ABSTRACT
Transplanting islets of Langerhans consists of implantation in the recipient’s hepatic portal system of endocrine pancreatic tissue, with a variable degree of purification. The field of islet transplantation has evolved significantly since the initial attempts by doctors Minkowski and von Mering in 1882, with remarkable acceleration over the last four decades, thanks to the incredible efforts of the research community worldwide, with continuous improvements in cell processing and transplantation techniques, patient management and development of specific immunotherapy protocols. Restoration of beta-cell function can be obtained by transplantation of allogeneic islets in both non-uremic (Islet Transplant Alone, ITA) and uremic (Simultaneous Islet and Kidney, SIK and Islet After Kidney, IAK) patients with diabetes, providing long-term sustained function and improved metabolic control even when requiring exogenous insulin (i.e., suboptimal islet mass transplanted or development of graft dysfunction). Preservation of beta-cell function is now attained in virtually all recipients of islet autografts, a therapeutic option that should be considered for individuals undergoing total pancreatectomy for non-malignant conditions and, as recently reported for selected cases with malignant conditions. In addition, islet transplantation represents an excellent platform toward the development of cellular therapies aimed at the restoration of beta-cell function using stem cells in the near future. In this chapter, we will review the state-of-the art of clinical islet transplantation.
INTRODUCTION
Diabetes affects 537 million adults (20-79 years) throughout the world (2021) and this number will rise to 643 million by 2030 and 783 million by 2045 (IDF Diabetes Atlas 10th edition, https://diabetesatlas.org/). Many cases of diabetes are successfully treated with life-long multiple daily injections of exogenous insulin and monitoring of blood glucose levels. In the last decades significant improvements in insulin therapy thanks to new preparations (i.e., ultrafast and long-lasting insulin analogues) and the adoption of intensive diabetes management (infusion pumps and continuous glucose monitoring system) have resulted in an overall improvement of patients’ glycemic control and a decreased incidence of chronic complications of diabetes (1,2). However, exogenous insulin administration cannot attain the desirable tight control in the majority of diabetics (3-5), cannot avoid the long-term complications of diabetes in all patients and the life expectancy of patients with diabetes is still shorter compared to that of the general population (6-8). A broad international assessment of treatment outcomes in children and adults with T1D (including 324,501 people from 19 countries in Australasia, Europe and North America) showed that the proportion of patients with HbA1c <7.5% (58 mmol/mol) varied from 15.7% to 46.4% among 44,058 people aged < 15 years, from 8.9% to 49.5% among 50,766 people aged 15-24 years and from 20.5% to 53.6% among 229 677 people aged ≥ 25 years (9). Diabetes is one of the leading causes of end-stage renal disease, blindness and amputation (10). In principle, the treatment for type 1 diabetes, type 3c diabetes and many cases of type 2 diabetes lies in the possibility of replacing destroyed or exhausted beta cell mass in order to restore two essential functions: sensing blood sugar levels and secreting appropriate amounts of insulin in the vascular bed, ideally into the portal system. Currently, the only available clinical approach of restoring beta cell mass in patients with diabetes is the allogenic/autologous transplantation of beta cells (i.e., pancreas or islet transplantation). Clinical trials performed in the last three decades have shown that restoration of beta-cell function via transplantation of isolated islet cells or vascularized pancreas allows reproducibly achievement of a more physiological release of endocrine hormones than exogenous insulin in subjects with diabetes (11). Transplanting islets of Langerhans consists of implantation in the recipient’s hepatic portal system of endocrine pancreatic tissue, with a variable degree of purification. Isolated islets are transplanted using minimally invasive techniques with lower morbidity than vascularized pancreas transplantation, which requires major surgery. The field of islet transplantation has evolved significantly since the initial attempts by doctors Minkowski andvon Mering in 1882 (12), with remarkable acceleration over the last three decades, thanks to the incredible efforts of the research community worldwide, with continuous improvements in cell processing and transplantation techniques, patient management and development of specific immunotherapy protocols. In addition, islet transplantation represents an excellent platform toward the development of cellular therapies aimed at the restoration of beta-cell function using stem cells in the near future. In this chapter, we will review the state-of-the art of clinical islet transplantation.
WHEN TO CONSIDER ISLET TRANSPLANTATION?
Transplantation of pancreatic islet may be considered as a therapeutic option in several conditions associated with loss of beta-cell function (Table 1). The procedure may be performed as Islet Transplant Alone (ITA) in non-uremic subjects, an option generally indicated for the treatment of iatrogenic (surgery-induced) diabetes and for non-uremic patients with Type 1 Diabetes. Subjects with end-stage renal disease (ESRD) may be considered for Simultaneous Islet-Kidney (SIK) or, if already undergone renal transplantation, Islet After Kidney (IAK) transplantation, respectively. In special situations, transplantation of islets may be considered in combination with other organs (i.e., in the context of multi-visceral transplantation following exenteration comprising the pancreas) (13).
The source of the islets for transplantation may be the patient’s own pancreas (autologous or auto-transplant) mainly when surgical removal of the gland is required due to different conditions. After total pancreatectomy, the subject develops surgery-induced (iatrogenic) insulin-requiring diabetes. Introduced in the early 1970’s (14), islet auto-transplantation allows achieving optimal metabolic control without the need for exogenous insulin in approximately 70% of the cases when adequate islet numbers can be recovered from the pancreas (generally >250,000 islet equivalents). More than 500 auto-transplant in patients with near-total or total pancreatectomy have been performed to date (15). The largest series were published by the University of Minnesota (16-19), the University of Cincinnati (20,21), and Leicester (22-25). Even when an inadequate islet mass to attain insulin-independence has been recovered, stable metabolic control and excellent management can be achieved in most subjects undergoing autologous islet transplantation (18,26-31). Islet auto-transplantation is currently reimbursed by health insurance in the United States. In the past auto-transplant has been performed almost exclusively in patients undergoing pancreatectomy because of chronic pancreatitis, successfully preserving β-cell mass and preventing diabetes after major pancreatic resections (15,16,32,33). Additional indications for auto-transplant other than chronic pancreatitis are still controversial (34), and have been limited to the procedure performed only in small case series (35-40) of benign enucleable tumors or pancreatic trauma. Recently, broader selection criteria for auto-transplant were published (39,41), exploring the possibility of extending auto-transplant to patients with known malignancy, either having completion pancreatectomy as treatment for severe pancreatic fistulae or extensive distal pancreatectomy for neoplasms of the pancreatic neck or pancreatoduodenectomy because at high risk of pancreatic fistula (Table 1). Of note, a randomized, open-label, controlled, bicentric trial (NCT01346098) aimed to compare pancreaticoduodenectomy (PD) and Total Pancreatectomy with Islet AutoTransplantation (TP-IAT) in patients at high risk of Post-Operative Pancreatic Fistula (POPF) was recently published. The results indicate that TP-IAT can be considered a valid alternative to PD in these patients, as it reduced complication number, severity and length of hospital stay. Of note, a trend toward a reduction of mortality, even for patients with malignancy was also evident. As expected, TPIAT was associated to a higher risk of diabetes, but IAT was able to preserve, at least in part, the endogenous insulin secretion, mitigating the impact of the pancreoprivic diabetes and assuring a good metabolic control without severe hypoglycemic episodes. In the field of islet transplantation, this study definitively confirmed IAT could be indicated for pancreas diseases other than chronic pancreatitis, suggesting the possibility to extend IAT indications (Milan protocol(42)). For the first time in a randomized prospective design, it was confirmed that IAT is feasible, safe and effective in patients with periampullary cancer, in agreement with previous series of patients undergoing IAT after pancreatic resection for a wide spectrum of disease besides chronic pancreatitis (43) (44) (37). This approach will be tested in further studies in the next years, such as the recently started TPIAT-01 trial (NCT05116072), which hypothesize that TPIAT rather than PD may improve the access to adjuvant chemotherapy in patients with adenocarcinoma.
In the case of subjects who lost islet function (mainly patients with Type 1 Diabetes or, more rarely, previous total pancreatectomy) the only option currently available for transplantable islet cells is allogeneic donor pancreata. These are generally obtained through multi-organ donation after cerebral death, following conventional donor:recipient ABO blood type matching. The use of a segment of the pancreas from living-related donors is technically feasible (45,46), but at the present time not preferred for islet transplantation due to the limited duration of graft function after transplantation of suboptimal islet numbers under standard immunosuppressive protocols, as well as the intrinsic risks for the donor (i.e., morbidity and risk to develop diabetes)(47).
Table 1. Indication for Islet Transplantation |
||
Condition |
Procedure |
Type of Transplant |
Diabetes Mellitus |
||
Type 1 |
ITA, SIK, IAK |
Allogeneic |
Type 2 |
ITA, SIK, IAK |
Allogeneic |
Surgery-Induced Diabetes |
||
Chronic pancreatitis |
ITA |
Autologous/Allogeneic |
Trauma |
ITA |
Autologous/Allogeneic |
Multi-visceral transplantation |
Different combinations: Liver-Islet Transplantation, Bowel-Liver-Islet Transplantation, etc. |
Allogeneic |
Cystic Fibrosis |
ITA Lung-Islet Transplantation |
Autologous/Allogeneic Allogeneic |
Benign enucleable tumors |
ITA |
Autologous |
Borderline/malignant pancreatic neoplasms |
ITA |
Autologous/Allogeneic
|
Grade C pancreatic fistula requiring completion pancreatectomy |
ITA |
Autologous/Allogeneic
|
ITA- Islet Transplant Alone; SIK- Simultaneous Islet-Kidney; IAK- Islet After Kidney
The current main indication for an allogeneic islet transplant is Type 1 Diabetes, which is characterized by the selective destruction of islet beta cells due to an autoimmune process. Ongoing clinical trials of allogeneic islet transplantation are recruiting subjects with unstable Type 1 Diabetes 18-65 years of age, either sex, with frequent metabolic instability requiring medical treatment (hypo-, hyper-glycemia, ketoacidosis) despite intensive insulin therapy; hypoglycemia unawareness (<54mg/dL); severe metabolic lability (mean amplitude of glycemic excursion >11,1 mmol/L or 200 mg/dl). The inadequate efficacy of medical therapy to attain the desirable metabolic control in this specific patient population with unstable diabetes justifies the use of transplantation of pancreatic islets (either isolated cellular graft or vascularized whole pancreas) (48). The main objective of the transplant is to correct the high susceptibility to severe hypoglycemia and glycemic imbalance that are associated with high mortality (8% in nonuremic subjects in the waiting list for 4 years to receive pancreas transplantation). Further indications for an islet transplant are presence of progressive complications of diabetes and psychological problems with insulin therapy that may compromise adherence to the therapeutic regimen. Islet transplant is indicated also for cases of subcutaneous insulin resistance requiring intraperitoneal or intravenous infusions, which are associated with substantial management hurdles and morbidity.
Table 2. Inclusion and Exclusion Criteria for Allogeneic Islet Transplantation in T1DM* |
Inclusion Criteria: |
· Mentally stable and able to comply with study procedures · Clinical history compatible with type 1 diabetes with onset of disease at <40 years of age, insulin dependence for at least 5 years at study entry, and a sum of age and insulin dependent diabetes duration of at least 28 · Absent stimulated C-peptide (<0.3 ng/ml) 60 and 90 minutes post-mixed-meal tolerance test · Involvement of intensive diabetes management, defined as: o Self-monitoring of glucose values no less than a mean of three times each day averaged over each week o Administration of three or more insulin injections each day or insulin pump therapy o Under the direction of an endocrinologist, diabetologist, or diabetes specialist with at least three clinical evaluations during the past 12 months prior to study enrollment · At least one episode of severe hypoglycemia in the past 12 months, defined as an event with one of the following symptoms: memory loss; confusion; uncontrollable behavior; irrational behavior; unusual difficulty in awakening; suspected seizure; seizure; loss of consciousness; or visual symptoms, compatible with hypoglycemia in which the individual required assistance of another subject was unable to treat him/herself person and which was associated with either a blood glucose level <54 mg/dl or prompt recovery after oral carbohydrate, intravenous glucose, or glucagon administration in the 12 months prior to study enrollment · Reduced awareness of hypoglycemia
|
Exclusion Criteria: |
· Body mass index (BMI) >30 kg/m2 or weight ≤50 kg · Insulin requirement of >1.0 IU/kg/day or <15 U/day · HbA1c >10% · Untreated proliferative diabetic retinopathy · Systolic blood pressure >160 mmHg or diastolic blood pressure >100 mmHg · Measured glomerular filtration rate using iohexol of <80 ml/min/1.73mm2. · Presence or history of macroalbuminuria (>300 mg/g creatinine) · Presence or history of panel-reactive anti-HLA antibody levels greater than background by flow cytometry. · Pregnant, breastfeeding, or unwilling to use effective contraception throughout the study and 4 months after study completion · Presence or history of active infection, including hepatitis B, hepatitis C, HIV, or tuberculosis. · Negative for Epstein-Barr virus by IgG determination · Invasive aspergillus, histoplasmosis, or coccidioidomycosis infection in the past year · History of malignancy except for completely resected squamous or basal cell carcinoma of the skin · Known active alcohol or substance abuse · Baseline Hgb below the lower limits of normal, lymphopenia, neutropenia, or thrombocytopenia · History of Factor V deficiency · Any coagulopathy or medical condition requiring long-term anticoagulant therapy after transplantation or individuals with an INR greater than 1.5 · Severe coexisting cardiac disease, characterized by any one of the following conditions: o Heart attack within the last 6 months o Evidence of ischemia on functional heart exam within the year prior to study entry o Left ventricular ejection fraction <30% · Persistent elevation of liver function tests at the time of study entry · Symptomatic cholecystolithiasis · Acute or chronic pancreatitis · Symptomatic peptic ulcer disease · Severe unremitting diarrhea, vomiting, or other gastrointestinal disorders that could interfere with the ability to absorb oral medications · Hyperlipidemia despite medical therapy, defined as fasting LDL cholesterol >130 mg/dl (treated or untreated) and/or fasting triglycerides >200 mg/dl · Currently receiving treatment for a medical condition that requires chronic use of systemic steroids except for the use of 5 mg or less of prednisone daily, or an equivalent dose of hydrocortisone, for physiological replacement only · Treatment with any antidiabetic medication other than insulin within the past 4 weeks · Use of any study medications within the past 4 weeks · Received a live attenuated vaccine(s) within the past 2 months · Any medical condition that, in the opinion of the investigator, might interfere with safe participation in the trial o Treatment with any immunosuppressive regimen at the time of enrollment. o A previous islet transplant. · A previous pancreas transplant, unless the graft failed within the first week due to thrombosis, followed by pancreatectomy and the transplant occurred more than 6 months prior to enrollment.
|
*Modified from the information relative to active trials from the Clinical Islet Transplant Consortium (www.citisletstudy.org/) as listed at http://clinicaltrials.gov/ct2/show/NCT00434811.
MULTIDISCIPLINARY TEAM
Islet Transplant Programs require the integration of multidisciplinary expertise. The endocrinologist expert in diabetes diagnosis and management is essential member of the team, and can identifying subjects who may benefit of beta-cell replacement therapy, and help with the evaluation of metabolic control during all phases of the follow-up. The psychologist is involved in the evaluation of islet transplant candidates to assess their motivation, mental fit to enroll in the trial, and ability to adhere to the therapy. Psychometric and psychological evaluations are performed during the follow-up period after transplantation. Transplant surgeons provide the expertise in organ procurement, with transplant procedures, overall management of patients and immunosuppression. A dedicated Cell Transplant Center with specialized experts in pancreatic cell isolation, purification, culture, potency assessment and quality assurance warrant that islet cell products are manufactured for clinical transplantation following cGMP standards and FDA regulations. The interventional radiologist performs the noninvasive cannulation of the portal vein and participates to the post-transplant monitoring of the liver using noninvasive imagine techniques. The organ procurement organizations and organ distribution networks (UNOS in the U.S.) contribute to the identification and allocation of donor organs matching the recipient’s characteristics. The ophthalmologist and nephrologist are involved to monitor and treat progressive diabetic complications (i.e., retinopathy and renal function, respectively).
ISLET ISOLATION AND TRANSPLANTATION
Islets are highly vascularized cell clusters ranging <50um to ~800um of diameter that constitute the endocrine component of the pancreas. It has been estimated that a healthy pancreas may contain approximately 106 islets scattered throughout the gland, and accounting for only ~1% of total pancreatic tissue. Each cluster comprises several thousands of endocrine cell subsets that are closely in touch with capillaries and with each other. Complex cell-cell interactions between different cell subsets, innervation, incretins and metabolites (sugar and amino acids, amongst other) in the blood and interstitial space all contribute to the proper control of glucose homeostasis (49). Preservation of the integrity of islet cell cluster is a prerequisite for their optimal function. The procedure currently used to extract islets from human pancreas is the so called automated method for isolation of the islets of Langerhans, established in 1987 by Ricordi and colleagues (50). Before the beginning of the isolation procedure, the spleen and the duodenum are removed from the pancreas and an accurate dissection and removal of the peripancretic fat, lymph nodes and vessels is performed. Then, the pancreas is divided at the neck and two 16-20 gauge angiocatheters are inserted into the main pancreatic ducts. The organ is then perfused with cooled perfusion solution containing collagenase and serine – protease inhibitor – dissolved in buffer at a pressure of 140-180 mmHg. After 10 minutes of cold perfusion, the distended pancreas is further cut into smaller sections, and placed into the Ricordi chamber. This chamber is composed of a superior and an inferior part, separated by a filter that has pores of about 700μm. Seven to nine stainless steel balls and the fragments of the pancreas are placed into the inferior part of the chamber, which is then filled with the digestion solution and closed together with the superior part of the chamber. A peristaltic pump connected to the system is activated creating a flow of 40 ml/min. The digestion runs in a closed circuit where warm Hank’s solution is pumped in the inferior chamber and the tissue released in the solution passes in the superior chamber through the filter. The collagenase is re-circulated at a temperature not exceeding 37°C and the chamber is agitated. When most of the islets are free of the surrounding acinar tissue, and intact islets are observed, the heating circuit is bypassed. The temperature is progressively decreased to 10°C and the collagenase diluted with cold RPMI. The free islets are then collected in containers, washed several times, re-suspended in cold organ preservation solution and purified with a continuous ficoll gradient using a Cobe 2991 cell separator. At the end of the procedure samples of the islet preparation are collected and evaluated through staining with dithizone (DTZ) which marks zinc in the insulin granules, resulting in a characteristic red stain. Adding few drops of DTZ solution to a sample allows easy evaluation of the morphology and number of isolated islets through computerized digital analysis. The islet manufacturing processes must be controlled by different assays and the islet batch product validated and characterized. Then safety testing is carried out for sterility and pyrogenicity, identity (insulin content), cell number (amount of tissue, counting of islets), purity (percentage of ductal, acinar, beta, and other cells), viability (islet nucleotide content), potency (insulin secretory response) and finally stability (storage in culture). Specific features of the final islet preparation are a required for islet preparations used in islet transplantation, in particular purity (> 20% of the preparation being islets), adequate number of islets (>5,000 islet equivalent recipient body weight for the first infusion, >3,000 for further infusions) and total tissue volume (< 5 ml). The infusion of the islets can be performed a few hours after the end of the isolation process or up to 72 hours thereafter. The implantation site is usually the hepatic parenchyma through the portal system of the recipient. Recently other implantation sites have been proposed (51) in the clinical setting, like the bone marrow (52,53), the subcutaneous site(54), the gastric submucosa (55), the omentum(56,57) or striated muscle (58,59), which in the future, may prove to be valid alternative sites for islet transplantation. The adequate amount of islets obtained is calculated with respect to the body weight of the recipient and re-suspended immediately before intrahepatic transplantation in 40-60 mL of a solution suitable for injection (Ringer Lactate, 1% Human Albumin and 2000 IE of heparin). Percutaneous trans hepatic catheterization is the most common access route, as well as a mini-laparotomy and cannulation of an omental or mesenteric vein, or recanalization of the umbilical vein. Access to the portal vein is usually provided by interventional radiologists. If the portal pressure is documented to be below 20 mmHg, the islet infusion bag is connected with the portal vein catheter and infused over a period of 15 to 60 minutes. Islet infusion is halted if the portal pressure exceeds 22 mmHg. After completion of the islet infusion, the catheter is withdrawn; coils and gelatin-sponges are deployed in the puncture tract to prevent bleeding. A schematic animation of the islet isolation and transplant procedures is available online [http://www.youtube.com/watch?v=aMNKu-ZVUls].
THE CONSORTIUM CONCEPT
A major development in the field of islet transplantation is the combination of individual centers into larger groups such as the GRAGIL network in France and Switzerland, the Nordic Network for Clinical Islet Transplantation (NNCIT) in the Scandinavian countries and the Clinical Islet Transplant Consortium (CITC) internationally but concentrated in North America. The need for dedicated infrastructures and personnel specialized in islet cell processing, quality assessment and cGMP standards impose an enormous financial burden on any Clinical Islet Transplant Program. Acquiring and maintaining the specialized expertise in islet cell processing requires a steep learning curve and continuous refinements and training that add to the costly procedure. Recent data have shown that the experience of the clinical islet transplant team in cell processing and management of immunosuppression are critically important in determining the success of a clinical trial (60). Based on these premises, the development of regional cell processing centers that are part of consortia that are integrated with distant transplant centers is increasingly being considered as a practical and cost-effective strategy (Figure 1). Initial reports of successful clinical trials carried on in the context of Consortia both in Europe and North America (61-64) support the feasibility of such an approach, which may be of assistance in reducing the operational costs while enhancing the success rate of clinical trials (i.e., better utilization of donor pancreata, more reproducible success in obtaining adequate numbers of functional islets from a donor pancreata, etc.).
Figure 1. Islet Transplant Consortium Models. A. The centralized (or ‘regional’) Cell Processing Facility receives the donor pancreas from a distant Transplant Center and isolates islet cell products that are sent back for implant. B. The centralized Cell Processing facility receives the donor pancreas from one of the Transplant Centers and distributes the isolated islets to any of the Transplant Center in the Consortium according to the best match of the cell product for the transplant candidate on the waiting list for transplant (that is, the islet cell product is not necessary returned to the center recovering the pancreas).
ISLET TRANSPLANT ACTIVITY
The Collaborative Islet Transplant Registry (CITR)
In 2001, the National Institute of Diabetes & Digestive & Kidney Diseases established the Collaborative Islet Transplant Registry (CITR) to compile data from all islet transplant programs in North America from 1999 to the present. The Juvenile Diabetes Research Foundation (JDRF) granted additional funding to include the participation of JDRF-funded European and Australian centers from 2006 through 2015. The cumulated North American, European and Australian data are pooled for analyses included in the annual report. CITR Annual Reports are publicly available as open access and can be downloaded or requested in hard copy at www.citregistry.org. From 1999 through 2020 – the cut-off for the last Eleventh Annual Report – CITR has collected data on the following groups of study subjects:
- Allogeneic islet transplantation (typically cadaveric donor), performed as either islet transplant alone (ITA) or islet-after-kidney (IAK). A small number of cases have been performed as islet simultaneous with kidney (SIK) or kidney-after-islet (KAI).
- Autologous islet transplantation, performed after total pancreatectomy (N=1,233) are also reported to CITR.
As of December 15, 2020, the CITR Registry included data on 1,399 allogeneic islet transplant recipients (1,108 islet transplant alone, ITA, and 236 islet after kidney, IAK, 49 simultaneous islet kidney, SIK, and 6 kidney after islet, KAI), who received 2,832 infusions from 3,326 donors. From 1999 through 2020, 28 National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) sponsored North American and 12 international Eurasian and Australian islet transplant centers (40 total) contributed data to the Collaborative Islet Transplant Registry (CITR). Combining the ITA and IAK recipients, 27.4% received a single islet infusion, 48.1% received two, 20.4% received three, and 4.1% received 4-6 infusions. Of 26 North American sites performing Auto-ITx from 1999 through December 2020, 15 reported data to CITR along with 5 European and Australian islet transplant centers. These sites registered 1233 autoislet transplant recipients. Of these, 1123 recipients were in North America, 98 in Europe, and
12 in Australia. One-hundred eight-five (185) were aged less than 18, and 1,057 were 18 or older at the time of their transplant.
Outside The Collaborative Islet Transplant Registry (CITR)
Although the CITR is an extraordinary source of valuable data, a recent publication indicates that it does not capture a major part of the international islet transplant activities and outcomes (65). In fact, a global online survey was recently administered to 69 islet transplantation programs. After integration of all data obtained, 103 islet transplant centers were identified, of which 94, in 25 countries, had reported allotransplantation activity during the 2000–2020 period: 15 in Asia (16%), 39 in Europe (42%), 34 North America (36%), 3 in Oceania (3%) and 3 in South America (3%) and between January 2000 and December 2020, 4,321 islet allotransplants in 2,149 patients were reported worldwide. Most islet transplants were performed in Europe (2,608, 59.7%), followed by North America (1,475, 33.8%), Asia (135, 3.1%), Oceania (119, 2.7%) and South America (28, 0.6%). Actually the ANZIPTR (Australia and New Zealand Islet and Pancreas Transplant Registry) and NHS-BT (UK National Health Service-Blood and Transplant) registry are publicly available registries containing a wealth of data on islet and pancreas transplantation in Australia/New Zealand and UK, respectively, including outcomes (66) (67). The European Pancreas and Islet Transplant Registry (EPITR) is a current effort from ESOT/EPITA aiming at covering these needs for Europe (https://esot.org/epita/epita-epitr/).
Clinical Management of Islet Transplant Recipients
The clinical management of islet transplant recipients requires the concerted effort of endocrinologist and transplant teams.
Immunosuppression
Preexisting and transplant-induced auto- and allo-specific cellular immune responses play a crucial role in the loss of islets and islet function infused in the liver (68-70) along with non-specific immune responses predominantly mediated by innate inflammatory processes related to mechanics and site (71-74). Islet graft rejection occurs without clinical symptoms. Neither guidelines nor formal consensus on the “best” or “standard” immunosuppressive strategy for human islet transplantation are currently available. Multiple induction and maintenance agents are administered peri- and post- every infusion in the same recipient. According to CITR data (75), a substantial shift in immunosuppression strategies has been documented during the last years.
Induction with interleukin-2 receptor antagonist (e.g., daclizumab) only, which comprised about 53% of all initial infusions in 1999-2002, was replaced or supplemented with regimens that included T-cell depletion with/without TNF antagonists in about 67% of the new infusions performed since 2015 (11,76-83). In 1999-2002, maintenance immunosuppression was predominantly (64%) calcineurin (CNI) +mTOR inhibitors (60). It was increasingly replaced or supplemented throughout the eras by a CNI and IMPDH-inhibitor combination (77,84-86).; in the most recent era, CNI+mTOR inhibitors were used in 15% of new infusions while CNI+IMPDH inhibitors were used in about 62%. Moreover, the use of alemtuzumab-induction therapy was recently reported and associated with encouraging longer-term function (87,88). New biologic agents with potentially lower islet cell and organ toxicity profiles are currently being evaluated in ongoing clinical trials. Amongst these are agents that target co-stimulation pathways in immune cells and/or adhesion molecules (CTLA4-Ig, LFA-1 PD-1/PD-L1 CD40 ) (89-95) or chemokine receptors (CXCR1/2) (71,96). Finally, calcineurin inhibitor-free immunosuppressive regimen was reported (97).
Antibiotic and Antiviral Prophylaxis
Subjects receiving immunosuppression therapy are more susceptible to opportunistic infections, as well as reactivation or de novo occurrence of viral infections. Antibiotic prophylaxis for Pneumocystis carinii consists in trimethoprim and sulfamethoxazole three times a week. Antiviral prophylaxis is aimed are reducing the risk, or treating the occurrence, of cytomegalovirus infections (which have been recognized increasing the risk of graft loss in solid organ transplantation) and of reactivation of Epstein - Barr virus infection (which has been associated with the dreadful post-transplant lymphoproliferative disease, PTLD). Current protocols utilize antiviral therapy with vanglancyclovir daily for the first trimester post-transplant. Monitoring of viremia in peripheral blood samples by PCR is becoming a routine during the follow-up as it allowed for the early detection of reactivation or de novo infections that may be treatable without compromising graft outcome (98,99).
Thromboembolism Prophylaxis
It has been recognized that isolated islets produce tissue factor and other pro-inflammatory molecules that may trigger an instant blood-mediated inflammatory reaction upon infusion into the blood stream. This, in turn, may enhance the generation of noxious stimuli after embolization in hepatic sinusoids of the liver, significantly reducing the mass of functional islets engrafting. An aggressive heparin treatment is generally implemented in the early period after transplant. Heparin is added to the transplant medium used during the islet infusion, while low molecular weight heparin injections are administered in the post-transplant period. This is aimed at enhancing islet engraftment in the hepatic portal system while reducing the risk of portal thrombosis.
Peri-transplant Insulin Management
Islet engraftment may take up a few weeks to allow for neovascularization of the clusters in the transplant microenvironment. The monitoring of glycemic control in the immediate post-transplant period should be intense to attain tight glycemic values in order to avoid excessive workload for the newly transplanted islets as well as to prevent hypoglycemic episodes. This is generally done by providing basal exogenous insulin that is then progressively reduced and withdrawn according to the glycemic values measured.
POST-TRANSPLANT CLINICAL MONITORING
Monitoring of cell blood counts (erythrocytes, white blood cells and differential), hemoglobin, platelets and coagulation markers is routinely performed in the post-transplant period. These tests allow assessing the myelosuppressive effects of anti-rejection drugs. In the case of anemia with clinical relevance, iron supplementation may be indicated, while for more severe cases erythropoietin treatment is implemented. In the case of severe neutropenia, marrow stimulation with granulocyte-colony stimulating factor (G-CSF) is promptly implemented.
Renal function is monitored periodically in the follow-up of islet transplant recipients to assess the impact of restoring beta-cell function on the progression of diabetic nephropathy, and also to identify and timely correct potential nephrotoxicity of anti-rejection drugs (i.e., CNI and mTOR inhibitors). Standard serological tests (serum creatinine, azotemia), urine tests (spot and 24-hr collections) are frequently performed during the follow-up and glomerular filtration rates (GFR) estimated using different algorithms (i.e., MDRD). The nephroprotective effect of ACE inhibitors and of antagonists of angiotensin-receptor (ARB) in subjects with diabetes has been recognized, and their use is particularly indicated in transplant recipients treated with immunosuppressive drugs known for their negative effects on renal function (100-104). Elevations of blood pressure from the range 130/80 mmHg are promptly treated pharmacologically.
Monitoring of lipid levels and prompt treatment of dyslipidemia are important in transplanted patients. Some of the anti-rejection drugs (i.e., mTOR inhibitors) are prone to induce dyslipidemia, which in turn may have toxic effects on beta-cells or contribute to creating an unfavorable environment (i.e., steatosis) in the liver (105). Prophylactic use of statins targeting LDL cholesterol levels <100mg/dL can be contemplated for islet transplant recipients.
Liver function is monitored in the post-transplant period. It is common to observe a transient and self-limited elevation of liver enzymes (transaminitis) because of the embolization of islets into the liver sinusoids (106,107). This is often associated with hyper-echoic pattern of the liver parenchyma at ultrasound evaluation in the early days post-transplant. This phenomenon resolves spontaneously without the need for medical treatment. Ultrasound evaluation of the liver and abdominal cavity in the days post-transplant also allows identifying timely possible procedural complication of the transplant, such as portal thrombosis, peritoneal hemorrhage and alterations of echogenicity of hepatic parenchyma (108).
The immune monitoring after islet transplantation does not differ much from that of any other organ transplant (109). Basal and serial evaluation of Panel Reactive Antibodies (PRA) is performed to determine possible allosensitization against Human Leukocyte Antigens (HLA) class I and II of the Histocompatibility complex of transplanted tissue. Generally, maintenance of an adequate immunosuppressive regimen can prevent the development of alloantibodies, thereby preventing their deleterious effects on graft survival and function (i.e., chronic rejection leading to graft loss) (110-112). Nonetheless, development of alloreactivity against donor or non-specific antigens may develop whenever reduction (i.e., during infections, toxicity and drug conversion, amongst other causes) or suspension (i.e., after complete graft loss) of immunosuppression is needed (112,113). The autoimmune process underlying Type 1 Diabetes is associated with the appearance of antibodies against self-antigens (autoantibodies; i.e., towards GAD, IA-1 and insulin). Serial titration of autoantibody levels during the follow-up period may enable detecting a reactivation of the autoimmune process, measured as conversion to positive values in previously negative subjects, or increase of antibody titers. These have been associated with a lower rate of insulin independence and shorter duration of graft function after islet transplantation (60,70). New assays for additional autoantibodies (i.e., ZnT8) and for autoreactive T cells are under evaluation to help enhancing the sensitivity of immune monitoring for early detection of recurrence of autoimmunity, which may enable implementation of timely immune interventions to rescue the transplanted cells (114-116).
MONITORING ISLET GRAFT FUNCTION
Several metabolic parameters allow monitoring the function of transplanted islets (Table 3). Since only subjects with Type 1 Diabetes who have undetectable stimulated c-peptide (<0.3 ng/dl) before transplant are recruited for an islet transplant, monitoring of basal and stimulated c-peptide offers an excellent biomarker of graft function, even when exogenous insulin is required. There is no consensus on which approach is most suited to accurately assess functional islet mass. Algorithms and indices that combine multiple parameters have been developed and proposed to help simplifying and obtaining objective functional assessment of islet transplant recipients (117-126). The main goal is to identify early changes that indicate propensity to graft dysfunction (i.e., functional impairment during an infection, drug-induced toxicity). Stimulation tests are performed before (at enrollment) and during the follow-up after transplant to evaluate the functional potency of the transplanted islets in response to different secretagogues (i.e., glucose, arginine, or mixed meal test). Insulin therapy is generally implemented when random glycemic sampling demonstrates on three subsequent occasions within the same week fasting values >140 mg/dl (7.8 mmol/L) and postprandial values >180 mg/dl (10.0 mmol/L), or after recording two consecutive A1c values >6.5%.
Table 3. Monitoring of Islet Graft Function |
||
Standard |
Stimulation |
Indices |
Glycosylated Hb (A1c) Fasting glycemia Postprandial glycemia MAGE* CGMS* Basal C-peptide Daily insulin requirement
|
Mixed Meal Intravenous glucose Intravenous arginine |
Hypo score Liability index Βeta score Beta 2 score Basal C-peptide/Glucose ratio HOMA-B* HOMA-IR* TEF* |
*Abbreviations. CGMS: Continuous Glucose Monitoring System. MAGE: Mean Amplitude of Glucose Excursions. HOMA-B: Homeostasis Model Assessment – functional beta cell mass. HOMA-IR: Homeostasis Model Assessment – Insulin-Resistance. TEF: Transplant Estimated Function |
The Igls Score
The lack of standardized definition of graft functional and clinical outcomes remains a source of concern in β-cell replacement influencing its recognition as a valid clinical option from the endocrinology community. In order to address this issue, the International Pancreas & Islet Transplant Association (IPITA) joined with the European Pancreas & Islet Transplant Association (EPITA) for a two-day workshop on “Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes” in January 2017 in Igls, Austria. The main objective was to develop consensus on the definition of function and failure of current and future forms of β-cell replacement therapies. As result of the workshop, an IPITA/EPITA Statement was recently published (127,128). This Statement introduces some relevant innovations in the field including a new classification for the definition of clinically successful outcome. The functional status and clinical success of a β-cell graft should be defined separately using the same components of assessment: the HbA1c, severe hypoglycemic events, insulin requirements, and C-peptide. Concordantly, a four-tiered system was proposed to classify the functional outcomes of β-cell replacement:
- optimal β-cell graft function: HbA1c ≤6.5%, the absence of any severe hypoglycemia, the absence of any requirement for exogenous insulin or other anti-diabetic drugs, and documentation of an increase over pre-transplant measurement of C-peptide
- good β-cell graft function: defined by: HbA1c <7.0%, the absence of any severe hypoglycemia, a reduction by more than 50% from baseline in insulin requirements or the use of non-insulin anti-diabetic drugs, and documentation of an increase over pre-transplant measurement of C-peptide.
- marginal β-cell graft function: no modification of HbA1c, the reduction of severe hypoglycemia, a reduction by less than 50% from baseline in insulin requirements, and documentation of an increase over pre-transplant measurement of C-peptide.
- failure β-cell graft function: absence of any evidence for a clinical impact (no modification of HbA1c, incidence of severe hypoglycemia and insulin requirement) and clinically insignificant levels of C-peptide.
Clinically successful outcomes includes both optimal and good functional outcomes, implying that the use of exogenous insulin or other anti-diabetic drugs is not synonymous with graft loss or failure. Neither a marginal β-cell graft nor a failed β-cell graft is considered a clinically successful. However, if documented impairment in hypoglycemia awareness, frequent occurrence or exposure to severe hypoglycemia, or marked glycemic variability/lability is convincingly improved, then it may be appropriate to consider that the β-cell graft is clinically impactful also in marginal function and the benefit of maintaining β-cell graft function may outweigh risks of maintaining immunosuppression. This implies that hypoglycemia awareness, serious hypoglycemia, and glycemic variability/lability must be evaluated at baseline for monitoring whether a marginally functioning graft is continuing to provide any clinical impact.
IPITA / EPITA Statement has the merit of having introduced a defined concept of clinical success based on easily measurable parameters over time and with a wide consensus of international experts Implementation of this new β-cell replacement outcome definition and its use in publication will be critical to improve the performance and to reliably compare the different β-cell replacement strategies (129).
In July 2019, a symposium at the 17th IPITA World Congress was held to examine the Igls criteria after 2 years in clinical practice, including validation against continuous glucose monitoring (CGM)-derived glucose targets, and to propose future refinements that would allow for comparison of outcomes with artificial pancreas system approaches. A new Igls 2.0 form composite criteria was suggested (130), in which clinical outcome based on glucose regulation is separated from β-cell graft function, with the latter considered only for further qualification of β-cell replacement modalities (Table 4-5).
Table 4. Proposed Igls Criteria 2.0 |
|||||
Treatment outcome |
Glycemic control |
Hypoglycemia |
Treatment success |
||
HbA1c, % (mmol/mol)a |
CGM, % time-in-range |
Severe hypoglycemia, events per y |
CGM, % time < 54 mg/dl (3.0 mmol/L) |
||
Optimal |
≤6.5 (48) |
≥80 |
None |
0 |
Yes |
Good |
<7.0 (53) |
≥70 |
None |
<1 |
Yes |
Marginal |
≤Baseline |
>Baseline |
<Baselineb |
<Baseline |
Noc |
Failure |
~Baseline |
~Baseline |
~Baselined |
~Baseline |
No |
Baseline, pretransplant assessment (not applicable to total pancreatectomy with islet autotransplantation patients).
Abbreviations: CGM, continuous glucose monitoring; HbA1c, glycated hemoglobin.
a Mean glucose should be used to provide an estimate of the HbA1c, termed the glucose management indicator, in the setting of disordered red blood cell life span.
b Should severe hypoglycemia occur following treatment, then continued benefit may require assessment of hypoglycemia awareness, exposure to serious hypoglycemia (<54 mg/dL [3.0 mmol/L]), and/or glycemic variability/lability with demonstration of improvement from baseline.
c Clinically, benefits of maintaining and monitoring β-cell graft function may outweigh risks of maintaining immunosuppression.
d If severe hypoglycemia was not present before β-cell replacement therapy, then a return to baseline measures of glycemic control used as the indication for treatment (6, 7) may be consistent with β-cell graft failure.
Table 5. Proposed Igls Criteria 2.0 |
||
β-cell graft functione |
C-peptide, ng/mL (nmol/L) f |
Insulin use or noninsulin antihyperglycemic therapy |
Optimal |
Any |
None |
Good |
>0.5 (0.17) stimulated |
Any |
Marginal |
0.3-0.5 (0.10-0.17) stimulated |
Any |
Failure |
<0.3 (0.10) stimulated |
Any |
e Categorization of β-cell graft function must first meet treatment outcome based on measures of glucose regulation.
f May not be reliable in uremic patients and/or in those patients with evidence of C-peptide production before β-cell replacement therapy.
IMPACT OF ISLET TRANSPLANTATION ON METABOLIC CONTROL (TABLE 6)
Four successful large-scale Phase 3 clinical trials in islet transplantation have been published recently: CIT-07 (multicenter, single-arm)(131), CIT06 (pivotal trial) (132), TRIMECO (multicenter, open-label, randomized) (133) and REP0211 (multicenter, Double blind, randomized) (134). All these studies demonstrate that human islets, when transplanted in patients with T1D with impaired awareness of hypoglycemia and severe hypoglycemic events, can safely and efficaciously maintain optimal glycemic control (135). The clinical experience confirms that the most remarkable effect of the islet transplant is the abrogation of severe hypoglycemia and the recovery of hypoglycemia awareness, which persists after development of graft dysfunction and even several months after graft failure (and loss of detectable c-peptide) (136,137). Following islet transplantation, the restoration of β cell responses to secretagogue stimulation is observed, with improved insulin secretion (‘first phase’) in response to intravenous glucose, as well as increased c-peptide secretion in response to oral glucose. Normalization of glycemic threshold triggering the release of counter-regulatory hormones can be demonstrated during hypoglycemic clamp studies, even if without reaching normalization of the magnitude of the vegetative response; furthermore, quasi-normal glucagon secretion in response to hypoglycemia can be observed (138-142). In addition to controlling hypoglycemia, insulin independence can be achieved when an adequate islet mass is transplanted (143). After islet transplantation, 5-year insulin independence may be as high as 50%. A quarter of patients may remain insulin independent, with HbA1c concentrations of less than or equal to 6·5%, for at least 10 years, with either islet transplantation alone or islet-after-kidney transplantation (144)(145). Moreover, the glucose control associated with excellent islet graft function closely matches glucose values measured in healthy adults: median glucose 103 mg/dl (95-112), glucose standard deviation around the mean value 14 (11-20), 0% time >180 mg/dl, 0% time <54 mg/dl, HbA1c 5.6 (5-5.8) (146). Additionally, a significant improvement of quality of life after islet transplant has been documented by using standardized psychometric instruments and interviews carried on by psychologists (147-155). Associated with the better glucose control and the evidence of islet function (c-peptide secretion), a positive impact on the microvascular complications of T1D has been described while is less evident on the macrovascular complications (156). More specifically, a stabilization/slower progression of retinopathy (104,157-159) and neuropathy (158,160-162) and an improvement of micro- and macroangiopathy (79,101,102,154,157,163-168) have been described. Some studies reported also a reduction of atherothrombotic profile paralleled by reduced incidence of cardiovascular accidents, an amelioration of cardiovascular and endothelial function, improved longevity of renal transplant (165) and a higher survival rates after islet transplantation in IAK recipients (162,165,169-173).
By combining donor selection criteria with improved isolation techniques and adequate immunomodulation of the recipient, insulin independence after single donor islet preparation is becoming more reproducibly possible to achieve. Islet preparations obtained from more than one donor pancreas can be transplanted at once after pooling them, or sequentially based on the metabolic needs of each subject. Data from the Clinical Islet Transplant Registry and independent trial reports have shown that insulin independence at one year from completion of the transplant is up to 70% with virtually 100% of the subjects maintaining graft function (c-peptide) if adequately immunosuppressed (75,82). A progressive loss of insulin independence with approximately 90% of subjects requiring reintroduction of exogenous insulin (most of them with detectable c-peptide) has been reported in recent clinical trials based on the ‘Edmonton protocol’ (induction with anti-IL2R antibody; maintenance with sirolimus and tacrolimus) and some variants of it (60,77,84,86,174). More recent trials using more potent lymphodepletion (i.e., thymoglobulin, anti-CD3 or anti-CD52 antibodies) and/or biologics (anti-IL2R, anti-TNF, anti-LFA-1 antibody or CTLA4Ig) have shown great promise with approximately 50% of insulin independence at 5 years after islet transplantation (86,175-179), which is comparable to some of the data in whole pancreas transplantation in subjects with Type 1 Diabetes (80,83,86,91,92). In light of the results of the last decade of clinical islet transplant trials, achievement of insulin independence, although desirable, no longer should be considered the main goal of islet transplantation. The sizable improvement of metabolic control in the absence of severe hypoglycemic events, the amelioration of diabetes complications and the achievement of sustained better quality of life, which are quite cumbersome to reproduce by the means of medical treatment, justify the risks associated with the islet transplant procedure and immunosuppression in this high-risk population of subjects with unstable diabetes.
Regarding auto transplantation the largest published series are from the University of Minnesota (16-19), University of Cincinnati (20,21), and Leicester (22-25,180). Overall, one-third of patients in the Minnesota series achieves insulin independence, and the majority have islet graft function, as documented by C-peptide positivity (16,22). Cincinnati, Leicester, and other centers have published similar results, with 22-40% of the patients being insulin independent after islet transplant (21,181,182). A significant association between insulin independence and the IEQ/kg transplanted (i.e., islet mass standardized by patient’s weight) was described. Bellin et al. (19) and White et al. (24) reported that insulin independence is related to the number of transplanted islet cells (>2,000 IEQ/kg and >3,000 IEQ/kg, respectively). Similarly, Sutherland et al. (183) reported that insulin independence at 1 year was observed in 63 % of the patients who received greater than 5,000 IE/kg. Moreover, pancreatectomy recipients benefit from an islet autograft ways apart from insulin independence. In fact, the major goal of IAT in these patients is a good glycemic control without brittle diabetes. Ninety percent of patients in the Minnesota series and 100% of those in the Leicester series were C-peptide positive after the procedure (16,22). The majority of patients receiving an islet auto transplant maintained good glycemic control, with 82% of all recipients having average HbA1c levels <7.0% (16).
IMPACT OF ISLET TRANSPLANTATION ON DIABETES COMPLICATIONS
Encouraging results have been reported in recent years on the multiple beneficial effects of islet transplantation on the progression of diabetes complications [reviewed in (184)]. Although based on nonrandomized pilot studies, which should be cautiously evaluated, they provide the proof of concept of the importance of restoring beta-cell function in patients with diabetes. In particular, improvement of micro- and macro-angiopathy (main causes of diabetic nephropathy) (79,101,102,154,157,163-168) and stabilization/reduced progression of retinopathy (104,157-159) and neuropathy (158,160-162) have been described. Amelioration of cardiovascular and endothelial function, reduction of atherothrombotic profile paralleled by reduced incidence of cardiovascular accidents and higher survival rates were reported In IAK recipients (169-172) (162,165,169,171,173). Furthermore, significantly improved longevity of a renal transplant was observed after islet transplantation (165). It is likely that these benefits are the consequence of improve metabolic control conferred by the islet transplant. In addition, it has been proposed a contribution of restored c-peptide secretion and its effects on multiple targets (185).
Table 6. Benefits of Islet Transplantation |
Metabolic control - Reduction of exogenous insulin requirements or insulin independence - Reduction of MAGE - Reduction or normalization of A1c - Absence of severe hypoglycemia |
Quality of Life - Reduced fear of hypoglycemia - Improvement of Diabetes Quality of Life |
Diabetes complications - Improvement of micro- and micro-angiopathy - Improvement of cardiovascular and endothelial function - Reduced incidence of acute cardiovascular events - Reduced nephropathy progression - Stabilization/slower neuropathy progression - Stabilization/slower retinopathy progression |
COMMON ADVERSE EVENTS AND THEIR MANAGEMENT (TABLE 7)
The procedure of islet transplantation has proven to be very safe, especially when compared with whole pancreas transplant (177,186,187). For allogenic islet transplantation bleeding, either intraperitoneal or liver subcapsular, is the most common procedure-related complication, occurring with an incidence as high as 13% (188). The use of fibrin tissue sealant and embolization coils in the hepatic catheter tract seems to effectively minimize the bleeding risk (188,189). Partial portal vein thrombosis complicates fewer than 5% of islet infusion procedures (174), and complete portal venous thrombosis is rare. The use of purer islet preparations, greater expertise in portal vein catheterization and new radiological devices (catheters medicated with anticoagulation) will continue reducing the risk of portal vein thrombosis, although the risk is unlikely be completely eliminated. Other complications of islet cell transplantation include transient liver enzyme elevation (50% incidence) (106), abdominal pain (50% incidence), focal hepatic steatosis (20% incidence) (190,191), and severe hypoglycemia (< 3% incidence). Another complication related to the intrahepatic islet transplantation procedure is portal hypertension that can occur acutely during the islet infusion, especially in the case of infusions other than the first one (192). Finally, severe hypoglycemia is a risk associated with the infusion of islets. Iatrogenic hypoglycemia in the immediate post-transplant period is a rare event. Frequent blood glucose monitoring immediately following islet transplantation is recommended to avoid severe unrecognized hypoglycemia in the early post-transplant period. The risk of transmission of CMV disease from donor to recipient has been surprisingly low in recipients of islet allografts, particularly in the most recent period with routine use of purified islet preparations (140-144). As with any allogeneic transplant, islet transplant recipients may become sensitized to islet donor histocompatibility antigens (HLA), leading to the development of panel reactive alloantibodies (PRA).Data on the development of cytotoxic antibodies against donor HLA in islet allotransplant recipients with failing grafts have been reported from several islet transplant centers (148-152). A potential consequence of high PRA levels in recipients of a failed islet transplants is that if these individuals develop diabetic nephropathy in the future, a high PRA may increase their time on a transplant list for a suitable kidney graft.
The need to implement anti-rejection therapy exposes transplant recipients to an increased risk of untoward side effects expected in any immunosuppressed subjects (Table 6) (107). Opportunistic infections of urinary tract, upper respiratory tract and skin are frequent, along with myelosuppressive and gastrointestinal effects of the immunosuppressive drugs. In the majority of the cases, these effects are not severe and resolve without sequel with medical treatment. Elevation of viremic titers for cytomegalovirus (CMV) or Epstein-Barr virus (EBV) in the presence of overt clinical symptoms (i.e., de novo infection or reactivation in seropositve subjects) imposes the implementation of anti-viral therapy and reduction of immunosuppressive drug dose (98). Timely intervention may result in faster resolution of the symptoms without compromising graft survival. Direct organ toxicity of immunosuppressive drugs has been recognized. Symptoms associated with neuro- and/or nephro-toxicity are relatively frequent in subjects receiving chronic immunosuppressive agents currently in use in the clinical arena. In these cases, modification of the anti-rejection regimen is indicated, with dose reduction or conversion to a different combination of drugs. In the majority of cases, these changes resolve the symptoms without compromising graft survival (193,194). Nephrotoxicity from sirolimus and/or tacrolimus has been described in patients with T1D undergoing islet transplantation, particularly when kidney function is already impaired because of pre-existing diabetic nephropathy (195,196).
As for the CITR 11th Allograft Data Report Scientific Summary, the decline in eGFR (CKD-Epi) after islet transplantation is both statistically significant and clinically important. IAK had much lower pre-transplant levels than ITA, which then declined at a slower rate. Initial levels were also lower in recipients age 35 and older and declined at a slower rate compared to younger recipients. Levels were generally lower among recipients managed with CNI+IMPDH compared to other maintenance immunosuppression regimens. Compared with an age-unadjusted cohort of 1,141 participants with T1D followed by the Diabetes Control and Complications Trial and then by the Epidemiology of Diabetes Interventions and Complications (EDIC) (The DCCT/EDIC Research Group, 2011) who started with mean eGFR levels of 126 ml/min/1.73m3, CITR allograft recipients had much lower mean eGFR (91±1SE for ITA and 62±2 for IAK) at their first transplant. CITR ITA recipients exhibited a decline in eGFR of 12 ml/min/1.73m3 and IAK experienced a mean decline of 2 ml/min/1.73m3 in 5 years from last infusion, compared to a mean decline of about 9 ml/min/1.73m3 over the first 5 years in the DCCT.
As of 2021, by decision of the Executive Committee, only serious adverse events (SAEs) are reportable to CITR. About 11% ITA and 14% of IAK allo-islet recipients experienced a serious adverse event in the first 30 days following transplantation. There was a sharp decline in the number of patients who experienced SAEs post-2010, with 15% or more of patients experiencing SAEs in early eras compared to ~5% in 2011-2014 and 2015-2018. In the first year after islet transplantation, which includes a majority of the reinfusions that were performed, about one-fourth of participants have experienced an SAE. SAE within 1-year was slightly more common in IAK (31%) than ITA (23%) and there was a significant decline post-2010 (>25% pre vs. <15% post). Life-threatening events have occurred in 13.4% of islet-alone, in 16.5% of IAK recipients, and in 20.4% of SIK recipients. Recent eras have seen a substantial decline in the incidence of life-threatening events. The most common life-threatening events reported were abnormal granulocytes (24 events) followed by abnormal liver function (23 events) and hypoglycemia (14 events). About 75% of patients who experienced a life-threatening event recovered fully, 12% recovered with sequelae, 5% did not recover, and 9% died as a result of the event.
A total of 189 instances of neoplasm have been diagnosed in 101 of the 1,399 islet recipients who collectively represent a total of 7,963 person-years of observed follow-up. This equates to about 0.02 neoplasms per person-year. Of the total 189 events, 61% were deemed possibly related to immunosuppression, and 12% definitely related. Of the total events, 69% recovered, 10% did not recover, 5% recovered with sequelae, and 3% resulted in fatality. There were 41 instances in 23 patients of basal carcinoma of the skin and 86 instances in 38 patients of squamous carcinoma of the skin. There were 56 instances in 39 recipients of non-skin cancers. Eleven deaths due to cancer occurred.
There have been 77 or 5.5% deaths; cumulative mortality rates differed significantly by transplant type (p<0.0001) but not by era. SIK transplant recipients were disproportionately represented among fatalities comprising only 3.5% of the allo-islet recipient population, but 15.6% percent of deaths. Of the reported deaths, ten were deemed possibly related or definitely related to islet transplantation or immunosuppression. The most common causes of death were (# cases): cardiovascular (15), neoplasm (11), infection (including pneumonia) (9), hemorrhage (4), and complications of diabetes (3). Twenty-four deaths did not have a cause specified.
An assessment of the surgical complication of islet auto transplantation was recently reported for the entire Minnesota series (n=413) (16). Surgical complications requiring reoperation during the initial admission occurred in 15.9% of the patients. The most common reason for reoperation was bleeding, occurring in 9.5% of the procedures. Anastomotic leaks occurred in 4.2 % of the patients, biliary in 1.4% and enteric in 2.8%. Intra-abdominal infection requiring reoperation occurred in 1.9% of patients, wound infections requiring operative debridement in 2.2%. Gastrointestinal issues, such as bowel obstruction, omental infarction, bowel ischemia, delayed reconstruction because of bowel edema, tube perforation, requiring reoperation in 4.7% of the patients. Two patients (<1%) required reoperation to remove an ischemic or bleeding spleen after spleen sparing pancreatectomy (done in 30% of patients).
Table 7. Most Frequent Complications in Islet Transplant Recipients |
Procedure-related - Hemorrhage - Portal thrombosis - Transient transaminitis |
Immunosuppression-related Hematological - Anemia - Leucopenia - Neutropenia Metabolic - Dyslipidemia Gastrointestinal - Oral ulcers (Sirolimus) - Diarrhea (Mycophenolic acid) - CMV colitis Respiratory tract - Upper respiratory infections - Interstitial pneumonitis (Sirolimus) Neurological - Neurotoxicity (Tacrolimus) Genitourinary - Urinary infections - Ovarian cysts - Dysmenorrhea - Nephropathy - Proteinuria Cutaneous - Infections - Cancer |
CURRENT CHALLENGES
There are many challenges that are currently limiting islet cell transplantation (Table 8) (197-199) While significant progress has been made in the islet transplantation field, several obstacles remain precluding its widespread use. The clinical experience of islet transplantation has been developed almost exclusively using the intra-hepatic infusion through the portal vein (60). It has been suggested that the loss of as many as 50-75% of islets during engraftment is the reason why a very large number of islets are needed to achieve normoglycemia (51,72). Moreover, two additional important limitations are the currently inadequate immunosuppression for preventing islet rejection (70) and the limited oxygen supply to islet in the engraftment site (200,201). Current immunosuppressive regimens are capable of preventing islet failure for months to years, but the agents used in these treatments may increase the risk for specific malignancies and opportunistic infections. In addition, the most commonly used agents (like calcineurin inhibitors and rapamycin) are also known to impair normal islet function and/or insulin action. Furthermore, like all medications, these agents have other associated toxicities, including the harmful effect of certain widely employed immunosuppressive agents on renal function. The second very significant factor for early and late loss of islet mass is the critical lack of immediate vascularization and chronic hypo-oxygenation. Physiological supply of oxygen and nutrients in native islets is maintained by a tight capillary network, destroyed by the islet isolation procedure, restricting supply to diffusion from the portal vein and hepatic arterial capillaries until the revascularization process is completed. Oxygen tension in the liver parenchyma decreases from approximately 40 to 5 mmHg, eight-fold lower compared to the intra-pancreatic levels, leading to severe hypoxia, and β-cell death. Revascularization of the islet graft in rodent transplant requires 10-14 days and much longer in non-human primates and human recipients. Even after the revascularization of the islets is completed, the capillary’ density is significantly lower compared to the physiological intra-pancreatic situation. Finally, one of the main challenges is the cost of the procedure and some regulatory issues, as recently demonstrated by the ongoing discussion in USA (202). In fact, on April 15, the FDA's Cellular, Tissue, and Gene Therapies Advisory Committee voted in favor of approval of the biologics license application (BLA) seeking to market allogeneic islets of Langerhans for the treatment of ‘brittle” type I diabetes mellitus in adults whose symptoms are not well controlled despite intensive insulin therapy. The FDA endorsement of islet transplantation adds to the list of national agencies in Europe, such as the Federal Office of Public Health in Switzerland, the National Health Service (NHS) in the UK, the Swedish Local Authorities and Regions, the Ministry of Health in Poland and Belgium and, more recently, the French National Authority for Health (HAS) in France that have approved islet transplantation as a reimbursed standard-of-care procedure. Unfortunately, the FDA has chosen to consider islets as a biologic that requires licensure, making the universal implementation of the procedure in the clinic very challenging.
Table 8. Current Challenges Faced for Islet Transplantation |
||
Challenge |
Possible impact |
Potential solutions |
Progressive graft dysfunction |
Reintroduction of exogenous insulin; Destabilization of metabolic control; Supplemental islet transplant. |
Incretin mimetics; Alternative islet implantation sites; Novel immunosuppressive protocols. |
Multiple islet donors |
Increased operational costs; Shortage of deceased donor pancreata for transplantation; Risk of allosensitization. |
Improved donor selection criteria; Optimized cell processing; Alternative sources of transplantable tissue (i.e., stem cells-derived or xenogeneic islets; Alternative implantation sites. |
Chronic immunosuppression |
Systemic toxicity; Increased risk of opportunistic infections; Islet cell toxicity. |
Use of biologics; Immune isolation techniques; Development of immune tolerance inducing protocols. |
Allosensitization |
Reduced graft survival; Preclude/worsen outcome of subsequent transplantation (i.e., islet or renal) |
Maximizing the success rate of single donor islet transplantation; Alternative sources of transplantable tissue; Immune isolation; Plasmapheresis / depletion of alloantibodies; Novel immunosuppressive protocols; Development of immune tolerance inducing protocols. |
Cumbersome graft monitoring |
Mainly rely on metabolic function tests, but cannot discriminate between immunological and metabolic causes of dysfunction; Liver needle biopsies do not provide adequate graft tissue; MRI and PET lack the resolution to detect islets scattered throughout the liver. |
Improved simple metabolic measures predictive of graft dysfunction; Improved sensitivity of noninvasive imaging techniques (functional MRI?); Improved immune monitoring techniques for early detection of immune events able to discriminate between rejection and autoimmunity. |
FUTURE DEVELOPMENTS IN BETA-CELL REPLACEMENT THERAPIES
The field of cellular therapies for the treatment of diabetes is rapidly evolving and a new exciting era has already begun (shown in Fig. 1). Efforts are ongoing to push to a broader dimension islet transplantation (143), including: (i) implementation of a scheme for donor and recipient selection and organ allocation to increase pancreas utilization (203-205); (ii) improvement and standardization of islet isolation process and its best codification by regulatory bodies (206-209) (iii) monitoring of transplanted islets by noninvasive imaging techniques (210,211); (iv) development of biomarkers to assess the efficacy of the immunosuppression/immunomodulation strategies (69,212-216); (v) identification of alternative transplantation sites (51); (vi) creation of an ideal bio-artificial niche for islet survival by bioengineering approaches (217,218) and (vii) use of immune-isolation techniques, such as hydrogel polymers that shield pancreatic islet from immune cell attack (219,220). On the other hand, there is increasing new excitement for the use of unlimited alternative sources of transplantable islets, such as xenogeneic (i.e., obtained from other species such as porcine islets) [reviewed in (221)] or derived from human stem cells (222-227). Pig islets may be available in plentiful amounts. Importantly, the ability to obtain genetically modified pigs that lack or overexpress specific molecules may be of assistance in developing cellular products with reduced immunogenicity for transplantation into humans. In turn, this technology may allow achieving long-term function under immunosuppressive regimens that are used for allogeneic cells or facilitating the induction of long-term acceptance of xenogeneic islet cells. Another area reporting great progress is that of regenerative medicine using human stem cells from embryonic or adult sources.
While adult stem cells, such as mesenchymal stem cells, have an immunomodulatory potential when infused at disease onset (228) (229) or as adjuvants to improve the outcomes of islet transplantation (230), the greatest enthusiasm lies in the possibility to use pluripotent stem cells to overcome the limits of islet transplantation (231,232). Human pluripotent stem cells (both embryonic stem [ES] and induced pluripotent stem [iPS] cells), are the best candidate for making β cells as they have unlimited potential for division and differentiation. Efficient protocols for the differentiation of pluripotent cells into β cells have been developed by several laboratories (233-241) and a great effort in the last year was concentrated on developing cellular products with consistent potency and safety profile for clinical application. Actually, six clinical trials using human pluripotent stem cells for the therapy of type 1 diabetes are registered in ClinicalTrial.gov: three active and recruiting (NCT03163511; NCT04678557; NCT04786262), one completed (NCT03162926), one enrolling by invitation (NCT02939118) and one active but not recruiting (NCT02239354). All these trials, except the NCT04786262, are using PEC-01 cells as a cell product. PEC-01 cells are a mixed cell population comprising pancreatic endoderm and poly-hormonal endocrine cells (233,242,243)differentiated by a pluripotent stem cell line called CyT49 (225). These pancreatic precursor cells are fully committed to further differentiate into mature endocrine pancreatic cells after their implantation and were tested within an encapsulation device in subcutaneous space. In December 2021, the interim results of some of these clinical trial appeared in two articles (244) . Over the follow-up period, which lasted up to 1 year, patients had 20% reduced insulin requirements, spent 13% more time in target blood glucose range, had stable average HbA1c <7.0%, had improved hypoglycemic awareness (average Clarke score decreased ∼1 point) associated with C-peptide levels that were, on average, ∼1/100th normal levels. Explanted grafts contained heterogeneous composition of pancreatic cells, including cells with mature β cell phenotype. In both the papers: (i) induction and maintenance immunosuppression appeared to be effective in preventing allogeneic and autoimmune destruction of the graft cells, (ii) the cell product appeared to be safe and well tolerated, since no teratoma formation was observed and the great majority of mild-to-moderate adverse effects was due to surgical procedure risks and side-effects of immunosuppression. These initial data reinforce the hope that pluripotent stem cells, differentiated into pancreatic endocrine cells, may be a renewable source of β cells for patients with T1D.
VX-880 is a second cell product approved in 2021 as investigational cell therapy for the treatment of type 1 diabetes. VX-880 consists of fully differentiated insulin-producing pancreatic islet cells obtained from pluripotent stem cells. A Phase 1/2, single-arm, open-label clinical trial was recently approved in patients who have T1D with impaired hypoglycemic awareness and severe hypoglycemia. VX-880 is infused in the portal vein and a chronic administration of concomitant immunosuppressive therapy is be required to protect the islet cells from immune rejection. Some preliminary results have already been shared in a press release in May 2022 and suggest that β cells fully differentiated from stem cells and transplanted into the liver may engraft and start secreting insulin early after infusion. In addition to ongoing clinical experiences, others commercial or academic organizations have announced their intention to conduct clinical trials of functional stem cell derived-islets (135). At this point, the need to shield the transplanted stem-cell-derived β-cells from immune rejection becomes more and more critical. In this direction, different strategies to reduce or avoid immune rejection are under evaluation (245) including (i) generation of universally compatible pluripotent stem cells by silencing or deleting HLA or genes essential for HLA expression or function and by expressing genes encoding immunosuppressive molecules (246), (ii) development of mild immunosuppressive regimens (e.g., monoclonal antibodies targeting NK cells and/or T cell subsets) sufficient to induce tolerance, (iii) improvement in encapsulation/containment of cell product and (iv) creation of a haplobank of stem cell lines (247).
A current limitation for islet transplantation is the inability to use non- or minimally-invasive predictive tests as well as biomarkers of early graft dysfunction to guide timely interventions aimed at preserving functional islet cell mass. Metabolic tests (i.e., glycemic control, insulin requirement, HbA1c, basal and stimulated c-peptide) remain the main indicators of graft function, the alteration of which may indicate underlying distress of the graft but cannot discriminate possible causes such as metabolic overload, immunity, or drug toxicity. In some cases, graft dysfunction may be reversible (i.e., transient metabolic overload due to an infection episode), but in many other cases at the time graft dysfunction is detected, a considerable mass of functional beta cells might already be irreversibly lost. Monitoring of transplanted islets by noninvasive imaging techniques (such as MRI, PET-CT, and US) is cumbersome, as they lack the resolution for the detection of cellular clusters the size of islets (~50-900um) that are scattered throughout the recipient’s liver [reviewed in (248)]. While encouraging preliminary studies have shown that preloading of aliquots of the islet graft with iron nanoparticles (for MRI) (249-253) or labeled glucose (for PET-CT) (254-257) can be used safely, these techniques do not allow assessing the whole mass of transplanted clusters and provide only passive and transient information on islet distribution in the transplant site. The progress in the field of functional MRI (fMRI) and toward the development of more sensitive beta-cell specific imaging techniques may allow a more objective assessment of islet cell mass over time in a near future. Detection of biomarkers [reviewed in (109)] in blood samples to determine immune cell function (i.e., cell surface expression of specific markers by flow cytometry and cytotoxic lymphocyte gene expression profiles, amongst other)(258-260) and autoimmunity reactivation (namely, autoantibody titers) is evaluated in ongoing clinical trials to identify means of assessing the efficacy of the immunomodulation strategies, detecting rejection episodes and reactivation of autoimmunity early enough to implement timely immune interventions to prevent graft loss (69,70,261-263). Unfortunately, some of the current tests lack adequate specificity as they may be affected also with underlying infections. With the rapid evolution of high throughput arrays, it is likely that new and more specific molecular biomarkers of islet cell distress and immune cell function will become available in the near future. Alternative transplantation sites [reviewed in (51)] are being currently explored that may contribute enhancing islet engraftment and attain sustained graft function long-term (52). Importantly, alternative sites may be modified using bioengineering approaches that could enable creating an ideal bio artificial endocrine pancreas [reviewed in (264)]. The use of immunoisolation techniques, such as using hydrogel polymers that shield islet cell clusters from immune cell attack, may contribute to achieve sustained function of transplanted cells without the need for life-long immunosuppression [reviewed in (265) and (264)].
CONCLUSIONS
In conclusion, islet transplantation as it is today cannot be the universal cure for type 1 diabetes. It represents a clinical option in few highly selected patients but it is the proof of principle that it is possible to replace efficiently β cells in patients with diabetes by a cell therapy. Restoration of physiologic metabolic control in patients with diabetes is highly desirable. Transplantation of islets of Langerhans allows the achievement of stable metabolic control in the most severe manifestations that cannot be matched with conventional medical therapies. The steady progress of clinical islet transplantation and the promising emerging new approaches that address immunity and beta cell sources justifies cautious optimism for the potential applicable of beta-cell replacement to all cases of insulin-dependent diabetes in the near future.
ACKNOWLEDGMENTS
This work was partially supported by the Italian Minister of Health (Ricerca Finalizzata RF-2009-1483387, RF-2009-1469691), Ministry of Education, University and Research (PRIN 2008, prot. 2008AFA7LC), Associazione Italiana per la Ricerca sul Cancro (AIRC, bando 5 x 1,000 N_12182 and Progetto IGN_ 11783), EU (HEALTH-F5-2009-241883-BetaCellTherapy) and the Juvenile Diabetes Research Foundation International.
The author alone is responsible for reporting and interpreting these data; the views expressed herein are those of the author and not necessarily those of the funding agencies.
ONLINE RESOURCES ON THE SUBJECT
Clinical Islet Transplant Consortium; Collaborative Islet Transplant Registry; Diabetes Research Institute Foundation; Health Resources and Services Administration; International Pancreas & Islet Transplant Association; The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK); Organ Procurement and Transplantation Network; The Cell Transplant Society; Scientific Registry of Transplant Recipients; United States Department of Health and Human Services; United Network For Organ Sharing (UNOS).
ADDITIONAL ONLINE RESOURCES IN RELATED TOPICS
American Diabetes Association; American Society of Transplantation; American Society of Transplant Surgeons; Beta Cell Biology Consortium; European Pancreas Club; European Society for Organ Transplantation; International Pancreas Transplant Registry; International Xenotransplantation Association; Juvenile Diabetes Research Foundation.
REFERENCES
- Gregg EW, Sattar N, Ali MK. The changing face of diabetes complications. Lancet Diabetes Endocrinol.2016;4(6):537-547.
- Harding JL, Pavkov ME, Magliano DJ, Shaw JE, Gregg EW. Global trends in diabetes complications: a review of current evidence. Diabetologia. 2019;62(1):3-16.
- Mannucci E, Monami M, Dicembrini I, Piselli A, Porta M. Achieving HbA1c targets in clinical trials and in the real world: a systematic review and meta-analysis. J Endocrinol Invest. 2014;37(5):477-495.
- Edelman SV, Polonsky WH. Type 2 Diabetes in the Real World: The Elusive Nature of Glycemic Control. Diabetes Care. 2017;40(11):1425-1432.
- Rodbard D. State of Type 1 Diabetes Care in the United States in 2016-2018 from T1D Exchange Registry Data. Diabetes Technol Ther. 2019;21(2):62-65.
- Livingstone SJ, Levin D, Looker HC, Lindsay RS, Wild SH, Joss N, Leese G, Leslie P, McCrimmon RJ, Metcalfe W, McKnight JA, Morris AD, Pearson DW, Petrie JR, Philip S, Sattar NA, Traynor JP, Colhoun HM. Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008-2010. Jama. 2015;313(1):37-44.
- Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371(21):1972-1982.
- Alva ML, Hoerger TJ, Zhang P, Cheng YJ. State-level diabetes-attributable mortality and years of life lost in the United States. Ann Epidemiol. 2018;28(11):790-795.
- McKnight JA, Wild SH, Lamb MJ, Cooper MN, Jones TW, Davis EA, Hofer S, Fritsch M, Schober E, Svensson J, Almdal T, Young R, Warner JT, Delemer B, Souchon PF, Holl RW, Karges W, Kieninger DM, Tigas S, Bargiota A, Sampanis C, Cherubini V, Gesuita R, Strele I, Pildava S, Coppell KJ, Magee G, Cooper JG, Dinneen SF, Eeg-Olofsson K, Svensson AM, Gudbjornsdottir S, Veeze H, Aanstoot HJ, Khalangot M, Tamborlane WV, Miller KM. Glycaemic control of Type 1 diabetes in clinical practice early in the 21st century: an international comparison. Diabet Med. 2015;32(8):1036-1050.
- Gregg EW, Li Y, Wang J, Burrows NR, Ali MK, Rolka D, Williams DE, Geiss L. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med. 2014;370(16):1514-1523.
- Marzorati S, Pileggi A, Ricordi C. Allogeneic islet transplantation. Expert Opin Biol Ther. 2007;7(11):1627-1645.
- Piemonti L PA. 25 Years of the Ricordi Automated Method for Islet Isolation. CellR4. 2013;1(1):e128.
- Tzakis AG, Ricordi C, Alejandro R, Zeng Y, Fung JJ, Todo S, Demetris AJ, Mintz DH, Starzl TE. Pancreatic islet transplantation after upper abdominal exenteration and liver replacement. Lancet. 1990;336(8712):402-405.
- Najarian JS, Sutherland DE, Matas AJ, Goetz FC. Human islet autotransplantation following pancreatectomy. Transplant Proc. 1979;11(1):336-340.
- Bramis K, Gordon-Weeks AN, Friend PJ, Bastin E, Burls A, Silva MA, Dennison AR. Systematic review of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Br J Surg. 2012;99(6):761-766.
- Sutherland DE, Radosevich DM, Bellin MD, Hering BJ, Beilman GJ, Dunn TB, Chinnakotla S, Vickers SM, Bland B, Balamurugan AN, Freeman ML, Pruett TL. Total pancreatectomy and islet autotransplantation for chronic pancreatitis. J Am Coll Surg. 2012;214(4):409-424; discussion 424-406.
- Bellin MD, Beilman GJ, Dunn TB, Pruett TL, Chinnakotla S, Wilhelm JJ, Ngo A, Radosevich DM, Freeman ML, Schwarzenberg SJ, Balamurugan AN, Hering BJ, Sutherland DE. Islet autotransplantation to preserve beta cell mass in selected patients with chronic pancreatitis and diabetes mellitus undergoing total pancreatectomy. Pancreas. 2013;42(2):317-321.
- Bellin MD, Freeman ML, Schwarzenberg SJ, Dunn TB, Beilman GJ, Vickers SM, Chinnakotla S, Balamurugan AN, Hering BJ, Radosevich DM, Moran A, Sutherland DE. Quality of life improves for pediatric patients after total pancreatectomy and islet autotransplant for chronic pancreatitis. Clin Gastroenterol Hepatol. 2011;9(9):793-799.
- Bellin MD, Carlson AM, Kobayashi T, Gruessner AC, Hering BJ, Moran A, Sutherland DE. Outcome after pancreatectomy and islet autotransplantation in a pediatric population. J Pediatr Gastroenterol Nutr.2008;47(1):37-44.
- Wilson GC, Sutton JM, Salehi M, Schmulewitz N, Smith MT, Kucera S, Choe KA, Brunner JE, Abbott DE, Sussman JJ, Ahmad SA. Surgical outcomes after total pancreatectomy and islet cell autotransplantation in pediatric patients. Surgery. 2013;154(4):777-783; discussion 783-774.
- Sutton JM, Schmulewitz N, Sussman JJ, Smith M, Kurland JE, Brunner JE, Salehi M, Choe KA, Ahmad SA. Total pancreatectomy and islet cell autotransplantation as a means of treating patients with genetically linked pancreatitis. Surgery. 2010;148(4):676-685; discussion 685-676.
- Webb MA, Illouz SC, Pollard CA, Gregory R, Mayberry JF, Tordoff SG, Bone M, Cordle CJ, Berry DP, Nicholson ML, Musto PP, Dennison AR. Islet auto transplantation following total pancreatectomy: a long-term assessment of graft function. Pancreas. 2008;37(3):282-287.
- Clayton HA, Davies JE, Pollard CA, White SA, Musto PP, Dennison AR. Pancreatectomy with islet autotransplantation for the treatment of severe chronic pancreatitis: the first 40 patients at the leicester general hospital. Transplantation. 2003;76(1):92-98.
- White SA, Davies JE, Pollard C, Swift SM, Clayton HA, Sutton CD, Weymss-Holden S, Musto PP, Berry DP, Dennison AR. Pancreas resection and islet autotransplantation for end-stage chronic pancreatitis. Ann Surg.2001;233(3):423-431.
- Garcea G, Weaver J, Phillips J, Pollard CA, Ilouz SC, Webb MA, Berry DP, Dennison AR. Total pancreatectomy with and without islet cell transplantation for chronic pancreatitis: a series of 85 consecutive patients. Pancreas.2009;38(1):1-7.
- Teuscher AU, Kendall DM, Smets YF, Leone JP, Sutherland DE, Robertson RP. Successful islet autotransplantation in humans: functional insulin secretory reserve as an estimate of surviving islet cell mass. Diabetes. 1998;47(3):324-330.
- Robertson RP, Lanz KJ, Sutherland DE, Kendall DM. Prevention of diabetes for up to 13 years by autoislet transplantation after pancreatectomy for chronic pancreatitis. Diabetes. 2001;50(1):47-50.
- Blondet JJ, Carlson AM, Kobayashi T, Jie T, Bellin M, Hering BJ, Freeman ML, Beilman GJ, Sutherland DE. The role of total pancreatectomy and islet autotransplantation for chronic pancreatitis. Surg Clin North Am.2007;87(6):1477-1501, x.
- Bellin MD, Sutherland DE. Pediatric islet autotransplantation: indication, technique, and outcome. Curr Diab Rep. 2010;10(5):326-331.
- Bellin MD, Sutherland DE, Beilman GJ, Hong-McAtee I, Balamurugan AN, Hering BJ, Moran A. Similar islet function in islet allotransplant and autotransplant recipients, despite lower islet mass in autotransplants. Transplantation. 2011;91(3):367-372.
- Robertson RP. Consequences on beta-cell function and reserve after long-term pancreas transplantation. Diabetes. 2004;53(3):633-644.
- Chinnakotla S, Bellin MD, Schwarzenberg SJ, Radosevich DM, Cook M, Dunn TB, Beilman GJ, Freeman ML, Balamurugan AN, Wilhelm J, Bland B, Jimenez-Vega JM, Hering BJ, Vickers SM, Pruett TL, Sutherland DE. Total pancreatectomy and islet autotransplantation in children for chronic pancreatitis: indication, surgical techniques, postoperative management, and long-term outcomes. Ann Surg. 2014;260(1):56-64.
- Dong M, Parsaik AK, Erwin PJ, Farnell MB, Murad MH, Kudva YC. Systematic review and meta-analysis: islet autotransplantation after pancreatectomy for minimizing diabetes. Clin Endocrinol (Oxf). 2011;75(6):771-779.
- Dudeja V, Beilman GJ, Vickers SM. Total pancreatectomy with islet autotransplantation in patients with malignancy: are we there yet? Ann Surg. 2013;258(2):219-220.
- Ris F, Niclauss N, Morel P, Demuylder-Mischler S, Muller Y, Meier R, Genevay M, Bosco D, Berney T. Islet autotransplantation after extended pancreatectomy for focal benign disease of the pancreas. Transplantation.2011;91(8):895-901.
- Jin SM, Oh SH, Kim SK, Jung HS, Choi SH, Jang KT, Lee KT, Kim JH, Lee MS, Lee MK, Kim KW. Diabetes-free survival in patients who underwent islet autotransplantation after 50% to 60% distal partial pancreatectomy for benign pancreatic tumors. Transplantation. 2013;95(11):1396-1403.
- Balzano G, Nano R, Maffi P, Mercalli A, Melzi R, Aleotti F, Gavazzi F, Berra C, De Cobelli F, Venturini M, Magistretti P, Scavini M, Capretti G, Del Maschio A, Secchi A, Zerbi A, Falconi M, Piemonti L. Salvage Islet Auto Transplantation After Relaparatomy. Transplantation. 2017;101(10):2492-2500.
- Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, Zerbi A, De Cobelli F, Gavazzi F, Magistretti P, Scavini M, Peccatori J, Secchi A, Ciceri F, Del Maschio A, Falconi M, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy: A Broader Spectrum of Indications Beyond Chronic Pancreatitis. Am J Transplant. 2016;16(6):1812-1826.
- Balzano G, Maffi P, Nano R, Zerbi A, Venturini M, Melzi R, Mercalli A, Magistretti P, Scavini M, Castoldi R, Carvello M, Braga M, Del Maschio A, Secchi A, Staudacher C, Piemonti L. Extending indications for islet autotransplantation in pancreatic surgery. Ann Surg. 2013;258(2):210-218.
- Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, De Cobelli F, Magistretti P, Scavini M, Secchi A, Falconi M, Piemonti L. Diabetes-free survival after extended distal pancreatectomy and islet auto transplantation for benign or borderline/malignant lesions of the pancreas. Am J Transplant. 2019;19(3):920-928.
- Balzano G, Piemonti L. Autologous islet transplantation in patients requiring pancreatectomy for neoplasm. Curr Diab Rep. 2014;14(8):512.
- Balzano G, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy for Neoplasm. Current Diabetes Reports. 2014;14(8):512.
- Balzano G, Maffi P, Nano R, Zerbi A, Venturini M, Melzi R, Mercalli A, Magistretti P, Scavini M, Castoldi R, Carvello M, Braga M, Del Maschio A, Secchi A, Staudacher C, Piemonti L. Extending Indications for Islet Autotransplantation in Pancreatic Surgery. Annals of Surgery. 2013;258(2):210-218.
- Balzano G, Maffi P, Nano R, Mercalli A, Melzi R, Aleotti F, Zerbi A, De Cobelli F, Gavazzi F, Magistretti P, Scavini M, Peccatori J, Secchi A, Ciceri F, Del Maschio A, Falconi M, Piemonti L. Autologous Islet Transplantation in Patients Requiring Pancreatectomy: A Broader Spectrum of Indications Beyond Chronic Pancreatitis. American Journal of Transplantation. 2016;16(6):1812-1826.
- Matsumoto S, Okitsu T, Iwanaga Y, Noguchi H, Nagata H, Yonekawa Y, Yamada Y, Nakai Y, Ueda M, Ishii A, Yabunaka E, Shapiro JA, Tanaka K. Insulin independence of unstable diabetic patient after single living donor islet transplantation. Transplant Proc. 2005;37(8):3427-3429.
- Matsumoto S, Tanaka K, Strong DM, Reems JA. Efficacy of human islet isolation from the tail section of the pancreas for the possibility of living donor islet transplantation. Transplantation. 2004;78(6):839-843.
- Matsumoto I, Shinzeki M, Asari S, Goto T, Shirakawa S, Ajiki T, Fukumoto T, Ku Y. Evaluation of glucose metabolism after distal pancreatectomy according to the donor criteria of the living donor pancreas transplantation guidelines proposed by the Japanese Pancreas and Islet Transplantation Association. Transplant Proc. 2014;46(3):958-962.
- Choudhary P, Rickels MR, Senior PA, Vantyghem MC, Maffi P, Kay TW, Keymeulen B, Inagaki N, Saudek F, Lehmann R, Hering BJ. Evidence-informed clinical practice recommendations for treatment of type 1 diabetes complicated by problematic hypoglycemia. Diabetes Care. 2015;38(6):1016-1029.
- Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Caicedo A. The unique cytoarchitecture of human pancreatic islets has implications for islet cell function. Proc Natl Acad Sci U S A. 2006;103(7):2334-2339.
- Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW. Automated method for isolation of human pancreatic islets. Diabetes. 1988;37(4):413-420.
- Cantarelli E, Piemonti L. Alternative transplantation sites for pancreatic islet grafts. Curr Diab Rep.2011;11(5):364-374.
- Maffi P, Balzano G, Ponzoni M, Nano R, Sordi V, Melzi R, Mercalli A, Scavini M, Esposito A, Peccatori J, Cantarelli E, Messina C, Bernardi M, Del Maschio A, Staudacher C, Doglioni C, Ciceri F, Secchi A, Piemonti L. Autologous pancreatic islet transplantation in human bone marrow. Diabetes. 2013;62(10):3523-3531.
- Maffi P, Nano R, Monti P, Melzi R, Sordi V, Mercalli A, Pellegrini S, Ponzoni M, Peccatori J, Messina C, Nocco A, Cardillo M, Scavini M, Magistretti P, Doglioni C, Ciceri F, Bloem SJ, Roep BO, Secchi A, Piemonti L. Islet Allotransplantation in the Bone Marrow of Patients With Type 1 Diabetes: A Pilot Randomized Trial. Transplantation. 2019;103(4):839-851.
- Yasunami Y, Nakafusa Y, Nitta N, Nakamura M, Goto M, Ono J, Taniguchi M. A Novel Subcutaneous Site of Islet Transplantation Superior to the Liver. Transplantation. 2018;102(6):945-952.
- Wszola M, Berman A, Ostaszewska A, Gorski L, Serwanska-Swietek M, Gozdowska J, Bednarska K, Krajewska M, Lipinska A, Chmura A, Kwiatkowski A. Islets Allotransplantation Into Gastric Submucosa in a Patient with Portal Hypertension: 4-year Follow-up. Transplant Proc. 2018;50(6):1910-1913.
- Berman DM, Molano RD, Fotino C, Ulissi U, Gimeno J, Mendez AJ, Kenyon NM, Kenyon NS, Andrews DM, Ricordi C, Pileggi A. Bioengineering the Endocrine Pancreas: Intraomental Islet Transplantation Within a Biologic Resorbable Scaffold. Diabetes. 2016;65(5):1350-1361.
- Baidal DA, Ricordi C, Berman DM, Alvarez A, Padilla N, Ciancio G, Linetsky E, Pileggi A, Alejandro R. Bioengineering of an Intraabdominal Endocrine Pancreas. N Engl J Med. 2017;376(19):1887-1889.
- Christoffersson G, Henriksnas J, Johansson L, Rolny C, Ahlstrom H, Caballero-Corbalan J, Segersvard R, Permert J, Korsgren O, Carlsson PO, Phillipson M. Clinical and experimental pancreatic islet transplantation to striated muscle: establishment of a vascular system similar to that in native islets. Diabetes. 2010;59(10):2569-2578.
- Bertuzzi F, Colussi G, Lauterio A, De Carlis L. Intramuscular islet allotransplantation in type 1 diabetes mellitus. Eur Rev Med Pharmacol Sci. 2018;22(6):1731-1736.
- Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, DiMercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006;355(13):1318-1330.
- Benhamou PY, Oberholzer J, Toso C, Kessler L, Penfornis A, Bayle F, Thivolet C, Martin X, Ris F, Badet L, Colin C, Morel P. Human islet transplantation network for the treatment of Type I diabetes: first data from the Swiss-French GRAGIL consortium (1999-2000). Groupe de Recherche Rhin Rhjne Alpes Geneve pour la transplantation d'Ilots de Langerhans. Diabetologia. 2001;44(7):859-864.
- Goss JA, Goodpastor SE, Brunicardi FC, Barth MH, Soltes GD, Garber AJ, Hamilton DJ, Alejandro R, Ricordi C. Development of a human pancreatic islet-transplant program through a collaborative relationship with a remote islet-isolation center. Transplantation. 2004;77(3):462-466.
- Goss JA, Schock AP, Brunicardi FC, Goodpastor SE, Garber AJ, Soltes G, Barth M, Froud T, Alejandro R, Ricordi C. Achievement of insulin independence in three consecutive type-1 diabetic patients via pancreatic islet transplantation using islets isolated at a remote islet isolation center. Transplantation. 2002;74(12):1761-1766.
- Lablanche S, Borot S, Wojtusciszyn A, Bayle F, Tetaz R, Badet L, Thivolet C, Morelon E, Frimat L, Penfornis A, Kessler L, Brault C, Colin C, Tauveron I, Bosco D, Berney T, Benhamou PY. Five-Year Metabolic, Functional, and Safety Results of Patients With Type 1 Diabetes Transplanted With Allogenic Islets Within the Swiss-French GRAGIL Network. Diabetes Care. 2015;38(9):1714-1722.
- Berney T, Andres A, Bellin MD, de Koning EJ, Johnson PR, Kay TW, Lundgren T, Rickels MR, Scholz H, Stock PG. A Worldwide Survey of Activities and Practices in Clinical Islet of Langerhans Transplantation. Transplant International. 2022:135.
- Blood N. Transplant. Organ and tissue donation and transplantation: activity report 2020/21. 2021. 2022.
- Webster AC, Hedley JA, Anderson PF, Hawthorne WJ, Radford T, Drogemuller C, Rogers N, Goodman D, Lee MH, Loudovaris T. Australia and New Zealand islet and pancreas transplant registry annual report 2019: islet donations, islet isolations, and islet transplants. Transplantation direct. 2020;6(7).
- Campbell PM, Salam A, Ryan EA, Senior P, Paty BW, Bigam D, McCready T, Halpin A, Imes S, Al Saif F, Lakey JR, Shapiro AM. Pretransplant HLA antibodies are associated with reduced graft survival after clinical islet transplantation. Am J Transplant. 2007;7(5):1242-1248.
- Hilbrands R, Huurman VA, Gillard P, Velthuis JH, De Waele M, Mathieu C, Kaufman L, Pipeleers-Marichal M, Ling Z, Movahedi B, Jacobs-Tulleneers-Thevissen D, Monbaliu D, Ysebaert D, Gorus FK, Roep BO, Pipeleers DG, Keymeulen B. Differences in baseline lymphocyte counts and autoreactivity are associated with differences in outcome of islet cell transplantation in type 1 diabetic patients. Diabetes. 2009;58(10):2267-2276.
- Piemonti L, Everly MJ, Maffi P, Scavini M, Poli F, Nano R, Cardillo M, Melzi R, Mercalli A, Sordi V, Lampasona V, Espadas de Arias A, Scalamogna M, Bosi E, Bonifacio E, Secchi A, Terasaki PI. Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes.2013;62(5):1656-1664.
- Citro A, Cantarelli E, Maffi P, Nano R, Melzi R, Mercalli A, Dugnani E, Sordi V, Magistretti P, Daffonchio L, Ruffini PA, Allegretti M, Secchi A, Bonifacio E, Piemonti L. CXCR1/2 inhibition enhances pancreatic islet survival after transplantation. J Clin Invest. 2012;122(10):3647-3651.
- Citro A, Cantarelli E, Piemonti L. Anti-inflammatory strategies to enhance islet engraftment and survival. Curr Diab Rep. 2013;13(5):733-744.
- Moberg L, Johansson H, Lukinius A, Berne C, Foss A, Kallen R, Ostraat O, Salmela K, Tibell A, Tufveson G, Elgue G, Nilsson Ekdahl K, Korsgren O, Nilsson B. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet. 2002;360(9350):2039-2045.
- Matsuoka N, Itoh T, Watarai H, Sekine-Kondo E, Nagata N, Okamoto K, Mera T, Yamamoto H, Yamada S, Maruyama I, Taniguchi M, Yasunami Y. High-mobility group box 1 is involved in the initial events of early loss of transplanted islets in mice. J Clin Invest. 2010;120(3):735-743.
- Barton FB, Rickels MR, Alejandro R, Hering BJ, Wease S, Naziruddin B, Oberholzer J, Odorico JS, Garfinkel MR, Levy M, Pattou F, Berney T, Secchi A, Messinger S, Senior PA, Maffi P, Posselt A, Stock PG, Kaufman DB, Luo X, Kandeel F, Cagliero E, Turgeon NA, Witkowski P, Naji A, O'Connell PJ, Greenbaum C, Kudva YC, Brayman KL, Aull MJ, Larsen C, Kay TW, Fernandez LA, Vantyghem MC, Bellin M, Shapiro AM. Improvement in outcomes of clinical islet transplantation: 1999-2010. Diabetes Care. 2012;35(7):1436-1445.
- Pileggi A, Ricordi C, Kenyon NS, Froud T, Baidal DA, Kahn A, Selvaggi G, Alejandro R. Twenty years of clinical islet transplantation at the Diabetes Research Institute--University of Miami. Clin Transpl. 2004:177-204.
- Hering BJ, Kandaswamy R, Ansite JD, Eckman PM, Nakano M, Sawada T, Matsumoto I, Ihm SH, Zhang HJ, Parkey J, Hunter DW, Sutherland DE. Single-donor, marginal-dose islet transplantation in patients with type 1 diabetes. Jama. 2005;293(7):830-835.
- Frank AM, Barker CF, Markmann JF. Comparison of whole organ pancreas and isolated islet transplantation for type 1 diabetes. Advances in surgery. 2005;39:137-163.
- Gerber PA, Pavlicek V, Demartines N, Zuellig R, Pfammatter T, Wuthrich R, Weber M, Spinas GA, Lehmann R. Simultaneous islet-kidney vs pancreas-kidney transplantation in type 1 diabetes mellitus: a 5 year single centre follow-up. Diabetologia. 2008;51(1):110-119.
- Froud T, Baidal DA, Faradji R, Cure P, Mineo D, Selvaggi G, Kenyon NS, Ricordi C, Alejandro R. Islet transplantation with alemtuzumab induction and calcineurin-free maintenance immunosuppression results in improved short- and long-term outcomes. Transplantation. 2008;86(12):1695-1701.
- Bellin MD, Kandaswamy R, Parkey J, Zhang HJ, Liu B, Ihm SH, Ansite JD, Witson J, Bansal-Pakala P, Balamurugan AN, Papas K, Sutherland DE, Moran A, Hering BJ. Prolonged insulin independence after islet allotransplants in recipients with type 1 diabetes. Am J Transplant. 2008;8(11):2463-2470.
- Alejandro R, Barton FB, Hering BJ, Wease S. 2008 Update from the Collaborative Islet Transplant Registry. Transplantation. 2008;86(12):1783-1788.
- Tan J, Yang S, Cai J, Guo J, Huang L, Wu Z, Chen J, Liao L. Simultaneous islet and kidney transplantation in seven patients with type 1 diabetes and end-stage renal disease using a glucocorticoid-free immunosuppressive regimen with alemtuzumab induction. Diabetes. 2008;57(10):2666-2671.
- Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343(4):230-238.
- Froud T, Ricordi C, Baidal DA, Hafiz MM, Ponte G, Cure P, Pileggi A, Poggioli R, Ichii H, Khan A, Ferreira JV, Pugliese A, Esquenazi VV, Kenyon NS, Alejandro R. Islet transplantation in type 1 diabetes mellitus using cultured islets and steroid-free immunosuppression: Miami experience. Am J Transplant. 2005;5(8):2037-2046.
- Vantyghem MC, Kerr-Conte J, Arnalsteen L, Sergent G, Defrance F, Gmyr V, Declerck N, Raverdy V, Vandewalle B, Pigny P, Noel C, Pattou F. Primary graft function, metabolic control, and graft survival after islet transplantation. Diabetes Care. 2009;32(8):1473-1478.
- Shapiro AM. Strategies toward single-donor islets of Langerhans transplantation. Curr Opin Organ Transplant.2011;16(6):627-631.
- Nijhoff MF, Engelse MA, Dubbeld J, Braat AE, Ringers J, Roelen DL, van Erkel AR, Spijker HS, Bouwsma H, van der Boog PJ, de Fijter JW, Rabelink TJ, de Koning EJ. Glycemic Stability Through Islet-After-Kidney Transplantation Using an Alemtuzumab-Based Induction Regimen and Long-Term Triple-Maintenance Immunosuppression. Am J Transplant. 2015.
- Badell IR, Russell MC, Thompson PW, Turner AP, Weaver TA, Robertson JM, Avila JG, Cano JA, Johnson BE, Song M, Leopardi FV, Swygert S, Strobert EA, Ford ML, Kirk AD, Larsen CP. LFA-1-specific therapy prolongs allograft survival in rhesus macaques. J Clin Invest. 2010;120(12):4520-4531.
- Turgeon NA, Avila JG, Cano JA, Hutchinson JJ, Badell IR, Page AJ, Adams AB, Sears MH, Bowen PH, Kirk AD, Pearson TC, Larsen CP. Experience with a novel efalizumab-based immunosuppressive regimen to facilitate single donor islet cell transplantation. Am J Transplant. 2010;10(9):2082-2091.
- Posselt AM, Bellin MD, Tavakol M, Szot GL, Frassetto LA, Masharani U, Kerlan RK, Fong L, Vincenti FG, Hering BJ, Bluestone JA, Stock PG. Islet transplantation in type 1 diabetics using an immunosuppressive protocol based on the anti-LFA-1 antibody efalizumab. Am J Transplant. 2010;10(8):1870-1880.
- Posselt AM, Szot GL, Frassetto LA, Masharani U, Tavakol M, Amin R, McElroy J, Ramos MD, Kerlan RK, Fong L, Vincenti F, Bluestone JA, Stock PG. Islet transplantation in type 1 diabetic patients using calcineurin inhibitor-free immunosuppressive protocols based on T-cell adhesion or costimulation blockade. Transplantation.2010;90(12):1595-1601.
- Fotino C, Pileggi A. Blockade of Leukocyte Function Antigen-1 (LFA-1) in Clinical Islet Transplantation. Curr Diab Rep. 2011;11(5):337-344.
- Li T, Ma R, Zhu JY, Wang FS, Huang L, Leng XS. PD-1/PD-L1 costimulatory pathway-induced mouse islet transplantation immune tolerance. Transplant Proc. 2015;47(1):165-170.
- Watanabe M, Yamashita K, Suzuki T, Kamachi H, Kuraya D, Koshizuka Y, Ogura M, Yoshida T, Aoyagi T, Fukumori D, Shimamura T, Okimura K, Maeta K, Miura T, Sakai F, Todo S. ASKP1240, a fully human anti-CD40 monoclonal antibody, prolongs pancreatic islet allograft survival in nonhuman primates. Am J Transplant.2013;13(8):1976-1988.
- Citro A, Cantarelli E, Piemonti L. The CXCR1/2 Pathway: Involvement in Diabetes Pathophysiology and Potential Target for T1D Interventions. Curr Diab Rep. 2015;15(10):638.
- Maffi P, Berney T, Nano R, Niclauss N, Bosco D, Melzi R, Mercalli A, Magistretti P, De Cobelli F, Battaglia M, Scavini M, Demuylder-Mischler S, Secchi A, Piemonti L. Calcineurin inhibitor-free immunosuppressive regimen in type 1 diabetes patients receiving islet transplantation: single-group phase 1/2 trial. Transplantation.2014;98(12):1301-1309.
- Cure P, Froud T, Leitao CB, Pileggi A, Tharavanij T, Bernetti K, Baidal DA, Selvaggi G, Ricordi C, Alejandro R. Late Epstein Barr virus reactivation in islet after kidney transplantation. Transplantation. 2008;86(9):1324-1325.
- Hafiz MM, Poggioli R, Caulfield A, Messinger S, Geiger MC, Baidal DA, Froud T, Ferreira JV, Tzakis AG, Ricordi C, Alejandro R. Cytomegalovirus prevalence and transmission after islet allograft transplant in patients with type 1 diabetes mellitus. Am J Transplant. 2004;4(10):1697-1702.
- Senior PA, Paty BW, Cockfield SM, Ryan EA, Shapiro AM. Proteinuria developing after clinical islet transplantation resolves with sirolimus withdrawal and increased tacrolimus dosing. Am J Transplant.2005;5(9):2318-2323.
- Maffi P, Bertuzzi F, De Taddeo F, Magistretti P, Nano R, Fiorina P, Caumo A, Pozzi P, Socci C, Venturini M, del Maschio A, Secchi A. Kidney function after islet transplant alone in type 1 diabetes: impact of immunosuppressive therapy on progression of diabetic nephropathy. Diabetes Care. 2007;30(5):1150-1155.
- Leitao CB, Cure P, Messinger S, Pileggi A, Lenz O, Froud T, Faradji RN, Selvaggi G, Kupin W, Ricordi C, Alejandro R. Stable renal function after islet transplantation: importance of patient selection and aggressive clinical management. Transplantation. 2009;87(5):681-688.
- Leitao CB, Froud T, Cure P, Tharavanij T, Pileggi A, Ricordi C, Alejandro R. Nonalbumin proteinuria in islet transplant recipients. Cell Transplant. 2010;19(1):119-125.
- Warnock GL, Thompson DM, Meloche RM, Shapiro RJ, Ao Z, Keown P, Johnson JD, Verchere CB, Partovi N, Begg IS, Fung M, Kozak SE, Tong SO, Alghofaili KM, Harris C. A multi-year analysis of islet transplantation compared with intensive medical therapy on progression of complications in type 1 diabetes. Transplantation.2008;86(12):1762-1766.
- Leitao CB, Bernetti K, Tharavanij T, Cure P, Lauriola V, Berggren PO, Ricordi C, Alejandro R. Lipotoxicity and decreased islet graft survival. Diabetes Care. 2010;33(3):658-660.
- Barshes NR, Lee TC, Goodpastor SE, Balkrishnan R, Schock AP, Mote A, Brunicardi FC, Alejandro R, Ricordi C, Goss JA. Transaminitis after pancreatic islet transplantation. J Am Coll Surg. 2005;200(3):353-361.
- Hafiz MM, Faradji RN, Froud T, Pileggi A, Baidal DA, Cure P, Ponte G, Poggioli R, Cornejo A, Messinger S, Ricordi C, Alejandro R. Immunosuppression and procedure-related complications in 26 patients with type 1 diabetes mellitus receiving allogeneic islet cell transplantation. Transplantation. 2005;80(12):1718-1728.
- Venturini M, Maffi P, Querques G, Agostini G, Piemonti L, Sironi S, De Cobelli F, Fiorina P, Secchi A, Del Maschio A. Hepatic steatosis after islet transplantation: Can ultrasound predict the clinical outcome? A longitudinal study in 108 patients. Pharmacol Res. 2015;98:52-59.
- Monti P, Vignali D, Piemonti L. Monitoring Inflammation, Humoral and Cell-mediated Immunity in Pancreas and Islet Transplants. Curr Diabetes Rev. 2015;11(3):135-143.
- Lobo PI, Spencer C, Simmons WD, Hagspiel KD, Angle JF, Deng S, Markmann J, Naji A, Kirk SE, Pruett T, Brayman KL. Development of anti-human leukocyte antigen class 1 antibodies following allogeneic islet cell transplantation. Transplant Proc. 2005;37(8):3438-3440.
- Rickels MR, Kearns J, Markmann E, Palanjian M, Markmann JF, Naji A, Kamoun M. HLA sensitization in islet transplantation. Clin Transpl. 2006:413-420.
- Cardani R, Pileggi A, Ricordi C, Gomez C, Baidal DA, Ponte GG, Mineo D, Faradji RN, Froud T, Ciancio G, Esquenazi V, Burke GW, 3rd, Selvaggi G, Miller J, Kenyon NS, Alejandro R. Allosensitization of islet allograft recipients. Transplantation. 2007;84(11):1413-1427.
- Campbell PM, Senior PA, Salam A, Labranche K, Bigam DL, Kneteman NM, Imes S, Halpin A, Ryan EA, Shapiro AM. High risk of sensitization after failed islet transplantation. Am J Transplant. 2007;7(10):2311-2317.
- Bosi E, Bottazzo GF, Secchi A, Pozza G, Shattock M, Saunders A, Gelet A, Touraine JL, Traeger J, Dubernard JM. Islet cell autoimmunity in type I diabetic patients after HLA-mismatched pancreas transplantation. Diabetes.1989;38 Suppl 1:82-84.
- Burke GW, 3rd, Vendrame F, Pileggi A, Ciancio G, Reijonen H, Pugliese A. Recurrence of autoimmunity following pancreas transplantation. Curr Diab Rep. 2011;11(5):413-419.
- Vendrame F, Pileggi A, Laughlin E, Allende G, Martin-Pagola A, Molano RD, Diamantopoulos S, Standifer N, Geubtner K, Falk BA, Ichii H, Takahashi H, Snowhite I, Chen Z, Mendez A, Chen L, Sageshima J, Ruiz P, Ciancio G, Ricordi C, Reijonen H, Nepom GT, Burke GW, 3rd, Pugliese A. Recurrence of type 1 diabetes after simultaneous pancreas-kidney transplantation, despite immunosuppression, is associated with autoantibodies and pathogenic autoreactive CD4 T-cells. Diabetes. 2010;59(4):947-957.
- Ryan EA, Paty BW, Senior PA, Lakey JR, Bigam D, Shapiro AM. Beta-score: an assessment of beta-cell function after islet transplantation. Diabetes Care. 2005;28(2):343-347.
- Faradji RN, Monroy K, Messinger S, Pileggi A, Froud T, Baidal DA, Cure PE, Ricordi C, Luzi L, Alejandro R. Simple measures to monitor beta-cell mass and assess islet graft dysfunction. Am J Transplant. 2007;7(2):303-308.
- Matsumoto S, Noguchi H, Hatanaka N, Shimoda M, Kobayashi N, Jackson A, Onaca N, Naziruddin B, Levy MF. SUITO index for evaluation of efficacy of single donor islet transplantation. Cell Transplant. 2009;18(5):557-562.
- Takita M, Matsumoto S, Qin H, Noguchi H, Shimoda M, Chujo D, Itoh T, Sugimoto K, Onaca N, Naziruddin B, Levy MF. Secretory Unit of Islet Transplant Objects (SUITO) Index can predict severity of hypoglycemic episodes in clinical islet cell transplantation. Cell Transplant. 2011.
- Ryan EA, Lakey JR, Rajotte RV, Korbutt GS, Kin T, Imes S, Rabinovitch A, Elliott JF, Bigam D, Kneteman NM, Warnock GL, Larsen I, Shapiro AM. Clinical outcomes and insulin secretion after islet transplantation with the Edmonton protocol. Diabetes. 2001;50(4):710-719.
- Ryan EA, Lakey JR, Paty BW, Imes S, Korbutt GS, Kneteman NM, Bigam D, Rajotte RV, Shapiro AM. Successful islet transplantation: continued insulin reserve provides long-term glycemic control. Diabetes.2002;51(7):2148-2157.
- Caumo A, Maffi P, Nano R, Luzi L, Hilbrands R, Gillard P, Jacobs-Tulleneers-Thevissen D, Secchi A, Keymeulen B, Pipeleers D, Piemonti L. Comparative evaluation of simple indices of graft function after islet transplantation. Transplantation. 2011;92(7):815-821.
- Caumo A, Maffi P, Nano R, Bertuzzi F, Luzi L, Secchi A, Bonifacio E, Piemonti L. Transplant estimated function: a simple index to evaluate beta-cell secretion after islet transplantation. Diabetes Care. 2008;31(2):301-305.
- Forbes S, Oram RA, Smith A, Lam A, Olateju T, Imes S, Malcolm AJ, Shapiro AM, Senior PA. Validation of the BETA-2 Score: An Improved Tool to Estimate Beta Cell Function After Clinical Islet Transplantation Using a Single Fasting Blood Sample. Am J Transplant. 2016;16(9):2704-2713.
- Golebiewska JE, Solomina J, Thomas C, Kijek MR, Bachul PJ, Basto L, Golab K, Wang LJ, Fillman N, Tibudan M, Cieply K, Philipson L, Debska-Slizien A, Millis JM, Fung J, Witkowski P. Comparative evaluation of simple indices using a single fasting blood sample to estimate beta cell function after islet transplantation. Am J Transplant. 2018;18(4):990-997.
- Rickels MR, Stock PG, Koning EJPd, Piemonti L, Pratschke J, Alejandro R, Bellin MD, Berney T, Choudhary P, Johnson PR, Kandaswamy R, Kay TWH, Keymeulen B, Kudva YC, Latres E, Langer RM, Lehmann R, Ludwig B, Markmann JF, Marinac M, Odorico JS, Pattou F, Senior PA, Shaw JAM, Vantyghem M-C, White S. Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes: a Consensus Report on the Igls Criteria from the IPITA/EPITA Opinion Leaders Workshop. Transplant International. 2018.
- Rickels MR, Stock PG, Koning EJPd, Piemonti L, Pratschke J, Alejandro R, Bellin MD, Berney T, Choudhary P, Johnson PR, Kandaswamy R, Kay TWH, Keymeulen B, Kudva YC, Latres E, Langer RM, Lehmann R, Ludwig B, Markmann JF, Marinac M, Odorico JS, Pattou F, Senior PA, Shaw JAM, Vantyghem M-C, White S. Defining Outcomes for β-Cell Replacement Therapy in the Treatment of Diabetes: a Consensus Report on the Igls Criteria from the IPITA/EPITA Opinion Leaders Workshop. Transplantation. 2018.
- Piemonti L, de Koning EJP, Berney T, Odorico JS, Markmann JF, Stock PG, Rickels MR. Defining outcomes for beta cell replacement therapy: a work in progress. Diabetologia. 2018;61(6):1273-1276.
- Landstra CP, Andres A, Chetboun M, Conte C, Kelly Y, Berney T, De Koning EJ, Piemonti L, Stock PG, Pattou F. Examination of the Igls Criteria for Defining Functional Outcomes of β-cell Replacement Therapy: IPITA Symposium Report. The Journal of Clinical Endocrinology & Metabolism. 2021;106(10):3049-3059.
- Hering BJ, Clarke WR, Bridges ND, Eggerman TL, Alejandro R, Bellin MD, Chaloner K, Czarniecki CW, Goldstein JS, Hunsicker LG, Kaufman DB, Korsgren O, Larsen CP, Luo X, Markmann JF, Naji A, Oberholzer J, Posselt AM, Rickels MR, Ricordi C, Robien MA, Senior PA, Shapiro AM, Stock PG, Turgeon NA. Phase 3 Trial of Transplantation of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia. Diabetes Care.2016;39(7):1230-1240.
- Markmann JF, Rickels MR, Eggerman TL, Bridges ND, Lafontant DE, Qidwai J, Foster E, Clarke WR, Kamoun M, Alejandro R, Bellin MD, Chaloner K, Czarniecki CW, Goldstein JS, Hering BJ, Hunsicker LG, Kaufman DB, Korsgren O, Larsen CP, Luo X, Naji A, Oberholzer J, Posselt AM, Ricordi C, Senior PA, Shapiro AMJ, Stock PG, Turgeon NA. Phase 3 trial of human islet-after-kidney transplantation in type 1 diabetes. Am J Transplant.2021;21(4):1477-1492.
- Lablanche S, Vantyghem MC, Kessler L, Wojtusciszyn A, Borot S, Thivolet C, Girerd S, Bosco D, Bosson JL, Colin C, Tetaz R, Logerot S, Kerr-Conte J, Renard E, Penfornis A, Morelon E, Buron F, Skaare K, Grguric G, Camillo-Brault C, Egelhofer H, Benomar K, Badet L, Berney T, Pattou F, Benhamou PY. Islet transplantation versus insulin therapy in patients with type 1 diabetes with severe hypoglycaemia or poorly controlled glycaemia after kidney transplantation (TRIMECO): a multicentre, randomised controlled trial. Lancet Diabetes Endocrinol.2018;6(7):527-537.
- Maffi P, Lundgren T, Tufveson G, Rafael E, Shaw JAM, Liew A, Saudek F, Witkowski P, Golab K, Bertuzzi F, Gustafsson B, Daffonchio L, Ruffini PA, Piemonti L. Targeting CXCR1/2 Does Not Improve Insulin Secretion After Pancreatic Islet Transplantation: A Phase 3, Double-Blind, Randomized, Placebo-Controlled Trial in Type 1 Diabetes. Diabetes Care. 2020;43(4):710-718.
- Piemonti L. Felix dies natalis, insulin... ceterum autem censeo "beta is better". Acta Diabetol. 2021.
- Johnson JA, Kotovych M, Ryan EA, Shapiro AJ. Reduced fear of hypoglycemia in successful islet transplantation. Diabetes Care. 2004;27(2):624-625.
- Barton FB, Rickels MR, Alejandro R, Hering BJ, Wease S, Naziruddin B, Oberholzer J, Odorico JS, Garfinkel MR, Levy M. Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care.2012;35(7):1436-1445.
- Paty BW, Ryan EA, Shapiro AJ, Lakey JR, Robertson RP. Intrahepatic islet transplantation in type 1 diabetic patients does not restore hypoglycemic hormonal counterregulation or symptom recognition after insulin independence. Diabetes. 2002;51(12):3428-3434.
- Rickels MR, Schutta MH, Markmann JF, Barker CF, Naji A, Teff KL. β-Cell function following human islet transplantation for type 1 diabetes. Diabetes. 2005;54(1):100-106.
- Rickels MR, Schutta MH, Mueller R, Markmann JF, Barker CF, Naji A, Teff KL. Islet cell hormonal responses to hypoglycemia after human islet transplantation for type 1 diabetes. Diabetes. 2005;54(11):3205-3211.
- Paty BW, Senior PA, Lakey JR, Shapiro AJ, Ryan EA. Assessment of glycemic control after islet transplantation using the continuous glucose monitor in insulin-independent versus insulin-requiring type 1 diabetes subjects. Diabetes technology & therapeutics. 2006;8(2):165-173.
- Rickels MR, Naji A, Teff KL. Acute insulin responses to glucose and arginine as predictors of β-cell secretory capacity in human islet transplantation. Transplantation. 2007;84(10):1357-1360.
- Vantyghem MC, de Koning EJP, Pattou F, Rickels MR. Advances in beta-cell replacement therapy for the treatment of type 1 diabetes. Lancet. 2019;394(10205):1274-1285.
- Vantyghem M-C, Chetboun M, Gmyr V, Jannin A, Espiard S, Le Mapihan K, Raverdy V, Delalleau N, Machuron F, Hubert T. Ten-year outcome of islet alone or islet after kidney transplantation in type 1 diabetes: A prospective parallel-arm cohort study. Diabetes Care. 2019;42(11):2042-2049.
- Marfil-Garza BA, Imes S, Verhoeff K, Hefler J, Lam A, Dajani K, Anderson B, O'Gorman D, Kin T, Bigam D. Pancreatic islet transplantation in type 1 diabetes: 20-year experience from a single-centre cohort in Canada. The lancet Diabetes & endocrinology. 2022.
- Vantyghem MC, Raverdy V, Balavoine AS, Defrance F, Caiazzo R, Arnalsteen L, Gmyr V, Hazzan M, Noel C, Kerr-Conte J, Pattou F. Continuous glucose monitoring after islet transplantation in type 1 diabetes: an excellent graft function (beta-score greater than 7) Is required to abrogate hyperglycemia, whereas a minimal function is necessary to suppress severe hypoglycemia (beta-score greater than 3). J Clin Endocrinol Metab.2012;97(11):E2078-2083.
- Johnson JA, Kotovych M, Ryan EA, Shapiro AM. Reduced fear of hypoglycemia in successful islet transplantation. Diabetes Care. 2004;27(2):624-625.
- Poggioli R, Faradji RN, Ponte G, Betancourt A, Messinger S, Baidal DA, Froud T, Ricordi C, Alejandro R. Quality of life after islet transplantation. Am J Transplant. 2006;6(2):371-378.
- Toso C, Shapiro AM, Bowker S, Dinyari P, Paty B, Ryan EA, Senior P, Johnson JA. Quality of life after islet transplant: impact of the number of islet infusions and metabolic outcome. Transplantation. 2007;84(5):664-666.
- Leitao CB, Tharavanij T, Cure P, Pileggi A, Baidal DA, Ricordi C, Alejandro R. Restoration of hypoglycemia awareness after islet transplantation. Diabetes Care. 2008;31(11):2113-2115.
- Tharavanij T, Betancourt A, Messinger S, Cure P, Leitao CB, Baidal DA, Froud T, Ricordi C, Alejandro R. Improved long-term health-related quality of life after islet transplantation. Transplantation. 2008;86(9):1161-1167.
- Radosevich DM, Jevne R, Bellin M, Kandaswamy R, Sutherland DE, Hering BJ. Comprehensive health assessment and five-yr follow-up of allogeneic islet transplant recipients. Clin Transplant. 2013;27(6):E715-724.
- Benhamou PY, Milliat-Guittard L, Wojtusciszyn A, Kessler L, Toso C, Baertschiger R, Debaty I, Badet L, Penfornis A, Thivolet C, Renard E, Bayle F, Morel P, Morelon E, Colin C, Berney T. Quality of life after islet transplantation: data from the GRAGIL 1 and 2 trials. Diabet Med. 2009;26(6):617-621.
- Cure P, Pileggi A, Froud T, Messinger S, Faradji RN, Baidal DA, Cardani R, Curry A, Poggioli R, Pugliese A, Betancourt A, Esquenazi V, Ciancio G, Selvaggi G, Burke GW, 3rd, Ricordi C, Alejandro R. Improved metabolic control and quality of life in seven patients with type 1 diabetes following islet after kidney transplantation. Transplantation. 2008;85(6):801-812.
- Barshes NR, Vanatta JM, Mote A, Lee TC, Schock AP, Balkrishnan R, Brunicardi FC, Goss JA. Health-related quality of life after pancreatic islet transplantation: a longitudinal study. Transplantation. 2005;79(12):1727-1730.
- Reid L, Baxter F, Forbes S. Effects of Islet Transplantation on Microvascular and Macrovascular Complications in Type 1 Diabetes. Diabet Med. 2021:e14570.
- Thompson DM, Meloche M, Ao Z, Paty B, Keown P, Shapiro RJ, Ho S, Worsley D, Fung M, Meneilly G, Begg I, Al Mehthel M, Kondi J, Harris C, Fensom B, Kozak SE, Tong SO, Trinh M, Warnock GL. Reduced progression of diabetic microvascular complications with islet cell transplantation compared with intensive medical therapy. Transplantation. 2011;91(3):373-378.
- Lee TC, Barshes NR, O'Mahony CA, Nguyen L, Brunicardi FC, Ricordi C, Alejandro R, Schock AP, Mote A, Goss JA. The effect of pancreatic islet transplantation on progression of diabetic retinopathy and neuropathy. Transplant Proc. 2005;37(5):2263-2265.
- Venturini M, Fiorina P, Maffi P, Losio C, Vergani A, Secchi A, Del Maschio A. Early increase of retinal arterial and venous blood flow velocities at color Doppler imaging in brittle type 1 diabetes after islet transplant alone. Transplantation. 2006;81(9):1274-1277.
- Del Carro U, Fiorina P, Amadio S, De Toni Franceschini L, Petrelli A, Menini S, Martinelli Boneschi F, Ferrari S, Pugliese G, Maffi P, Comi G, Secchi A. Evaluation of polyneuropathy markers in type 1 diabetic kidney transplant patients and effects of islet transplantation: neurophysiological and skin biopsy longitudinal analysis. Diabetes Care. 2007;30(12):3063-3069.
- Vantyghem MC, Quintin D, Caiazzo R, Leroy C, Raverdy V, Cassim F, Glowacki F, Hubert T, Gmyr V, Noel C, Kerr-Conte J, Pattou F. Improvement of electrophysiological neuropathy after islet transplantation for type 1 diabetes: a 5-year prospective study. Diabetes Care. 2014;37(6):e141-142.
- D'Addio F, Maffi P, Vezzulli P, Vergani A, Mello A, Bassi R, Nano R, Falautano M, Coppi E, Finzi G, D'Angelo A, Fermo I, Pellegatta F, La Rosa S, Magnani G, Piemonti L, Falini A, Folli F, Secchi A, Fiorina P. Islet transplantation stabilizes hemostatic abnormalities and cerebral metabolism in individuals with type 1 diabetes. Diabetes Care. 2014;37(1):267-276.
- Toso C, Baertschiger R, Morel P, Bosco D, Armanet M, Wojtusciszyn A, Badet L, Philippe J, Becker CD, Hadaya K, Majno P, Buhler L, Berney T. Sequential kidney/islet transplantation: efficacy and safety assessment of a steroid-free immunosuppression protocol. Am J Transplant. 2006;6(5 Pt 1):1049-1058.
- Fiorina P, Folli F, Zerbini G, Maffi P, Gremizzi C, Di Carlo V, Socci C, Bertuzzi F, Kashgarian M, Secchi A. Islet transplantation is associated with improvement of renal function among uremic patients with type I diabetes mellitus and kidney transplants. J Am Soc Nephrol. 2003;14(8):2150-2158.
- Fiorina P, Venturini M, Folli F, Losio C, Maffi P, Placidi C, La Rosa S, Orsenigo E, Socci C, Capella C, Del Maschio A, Secchi A. Natural history of kidney graft survival, hypertrophy, and vascular function in end-stage renal disease type 1 diabetic kidney-transplanted patients: beneficial impact of pancreas and successful islet cotransplantation. Diabetes Care. 2005;28(6):1303-1310.
- Senior PA, Zeman M, Paty BW, Ryan EA, Shapiro AM. Changes in renal function after clinical islet transplantation: four-year observational study. Am J Transplant. 2007;7(1):91-98.
- Gillard P, Rustandi M, Efendi A, Lee DH, Ling Z, Hilbrands R, Kuypers D, Mathieu C, Jacobs-Tulleneers-Thevissen D, Gorus F, Pipeleers D, Keymeulen B. Early Alteration of Kidney Function in Nonuremic Type 1 Diabetic Islet Transplant Recipients Under Tacrolimus-Mycophenolate Therapy. Transplantation. 2014.
- Fung MA, Warnock GL, Ao Z, Keown P, Meloche M, Shapiro RJ, Ho S, Worsley D, Meneilly GS, Al Ghofaili K, Kozak SE, Tong SO, Trinh M, Blackburn L, Kozak RM, Fensom BA, Thompson DM. The effect of medical therapy and islet cell transplantation on diabetic nephropathy: an interim report. Transplantation. 2007;84(1):17-22.
- Fiorina P, Folli F, Bertuzzi F, Maffi P, Finzi G, Venturini M, Socci C, Davalli A, Orsenigo E, Monti L, Falqui L, Uccella S, La Rosa S, Usellini L, Properzi G, Di Carlo V, Del Maschio A, Capella C, Secchi A. Long-term beneficial effect of islet transplantation on diabetic macro-/microangiopathy in type 1 diabetic kidney-transplanted patients. Diabetes Care. 2003;26(4):1129-1136.
- Fiorina P, Folli F, Maffi P, Placidi C, Venturini M, Finzi G, Bertuzzi F, Davalli A, D'Angelo A, Socci C, Gremizzi C, Orsenigo E, La Rosa S, Ponzoni M, Cardillo M, Scalamogna M, Del Maschio A, Capella C, Di Carlo V, Secchi A. Islet transplantation improves vascular diabetic complications in patients with diabetes who underwent kidney transplantation: a comparison between kidney-pancreas and kidney-alone transplantation. Transplantation.2003;75(8):1296-1301.
- Fiorina P, Gremizzi C, Maffi P, Caldara R, Tavano D, Monti L, Socci C, Folli F, Fazio F, Astorri E, Del Maschio A, Secchi A. Islet transplantation is associated with an improvement of cardiovascular function in type 1 diabetic kidney transplant patients. Diabetes Care. 2005;28(6):1358-1365.
- Del Carro U, Fiorina P, Amadio S, De Toni Franceschini L, Petrelli A, Menini S, Boneschi FM, Ferrari S, Pugliese G, Maffi P, Comi G, Secchi A. Evaluation of polyneuropathy markers in type 1 diabetic kidney transplant patients and effects of islet transplantation: neurophysiological and skin biopsy longitudinal analysis. Diabetes Care. 2007;30(12):3063-3069.
- Danielson KK, Hatipoglu B, Kinzer K, Kaplan B, Martellotto J, Qi M, Mele A, Benedetti E, Oberholzer J. Reduction in carotid intima-media thickness after pancreatic islet transplantation in patients with type 1 diabetes. Diabetes Care. 2013;36(2):450-456.
- Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, Lakey JR, Shapiro AM. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54(7):2060-2069.
- Vantyghem MC, Defrance F, Quintin D, Leroy C, Raverdi V, Prevost G, Caiazzo R, Kerr-Conte J, Glowacki F, Hazzan M, Noel C, Pattou F, Diamenord AS, Bresson R, Bourdelle-Hego MF, Cazaubiel M, Cordonnier M, Delefosse D, Dorey F, Fayard A, Fermon C, Fontaine P, Gillot C, Haye S, Le Guillou AC, Karrouz W, Lemaire C, Lepeut M, Leroy R, Mycinski B, Parent E, Siame C, Sterkers A, Torres F, Verier-Mine O, Verlet E, Desailloud R, Durrbach A, Godin M, Lalau JD, Lukas-Croisier C, Thervet E, Toupance O, Reznik Y, Westeel PF. Treating diabetes with islet transplantation: Lessons from the past decade in Lille. Diabetes Metab. 2014;40(2):108-119.
- Bellin MD, Kandaswamy R, Parkey J, Zhang HJ, Liu B, Ihm SH, Ansite JD, Witson J, Bansal-Pakala P, Balamurugan AN, Papas KK, Sutherland DE, Moran A, Hering BJ. Prolonged insulin independence after islet allotransplants in recipients with type 1 diabetes. Am J Transplant. 2008;8(11):2463-2470.
- Maffi P, Scavini M, Socci C, Piemonti L, Caldara R, Gremizzi C, Melzi R, Nano R, Orsenigo E, Venturini M, Staudacher C, Del Maschio A, Secchi A. Risks and benefits of transplantation in the cure of type 1 diabetes: whole pancreas versus islet transplantation. A single center study. Rev Diabet Stud. 2011;8(1):44-50.
- Shapiro AT, C; Imes, S; Koh, A; Kin, T; O'Gorman, D; Malcolm, A; Dinyari, P; Owen, R; Kneteman, RN; Bigam, DL; Calne, RY; Senior, PA; Roep, BO. Five-Year Results of Islet-Alone Transplantation Match Pancreas-Alone Transplantation with Alemtuzumab, Tac/MMF, with Strong Suppression of Auto and Alloreactivity. Paper presented at: 13th World Congressof the International Pancreas and Islet Transplant Association (IPITA)2011; Prague
- Bellin MD, Barton FB, Heitman A, Harmon JV, Kandaswamy R, Balamurugan AN, Sutherland DE, Alejandro R, Hering BJ. Potent induction immunotherapy promotes long-term insulin independence after islet transplantation in type 1 diabetes. Am J Transplant. 2012;12(6):1576-1583.
- Johnson PR, White SA, Robertson GS, Koppiker N, Burden AC, Dennison AR, London NJ. Pancreatic islet autotransplantation combined with total pancreatectomy for the treatment of chronic pancreatitis--the Leicester experience. J Mol Med (Berl). 1999;77(1):130-132.
- Ahmad SA, Lowy AM, Wray CJ, D'Alessio D, Choe KA, James LE, Gelrud A, Matthews JB, Rilo HL. Factors associated with insulin and narcotic independence after islet autotransplantation in patients with severe chronic pancreatitis. J Am Coll Surg. 2005;201(5):680-687.
- Morgan KA, Nishimura M, Uflacker R, Adams DB. Percutaneous transhepatic islet cell autotransplantation after pancreatectomy for chronic pancreatitis: a novel approach. HPB (Oxford). 2011;13(7):511-516.
- Sutherland DE, Gruessner AC, Carlson AM, Blondet JJ, Balamurugan AN, Reigstad KF, Beilman GJ, Bellin MD, Hering BJ. Islet autotransplant outcomes after total pancreatectomy: a contrast to islet allograft outcomes. Transplantation. 2008;86(12):1799-1802.
- Bassi R, Fiorina P. Impact of islet transplantation on diabetes complications and quality of life. Curr Diab Rep.2011;11(5):355-363.
- Hansen A, Johansson BL, Wahren J, von Bibra H. C-peptide exerts beneficial effects on myocardial blood flow and function in patients with type 1 diabetes. Diabetes. 2002;51(10):3077-3082.
- Ryan EA, Paty BW, Senior PA, Shapiro AM. Risks and side effects of islet transplantation. Curr Diab Rep.2004;4(4):304-309.
- Gaba RC, Garcia-Roca R, Oberholzer J. Pancreatic islet cell transplantation: an update for interventional radiologists. J Vasc Interv Radiol. 2012;23(5):583-594; quiz 594.
- Villiger P, Ryan EA, Owen R, O'Kelly K, Oberholzer J, Al Saif F, Kin T, Wang H, Larsen I, Blitz SL, Menon V, Senior P, Bigam DL, Paty B, Kneteman NM, Lakey JR, Shapiro AM. Prevention of bleeding after islet transplantation: lessons learned from a multivariate analysis of 132 cases at a single institution. Am J Transplant. 2005;5(12):2992-2998.
- Froud T, Yrizarry JM, Alejandro R, Ricordi C. Use of D-STAT to prevent bleeding following percutaneous transhepatic intraportal islet transplantation. Cell Transplant. 2004;13(1):55-59.
- Bhargava R, Senior PA, Ackerman TE, Ryan EA, Paty BW, Lakey JR, Shapiro AM. Prevalence of hepatic steatosis after islet transplantation and its relation to graft function. Diabetes. 2004;53(5):1311-1317.
- Maffi P, Angeli E, Bertuzzi F, Paties C, Socci C, Fedeli C, De Taddeo F, Nano R, Di Carlo V, Del Maschio A, Secchi A. Minimal focal steatosis of liver after islet transplantation in humans: a long-term study. Cell Transplant.2005;14(10):727-733.
- Casey JJ, Lakey JR, Ryan EA, Paty BW, Owen R, O'Kelly K, Nanji S, Rajotte RV, Korbutt GS, Bigam D, Kneteman NN, Shapiro AM. Portal venous pressure changes after sequential clinical islet transplantation. Transplantation. 2002;74(7):913-915.
- Molinari M, Al-Saif F, Ryan EA, Lakey JR, Senior PA, Paty BW, Bigam DL, Kneteman NM, Shapiro AM. Sirolimus-induced ulceration of the small bowel in islet transplant recipients: report of two cases. Am J Transplant. 2005;5(11):2799-2804.
- Ponte GM, Baidal DA, Romanelli P, Faradji RN, Poggioli R, Cure P, Froud T, Selvaggi G, Pileggi A, Ricordi C, Alejandro R. Resolution of severe atopic dermatitis after tacrolimus withdrawal. Cell transplantation.2007;16(1):23-30.
- Andres A, Toso C, Morel P, Demuylder-Mischler S, Bosco D, Baertschiger R, Pernin N, Bucher P, Majno PE, Buhler LH, Berney T. Impairment of renal function after islet transplant alone or islet-after-kidney transplantation using a sirolimus/tacrolimus-based immunosuppressive regimen. Transpl Int. 2005;18(11):1226-1230.
- Gala-Lopez BL, Senior PA, Koh A, Kashkoush SM, Kawahara T, Kin T, Humar A, Shapiro AM. Late cytomegalovirus transmission and impact of T-depletion in clinical islet transplantation. Am J Transplant.2011;11(12):2708-2714.
- Pileggi A, Cobianchi L, Inverardi L, Ricordi C. Overcoming the challenges now limiting islet transplantation: a sequential, integrated approach. Ann N Y Acad Sci. 2006;1079:383-398.
- Mineo D, Pileggi A, Alejandro R, Ricordi C. Point: steady progress and current challenges in clinical islet transplantation. Diabetes Care. 2009;32(8):1563-1569.
- Pellegrini S, Cantarelli E, Sordi V, Nano R, Piemonti L. The state of the art of islet transplantation and cell therapy in type 1 diabetes. Acta Diabetol. 2016.
- Lo JF, Wang Y, Li Z, Zhao Z, Hu D, Eddington DT, Oberholzer J. Quantitative and temporal control of oxygen microenvironment at the single islet level. J Vis Exp. 2013(81):e50616.
- Barkai U, Weir GC, Colton CK, Ludwig B, Bornstein SR, Brendel MD, Neufeld T, Bremer C, Leon A, Evron Y, Yavriyants K, Azarov D, Zimermann B, Maimon S, Shabtay N, Balyura M, Rozenshtein T, Vardi P, Bloch K, de Vos P, Rotem A. Enhanced oxygen supply improves islet viability in a new bioartificial pancreas. Cell Transplant.2013;22(8):1463-1476.
- Piemonti L, Andres A, Casey J, de Koning E, Engelse M, Hilbrands R, Johnson P, Keymeulen B, Kerr‐Conte J, Korsgren O. US Food and Drug Administration (FDA) Panel Endorses Islet Cell Treatment for Type 1 Diabetes: A Pyrrhic victory? Transplant International. 2021.
- Hudson A, Bradbury L, Johnson R, Fuggle SV, Shaw JA, Casey JJ, Friend PJ, Watson CJ. The UK Pancreas Allocation Scheme for Whole Organ and Islet Transplantation. Am J Transplant. 2015;15(9):2443-2455.
- Cornateanu SM, O'Neill S, Dholakia S, Counter CJ, Sherif AE, Casey JJ, Friend P, Oniscu GC. Pancreas utilization rates in the UK - an 11-year analysis. Transpl Int. 2021;34(7):1306-1318.
- Nordheim E, Lindahl JP, Carlsen RK, Asberg A, Birkeland KI, Horneland R, Boye B, Scholz H, Jenssen TG. Patient selection for islet or solid organ pancreas transplantation: experiences from a multidisciplinary outpatient-clinic approach. Endocr Connect. 2021;10(2):230-239.
- Nano R, Kerr-Conte JA, Scholz H, Engelse M, Karlsson M, Saudek F, Bosco D, Antonioli B, Bertuzzi F, Johnson PRV, Ludwing B, Ling Z, De Paep DL, Keymeulen B, Pattou F, Berney T, Korsgren O, de Koning E, Piemonti L. Heterogeneity of Human Pancreatic Islet Isolation Around Europe: Results of a Survey Study. Transplantation.2020;104(1):190-196.
- Perrier Q, Lavallard V, Pernin N, Wassmer CH, Cottet-Dumoulin D, Lebreton F, Bellofatto K, Andres A, Berishvili E, Bosco D, Berney T, Parnaud G. Failure mode and effect analysis in human islet isolation: from the theoretical to the practical risk. Islets. 2021;13(1-2):1-9.
- Witkowski P, Philipson LH, Kaufman DB, Ratner LE, Abouljoud MS, Bellin MD, Buse JB, Kandeel F, Stock PG, Mulligan DC, Markmann JF, Kozlowski T, Andreoni KA, Alejandro R, Baidal DA, Hardy MA, Wickrema A, Mirmira RG, Fung J, Becker YT, Josephson MA, Bachul PJ, Pyda JS, Charlton M, Millis JM, Gaglia JL, Stratta RJ, Fridell JA, Niederhaus SV, Forbes RC, Jayant K, Robertson RP, Odorico JS, Levy MF, Harland RC, Abrams PL, Olaitan OK, Kandaswamy R, Wellen JR, Japour AJ, Desai CS, Naziruddin B, Balamurugan AN, Barth RN, Ricordi C. The demise of islet allotransplantation in the United States: A call for an urgent regulatory update. Am J Transplant. 2021;21(4):1365-1375.
- Piemonti L, Andres A, Casey J, de Koning E, Engelse M, Hilbrands R, Johnson P, Keymeulen B, Kerr-Conte J, Korsgren O, Lehmann R, Lundgren T, Maffi P, Pattou F, Saudek F, Shaw J, Scholz H, White S, Berney T. US food and drug administration (FDA) panel endorses islet cell treatment for type 1 diabetes: A pyrrhic victory? Transpl Int. 2021;34(7):1182-1186.
- Arifin DR, Bulte JWM. In Vivo Imaging of Pancreatic Islet Grafts in Diabetes Treatment. Front Endocrinol (Lausanne). 2021;12:640117.
- Murakami T, Fujimoto H, Inagaki N. Non-invasive Beta-cell Imaging: Visualization, Quantification, and Beyond. Front Endocrinol (Lausanne). 2021;12:714348.
- Piemonti L, Everly MJ, Maffi P, Scavini M, Poli F, Nano R, Cardillo M, Melzi R, Mercalli A, Sordi V, Lampasona V, De Arias AE, Scalamogna M, Bosi E, Bonifacio E, Secchi A, Terasaki PI. Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes. 2013;62(5):1656-1664.
- Sabbah S, Liew A, Brooks AM, Kundu R, Reading JL, Flatt A, Counter C, Choudhary P, Forbes S, Rosenthal MJ, Rutter MK, Cairns S, Johnson P, Casey J, Peakman M, Shaw JA, Tree TIM. Autoreactive T cell profiles are altered following allogeneic islet transplantation with alemtuzumab induction and re-emerging phenotype is associated with graft function. Am J Transplant. 2021;21(3):1027-1038.
- Buron F, Reffet S, Badet L, Morelon E, Thaunat O. Immunological Monitoring in Beta Cell Replacement: Towards a Pathophysiology-Guided Implementation of Biomarkers. Curr Diab Rep. 2021;21(6):19.
- Anteby R, Lucander A, Bachul PJ, Pyda J, Grybowski D, Basto L, Generette GS, Perea L, Golab K, Wang LJ, Tibudan M, Thomas C, Fung J, Witkowski P. Evaluating the Prognostic Value of Islet Autoantibody Monitoring in Islet Transplant Recipients with Long-Standing Type 1 Diabetes Mellitus. J Clin Med. 2021;10(12).
- Suwandi JS, Nikolic T, Roep BO. Translating Mechanism of Regulatory Action of Tolerogenic Dendritic Cells to Monitoring Endpoints in Clinical Trials. Front Immunol. 2017;8:1598.
- Tremmel DM, Odorico JS. Rebuilding a better home for transplanted islets. Organogenesis. 2018;14(4):163-168.
- Citro A, Moser PT, Dugnani E, Rajab TK, Ren X, Evangelista-Leite D, Charest JM, Peloso A, Podesser BK, Manenti F, Pellegrini S, Piemonti L, Ott HC. Biofabrication of a vascularized islet organ for type 1 diabetes. Biomaterials. 2019;199:40-51.
- Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater. 2021.
- Basta G, Montanucci P, Calafiore R. Microencapsulation of cells and molecular therapy of type 1 diabetes mellitus: The actual state and future perspectives between promise and progress. J Diabetes Investig.2021;12(3):301-309.
- Marigliano M, Bertera S, Grupillo M, Trucco M, Bottino R. Pig-to-nonhuman primates pancreatic islet xenotransplantation: an overview. Curr Diab Rep. 2011;11(5):402-412.
- Pagliuca FW, Millman JR, Gurtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of Functional Human Pancreatic beta Cells In Vitro. Cell. 2014;159(2):428-439.
- Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O'Dwyer S, Quiskamp N, Mojibian M, Albrecht T, Yang YH, Johnson JD, Kieffer TJ. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121-1133.
- Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D'Amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol.2008;26(4):443-452.
- Schulz TC, Young HY, Agulnick AD, Babin MJ, Baetge EE, Bang AG, Bhoumik A, Cepa I, Cesario RM, Haakmeester C, Kadoya K, Kelly JR, Kerr J, Martinson LA, McLean AB, Moorman MA, Payne JK, Richardson M, Ross KG, Sherrer ES, Song X, Wilson AZ, Brandon EP, Green CE, Kroon EJ, Kelly OG, D'Amour KA, Robins AJ. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS One. 2012;7(5):e37004.
- Pellegrini S, Piemonti L, Sordi V. Pluripotent stem cell replacement approaches to treat type 1 diabetes. Curr Opin Pharmacol. 2018;43:20-26.
- Odorico J, Markmann J, Melton D, Greenstein J, Hwa A, Nostro C, Rezania A, Oberholzer J, Pipeleers D, Yang L, Cowan C, Huangfu D, Egli D, Ben-David U, Vallier L, Grey ST, Tang Q, Roep B, Ricordi C, Naji A, Orlando G, Anderson DG, Poznansky M, Ludwig B, Tomei A, Greiner DL, Graham M, Carpenter M, Migliaccio G, D'Amour K, Hering B, Piemonti L, Berney T, Rickels M, Kay T, Adams A. Report of the Key Opinion Leaders Meeting on Stem Cell-derived Beta Cells. Transplantation. 2018;102(8):1223-1229.
- Carlsson P-O, Schwarcz E, Korsgren O, Le Blanc K. Preserved β-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes. 2015;64(2):587-592.
- Madani S, Setudeh A, Aghayan HR, Alavi-Moghadam S, Rouhifard M, Rezaei N, Rostami P, Mohsenipour R, Amirkashani D, Bandarian F. Placenta derived Mesenchymal Stem Cells transplantation in Type 1 diabetes: preliminary report of phase 1 clinical trial. Journal of Diabetes & Metabolic Disorders. 2021;20(2):1179-1189.
- Arzouni AA, Vargas-Seymour A, Nardi N, JF King A, Jones PM. Using mesenchymal stromal cells in islet transplantation. Stem Cells Translational Medicine. 2018;7(8):559-563.
- Pellegrini S, Piemonti L, Sordi V. Pluripotent stem cell replacement approaches to treat type 1 diabetes. Current Opinion in Pharmacology. 2018;43:20-26.
- Migliorini A, Nostro MC, Sneddon JB. Human pluripotent stem cell-derived insulin-producing cells: A regenerative medicine perspective. Cell Metabolism. 2021;33(4):721-731.
- D'Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter MK, Baetge EE. Production of pancreatic hormone–expressing endocrine cells from human embryonic stem cells. Nature biotechnology. 2006;24(11):1392-1401.
- Zhang D, Jiang W, Liu M, Sui X, Yin X, Chen S, Shi Y, Deng H. Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell research. 2009;19(4):429-438.
- Tateishi K, He J, Taranova O, Liang G, D'Alessio AC, Zhang Y. Generation of insulin-secreting islet-like clusters from human skin fibroblasts. Journal of Biological Chemistry. 2008;283(46):31601-31607.
- Nostro MC, Sarangi F, Ogawa S, Holtzinger A, Corneo B, Li X, Micallef SJ, Park I-H, Basford C, Wheeler MB. Stage-specific signaling through TGFβ family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development. 2011;138(5):861-871.
- Hrvatin S, O’Donnell CW, Deng F, Millman JR, Pagliuca FW, DiIorio P, Rezania A, Gifford DK, Melton DA. Differentiated human stem cells resemble fetal, not adult, β cells. Proceedings of the National Academy of Sciences. 2014;111(8):3038-3043.
- Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D, Melton DA. Generation of functional human pancreatic β cells in vitro. Cell. 2014;159(2):428-439.
- Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, O'dwyer S, Quiskamp N, Mojibian M, Albrecht T. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nature biotechnology. 2014;32(11):1121.
- Russ HA, Parent AV, Ringler JJ, Hennings TG, Nair GG, Shveygert M, Guo T, Puri S, Haataja L, Cirulli V. Controlled induction of human pancreatic progenitors produces functional beta‐like cells in vitro. The EMBO journal. 2015;34(13):1759-1772.
- Sambathkumar R, Migliorini A, Nostro MC. Pluripotent stem cell-derived pancreatic progenitors and β-Like cells for Type 1 diabetes treatment. Physiology. 2018;33(6):394-402.
- Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nature biotechnology. 2008;26(4):443-452.
- Kelly OG, Chan MY, Martinson LA, Kadoya K, Ostertag TM, Ross KG, Richardson M, Carpenter MK, D'amour KA, Kroon E. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nature biotechnology. 2011;29(8):750-756.
- Ramzy A, Thompson DM, Ward-Hartstonge KA, Ivison S, Cook L, Garcia RV, Loyal J, Kim PTW, Warnock GL, Levings MK, Kieffer TJ. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell. 2021;28(12):2047-2061.e2045.
- Sordi V, Pellegrini S, Piemonti L. Immunological Issues After Stem Cell-Based beta Cell Replacement. Curr Diab Rep. 2017;17(9):68.
- Zheng D, Wang X, Xu RH. Concise review: one stone for multiple birds: generating universally compatible human embryonic stem cells. Stem Cells. 2016;34(9):2269-2275.
- Barry J, Hyllner J, Stacey G, Taylor CJ, Turner M. Setting up a haplobank: issues and solutions. Current stem cell reports. 2015;1(2):110-117.
- Borot S, Crowe LA, Toso C, Vallee JP, Berney T. Noninvasive imaging techniques in islet transplantation. Curr Diab Rep. 2011;11(5):375-383.
- Wang P, Schuetz C, Vallabhajosyula P, Medarova Z, Tena A, Wei L, Yamada K, Deng S, Markmann JF, Sachs DH, Moore A. Monitoring of Allogeneic Islet Grafts in Nonhuman Primates Using MRI. Transplantation.2015;99(8):1574-1581.
- Malosio ML, Esposito A, Brigatti C, Palmisano A, Piemonti L, Nano R, Maffi P, De Cobelli F, Del Maschio A, Secchi A. Mr Imaging Monitoring of Iron Labeled Pancreatic Islets in a Small Series of Patients: Islets Fate in Successful, Unsuccessful and Auto-Transplantation. Cell Transplant. 2014.
- Esposito A, Palmisano A, Maffi P, Malosio ML, Nano R, Canu T, De Cobelli F, Piemonti L, Ironi G, Secchi A, Del Maschio A. Liver perfusion changes occurring during pancreatic islet engraftment: a dynamic contrast-enhanced magnetic resonance study. Am J Transplant. 2014;14(1):202-209.
- Saudek F, Jirak D, Girman P, Herynek V, Dezortova M, Kriz J, Peregrin J, Berkova Z, Zacharovova K, Hajek M. Magnetic resonance imaging of pancreatic islets transplanted into the liver in humans. Transplantation.2010;90(12):1602-1606.
- Toso C, Vallee JP, Morel P, Ris F, Demuylder-Mischler S, Lepetit-Coiffe M, Marangon N, Saudek F, James Shapiro AM, Bosco D, Berney T. Clinical magnetic resonance imaging of pancreatic islet grafts after iron nanoparticle labeling. Am J Transplant. 2008;8(3):701-706.
- Toso C, Zaidi H, Morel P, Armanet M, Andres A, Pernin N, Baertschiger R, Slosman D, Buhler LH, Bosco D, Berney T. Positron-emission tomography imaging of early events after transplantation of islets of Langerhans. Transplantation. 2005;79(3):353-355.
- Eriksson O, Alavi A. Imaging the islet graft by positron emission tomography. Eur J Nucl Med Mol Imaging.2012;39(3):533-542.
- Kriz J, Jirak D, Berkova Z, Herynek V, Lodererova A, Girman P, Habart D, Hajek M, Saudek F. Detection of pancreatic islet allograft impairment in advance of functional failure using magnetic resonance imaging. Transpl Int. 2012;25(2):250-260.
- Eich T, Eriksson O, Lundgren T. Visualization of early engraftment in clinical islet transplantation by positron-emission tomography. N Engl J Med. 2007;356(26):2754-2755.
- Han D, Leith J, Alejandro R, Bolton W, Ricordi C, Kenyon NS. Peripheral blood cytotoxic lymphocyte gene transcript levels differ in patients with long-term type 1 diabetes compared to normal controls. Cell Transplant.2005;14(6):403-409.
- Han D, Xu X, Baidal D, Leith J, Ricordi C, Alejandro R, Kenyon NS. Assessment of cytotoxic lymphocyte gene expression in the peripheral blood of human islet allograft recipients: elevation precedes clinical evidence of rejection. Diabetes. 2004;53(9):2281-2290.
- Han D, Xu X, Pastori RL, Ricordi C, Kenyon NS. Elevation of cytotoxic lymphocyte gene expression is predictive of islet allograft rejection in nonhuman primates. Diabetes. 2002;51(3):562-566.
- Huurman VA, Velthuis JH, Hilbrands R, Tree TI, Gillard P, van der Meer-Prins PM, Duinkerken G, Pinkse GG, Keymeulen B, Roelen DL, Claas FH, Pipeleers DG, Roep BO. Allograft-specific cytokine profiles associate with clinical outcome after islet cell transplantation. Am J Transplant. 2009;9(2):382-388.
- Roep BO, Stobbe I, Duinkerken G, van Rood JJ, Lernmark A, Keymeulen B, Pipeleers D, Claas FH, de Vries RR. Auto- and alloimmune reactivity to human islet allografts transplanted into type 1 diabetic patients. Diabetes. 1999;48(3):484-490.
- Stobbe I, Duinkerken G, van Rood JJ, Lernmark A, Keymeulen B, Pipeleers D, De Vries RR, Glass FH, Roep BO. Tolerance to kidney allograft transplanted into Type I diabetic patients persists after in vivo challenge with pancreatic islet allografts that express repeated mismatches. Diabetologia. 1999;42(11):1379-1380.
- Hallé JP, deVos P, Rosenberg L. The bioartificial pancreas and other biohybrid therapies. Kerala, India: Transworld Research Network.
- Basta G, Calafiore R. Immunoisolation of pancreatic islet grafts with no recipient's immunosuppression: actual and future perspectives. Curr Diab Rep. 2011;11(5):384-391.