som påverkar transplantationsresultatet. En faktor var relaterad till bukspottkörtelns transporttid, en till ö-cellsfunktion, och två till den transplanterade vävnadsvolymen. När dessa fyra lättillgängliga fakto- rer sattes i en prediktiv modell, förutspådde modellen ca 40% av variationen i transplantationsresultaten. Modellen kommer att kunna användas för att avgöra om öarna har tillräckligt hög kvalitet för att transplanteras. Arbetet i denna avhandling identifierar och förbättrar ett antal kritiska de- lar av ö-isoleringsproceduren. Med ännu mer standardisering kommer ö- isolering och transplantation att bli en mer kostnadseffektiv och tillgänglig behandling för många typ 1-diabetiker.

54

Acknowledgments

After the thrombosis of sitting in a chair for a few months to write what will likely remain unread by the great majority of you, I would like to take the opportunity, ink, and page space to thank those of you who have made this thesis (and my life) so nice. It is a bit of a “light take” on things so don’t get all riled up if I’m a bit sarcas- tic at times. Thanks and appreciation go to;

Prof. Olle Korsgren, many thanks go to you for starting your program, being such a good application writer and for giving me an opportunity to help you in the quest to make some people’s lives better through islet transplantation. Your seemingly effort- less knack for inventing novel solutions to challenges in this field makes for an ex- citing environment.

Dr. Daniel Brandhorst, the walking islet isolation encyclopedia, thank you for your frank advice and for sharing your seemingly boundless knowledge of the literature. I was very happy to hear you were coming to the lab in Uppsala in 2005 and very sad to hear you were leaving for opportunities elsewhere. I learned a lot from you and wish you the best.

Gunilla Lewén, for your putting up with me, listening and being concerned about the routine lab perssonel’s well-being. You are an effective leader and dedicated boss. I should probably be at work right now actually…by the way I’ll tell Christi- na that you miss her.

To my coauthors; Dr. Heide Brandhorst, your cheery, calm attitude and sound analysis made for pleasant collaborations. Also, the stories about a former boss standing over you in the middle of the night to wake you up for an isolation, or booking an ambulance to bring you back to the lab when you were on vacation, etc. gave me a good laugh and made isolations go that much quicker. Dr. Torbjörn Lundgren, thanks for the nice collaboration on the last manuscript of this thesis, something we can share as thesis-brothers (‘sis-bros?). To Drs. Annika Tibell, Gunnar Tufveson Peter Buchwald, Camillo Ricordi, Axel Foss, Trond Jenssen, Lauri Kylönnen, Kaija Salmela, Marie Felldin, and Jugonna Feelgood thank you for productive collaborations and for the efforts you have invested in producing the work for this thesis. Especially you, Dr. Feelgood, there’s just something about you that makes me feel alright.

55

To Prof. Bo Nilsson and Dr. Masafumi Goto, thanks for helping build such a great lab here in Uppsala. It would be a much different place without you…likely worse.

Magnus Ståhle, not only a co-worker and co-author but also a co-friend. Thanks for keeping the lab updated with the newest SOPs, giving me chocolate, creating gra- dients and counting thousands of microspheres. It was fun torturing you. By the way, I have some exciting projects lined up…

To the rest of The Islet Isolation Superteam; Bumsan the workhorse who’s been here since the beginning, it’s been a pleasure working with you and I’m sorry for the technical difficulties I may have introduced to the lab. To the old lab rats Drs.; Ulri- ka Johansson, your thesis kept me warm at night during the writing of this thesis (‘sis-sis?), I hope it is half as perfect as yours. Also, thanks for picking me up at the airport when I moved to Sweden, you were so nice and even laughed at my terrible observation that “fart” (utfart, infart) was written on the signs everywhere, Ki-Jah! My initial roommates Annika Möell and Sanja Cabric impressed on me the impor- tance of fluor and ability to talk about anything and everything (Sanja, you should still be a talkshow host). Peter Schmidt, your witty commentary is always welcome and makes for a fun time, especially in Queen’s English, Lisa Moberg, great work with nicotinaMIDE and IBMIR and thanks for teaching me the word grymt, Anna Karin Lidehäll, for being so nice and pleasant to talk to, I will try to arrange for more cinnamon chewing gum, Fredrick Carlsson, for your off-the-wall comments and for brewing great beers, Annette vM, for showing me around the place when I started, Naomi Forslund, I learned a lot from you even though you might not be- lieve it, Nina Bodin, Seta Kurt, and Anna Svensson it was a real pleasure and I wish you the best in your new, exciting careers as moose shovelers. To the current lab rats Drs.; Dr. Torsten Eich, for taking the performance pressure off me when the COBE exploded after I was here for a month, proof that it can happen to the best of us . Also, thanks for the intellectual conversations and for welcoming me to the lab so warmly, Viola Selling, I just needed to learn that once you got your coffee that everything would be alright during the isolations…thanks, also for the nice conversations, Karin Andersson, for being great in the lab and for using words most Swedes don’t usually use, Sofie Ingvast and Hanna Åkerström, looking forward to working with you in this exciting field and interesting lab environment. To the current/recent MDs, PhDs and PhD wannabes; Dr. Olof Eriksson, for your good work with the isolations and interesting and enlightening mediumbyte discus- sions, you have it all, including great taste of music, Dr. Johan Bränström, for being a capable technician and soon isolation leader, you have the best type of ideas, those that spawn new ideas, Oskar Skog, for being a good teacher of virus theory, sound thinker (sarcastic or not? Who knows…) and for not pushing the snuss on me too hard, Monika Hodik for your kind ear, soft-spoken manner and giving me chocolate, Drs. Pepe and Ana Caballero, for the wonderful saunas, food and wine and for the name Andrés del Monte Librè, you forced me to think about why we do what we do when we do what we do, Sana Asif, for your kind demeanor, Dr.

56

“Snygg David” Berglund, the newbie, for being a fine example of what an Ameri- can should be, together we might combine to make one whole Swede and one whole American, looking forward to shared fun, Dr. Arian Sadeghi, the other ö-lab new- bie, love your film roles and enjoy your sense of humor, the islet lab welcomes you with its sarlacc-like tentacles, next person is not a PhD wannabe but worth a mention I guess, Minna Maria Honkanen-Scott, an islet specialist from MN or Finland? I get mixed up, thanks for being my soundboard and sharing a beer every now and then, your sense of humor is priceless, well, maybe very expensive anyway!

Peetra Magnusson and ALo, for keeping the lid on this place, your advice and support are very welcome and I’m impressed with the work you’re doing.

Other lab personnel; great work on the assays! We’ve used a lot of what you’ve done in the papers presented in this thesis! Special thanks to Elisabeth Wijkström, Anna Karlsson and Susanne Lindblom for their dedication and professionalism in the lab, Anna Ståhle, for keeping Magnus in line and for having such cute kids, also for being a good friend and making nice baked goods, Rutinen, för resetipsen!

GIG gruppen; Lisa Christiansson, the perfect lunch conversation, are we human or are we dancer?, Sara Mangsbo, I hope I have your respect, I will never bake a sugarless brownie for you, Moa Fransson, your witty comments at lunch are a re- freshing break from all the Nobel Prize winning research we’re all performing, Lin- da and hot, hot, hot, hot Fredrik (aka Ninja puker Sandin), you’ve been great friends and it was fun showing you around my hood on the mean streets of Minne- apolis and Duluth, Hannah Karlsson, for being such a good dancer, it’s a joy just to watch, Daniel Jarblad, for tips on how to Lindy-hop and keeping Justyna in Swe- den, Justyna Leja, somehow it’s like you are my child and at the same time my parent, in any case you’re great and if you put your mind to it you can do it!, Lina Liljenfeldt, I’m always impressed how your shirt can match your jewelry can match your make-up can match your nails without looking like you’re trying to match everything, Pella and Camilla, thanks for the great cake, de-lish! Di Yu and Chuan Jin, for help with the reference in the history section of the thesis, Björn Carlsson, for your assessments of the lab and your knowledge of Star Wars, To the others, thanks for making the lab a fun place to work and eat.

Ole Forsberg, for taking me with you to hockey, showing me kayaking and for advice and opinions about lab and life, Jennie Bäck, for smiling so much, interest- ing conversations about coagulation, and helping with my Swedish, Graciela Elgue¸ for reminding me there is oceans of time, Javier Sanchez, for helping me under- stand to some degree what serum can offer our friends the islets.

Admin perssonel; Mona Persson, Ann-Sofie and Nettan Lindberg, Elin Ekberg, Karin Erlandsson, Anna-Maria Andersson, and the new (for us Clinimmers) IGP admin staff, thanks for doing the jobs I have no patience for. Stort TACK!

57

My roomies and associates; Alexander Boikov, for introducing me to adjustable beds, Maria Hårdstedt and Per Johansson, for fixing my first digs in Valsätra and keeping the rent low, Maria, for your thoughtfulness and listening, Frauke Fich- tner and Cathrin Schulz (Mueller)¸ for teaching me the most interesting German phrases to say to your friends when they came to visit, ich habe einen kater.

Julia Paraskova, my favorite Swedish/American/Russian-speaking, GAC- attending, game-playing, Guitar Hero-rocking, chocolate cake-making friend, I’m so happy that you can play the piano, Dr. Daniel Lundberg, oh how I Love To Hate You but must admit with your GAC roots, MST3K collection, and chemical imbal- ances I kind of like you, too, Pharmen-gänget, Dr. Mattias Norby, Dr. Johanna Dernfalk, Dr. Joris Van Schaik, and Dr. Anna Olofsson for the pleasant company and good times at the old Pharmen dinners.

To my US friends whom I miss, you are all welcome whenever; Seth Nelson, we were on the same hockey team for our entire career until college, hard to imagine life without all the fun times we’ve shared (e.g. Branson hostess, hunting, hockey trips, ping pong, crazy boat rides, hitting balls onto the frozen Crystal Lake, biking, bonfires on Deer Lake), David Rajala, for being a great example, sharing trips and fun times at the lake, strobe-light fighting, Nwahs Nacnud Nosettam, for being hilarious, the human pop culture king, always laughing when you’re around, Dan and Susie Miller, Dan, you’ve been a great friends since almost the beginning, “…Dan and Andrew rule, HUH!”, Susie, IKEA stole my thunder by selling Mara- bou in the US, now I’m worthless to you, Cory and Sara Rehnstrand, for all the laughs and good times, hot tubs, cheering in Lantinen’s sauna, being good friends, love and miss your sense of humor, Jeff and Amy Wagner, for fixing tix to some hockey games and beating all of us at golf regularly, kept us humble, Moose Ben- ner, for all the Chris Farley impressions, skunky, golf and hilarious times we’ve shared, Aaron Pulskamp, for being a great friend during our younger years, blam- ing me for when you drove the three-wheeler into the fence, pulling me on the field of stones on the sled, tossing frozen cats into the swamp, watching your dad swear at the calves when taking care of them, speaking alien, attempting what would have been an Oscar winning film “The Fly”, and being a generally upstanding citizen, Peter Nerothin, for keeping it real and starting Insulindependence, love your take on life, Scottie Morris and Wickum, good times shared in college and it’s always good to catch up when I’m home.

Dancing friends; Martin och Mia Tengstrand, for the wonderful times on and off the dance floor, I finally got the dishwasher installed, Sofia Forsberg, for giving me my first dance where I really fell in love with dance, Ulrika Kulin, Matilda Stigell, Hanna Persson, Salina Jerbi, Titti Geschwindt, Jana Larsson, Knapp Maria Pettersson, Erik Bergstrom, Stephan Jonsson, Mats Hjelmstedt, Sara Bodeby, Madde o Patrik, och många fler för att ni gör livet så värt att leva genom dans!

58

My toughest hockey coach, Ted Brill for being a goal-oriented, hard as nails, moti- vator. I was sick during a game and his response to my prediction I would lose con- trol of bodily functions, “Well, shit on them! Now get out there!” (I sat out the next game) You are missed.

To Allison Elmore, for letting me visit you for a month every summer when we grew up and assault you with my cheesy jokes and steal your Pringles. (I just rea- lized Word autocorrects for lowercase Pringles…scary) I still have your letters, you have to come here to get them.

To Dr. Angelika Danielsson, for inviting me to take that first Bugg dance class, for helping out on this thesis and calming me down when I needed it. You know what to say to keep me in line and you’ve made my time here memorable and fruitful.

To Dr. Dieter Fuchs, Dr. Ania P aczek, Mari Sandrup, Olof and Dr. Maria Andrén, Olle Pallars and Itti Andersson, you are great friends, like family for me. I appreciate so much all the fun times, trips, food, evening teas, fikas, conversations about life, and support. To Dieter and Itti, I’m so glad I met you early on in my time here, we’ve really shared a lot of fun times together and you two know me best here. Special thanks to Dieter for your great suggestions for the thesis. Itti, just so you know, I consider you another sister, at the same time you’re an uncomfortably good friend.

Kerstin o Åke Andersson, stort tack för att jag har känt mig så välkommen i det här landet. Ni har bidraget stort till att jag trivs så mycket. Och presenten har gjort sitt uppdrag utmärkt! Tusen tack!

Dar and Mike Nielsen and family, you’ve been great friends to my family and taught me many things about life, how to live and succeed. Thank you for all the times we’ve shared and for your support. We love you.

Jim Pirius, the godfather, probably the farthest thing from the movie…you’re kind, thoughtful, fun, smart and have given me many wonderful memories. You also feel like a part of the family and I’m so happy it turned out like that. Love you, too.

To my family, all my aunts and uncles and cousins, in Swedish they say “none named, none forgotten”, I’m running with that theory on this one but know I love you all and cherish the times when we’re together. I’m so lucky to have access to so many wonderful people who can’t get rid of me easily. My best memories are when we’ve been together. The food, the laughs, the impersonations, the hockey, conver- sations and trips together have all been wonderful. Why do we have to be so far apart, WHY?! BOO-hooo…I love you.

Jessica Friberg, happy to see you’ve got a nice place to call home.

59

A special thanks to Grandpa Lutz, I love hearing your stories, thanks for helping renovate my house in Minneapolis, all car help and all home grown corn etc. You are wonderful and I’m so happy for the time we get to spend together.

Meredith and Sean Long, I am proud to call you sister and brother. You have four gorgeous children, and I apologize I am not around more for them. To the kids, it’s been a lot of fun being your jungle gym, I just love seeing your smiling faces, Han- nah, keep dancing, you’re so good at it, Parker, thanks for being so nice, some- times I remember how nice you are so I can keep things in perspective for myself, Matthew, I hope we can toss the football around when I am around, I will teach you some dance moves, Jace, what a joy you are to have around, always smiling!

Those family members who are not here; Grandma and Grandpa Friberg, Grandma Lutz, Dad, and Joel, I miss you all so much and wish you could be here to see what I’ve done and who I’ve become.

Mom, I hope you’re proud. You were the one who got up at 5:30 in the morning to take me to hockey practice, who always thought I was the best player, you make great spaghetti sauce, and always have an ear for me even though you’ve heard my problems a thousand times. Hope we have a “totally relaxing” time when you’re here this go ‘round. I love you.

Special thanks to all ORGAN DONORS, without your precious gifts we would not be able to help others live better, longer lives.

60

References

1. J. B. Ferguson, personal communication. 2. G. D. Dwivedi, S., Sushruta - the Clinician - Teacher par Excellence. Indian J Chest Dis Allied Sci 49, 243 (2007). 3. T. Willis, Pharmaceutic rationalis sive diatriba de medicamentorum operationibus in humano corpore. 2 vols. London, (1674-1675). 4. M. Dobson, Experiments and observations on the urine in diabetes. Med Obs Inq 5, 298 (1776). 5. T. Cawley, A singular case of diabetes, consisting entirely in the quality of the urine; with an inquiry into the different theories of that disease. London Med J, 286 (1788). 6. J. Rollo, An account of Two Cases of the Diabetes Mellitus, With Remarks as They Arose During the Progress of the Cure. London: C. Dilly, (1797). 7. P. Langerhans, Friedrich Wilhelm Universität (1869). 8. E. Laguesse, Sur la formation des ilots de Langerhans dans le pancréas. Comptes rendus hebdomadaires des séances et memoires de la société de biologie 5, 819 (1893). 9. J. Mering, Minkowski, O., Diabetes Mellitus nach Pankeasexstirpation. Archiv fur experimentellen Pathologie und Pharmakologie 26, 370 (1890). 10. P. W. Williams, Notes on diabetes treated with extract and by grafts of sheep's pancreas. The British Medical Journal 19, 1303 (1894). 11. E. L. Opie, On the relation of chronic interstitial pancreatitis to the islands of Langerhans and to diabetes mellitus. J Exp Med 5, 393 (1901). 12. J. De Meyer, Sur la signification physiologique de la sécrétion interne du pancréas. Zentralbl Physiol 18, S826 (1904). 13. C. H. Best, The internal secretion of the pancreas. Can Med Assoc J 87, 1046 (Nov 17, 1962). 14. F. G. Banting, Best, C.H., The internal secretion of the pancreas. J Lab Clin Med 7, 251 (1922). 15. L. Rosenfeld, Insulin: discovery and controversy. Clin Chem 48, 2270 (Dec, 2002). 16. Banting and Best develop the 'miracle drug'. http://archives.cbc.ca/health/medical_research/clips/4060/. accessed March 1, 2011, (Broadcast Date: Sept. 26 1977). 17. E. Lanceraux, Le diabète maigre: ses symptomes, son évolution, son pronostic et son traitement. Union Méd Paris 20, 205 (1880).

61

18. Classification and diagnosis of diabetes mellitus and other categories of glucose intolerance. National Diabetes Data Group. Diabetes 28, 1039 (Dec, 1979). 19. W. Gepts, Pathologic anatomy of the pancreas in juvenile diabetes mellitus. Diabetes 14, 619 (Oct, 1965). 20. J. Ludvigsson, Why diabetes incidence increases--a unifying theory. Annals of the New York Academy of Sciences 1079, 374 (Oct, 2006). 21. M. Karvonen et al., Incidence of childhood type 1 diabetes worldwide. Diabetes Mondiale (DiaMond) Project Group. Diabetes Care 23, 1516 (Oct, 2000). 22. P. Onkamo, S. Vaananen, M. Karvonen, J. Tuomilehto, Worldwide increase in incidence of Type I diabetes--the analysis of the data on published incidence trends. Diabetologia 42, 1395 (Dec, 1999). 23. P. Jarosz-Chobot et al., Rapid increase in the incidence of type 1 diabetes in Polish children from 1989 to 2004, and predictions for 2010 to 2025. Diabetologia 54, 508 (Mar, 2011). 24. G. Bruno et al., Age-period-cohort analysis of 1990-2003 incidence time trends of childhood diabetes in Italy: the RIDI study. Diabetes 59, 2281 (Sep, 2010). 25. Incidence and trends of childhood Type 1 diabetes worldwide 1990- 1999. Diabetic medicine : a journal of the British Diabetic Association 23, 857 (Aug, 2006). 26. E. V. Moltchanova, N. Schreier, N. Lammi, M. Karvonen, Seasonal variation of diagnosis of Type 1 diabetes mellitus in children worldwide. Diabetic medicine : a journal of the British Diabetic Association 26, 673 (Jul, 2009). 27. Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. Atlanta, GA: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, 2011. 28. G. Soltesz, C. C. Patterson, G. Dahlquist, Worldwide childhood type 1 diabetes incidence--what can we learn from epidemiology? Pediatr Diabetes 8 Suppl 6, 6 (Oct, 2007). 29. V. Harjutsalo, L. Sjoberg, J. Tuomilehto, Time trends in the incidence of type 1 diabetes in Finnish children: a cohort study. Lancet 371, 1777 (May 24, 2008). 30. N. Lammi et al., Marked temporal increase in the incidence of type 1 and type 2 diabetes among young adults in Finland. Diabetologia 51, 897 (May, 2008). 31. Y. Berhan, I. Waernbaum, T. Lind, A. Mollsten, G. Dahlquist, Thirty years of prospective nationwide incidence of childhood type 1 diabetes: the accelerating increase by time tends to level off in Sweden. Diabetes 60, 577 (Feb, 2011). 32. T. Deckert, J. E. Poulsen, M. Larsen, Prognosis of diabetics with diabetes onset before the age of thirty-one. I. Survival, causes of death, and complications. Diabetologia 14 , 363 (Jun, 1978).

62

33. A. S. Go, G. M. Chertow, D. Fan, C. E. McCulloch, C. Y. Hsu, Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. The New England journal of medicine 351, 1296 (Sep 23, 2004). 34. Diabetes mellitus: a major risk factor for cardiovascular disease. A joint editorial statement by the American Diabetes Association; The National Heart, Lung, and Blood Institute; The Juvenile Diabetes Foundation International; The National Institute of Diabetes and Digestive and Kidney Diseases; and The American Heart Association. Circulation 100, 1132 (Sep 7, 1999). 35. I. Svenningsson, B. Marklund, S. Attvall, B. Gedda, Type 2 diabetes: perceptions of quality of life and attitudes towards diabetes from a gender perspective. Scand J Caring Sci, (Mar 1, 2011). 36. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 329, 977 (Sep 30, 1993). 37. N. H. White et al., Identification of type I diabetic patients at increased risk for hypoglycemia during intensive therapy. The New England journal of medicine 308, 485 (Mar 3, 1983). 38. G. Bruno, A. Landi, Epidemiology and costs of diabetes. Transplantation proceedings 43, 327 (Jan-Feb, 2011). 39. A. A. Irvine, D. Cox, L. Gonder-Frederick, Fear of hypoglycemia: relationship to physical and psychological symptoms in patients with insulin-dependent diabetes mellitus. Health Psychol 11, 135 (1992). 40. W. D. Kelly, R. C. Lillehei, F. K. Merkel, Y. Idezuki, F. C. Goetz, Allotransplantation of the pancreas and duodenum along with the kidney in diabetic nephropathy. Surgery 61, 827 (Jun, 1967). 41. A. C. Gruessner, D. E. Sutherland, Pancreas transplant outcomes for United States (US) cases as reported to the United Network for Organ Sharing (UNOS) and the International Pancreas Transplant Registry (IPTR). Clin Transpl, 45 (2008). 42. T. M. Sasaki et al., Successful long-term kidney-pancreas transplants in diabetic patients with high C-peptide levels. Transplantation 65, 1510 (Jun 15, 1998). 43. M. R. Rickels, R. Mueller, K. L. Teff, A. Naji, {beta}-Cell secretory capacity and demand in recipients of islet, pancreas, and kidney transplants. The Journal of clinical and metabolism 95, 1238 (Mar, 2010). 44. A. Humar et al., The impact of donor obesity on outcomes after cadaver pancreas transplants. Am J Transplant 4, 605 (Apr, 2004). 45. A. M. Shapiro et al., International trial of the Edmonton protocol for islet transplantation. N Engl J Med 355, 1318 (Sep 28, 2006). 46. E. A. Ryan et al., Five-year follow-up after clinical islet transplantation. Diabetes 54, 2060 (Jul, 2005). 47. L. C. Stene et al., Normal but increasing hemoglobin A1c levels predict progression from islet autoimmunity to overt type 1 diabetes:

63

Diabetes Autoimmunity Study in the Young (DAISY). Pediatr Diabetes 7, 247 (Oct, 2006). 48. P. Fiorina et al., Long-term beneficial effect of islet transplantation on diabetic macro-/microangiopathy in type 1 diabetic kidney- transplanted patients. Diabetes Care 26, 1129 (Apr, 2003). 49. K. I. Rother, D. M. Harlan, Challenges facing islet transplantation for the treatment of type 1 diabetes mellitus. J Clin Invest 114, 877 (Oct, 2004). 50. M. Elashoff, A. V. Matveyenko, B. Gier, R. Elashoff, P. C. Butler, Increased Incidence of Pancreatitis and Cancer Among Patients Given Glucagon Like Peptide-1 Based Therapy. Gastroenterology, (Feb 17, 2011). 51. T. Tharavanij et al., Improved long-term health-related quality of life after islet transplantation. Transplantation 86, 1161 (Nov 15, 2008). 52. T. Berney, P. R. Johnson, Donor pancreata: evolving approaches to organ allocation for whole pancreas versus islet transplantation. Transplantation 90, 238 (Aug 15, 2010). 53. A. Humar et al., Technical failures after pancreas transplants: why grafts fail and the risk factors--a multivariate analysis. Transplantation 78, 1188 (Oct 27, 2004). 54. I. Matsumoto et al., Improvement in islet yield from obese donors for human islet transplants. Transplantation 78, 880 (Sep 27, 2004). 55. J. Beckwith, J. A. Nyman, B. Flanagan, R. Schrover, H. J. Schuurman, A health economic analysis of clinical islet transplantation. Clin Transplant, (Feb 16, 2011). 56. A. P. Guignard et al., Cost analysis of human islet transplantation for the treatment of type 1 diabetes in the Swiss-French Consortium GRAGIL. Diabetes Care 27, 895 (Apr, 2004). 57. G. L. Szot et al., Successful clinical islet isolation using a GMP- manufactured collagenase and neutral protease. Transplantation 88, 753 (Sep 27, 2009). 58. D. O'Gorman et al., The standardization of pancreatic donors for islet isolation. Transplant Proc 37, 1309 (Mar, 2005). 59. M. Goto et al., Refinement of the automated method for human islet isolation and presentation of a closed system for in vitro islet culture. Transplantation 78, 1367 (Nov 15, 2004). 60. B. Hering, Wijkstrom, M., Eckman, P.M., in Islet Transplantation: Status, Protocols and perspectives, Collaborative Islet Transplnat Registry (CITR), R. W. G. Gruessner, Sutherland, D.E.R., Ed. (Springer, 2004). 61. J. J. Blondet et al., The role of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Surg Clin North Am 87, 1477 (Dec, 2007). 62. D. C. Wahoff et al., Autologous islet transplantation to prevent diabetes after pancreatic resection. Ann Surg 222, 562 (Oct, 1995).

64

63. M. A. Webb et al., Islet auto transplantation following total pancreatectomy: a long-term assessment of graft function. Pancreas 37, 282 (Oct, 2008). 64. E. Rafael et al., Intramuscular autotransplantation of pancreatic islets in a 7-year-old child: a 2-year follow-up. Am J Transplant 8, 458 (Feb, 2008). 65. S. Illouz et al., Islet autotransplantation restores normal glucose tolerance in a patient with chronic pancreatitis. Diabetes Care 30, e130 (Dec, 2007). 66. P. R. Johnson et al., Pancreatic islet autotransplantation combined with total pancreatectomy for the treatment of chronic pancreatitis-- the Leicester experience. J Mol Med 77, 130 (Jan, 1999). 67. F. Ris et al., Islet autotransplantation after extended pancreatectomy for focal benign disease of the pancreas. Transplantation 91, 895 (Apr 27, 2011). 68. M. D. Bellin et al., Similar islet function in islet allotransplant and autotransplant recipients, despite lower islet mass in autotransplants. Transplantation 91, 367 (Feb 15, 2011). 69. V. A. Huurman et al., Cellular islet autoimmunity associates with clinical outcome of islet cell transplantation. PLoS One 3, e2435 (2008). 70. B. O. Roep et al., Auto- and alloimmune reactivity to human islet allografts transplanted into type 1 diabetic patients. Diabetes 48, 484 (Mar, 1999). 71. C. Jaeger, M. D. Brendel, B. J. Hering, M. Eckhard, R. G. Bretzel, Progressive islet graft failure occurs significantly earlier in autoantibody-positive than in autoantibody-negative IDDM recipients of intrahepatic islet allografts. Diabetes 46, 1907 (Nov, 1997). 72. C. A. van Kampen et al., Alloreactivity against repeated HLA mismatches of sequential islet grafts transplanted in non-uremic type 1 diabetes patients. Transplantation 80, 118 (Jul 15, 2005). 73. V. A. Huurman et al., Allograft-specific cytokine profiles associate with clinical outcome after islet cell transplantation. Am J Transplant 9, 382 (Feb, 2009). 74. J. L. Contreras et al., Brain death significantly reduces isolated pancreatic islet yields and functionality in vitro and in vivo after transplantation in rats. Diabetes 52, 2935 (Dec, 2003). 75. P. E. Cryer, S. N. Davis, H. Shamoon, Hypoglycemia in diabetes. Diabetes Care 26, 1902 (Jun, 2003). 76. J. A. Johnson, M. Kotovych, E. A. Ryan, A. M. Shapiro, Reduced fear of hypoglycemia in successful islet transplantation. Diabetes Care 27, 624 (Feb, 2004). 77. B. W. Paty, P. A. Senior, J. R. Lakey, A. M. Shapiro, E. A. Ryan, Assessment of glycemic control after islet transplantation using the continuous glucose monitor in insulin-independent versus insulin-

65

requiring type 1 diabetes subjects. Diabetes Technol Ther 8, 165 (Apr, 2006). 78. L. Luzi et al., Metabolic effects of restoring partial beta-cell function after islet allotransplantation in type 1 diabetic patients. Diabetes 50, 277 (Feb, 2001). 79. R. Lehmann, G. A. Spinas, W. Moritz, M. Weber, Has time come for new goals in human islet transplantation? Am J Transplant 8, 1096 (Jun, 2008). 80. J. S. Najarian et al., Human islet transplantation: a preliminary report. Transplantation proceedings 9, 233 (Mar, 1977). 81. S. A. White, R. F. James, S. M. Swift, R. M. Kimber, M. L. Nicholson, Human islet cell transplantation--future prospects. Diabetic medicine : a journal of the British Diabetic Association 18, 78 (Feb, 2001). 82. A. M. Shapiro et al., Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 343, 230 (Jul 27, 2000). 83. The CITR Coordinating Center and Investigators. The Collaborative Islet Transplant Registry 2009 Annual Report. www.citregistry.org/reports/reports.htm., (2009). 84. . (U.S. Food and Drug Administration, http://www.fda.gov/AboutFDA/Transparency/Basics/ucm194879.ht m Access date: April 9,2011). 85. E. Linetsky, C. Ricordi, Regulatory challenges in manufacturing of pancreatic islets. Transplant Proc 40, 424 (Mar, 2008). 86. E. D. Sotta et al., Establishing an islet transplantation program in a developing country. Transplant Proc 36, 1700 (Jul-Aug, 2004). 87. H. Rouard et al., A closed and single-use system for monocyte enrichment: potential for dendritic cell generation for clinical applications. Transfusion 43, 481 (Apr, 2003). 88. T. Sun, D. Norton, J. W. Haycock, A. J. Ryan, S. MacNeil, Development of a closed bioreactor system for culture of tissue- engineered skin at an air-liquid interface. Tissue Eng 11, 1824 (Nov- Dec, 2005). 89. S. J. Wagner, A. C. Myrup, Toward closed-system culture of blood origin endothelial cells. Transfusion 45, 1201 (Jul, 2005). 90. G. D. Taylor, T. Kirkland, J. Lakey, R. Rajotte, G. L. Warnock, Bacteremia due to transplantation of contaminated cryopreserved pancreatic islets. Cell Transplant 3, 103 (Jan-Feb, 1994). 91. J. R. Lakey, R. V. Rajotte, G. L. Warnock, Microbial surveillance of human islet isolation, in vitro culture, and cryopreservation. Clin Invest Med 18, 168 (Jun, 1995). 92. P. Bucher et al., Microbial surveillance during human pancreatic islet isolation. Transplant Proc 36, 1147 (May, 2004). 93. T. Kin, S. Rosichuk, A. M. Shapiro, J. R. Lakey, Detection of microbial contamination during human islet isolation. Cell Transplant 16, 9 (2007).

66

94. J. Lloveras et al., Significance of contaminated islet preparations in clinical islet transplantation. Transplant Proc 26, 579 (Apr, 1994). 95. J. R. Lakey et al., Development of an automated computer- controlled islet isolation system. Cell Transplant 6, 47 (Jan-Feb, 1997). 96. B. J. Hering et al., The evaluation of neutral density separation utilizing Ficoll-sodium diatrizoate and Nycodenz and centrifugal elutriation in the purification of bovine and canine islet preparations. Horm Metab Res Suppl 25, 57 (1990). 97. R. A. Klaffschenkel et al., A closed system for islet isolation and purification using the COBE2991 cell processor may reduce the need of clean room facilities. Cell Transplant 16, 587 (2007). 98. T. Anazawa et al., Prediction of pancreatic tissue densities by an analytical test gradient system before purification maximizes human islet recovery for islet autotransplantation/allotransplantation. Transplantation 91, 508 (Mar 15, 2011). 99. S. P. Lake et al., Large-scale purification of human islets utilizing discontinuous albumin gradient on IBM 2991 cell separator. Diabetes 38 Suppl 1, 143 (Jan, 1989). 100. S. Matsumoto et al., Insulin independence after living-donor distal pancreatectomy and islet allotransplantation. Lancet 365, 1642 (May 7-13, 2005). 101. H. A. Tons, O. T. Terpstra, E. Bouwman, Heterogeneity of human pancreata in perspective of the isolation of the islets of langerhans. Transplantation proceedings 40, 367 (Mar, 2008). 102. D. Brandhorst, H. Brandhorst, B. J. Hering, K. Federlin, R. G. Bretzel, Large variability of the intracellular ATP content of human islets isolated from different donors. J Mol Med 77, 93 (Jan, 1999). 103. P. Witkowski et al., Validation of the scoring system for standardization of the pancreatic donor for islet isolation as used in a new islet isolation center. Transplantation proceedings 38, 3039 (Nov, 2006). 104. C. Rastellini et al., The effect of donor age on the outcome of islets isolated from human pancreata. Transplantation proceedings 27, 3257 (Dec, 1995). 105. R. Nano et al., Islet isolation for allotransplantation: variables associated with successful islet yield and graft function. Diabetologia 48, 906 (May, 2005). 106. J. R. Lakey et al., Variables in organ donors that affect the recovery of human islets of Langerhans. Transplantation 61, 1047 (Apr 15, 1996). 107. D. Brandhorst, B. J. Hering, H. Brandhorst, K. Federlin, R. G. Bretzel, Influence of donor data and organ procurement on human islet isolation. Transplant Proc 26, 592 (Apr, 1994). 108. R. J. Ketchum et al., Analysis of donor age and cold ischemia time as factors in cadaveric human islet isolation. Transplantation proceedings 26, 596 (Apr, 1994).

67

109. N. Niclauss, Bosco, D, Morel P., Demuylder-Mischler, S., Brault, C., Milliat-Guittard,L., Colin, C., Parnaud G.,Muller, Y. D., Giovannoni, L., Meier R., Toso, C., Badet, L., Benhamou, PY., Berney, T., Influence of Donor Age on Islet Isolation and Transplantation Outcome. Transplantation 90, (2010). 110. Y. Zeng, M. A. Torre, T. Karrison, J. R. Thistlethwaite, The correlation between donor characteristics and the success of human islet isolation. Transplantation 57, 954 (Mar 27, 1994). 111. P. Y. Benhamou et al., Human islet isolation in 104 consecutive cases. Factors affecting isolation success. Transplantation 57, 1804 (Jun 27, 1994). 112. J. S. Kaddis, J. S. Danobeitia, J. C. Niland, T. Stiller, L. A. Fernandez, Multicenter analysis of novel and established variables associated with successful human islet isolation outcomes. Am J Transplant 10, 646 (Mar, 2010). 113. M. Takita et al., One hundred human pancreatic islet isolations at Baylor Research Institute. Proc (Bayl Univ Med Cent) 23, 341 (Oct, 2010). 114. J. R. Lakey et al., Effect of core pancreas temperature during cadaveric procurement on human islet isolation and functional viability. Transplantation 73, 1106 (Apr 15, 2002). 115. P. C. Watt et al., Donor factors affecting successful isolation of islets from the human pancreas. Transplantation proceedings 26, 594 (Apr, 1994). 116. O. Sabek, D. Hathaway, D. Fraga, A. O. Gaber, Influence of human donor factors on pancreatic collagenase digestion. Transplantation proceedings 30, 353 (Mar, 1998). 117. T. Hubert et al., Acute insulin response to arginine in deceased donors predicts the outcome of human islet isolation. Am J Transplant 8, 872 (Apr, 2008). 118. B. J. Hering et al., Transplantation of cultured islets from two-layer preserved pancreases in type 1 diabetes with anti-CD3 antibody. Am J Transplant 4, 390 (Mar, 2004). 119. T. Tsujimura et al., Human islet transplantation from pancreases with prolonged cold ischemia using additional preservation by the two-layer (UW solution/perfluorochemical) cold-storage method. Transplantation 74, 1687 (Dec 27, 2002). 120. Y. Kuroda et al., A new, simple method for cold storage of the pancreas using perfluorochemical. Transplantation 46, 457 (Sep, 1988). 121. J. Caballero-Corbalan et al., No beneficial effect of two-layer storage compared with UW-storage on human islet isolation and transplantation. Transplantation 84, 864 (Oct 15, 2007). 122. T. Kin et al., Islet isolation and transplantation outcomes of pancreas preserved with University of Wisconsin solution versus two-layer method using preoxygenated perfluorocarbon. Transplantation 82, 1286 (Nov 27, 2006).

68

123. C. Hellerstroem, A Method for the Microdissection of Intact Pancreatic Islets of Mammals. Acta Endocrinol (Copenh) 45, 122 (Jan, 1964). 124. S. Moskalewski, Isolation and culture of the islets of Langerhans of the guinea pig. Gen Comp Endocrinol, 342 (1965). 125. P. E. Lacy, M. Kostianovsky, Method for the isolation of intact islets of Langerhans from the rat pancreas. Diabetes 16, 35 (Jan, 1967). 126. A. Horaguchi, R. C. Merrell, Preparation of viable islet cells from dogs by a new method. Diabetes 30, 455 (May, 1981). 127. D. W. Gray, P. McShane, A. Grant, P. J. Morris, A method for isolation of islets of Langerhans from the human pancreas. Diabetes 33, 1055 (Nov, 1984). 128. C. Ricordi, P. E. Lacy, E. H. Finke, B. J. Olack, D. W. Scharp, Automated method for isolation of human pancreatic islets. Diabetes 37, 413 (Apr, 1988). 129. E. Linetsky et al., Improved human islet isolation using a new enzyme blend, liberase. Diabetes 46, 1120 (Jul, 1997). 130. T. J. Fetterhoff et al., Human pancreatic dissociation using a purified enzyme blend. Transplant Proc 27, 3282 (Dec, 1995). 131. G. H. Wolters, G. H. Vos-Scheperkeuter, H. C. Lin, R. van Schilfgaarde, Different roles of class I and class II Clostridium histolyticum collagenase in rat pancreatic islet isolation. Diabetes 44, 227 (Feb, 1995). 132. G. H. Wolters, G. H. Vos-Scheperkeuter, J. H. van Deijnen, R. van Schilfgaarde, An analysis of the role of collagenase and protease in the enzymatic dissociation of the rat pancreas for islet isolation. Diabetologia 35, 735 (Aug, 1992). 133. T. Kin, D. O'Gorman, P. Senior, A. M. Shapiro, Experience of islet isolation without neutral protease supplementation. Islets 2, 278 (Sep-Oct, 2010). 134. P. R. Johnson, S. A. White, N. J. London, Collagenase and human islet isolation. Cell Transplant 5, 437 (Jul-Aug, 1996). 135. H. Jahr, G. Pfeiffer, B. J. Hering, K. Federlin, R. G. Bretzel, Endotoxin-mediated activation of cytokine production in human PBMCs by collagenase and Ficoll. J Mol Med 77, 118 (Jan, 1999). 136. T. Eckhardt, H. Jahr, K. Federlin, R. G. Bretzel, Endotoxin impairs the engraftment of rat islets transplanted beneath the kidney capsule of C57BL/6-mice. J Mol Med 77, 123 (Jan, 1999). 137. M. J. Barnett et al., Quantitative assessment of collagenase blends for human islet isolation. Transplantation 80, 723 (Sep 27, 2005). 138. H. Brandhorst, M. D. Brendel, M. Eckhard, R. G. Bretzel, D. Brandhorst, Influence of neutral protease activity on human islet isolation outcome. Transplant Proc 37, 241 (Jan-Feb, 2005). 139. T. Kin, P. R. Johnson, A. M. Shapiro, J. R. Lakey, Factors influencing the collagenase digestion phase of human islet isolation. Transplantation 83, 7 (Jan 15, 2007).

69

140. D. O'Gorman, T. Kin, D. McGhee-Wilson, A. M. Shapiro, J. R. Lakey, Multi-lot analysis of custom collagenase enzyme blend in human islet isolations. Transplant Proc 37, 3417 (Oct, 2005). 141. H. Brandhorst et al., The ratio between class II and class I collagenase determines the amount of neutral protease activity required for efficient islet release from the rat pancreas. Transplant Proc 37, 215 (Jan-Feb, 2005). 142. H. Brandhorst et al., The ratio between collagenase class I and class II influences the efficient islet release from the rat pancreas. Transplantation 85, 456 (Feb 15, 2008). 143. T. Kin, X. Zhai, D. O'Gorman, A. M. Shapiro, Detrimental effect of excessive collagenase class II on human islet isolation outcome. Transpl Int 21, 1059 (Nov, 2008). 144. H. Brandhorst et al., Large-scale comparison of Liberase HI and collagenase NB1 utilized for human islet isolation. Cell transplantation 19, 3 (2010). 145. B. Antonioli et al., Characterization of collagenase blend enzymes for human islet transplantation. Transplantation 84, 1568 (Dec 27, 2007). 146. G. Klock et al., Fractions from commercial collagenase preparations: use in enzymic isolation of the islets of Langerhans from porcine pancreas. Cell Transplant 5, 543 (Sep-Oct, 1996). 147. D. O'Gorman et al., Comparison of human islet isolation outcomes using a new mammalian tissue-free enzyme versus collagenase NB- 1. Transplantation 90, 255 (Aug 15, 2010). 148. A. N. Balamurugan et al., Successful human islet isolation and transplantation indicating the importance of class 1 collagenase and collagen degradation activity assay. Transplantation 89, 954 (Apr 27, 2010). 149. J. Caballero-Corbalan et al., Mammalian tissue-free liberase: a new GMP-graded enzyme blend for human islet isolation. Transplantation 90, 332 (Aug 15, 2010). 150. J. Caballero-Corbalan et al., Vitacyte collagenase HA: a novel enzyme blend for efficient human islet isolation. Transplantation 88, 1400 (Dec 27, 2009). 151. R. C. McCarthy et al., Development and characterization of a collagen degradation assay to assess purified collagenase used in islet isolation. Transplant Proc 40, 339 (Mar, 2008). 152. H. Brandhorst et al., The effect of truncated collagenase class I isomers on human islet isolation outcome. Transplantation 90, 334 (Aug 15, 2010). 153. O. Korsgren, Nilsson, B., Current status of clinical islet transplantation. Transplantation 79, 1289 (May 27, 2005). 154. J. R. T. Lakey, P. W. Burridge, A. M. J. Shapiro, Technical aspects of islet preparation and transplantation. Transplant International 16, 613 (2003).

70

155. G. S. Robertson, D. R. Chadwick, H. Contractor, R. F. James, N. J. London, The optimization of large-scale density gradient isolation of human islets. Acta Diabetol 30, 93 (1993). 156. B. J. Hering et al., Single-donor, marginal-dose islet transplantation in patients with type 1 diabetes. Jama 293, 830 (Feb 16, 2005). 157. G. S. Robertson et al., Storage of human pancreatic digest in University of Wisconsin solution significantly improves subsequent islet purification. Br J Surg 79, 899 (Sep, 1992). 158. D. Brandhorst, H. Brandhorst, B. J. Hering, K. Federlin, R. G. Bretzel, Islet isolation from the pancreas of large mammals and humans: 10 years of experience. Exp Clin Endocrinol Diabetes 103 Suppl 2, 3 (1995). 159. H. L. Rilo et al., Comparison of human islet number and viability obtained during purification on Euro-Collins-Ficoll gradients processed on a refrigerated or nonrefrigerated COBE 2991 cell separator. Transplant Proc 26, 3430 (Dec, 1994). 160. M. Eckhard, D. Brandhorst, H. Brandhorst, M. D. Brendel, R. G. Bretzel, Optimization in osmolality and range of density of a continuous ficoll-sodium-diatrizoate gradient for isopycnic purification of isolated human islets. Transplant Proc 36, 2849 (Nov, 2004). 161. D. R. Chadwick et al., Pancreatic islet purification using bovine serum albumin: the importance of density gradient temperature and osmolality. Cell Transplant 2, 355 (Jul-Aug, 1993). 162. B. Barbaro et al., Improved human pancreatic islet purification with the refined UIC-UB density gradient. Transplantation 84, 1200 (Nov 15, 2007). 163. J. M. Anderson, M. C. Deeds, A. S. Armstrong, D. A. Gastineau, Y. C. Kudva, Utilization of a test gradient enhances islet recovery from deceased donor pancreases. Cytotherapy 9, 630 (2007). 164. S. Matsumoto et al., Successful islet transplantation from nonheartbeating donor pancreata using modified Ricordi islet isolation method. Transplantation 82, 460 (Aug 27, 2006). 165. Y. Yonekawa et al., The use of test gradients to determine the bottom layer density for subsequent isopycnic purification of human islets on a COBE 2991 cell processor. Xenotransplantation 14, 480 (Sept, 2007). 166. S. Deng et al., Islet alone versus islet after kidney transplantation: metabolic outcomes and islet graft survival. Transplantation 88, 820 (Sep 27, 2009). 167. U. Johansson, A. Olsson, S. Gabrielsson, B. Nilsson, O. Korsgren, Inflammatory mediators expressed in human islets of Langerhans: implications for islet transplantation. Biochemical and biophysical research communications 308, 474 (Aug 29, 2003). 168. R. G. Bretzel et al., Improved survival of intraportal pancreatic islet cell allografts in patients with type-1 diabetes mellitus by refined peritransplant management. J Mol Med 77, 140 (Jan, 1999).

71

169. S. Marzorati et al., Culture medium modulates proinflammatory conditions of human pancreatic islets before transplantation. Am J Transplant 6, 2791 (Nov, 2006). 170. J. Ono, R. Takaki, H. Okano, M. Fukuma, Long-term culture of pancreatic islet cells with special reference to the beta-cell function. In Vitro 15, 95 (Feb, 1979). 171. R. Lehmann et al., Superiority of small islets in human islet transplantation. Diabetes 56, 594 (Mar, 2007). 172. K. K. Papas et al., High-density culture of human islets on top of silicone rubber membranes. Transplantation proceedings 37, 3412 (Oct, 2005). 173. D. A. Baidal et al., The bag method for islet cell infusion. Cell Transplant 12, 809 (2003). 174. C. Ricordi et al., Islet isolation assessment in man and large animals. Acta Diabetol Lat 27, 185 (Jul-Sep, 1990). 175. P. Bucher et al., Assessment of a novel two-component enzyme preparation for human islet isolation and transplantation. Transplantation 79, 91 (Jan 15, 2005). 176. T. Kin et al., Risk factors for islet loss during culture prior to transplantation. Transpl Int 21, 1029 (Nov, 2008). 177. P. Buchwald et al., Quantitative Assessment of Islet Cell Products: Estimating the Accuracy of the Existing Protocol and Accounting for Islet Size Distribution. Cell Transplant 18, 1223 (2009). 178. H. J. Kissler et al., Validation of methodologies for quantifying isolated human islets: an islet cell resources study. Clin Transplant 24, 236 (Aug 30, 2009). 179. T. J. Fetterhoff, K. J. Wile, D. Coffing, T. Cavanagh, M. J. Wright, Quantitation of isolated pancreatic islets using imaging technology. Transplant Proc 26, 3351 (Dec, 1994). 180. M. P. van der Burg et al., Morphometry of native and isolated canine islets: a new approach to isolation assessment. Transplant Proc 26, 632 (Apr, 1994). 181. K. J. Wile, T. J. Fetterhoff, D. Coffing, T. J. Cavanagh, M. J. Wright, Morphologic analysis of pancreatic islets automated image analysis. Transplant Proc 26, 3441 (Dec, 1994). 182. T. J. Cavanagh, J. R. Lakey, M. J. Wright, T. Fetterhoff, K. Wile, Crude collagenase loses islet-isolating efficacy regardless of storage conditions. Transplant Proc 29, 1942 (Jun, 1997). 183. R. Lehmann et al., Evaluation of islet isolation by a new automated method (Coulter Multisizer Ile) and manual counting. Transplant Proc 30, 373 (Mar, 1998). 184. J. P. Stegemann, J. J. O'Neil, D. T. Nicholson, C. J. Mullon, Improved assessment of isolated islet tissue volume using digital image analysis. Cell Transplant 7, 469 (Sep-Oct, 1998). 185. N. Lembert et al., Areal density measurement is a convenient method for the determination of porcine islet equivalents without counting and sizing individual islets. Cell Transplant 12, 33 (2003).

72

186. J. H. Kim et al., Influence of strain and age differences on the yields of porcine islet isolation: extremely high islet yields from SPF CMS miniature pigs. Xenotransplantation 14, 60 (Jan, 2007). 187. P. Girman, Z. Berkova, E. Dobolilova, F. Saudek, How to use image analysis for islet counting. Rev Diabet Stud 5, 38 (Spring, 2008). 188. L. A. Fernandez et al., Validation of large particle flow cytometry for the analysis and sorting of intact pancreatic islets. Transplantation 80, 729 (Sep 27, 2005). 189. N. Niclauss et al., Computer-assisted digital image analysis to quantify the mass and purity of isolated human islets before transplantation. Transplantation 86, 1603 (December 15, 2008). 190. A. Friberg et al., Digital image analysis of islet samples yields consistent, reproducible results. Xenotransplantation 14, 389 (2007). 191. M. Brissova et al., Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy. J Histochem Cytochem 53, 1087 (Sep, 2005). 192. P. M. Jones, Persaud, S. J., Ed., Islet function and insulin secretion, (Blackwell Publishing, ed. 4, 2010), 4, pp. 85-103. 193. F. M. Matschinsky, Banting Lecture 1995. A lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes 45, 223 (Feb, 1996). 194. T. K. Bratanova-Tochkova et al., Triggering and augmentation mechanisms, granule pools, and biphasic insulin secretion. Diabetes 51 Suppl 1, S83 (Feb, 2002). 195. H. Brandhorst, D. Brandhorst, M. D. Brendel, B. J. Hering, R. G. Bretzel, Assessment of intracellular insulin content during all steps of human islet isolation procedure. Cell Transplant 7, 489 (Sep-Oct, 1998). 196. K. K. Papas et al., Human islet oxygen consumption rate and DNA measurements predict diabetes reversal in nude mice. Am J Transplant 7, 707 (Mar, 2007). 197. I. R. Sweet et al., Glucose-stimulated increment in oxygen consumption rate as a standardized test of human islet quality. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons 8, 183 (Jan, 2008). 198. C. Fraker et al., The use of the BD oxygen biosensor system to assess isolated human islets of langerhans: oxygen consumption as a potential measure of islet potency. Cell transplantation 15, 745 (2006). 199. M. Goto et al., A novel predictive method for assessing the quality of isolated pancreatic islets using scanning electrochemical microscopy. Transplant Proc 41, 311 (Jan-Feb, 2009). 200. M. Goto, J. Holgersson, M. Kumagai-Braesch, O. Korsgren, The ADP/ATP ratio: A novel predictive assay for quality assessment of isolated pancreatic islets. Am J Transplant 6, 2483 (Oct, 2006).

73

201. M. S. Hanson et al., A simplified approach to human islet quality assessment. Transplantation 89, 1178 (May 27, 2010). 202. F. Bertuzzi et al., Lessons from in vitro perifusion of pancreatic islets isolated from 80 human pancreases. Cell Transplant 8, 709 (Nov-Dec, 1999). 203. H. Ichii et al., A novel method for the assessment of cellular composition and beta-cell viability in human islet preparations. Am J Transplant 5, 1635 (Jul, 2005). 204. M. Goto et al., Key factors for human islet isolation and clinical transplantation. Transplant Proc 37, 1315 (Mar, 2005). 205. F. Bertuzzi et al., Tissue factor and CCL2/monocyte chemoattractant protein-1 released by human islets affect islet engraftment in type 1 diabetic recipients. J Clin Endocrinol Metab 89, 5724 (Nov, 2004). 206. H. Johansson et al., Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes 54, 1755 (Jun, 2005). 207. S. E. Choi et al., IL-6 protects pancreatic islet beta cells from pro- inflammatory cytokines-induced cell death and functional impairment in vitro and in vivo. Transplant immunology 13, 43 (Jun-Jul, 2004). 208. O. P. Kristiansen, T. Mandrup-Poulsen, Interleukin-6 and diabetes: the good, the bad, or the indifferent? Diabetes 54 Suppl 2, S114 (Dec, 2005). 209. U. Johansson, Uppsala University (2007). 210. F. Bertuzzi, C. Ricordi, Prediction of clinical outcome in islet allotransplantation. Diabetes Care 30, 410 (Feb, 2007). 211. T. Froud et al., Islet transplantation in type 1 diabetes mellitus using cultured islets and steroid-free immunosuppression: Miami experience. Am J Transplant 5, 2037 (Aug, 2005). 212. E. A. Ryan et al., Beta-score: an assessment of beta-cell function after islet transplantation. Diabetes Care 28, 343 (Feb, 2005). 213. H. Noguchi et al., Secretory unit of islet in transplantation (SUIT) and engrafted islet rate (EIR) indexes are useful for evaluating single islet transplantation. Cell Transplant 17, 121 (2008). 214. R. N. Faradji et al., Simple measures to monitor beta-cell mass and assess islet graft dysfunction. Am J Transplant 7, 303 (Feb, 2007). 215. M. R. Rickels et al., {beta}-Cell function following human islet transplantation for type 1 diabetes. Diabetes 54, 100 (Jan, 2005). 216. M. R. Rickels, A. Naji, K. L. Teff, Acute insulin responses to glucose and arginine as predictors of beta-cell secretory capacity in human islet transplantation. Transplantation 84, 1357 (Nov 27, 2007). 217. A. U. Teuscher et al., Successful islet autotransplantation in humans: functional insulin secretory reserve as an estimate of surviving islet cell mass. Diabetes 47, 324 (Mar, 1998). 218. J. S. Petersen et al., Glutamic acid decarboxylase (GAD65) autoantibodies in prediction of beta-cell function and remission in

74

recent-onset IDDM after cyclosporin treatment. The Canadian- European Randomized Control Trial Group. Diabetes 43, 1291 (Nov, 1994). 219. M. R. Rickels et al., Islet cell hormonal responses to hypoglycemia after human islet transplantation for type 1 diabetes. Diabetes 54, 3205 (Nov, 2005). 220. J. P. Palmer et al., C-peptide is the appropriate outcome measure for type 1 diabetes clinical trials to preserve beta-cell function: report of an ADA workshop, 21-22 October 2001. Diabetes 53, 250 (Jan, 2004). 221. O. K. Faber et al., Kinetics of human connecting peptide in normal and diabetic subjects. The Journal of clinical investigation 62, 197 (Jul, 1978). 222. C. J. Greenbaum et al., Mixed-meal tolerance test versus glucagon stimulation test for the assessment of beta-cell function in therapeutic trials in type 1 diabetes. Diabetes Care 31, 1966 (Oct, 2008). 223. P. A. Senior, M. Zeman, B. W. Paty, E. A. Ryan, A. M. Shapiro, Changes in renal function after clinical islet transplantation: four- year observational study. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons 7, 91 (Jan, 2007). 224. S. H. Ihm et al., Effect of donor age on function of isolated human islets. Diabetes 55, 1361 (May, 2006). 225. B. Keymeulen et al., Correlation between beta cell mass and glycemic control in type 1 diabetic recipients of islet cell graft. Proc Natl Acad Sci U S A 103, 17444 (Nov 14, 2006). 226. E. A. Ryan et al., Successful islet transplantation: continued insulin reserve provides long-term glycemic control. Diabetes 51, 2148 (Jul, 2002). 227. M. J. Mihatsch et al., The side-effects of ciclosporine-A and tacrolimus. Clin Nephrol 49, 356 (Jun, 1998). 228. Z. A. Latif, J. Noel, R. Alejandro, A simple method of staining fresh and cultured islets. Transplantation 45, 827 (Apr, 1988). 229. O. M. Sabek, P. Cowan, D. W. Fraga, A. O. Gaber, The effect of isolation methods and the use of different enzymes on islet yield and in vivo function. Cell Transplant 17 , 785 (2008). 230. T. Kin et al., Enhancing the success of human islet isolation through optimization and characterization of pancreas dissociation enzyme. Am J Transplant 7, 1233 (May, 2007). 231. M. Shimoda et al., Assessment of human islet isolation with four different collagenases. Transplantation proceedings 42, 2049 (Jul- Aug, 2010). 232. T. A. Blinman et al., Activation of pancreatic acinar cells on isolation from tissue: cytokine upregulation via p38 MAP kinase. Am J Physiol Cell Physiol 279, C1993 (Dec, 2000).

75

233. B. Movahedi, C. Gysemans, D. Jacobs-Tulleneers-Thevissen, C. Mathieu, D. Pipeleers, Pancreatic duct cells in human islet cell preparations are a source of angiogenic cytokines interleukin-8 and vascular endothelial growth factor. Diabetes 57, 2128 (Aug, 2008). 234. C. Ricordi, J. R. Lakey, B. J. Hering, Challenges toward standardization of islet isolation technology. Transplant Proc 33, 1709 (Feb-Mar, 2001). 235. A. Kim et al., Computer-assisted Large-scale Visualization and Quantification of Pancreatic Islet Mass, Size Distribution and Architecture. Journal of visualized experiments : JoVE, (2011). 236. K. J. Rydgard et al., Procurement of human pancreases for islet isolation-the initiation of a Scandinavian collaborative network. Transplant Proc 33, 2538 (Jun, 2001). 237. H. Brandhorst et al., A new oxygen carrier for improved long-term storage of human pancreata before islet isolation. Transplantation 89, 155 (Jan 27, 2010). 238. J. Kerr-Conte, F. Pattou, Y. Xia, C. Proye, J. Lefebvre, Simple dithizone-stained multilayer test for selection of density gradient before human islet mass purification. Transplant Proc 26, 3413 (Dec, 1994). 239. J. Kerr-Conte et al., Upgrading pretransplant human islet culture technology requires human serum combined with media renewal. Transplantation 89, 1154 (May 15, 2010). 240. A. Sadeghi et al., Large-scale bioreactor expansion of tumor- infiltrating lymphocytes. J Immunol Methods 364, 94 (Feb 1, 2011).

76