Abstract

Pancreatic islet transplantation has become an established approach to β-cell replacement therapy for the treatment of insulin-deficient diabetes. Recent progress in techniques for islet isolation, islet culture, and peritransplant management of the islet transplant recipient has resulted in substantial improvements in metabolic and safety outcomes for patients. For patients requiring total or subtotal pancreatectomy for benign disease of the pancreas, isolation of islets from the diseased pancreas with intrahepatic transplantation of autologous islets can prevent or ameliorate postsurgical diabetes, and for patients previously experiencing painful recurrent acute or chronic pancreatitis, quality of life is substantially improved. For patients with type 1 diabetes or insulin-deficient forms of pancreatogenic (type 3c) diabetes, isolation of islets from a deceased donor pancreas with intrahepatic transplantation of allogeneic islets can ameliorate problematic hypoglycemia, stabilize glycemic lability, and maintain on-target glycemic control, consequently with improved quality of life, and often without the requirement for insulin therapy. Because the metabolic benefits are dependent on the numbers of islets transplanted that survive engraftment, recipients of autoislets are limited to receive the number of islets isolated from their own pancreas, whereas recipients of alloislets may receive islets isolated from more than one donor pancreas. The development of alternative sources of islet cells for transplantation, whether from autologous, allogeneic, or xenogeneic tissues, is an active area of investigation that promises to expand access and indications for islet transplantation in the future treatment of diabetes.

Essential Points
  • Islet autotransplantation following total pancreatectomy involves complete resection of the pancreas, most commonly in the setting of recurrent acute or chronic pancreatitis, with islets isolated from the diseased pancreas returned to the patient via an intraportal infusion for intrahepatic engraftment

  • Islet allotransplantation involves procurement of a deceased donor pancreas, most commonly for a recipient with type 1 diabetes, with islets isolated from the donated pancreas returned to the patient via an intraportal infusion for intrahepatic engraftment

  • Care of the islet recipient requires attention to reduce inflammation and minimize thrombotic and bleeding risks while maintaining normoglycemia that is critical for islet survival until revascularization and engraftment are complete

  • Autoislet recipients benefit from amelioration of pain, improved quality of life, on-target glycemic control, and in about a third of patients insulin independence; some patients may experience postprandial hypoglycemia

  • Alloislet recipients benefit from amelioration of hypoglycemia, on-target glycemic control, improved quality of life, and in about half of patients insulin independence; islets from more than one donor pancreas are often required to achieve or maintain insulin independence

  • Metabolic outcomes for both autoislet and alloislet recipients are dependent on the numbers of islets transplanted and the functional islet β-cell mass surviving engraftment

  • Novel approaches are under evaluation to further reduce the detrimental consequences of peritransplant inflammation on intrahepatic islet engraftment efficiency and, for alloislets, calcineurin inhibitor–free immunosuppression that may have less potential for adverse effects on kidney function

Islet transplantation has been a long sought-after solution for reestablishing glucose homeostasis in diabetes caused by islet β-cell loss via the autoimmune attack of type 1 diabetes or from surgical resection or pancreatic fibrosis in pancreatogenic (type 3c) forms of diabetes. Initial work in rat models of diabetes published in the early 1970s largely from the laboratories of Paul Lacy at Washington University (1) and Clyde Barker at the University of Pennsylvania (2) established the efficacy of islet transplantation in normalizing glycemia in both syngeneic and allogeneic settings, and such work pointed toward the liver as the most efficient site of engraftment following portal vein infusion (3). Following validation of intrahepatic autologous islet transplantation for the prevention of surgical diabetes following pancreatectomy in dogs (4), John Najarian and David Sutherland at the University of Minnesota published in 1980 the first patient series of total pancreatectomy with islet autotransplantation for the treatment of chronic pancreatitis, demonstrating in some cases prevention or amelioration of postpancreatectomy diabetes (5). This proof of principle for the use of the liver for transplantation of isolated autoislets established the possibility for use of alloislets for treatment of humans with type 1 diabetes.

Efforts to enhance the purity and viability of isolated islets for transplantation by a number of groups (6, 7) led to the publication in 1988 of an automated method for isolation of human pancreatic islets by Camillo Ricordi (8) that was applied by David Scharp and Paul Lacy to the first case of allogeneic islet transplantation for type 1 diabetes resulting in a short period of insulin independence and published in 1990 (9). However, it was not until 2000 that the Edmonton group, led by James Shapiro, reported consistent achievement of insulin independence in seven out of seven consecutive patients with type 1 diabetes who received islets isolated from an average of two donor pancreases (10). This established the need for transplantation of a sufficient islet β-cell mass to attain insulin independence and near-normal glycemic control in humans with type 1 diabetes. It also required the avoidance of glucocorticoid use in the immunosuppression regimen to avoid islet toxicity and insulin resistance.

During the time since publication of the Edmonton protocol, many countries internationally have advanced allogeneic islet transplantation to the treatment of type 1 diabetes patients experiencing severe hypoglycemia and hypoglycemia unawareness or marked glycemic lability (11), or those already committed to chronic immunosuppression in support of a kidney transplant (12–14). Meanwhile, efforts in the United States have focused on improving the efficiency of both islet isolation and posttransplant islet engraftment to minimize the number of donor pancreases required to stabilize metabolic control and, when possible, eliminate the requirement for insulin therapy (15–17). In 2005 the Minnesota group led by Bernard Hering introduced several modifications to peritransplantation management and reported consistent achievement of insulin independence in eight out of eight consecutive patients with type 1 diabetes who received islets isolated from a single donor pancreas (18). This enhanced efficiency for allogeneic islet transplantation has been further evaluated by a phase 3 multicenter licensure trial conducted by the National Institutes of Health–sponsored Clinical Islet Transplantation Consortium and published in 2016 (19). Importantly, this trial selected patients with type 1 diabetes experiencing severe hypoglycemia accompanied by hypoglycemia unawareness or marked glycemic lability and achieved an HbA1c ≤6.5% in the absence of severe hypoglycemia, and thereby met the US Food and Drug Administration’s primary outcome of achieving an HbA1c <7.0% in the absence of severe hypoglycemia. A subsequent trial in Europe confirmed these benefits in comparison with intensive insulin therapy using a randomized controlled design (20).

This review is focused on updating our current knowledge about human islet transplantation, including the present clinical indications and outcomes for both islet autotransplantation and allotransplantation, as well as considering directions for future research. Less extensive information is included about successful pancreas transplantation, which was reviewed by Jennifer Larsen in Endocrine Reviews in 2004 (21), but nonetheless remains the most appropriate comparator for assessing the degree of success for islet transplantation. Lastly, novel sources of islet tissue for transplantation are considered, as well as the requirements they must meet to join pancreas and islet transplantation as approaches to β-cell replacement therapy in the treatment of diabetes.

Procedures for Islet Transplantation

Islet autotransplantation following total pancreatectomy

Islets transplanted in patients after undergoing total pancreatectomy and islet autotransplantation (TPIAT) are autologous, and as such, are not considered a manufactured biologic product by the US Food and Drug Administration. Thus, the practice of patients receiving their own islets requires processing under current good tissue practice (cGTP), but not the more stringent current good manufacturing practice (cGMP), regulations governing the processing of allogeneic islets for transplantation. Because a patient receives only those islets that can be isolated from their own pancreas, the approach to processing aims to maximize islet yield rather than islet purity.

The standard approach involves complete resection of the pancreas, most commonly in the setting of recurrent acute or chronic pancreatitis, together with the duodenum and the spleen, which both share their arterial blood supply with the pancreas (Fig. 1). When technically feasible, some centers will attempt to preserve the spleen in situ (22). The spleen may be preserved during total pancreatectomy either by separating the splenic artery and vein from the pancreas or by maintaining the short gastric vessels. The first approach affects in situ perfusion of the pancreas and is not recommended when islets will be isolated, whereas the second approach may lead to sinusoidal hypertension and the development of gastric varices. While gastrojejunostomy and choledochojejunostomy Roux-en-Y reconstruction is performed, the pancreas is brought to a cGTP, or where available cGMP, islet isolation facility where it is digested using a combination of chemical [collagenase and neutral protease blend (23, 24)] and mechanical (Ricordi chamber) methods, and it undergoes variable centrifuge purification that separates the acinar and ductal tissue from the islets (25, 26). Purification results in loss of some islets, so preparations remain rather impure to maximize islet recovery while minimizing by centrifugation the transplanted tissue volume. The final islet product is transplanted into the liver by infusing it intraportally via the splenic vein stump, a mesenteric vein, or recannulation of the umbilical vein (the latter directs islets only into the left portal vein). If excessive portal pressure is caused by a high tissue volume, a portion of the islet product may be placed in the peritoneal cavity or other nonhepatic sites. From the portal venous circulation the islets distribute throughout the liver wherein the islets lodge in the hepatic sinusoids that provide a natural bio-scaffold important to promote cell survival (27).

Procedure for total pancreatectomy with islet autotransplantation. The diseased pancreas is resected during total pancreatectomy together with the duodenum and the spleen, which both share their arterial blood supply with the pancreas. While gastrojejunostomy and choledochojejunostomy Roux-en-Y reconstruction is performed, the pancreas is brought to a cGMP or cGTP islet isolation facility where it is digested by collagenases and undergoes variable centrifuge purification that separates the acinar from the endocrine cells with the goal of maximizing islet recovery while minimizing the transplanted tissue volume. The final islet product is transplanted intraportally via the splenic vein stump, a mesenteric vein, or recannulation of the umbilical vein. If excessive portal pressure is caused by a high tissue volume, a portion of the islet product may be placed in the peritoneal cavity or other nonhepatic site.
Figure 1.

Procedure for total pancreatectomy with islet autotransplantation. The diseased pancreas is resected during total pancreatectomy together with the duodenum and the spleen, which both share their arterial blood supply with the pancreas. While gastrojejunostomy and choledochojejunostomy Roux-en-Y reconstruction is performed, the pancreas is brought to a cGMP or cGTP islet isolation facility where it is digested by collagenases and undergoes variable centrifuge purification that separates the acinar from the endocrine cells with the goal of maximizing islet recovery while minimizing the transplanted tissue volume. The final islet product is transplanted intraportally via the splenic vein stump, a mesenteric vein, or recannulation of the umbilical vein. If excessive portal pressure is caused by a high tissue volume, a portion of the islet product may be placed in the peritoneal cavity or other nonhepatic site.

Because the source of islets is a diseased pancreas, several important challenges arise in optimizing islet yield and minimizing risk due to impure or contaminated preparations. Effective delivery of a collagenase enzyme blend to the pancreatic parenchyma is best achieved by transection of the midportion of the gland and cannulation of each half of the main pancreatic duct toward the pancreatic head and tail, respectively, for enzyme instillation. Dilated ducts require larger gauge catheters to prevent leakage of the collagenase solution during perfusion (26). This approach is markedly compromised by prior pancreatic surgery involving drainage procedures that open the main pancreatic duct along its length, and by extensive intraductal calcification that may develop in the course of chronic pancreatitis. Without effective delivery of collagenase enzymes for pancreas digestion, islets cannot be liberated from the exocrine compartment. Additionally, prior pancreatic surgery involving resection of pancreatic tissue, and especially distal pancreatectomy, affects the starting material available for digestion and may further compromise the potential islet yield following processing (28, 29). Finally, sterility of the islet product is often compromised by prior endoscopic procedures that breach the integrity of the sphincter of Oddi, including endoscopic retrograde cholangiopancreatography, sphincterotomy, and intraductal stenting, which allows access and colonization of small intestinal bacterial in the pancreatic ductal tree (26). Unlike with islet allotransplantaton, concern over bacterial contamination and a desire to complete the islet infusion during the same operation as the pancreatectomy has prevented more formal evaluation of islet culture prior to islet autotransplantation.

Given the complexities involved in isolating islets for autologous transplantation, and the relative rarity of an indication for TPIAT, there are far fewer clinical facilities capable of offering high-quality islet processing than pancreatic surgery. To provide islet autotransplantation to patients requiring total pancreatectomy closer to their homes, a few pancreatic surgery programs have partnered with regional or distant institutions offering islet isolation (30–32). The largest reported series (32) included 36 consecutive patients using a remote islet isolation laboratory with the time from pancreatectomy to receipt of islets for transplantation limited to ~8 to 9 hours, and it demonstrated similar clinical outcomes as prior reports using on-site islet processing (25, 33–36) (Table 1). This demonstrated feasibility of remote islet processing supports that lack of an on-site islet isolation facility should not discourage the use of islet autotransplantation for the prevention or amelioration of postsurgical diabetes in patients requiring pancreatectomy (37).

Table 1.

Comparison of Published High-Volume Center Experiences With TPIAT

YearsUniversity of Minnesota (25)
University of Cincinnati (33, 34)
Leicester General Hospital (35, 36)
Cleveland Clinic(32)
1997–20072000–20041994–20062008–2014
Number of patients173455036
Time until islet infusion, h2–73–42–48–9
Median islet yield, IE/kg30544933a22454308
HbA1c, 1-y median, %NRbNR7.0NR
HbA1c, 2-y median, %NRbNR6.76.8
Insulin independence, 1-y median, %32NR26NR
Insulin independence, 2-y median, %24402433
YearsUniversity of Minnesota (25)
University of Cincinnati (33, 34)
Leicester General Hospital (35, 36)
Cleveland Clinic(32)
1997–20072000–20041994–20062008–2014
Number of patients173455036
Time until islet infusion, h2–73–42–48–9
Median islet yield, IE/kg30544933a22454308
HbA1c, 1-y median, %NRbNR7.0NR
HbA1c, 2-y median, %NRbNR6.76.8
Insulin independence, 1-y median, %32NR26NR
Insulin independence, 2-y median, %24402433

[Adapted with permission from Johnston PC, Lin YK, Walsh RM, et al. Factors associated with islet yield and insulin independence after total pancreatectomy and islet cell autotransplantation in patients with chronic pancreatitis utilizing off-site islet isolation: Cleveland Clinic experience. J Clin Endocrinol Metab 2015;100:1765-1770. Illustration presentation copyright by the Endocrine Society.]

Abbreviations: IE/kg, IE/kg recipient body weight, whereby an IE approximates a standard islet diameter of 150 μm; NR, not reported.

a

Mean reported (34).

b

A subgroup (n = 8) in whom insulin independence was tested all had HbA1c <7.0% (25).

Table 1.

Comparison of Published High-Volume Center Experiences With TPIAT

YearsUniversity of Minnesota (25)
University of Cincinnati (33, 34)
Leicester General Hospital (35, 36)
Cleveland Clinic(32)
1997–20072000–20041994–20062008–2014
Number of patients173455036
Time until islet infusion, h2–73–42–48–9
Median islet yield, IE/kg30544933a22454308
HbA1c, 1-y median, %NRbNR7.0NR
HbA1c, 2-y median, %NRbNR6.76.8
Insulin independence, 1-y median, %32NR26NR
Insulin independence, 2-y median, %24402433
YearsUniversity of Minnesota (25)
University of Cincinnati (33, 34)
Leicester General Hospital (35, 36)
Cleveland Clinic(32)
1997–20072000–20041994–20062008–2014
Number of patients173455036
Time until islet infusion, h2–73–42–48–9
Median islet yield, IE/kg30544933a22454308
HbA1c, 1-y median, %NRbNR7.0NR
HbA1c, 2-y median, %NRbNR6.76.8
Insulin independence, 1-y median, %32NR26NR
Insulin independence, 2-y median, %24402433

[Adapted with permission from Johnston PC, Lin YK, Walsh RM, et al. Factors associated with islet yield and insulin independence after total pancreatectomy and islet cell autotransplantation in patients with chronic pancreatitis utilizing off-site islet isolation: Cleveland Clinic experience. J Clin Endocrinol Metab 2015;100:1765-1770. Illustration presentation copyright by the Endocrine Society.]

Abbreviations: IE/kg, IE/kg recipient body weight, whereby an IE approximates a standard islet diameter of 150 μm; NR, not reported.

a

Mean reported (34).

b

A subgroup (n = 8) in whom insulin independence was tested all had HbA1c <7.0% (25).

Islet engraftment in the liver has been essential for the success of clinical islet transplantation to date. As a site, the liver provides transplanted islets with oxygenation via the portal circulation until revascularization by the hepatic arterial system occurs (38, 39). With revascularization there is also evidence for reinnervation of intrahepatic islets by the sympathetic nervous system in rodent models that is important for modulating islet activity and hormone secretion (40). Additionally, insulin and glucagon secreted from islets normally enter the portal circulation and are ~50% extracted by the liver where insulin suppresses and glucagon activates hepatic glucose production. The insulin delivered to the liver is secreted in coordinate pulses, the amplitude of which contributes to insulin action on the liver, and is directly dependent on the functional islet β-cell mass. When transplanted in the liver via portal vein infusion, human islets reestablish coordinated pulsatile secretion of insulin, and although the islets ultimately drain into the hepatic venous circulation, direct hepatic vein catheterization studies in two autoislet recipients indicated normal first-pass hepatic extraction, supporting that intrahepatic islets deliver insulin directly to the hepatic sinusoids (41). Indeed, the liver is the site deemed most efficacious in providing survival and function of transplanted islets to consistently reverse diabetes and achieve insulin independence in large animal models (42). Maximizing islet survival until engraftment is particularly crucial for autologous transplantation when there is no additional source of islet tissue.

As already mentioned, there is a trade-off with islet purification in that the quantity of islets recovered decreases with further purification. Thus, for the sake of maximizing islet recovery for transplantation, preparations of islets for autologous transplantation have lower purity than do preparations for allogenic transplantation, and the higher tissue volume associated with impure islet preparations increases the risk for portal hypertension and portal vein thrombosis, complicating intraportal islet delivery to the intrahepatic site. Purification should be used to reduce the final tissue volume for transplantation to <0.2 to 0.25 mL/kg recipient body weight up to a maximum of 15 to 20 mL (25, 43–45). For higher tissue volumes, or when excessive portal pressure develops during intraportal islet infusion (>25 cm H2O) (45), a portion of the islet product may be placed in the peritoneal cavity or other nonhepatic site (29). Alternative sites for islet transplantation should only be used judiciously when it is unsafe to deliver the entire preparation to the liver given the limited experience and uncertain efficacy with nonhepatic islet transplantation. Attempts at placing the entire islet product in a nonhepatic site have rarely been attempted and to date have proved less successful. In three adult patients receiving autoislets in the brachioradialis muscle of the forearm, none maintained posttransplantation HbA1c <7.0% despite evidence of stimulated C-peptide production (46), whereas the same approach in a 7-year-old child did normalize glycemia and support growth with the provision of exogenous insulin during 2 years of reported follow-up (47). The intramuscular site has not been able to support the survival of alloislets (48). Four patients with contraindication to intraportal islet infusion underwent transplantation of autoislets to the bone marrow of the iliac crest that resulted in very marginal graft function (49). The bone marrow site is also expected to be more immunogenic than the liver for alloislets (50). The peritoneal cavity has been successful using the entire islet preparation in dogs (51) and, as mentioned above, this site is used in humans when excess islets remain after using the liver site (29, 52).

Islet allotransplantation for the treatment of type 1 diabetes

Islets transplanted in patients with type 1 diabetes are allogeneic, and as such are regulated as a manufactured biologic product by the US Food and Drug Administration, with islet production requiring cGMP quality standards. In other countries such as Canada, Australia, and several in Europe with national health services, islet transplantation is available as standard of care for selected patients (see “Indications for Islet Transplantation” below). Importantly, the selection of a donor pancreas for islet isolation must adhere to ABO blood group antigen compatibility, avoid any unacceptable human leukocyte antigens (HLAs) that could react with preformed anti-HLA antibodies present in the recipient, and pass a T lymphocyte and B lymphocyte crossmatch between donor cells and recipient blood. Preexisting immunity against HLAs develops from previous exposure to nonself HLAs as occurs with pregnancy (the father’s HLA haplotype expressed by the fetus), red blood cell transfusions (that are ABO but not HLA matched), solid organ transplantation (e.g., prior kidney transplantation for diabetic nephropathy), human bone and tendon graphs commonly used in orthopedics, and human heart valves and ventricular assist devices sometimes required in cardiac surgery. Thus, close communication with both the organ procurement organization and institution tissue typing laboratory is essential for minimizing the risk of antibody–antigen recognition leading to acute rejection of the allogeneic product.

The standard approach involves procurement of a pancreas from a deceased donor without diabetes (Fig. 2). Procurement, preservation, and transport of the deceased donor pancreas for islet transplantation requires the same care and speed as that for a pancreas used for whole organ transplantation (53), with cold ischemia time kept to a minimum (<8 hours) (17). Although younger donors (<40 to 45 years of age) provide higher quality pancreases and islets (54, 55), and so may be optimal for both whole organ and isolated islet transplantation (56), fatty pancreases common in our increasingly overweight and obese population carry an increased risk for technical failure following whole pancreas transplantation. At the same time, organs procured from higher body mass index donors lend to higher islet yields following isolation (57), and so islet transplantation can make use of high-quality organs that would otherwise be discarded (58) and more often yield sufficient numbers of islets important for posttransplant outcomes (59). The successful use of nonheart-beating donors for clinical islet transplantation has been reported (60, 61), but such donation after circulatory death donors are infrequently used as a source of islets outside Japan where (heart-beating) brain death donation is extremely limited. Recent guidelines have been proposed on the procurement of pancreases from donation after circulatory death donors for use in transplantation, including a limit on warm ischemia time (<30 to 60 minutes) (62), but no comparative data on clinical outcomes with the standard use of brain death donors are available.

Procedure for islet allotransplantation. A pancreas is procured from a deceased donor without diabetes and brought to a cGMP islet isolation facility where it is digested by collagenases and purified by centrifugation with the final islet product placed in culture for up to 72 h prior to intraportal delivery in a recipient with type 1 diabetes. Shown here is percutaneous, transhepatic access to the main portal vein via a right portal branch vein identified under ultrasound and fluoroscopic guidance. Alternatively, access to the main portal vein can be achieved via a mesenteric vein identified by direct visualization during minilaparotomy.
Figure 2.

Procedure for islet allotransplantation. A pancreas is procured from a deceased donor without diabetes and brought to a cGMP islet isolation facility where it is digested by collagenases and purified by centrifugation with the final islet product placed in culture for up to 72 h prior to intraportal delivery in a recipient with type 1 diabetes. Shown here is percutaneous, transhepatic access to the main portal vein via a right portal branch vein identified under ultrasound and fluoroscopic guidance. Alternatively, access to the main portal vein can be achieved via a mesenteric vein identified by direct visualization during minilaparotomy.

Once brought to the cGMP islet isolation facility, the deceased donor pancreas is digested by intraductal collagenase delivery (23, 24), mechanically separated using the Ricordi method (8), and purified by centrifugation with the final islet product placed in culture for 36 to 72 hours prior to intraportal delivery in a recipient with type 1 diabetes (63) (Fig. 2). Unlike the autoislet procedure, a period of islet culture may be particularly helpful for allogeneic transplantation because islet culture has been shown to decrease the number of passenger leukocytes and islet expression of class I HLA (64), as well as islet production of CCL2/MCP-1 and tissue factor (65–68), and so may result in a less immunogenic and inflammatory graft. Although direct comparisons of clinical outcomes from islet preparations with or without culture are not available, improved outcomes have been associated with the adoption of islet culture in the Collaborative Islet Transplant Registry (69), and a period of islet culture allows for the induction of immunosuppression in the recipient before exposure to allogeneic tissue. Access to the main portal vein for islet infusion can be achieved either via percutaneous, transhepatic access of a portal branch vein identified under ultrasound and fluoroscopic guidance (70, 71), or via a mesenteric vein identified by direct visualization during minilaparotomy (72).

Similar to the use of regional islet isolation facilities to facilitate access to autologous islet transplantation, several regional networks have developed in support of providing allogeneic islet transplantation for type 1 diabetes (58, 73–76). This approach not only brings the treatment closer to where a patient might live, but also takes advantage of the procurement of organs by teams outside the procurement area of the isolation facility who are committed to high-quality recovery and rapid transport of pancreata to the processing institution and appropriate preparation of the recipient while awaiting the return of the purified human pancreatic islet product for transplantation. Similar clinical outcomes have been reported for recipients of islets processed on-site or off-site following the same study protocol (58, 76).

In addition to the liver as a transplant site providing islets with oxygenation via the portal circulation until revascularization by the hepatic arterial systems occurs (38, 39), also important for allogeneic islet transplantation where islets from more than one donor pancreas may be transplanted is the capacity of the liver to accommodate sequential infusion of islets from three or even more donor pancreases (77). Nonetheless, exposure to more donor pancreases increases the risk of sensitization (development of alloantibody) to HLAs, and so an optimal approach for the patient with type 1 diabetes minimizes the number of donors required to achieve metabolic benefit. The major limitation with the liver as a site for allogeneic islet transplantation is the inability for biopsy to reliably include islet tissue necessary for pathologic evaluation (78), and so precludes accurate attribution of the cause for islet graft dysfunction or failure. The only report of successful islet engraftment outside of the liver is a case using an omental pocket created with a bioartificial thrombin scaffold (79). Although this approach may benefit patients with liver disease precluding intrahepatic transplantation, currently its use is limited to one time therapy and is not any more accessible to biopsy monitoring.

Indications for Islet Transplantation

Islet autotransplantation

Recurrent acute and chronic pancreatitis

Islet autotransplantation is indicated when total pancreatectomy is required for treatment of recurrent acute or chronic pancreatitis associated with frequent debilitating exacerbations and/or chronic, unrelentingly pain. The rationale for total pancreatectomy is removal of the source of pain and disease exacerbations to improve a very poor quality of life, reduce or eliminate chronic narcotic use, and facilitate the ability to maintain work and/or child care responsibilities. For most patients with chronic pancreatitis there is the eventuality of developing pancreatogenic (type 3c) diabetes due to progressive destruction of the pancreatic microarchitecture by fibrosis leading to islet damage and loss (80, 81). However, with total pancreatectomy the development of insulin-dependent diabetes is certain and immediate and must be understood as acceptable to patients considering surgery (29, 36). The rationale for transplanting the patients’ own islets into the liver is to prevent or ameliorate the diabetes caused by pancreatectomy. Islet autotransplantation does not decrease the risk of type 3c diabetes by relocating healthy islets from a diseased pancreas to the liver, but recovery of a high quantity of healthy islets is more likely to result in satisfactory control of the postpancreatectomy diabetes. For patients already experiencing diabetes, some institutions still offer islet autotransplantation as long as there is preoperative evidence of some preserved β-cell capacity for secretion assessed by stimulated C-peptide (82); however, evidence that this approach benefits control of the postoperative diabetes is lacking.

The causes of chronic pancreatitis in 409 adults and children 5 to 69 years of age treated by TPIAT at the University of Minnesota include idiopathic (41%), pancreas divisum (17%), hereditary [PRSS1 mutations (83)] and other genetic (14%), sphincter of Oddi dysfunction (9%), and alcohol abuse (7%) (29). It is now clear that interactions between obstructive, genetic [e.g., SPINK1 and CFTR mutations (84)], environmental, and metabolic factors contribute to the development of recurrent acute and chronic pancreatitis (85). Historically, patients contend with chronic pancreatitis and attendant interventional attempts, including cholecystectomy, sphincterotomy, pancreatic duct stenting, and pancreatic surgery, for an average of almost 10 years before pancreatectomy is performed as a last resort (29). Although pancreatic drainage procedures are indicated for decompression of a large, dilated main duct in the setting of pancreatitis (86), these approaches have proven ineffective in the treatment of patients with “small duct” disease. Currently, earlier consideration of pancreatectomy is being advocated to shorten the seemingly inevitable progressive decline in quality of life, with possible advantages of preventing opioid addiction and increasing the yield of islets recovered for transplantation (37). Resolution of pain following TPIAT is more much likely in the setting of recurrent acute than once chronic pancreatitis has become established (87).

Trauma and benign neoplasms of the pancreas

Even less common indications for islet autotransplantation include following pancreatic resection for trauma or other benign neoplasms of the pancreas (88–90). Typically, when trauma or benign tumors present in the neck of the pancreas, including insulinomas not amenable to enucleation (91), the ~70% to 80% pancreatectomy required for mass resection normally carries a high risk for postsurgical diabetes that can be prevented by islet autotransplantation. For glands that have not undergone previous instrumentation and are unlikely to have bacterial colonization, a period of islet culture can be applied while pathologic assessment is performed to ensure that any resected lesion is benign (89, 90). Remarkably, a case of successful islet autotransplantation following completion pancreatectomy for gunshot wound trauma sustained by a US airman with remote islet isolation and infusion within 24 hours of surgery has been reported (92, 93).

Successful islet autotransplantation cases have been reported following total pancreatectomy for indolent renal cell carcinoma metastases (94) and following salvage total pancreatectomy for pancreatic or ampullary adenocarcinoma (95–97); however, the risk of potentially disseminating otherwise resected malignant cells to the liver outweighs the potential benefit for glycemic control in patients with known malignant disease (98). In a series of 31 patients with known malignancy who underwent TPIAT, 3 developed de novo liver metastases during a median 2.5 years of follow-up (90).

Islet allotransplantation

Type 1 diabetes

Islet allotransplantation is indicated to treat type 1 diabetes complicated by hypoglycemia unawareness associated with severe hypoglycemia events and/or excessive glycemic lability, or by hyperglycemia associated with a stable functioning kidney graft. All patients should have completed a structured education program on basal-bolus insulin delivery with flexible dosing of modern insulin analogs using pump or multidose injection delivery based on frequent self-monitoring of blood glucose with or without continuous glucose monitoring.

The rationale for islet transplantation in hypoglycemia unawareness is that in long-standing type 1 diabetes with near-total destruction of islet β-cells and the accompanying loss of the α-cell response to hypoglycemia (99), replacement of islet function by transplantation addresses the underlying pathophysiology and allows for avoidance of hypoglycemia and stabilization of glycemic lability that otherwise contribute to the development of impaired awareness of hypoglycemia and risk for experiencing severe hypoglycemia events. Current evidence-informed guidelines recommend that patients with problematic hypoglycemia, defined by experiencing two or more episodes per year of severe hypoglycemia or by experiencing one episode in the context of impaired awareness of hypoglycemia, extreme glycemic lability, or major fear and maladaptive behavior, should be considered for either pancreas or islet transplantation (100). What effect recently available continuous glucose monitoring communicating pumps with automated insulin delivery (hybrid closed loop) may have on type 1 diabetes patients with problematic hypoglycemia is not yet known, as patients experiencing severe hypoglycemia have been excluded from registration trials of artificial pancreas technologies (101).

The rationale for islet transplantation when hyperglycemia is associated with a stable functioning kidney graft is that improvement in glycemic control may protect the transplanted kidney from recurrence of diabetic nephropathy with minimal added risk because immunosuppression is already required. Some institutions consider this simultaneously with kidney transplantation from a deceased donor, which has the advantage of minimizing exposure to additional HLAs because the islets and kidney come from the same donor, but the disadvantage of being limited to the number of islets isolated from the paired pancreas. Other patients with type 1 diabetes considered for islet transplantation are those with problematic hyperglycemia, defined by the presence of recurrent episodes of diabetic ketoacidosis or severe, rapidly progressing secondary complications of diabetes (102, 103). A rare but important indication is for patients with type 1 diabetes complicated by allergy or resistance to subcutaneously administered insulin (104). Finally, retransplantation of alloislets prepared from a viable allograft pancreatectomy has been performed successfully in a patient with type 1 diabetes experiencing recurrent episodes of graft pancreatitis who initially received the pancreas transplant for hypoglycemia unawareness (105), and in a patient with type 1 diabetes who underwent simultaneous pancreas/kidney transplantation that was complicated by bleeding at the pancreas graft arterial anastomosis (106).

Pancreatogenic (type 3c) diabetes

In addition to type 1 diabetes, any cause of insulin-deficient diabetes such as cystic fibrosis–related diabetes or other pancreatogenic forms of diabetes (e.g., chronic pancreatitis or following pancreatectomy) may be considered for islet transplantation when β-cell failure is associated with glycemic instability and either problematic hypoglycemia or hyperglycemia despite availability of, and adherence to, optimized medical care (102, 103). Although it is expected that C-peptide levels in such individuals would be low, undetectable levels of C-peptide are not required for determination of eligibility (102, 103). Islet allotransplantation for cystic fibrosis–related diabetes with unstable glycemic control and frequent hypoglycemia has been reported simultaneously with bilateral lung transplantation in four cases using islets isolated from the same donor and transplanted after as much as 7 days of islet culture to enable initial recovery from the lung transplantation (107), in a case simultaneously with and after bilateral lung and liver transplantation (108), and in a case after lung transplantation that resulted in a prolonged period of insulin independence (109). Whether the observed improvements in glycemic control and nutritional status noted following islet transplantation may impact lung function and mortality in cystic fibrosis requires further study (110).

Peritransplant Management of the Islet Recipient

Care of the autoislet recipient

At the time of pancreatectomy, insulin therapy is initiated IV per a modified hospital protocol targeting normoglycemia (32, 35) that serves to “rest” the islets at the time of transplantation when they have been stressed by the isolation procedure and are devascularized and so relatively underoxygenated (Fig. 3). Exposure of devascularized, underoxygenated islets to hyperglycemia has been shown to induce β-cell apoptosis (111) and should be strictly avoided (112). Furthermore, hyperglycemia has detrimental effects on both the function (113) and revascularization (114) of isolated human islets. Importantly, the degree of glycemic control achieved during the first 5 postoperative days is associated with the likelihood of achieving long-term insulin independence (32). At the time of islet infusion, anticoagulation is initiated with unfractionated heparin, usually at a dose of 50 to 70 U/kg recipient body weight (25, 28, 29, 32, 35, 43), which is either divided equally among the islet product bags [each composed of no more than 5 mL of tissue per 200 mL of infusion medium (26)] or between the islet product and a peripheral venous infusion. As soon as hemostasis is assured postoperatively, a continuous infusion of heparin is given either at a fixed rate (e.g., 500 U/h) or titrated to achieve and maintain the partial thromboplastin time (PTT) of ~50 seconds for the first 48 to 72 hours. The initial goal for anticoagulation is to prevent portal vein thrombosis, and because there is a balance between thrombosis risk and surgical bleeding risk, Doppler ultrasound of the liver on the first postoperative day assessing blood flow in the main as well as right and left branches of the portal vein is helpful to determine whether more aggressive anticoagulation is indicated, or whether less aggressive anticoagulation may be tolerated should there be an observed increase in surgical drain output or decrease in hematocrit suggestive of surgical site bleeding. In addition to serially monitoring of the hematocrit and PTT, hepatic transaminase levels are assessed as indicators of transient ischemic injury to the hepatic parenchyma following intraportal islet infusion, and in the absence of a resolving trend after the first few days may prompt repeat ultrasonography of the liver. Perioperative antibiotic prophylaxis should follow institutional practice for coverage against skin as well as upper gastrointestinal tract flora, and so have activity against both Gram-positive cocci and enteric Gram-negative bacilli until culture results are available from the transplanted islet product.

Peritransplant management for islet autotransplantation. At the time of pancreatectomy, insulin therapy is initiated IV according to a modified hospital protocol targeting normoglycemia, and at the time of islet infusion anticoagulation is initiated with unfractionated heparin. Once stable postoperatively and as oral intake is advanced, IV insulin and heparin are transitioned to subcutaneous regimens, antiplatelet therapy with aspirin is added, instruction in a carbohydrate-controlled diet is provided, and pancreatic enzyme replacement and acid-blocking therapy is initiated or resumed.
Figure 3.

Peritransplant management for islet autotransplantation. At the time of pancreatectomy, insulin therapy is initiated IV according to a modified hospital protocol targeting normoglycemia, and at the time of islet infusion anticoagulation is initiated with unfractionated heparin. Once stable postoperatively and as oral intake is advanced, IV insulin and heparin are transitioned to subcutaneous regimens, antiplatelet therapy with aspirin is added, instruction in a carbohydrate-controlled diet is provided, and pancreatic enzyme replacement and acid-blocking therapy is initiated or resumed.

Once stable postoperatively and as oral intake is advanced, IV insulin and heparin are transitioned to subcutaneous regimens. Exogenous insulin injections are given for at least several weeks after the islet infusion to allow time for establishment of arterialization and adequate oxygenation of the intrahepatic islets (115, 116). Thus, education is required in the provision of intensive insulin therapy with basal-bolus insulin dosing based on at least four times daily glucose monitoring, and in adherence to a carbohydrate controlled diet that is also low in fat and accompanied with chronic treatment with oral preparations of pancreatic enzymes required to assist with digestion and absorption of nutrients and avoidance of diarrhea (117). Acid-blocking therapy is initiated or resumed both to prevent gastric inactivation of the administered pancreatic enzymes and to prevent the development of marginal ulceration around the gastrojejunal anastomosis. Protein supplements including high-protein shakes are often required to meet nutrient requirements and mitigate weight loss. Total enteral or parenteral nutrition is sometimes required in the postoperative period for protein-calorie malnutrition, and hyperalimentation can result in hyperglycemia and hepatic toxicity (118). Care should be taken to minimize the calories provided by simple sugars and adjust insulin doses ahead of planned changes in nutrition therapy to maintain normoglycemia during this critical period of intrahepatic islet engraftment.

After 48 to 72 hours of anticoagulation with heparin, in the absence of evidence for portal main or branch vein thrombosis, heparin may be transitioned to subcutaneous administration at high prophylactic dosing for at least the first 1 or 2 weeks (43) or until hospital discharge, which also serves to prevent microthrombi formation that can interfere with islet engraftment and revascularization. As a consequence of splenectomy, thrombocytosis is frequently observed, and antiplatelet therapy with full-dose aspirin is added as soon as surgically appropriate, or with low-dose aspirin when prolonged anticoagulation is planned. By discharge, those with splenectomy require immunization against encapsulated bacteria, including pneumococci, meningococci, and Haemophilus influenza type b, to reduce the risk of future postsplenectomy sepsis (119). Children should complete vaccination at least 2 weeks before splenectomy, and they require antibiotic prophylaxis for the first year postoperatively (120). Annual influenza vaccination should be performed using recommended inactivated and recombinant vaccines. Because the islets are autologous, no immunosuppressive drugs are required.

Care of the alloislet recipient

At the time a compatible islet preparation is available and enters culture (Fig. 4), the intended recipient receives induction and maintenance immunosuppression in the hospital where intensive insulin therapy is maintained targeting normoglycemia that again serves to “rest” the islets at the time of transplantation when they have been stressed by the isolation procedure and are devascularized and so relatively underoxygenated and susceptible to glucotoxicity (121). While practice varies in terms of islet culture time and choice and timing of induction agents, in the phase 3 study conducted by the Clinical Islet Transplantation Consortium (B7), the T lymphocyte–depleting polyclonal antibody thymoglobulin was initiated with the standard 1.5 mg/kg dose divided as 0.5 mg/kg on day −2 and 1.0 mg/kg on day −1, with additional 1.5 mg/kg doses on days 0, 1, and 2 when tolerated per prescribing guidelines. First piloted by Hering et al. (18), this approach allows for glucocorticoid premedication prior to the first dose on day −2 and dissipation of effects from the administered glucocorticoid and cytokines released from lysed T lymphocytes that are toxic to β-cells by allowing at least 36 hours to pass from the first dose of thymoglobulin to the islet infusion. Because the cytokine TNF-α is particularly toxic to β-cells and further may contribute to the induced expression of class II HLAs on alloislets (122), slow-release pentoxifylline is administered from day −2 to day 7 because it can inhibit TNF-α production (123), and immediately prior to the islet infusion the TNF-α inhibitor etanercept is administered IV followed by three additional doses subcutaneously on days 3, 7, and 10. Anticoagulation is again initiated with unfractionated heparin at a dose of 70 U/kg recipient body weight that is divided equally among the islet product bags. As soon as hemostasis is assured after the procedure, a continuous infusion of heparin is titrated to achieve and maintain the PTT at ~50 to 60 seconds for the first 48 hours, after which unfractionated heparin is transitioned to a subcutaneous regimen consistent with high prophylactic dosing for at least the first week (124), and it is discontinued only with Doppler ultrasound of the liver demonstrating normal blood flow in the main as well as right and left branches of the portal vein. In the absence of any procedurally related bleeding complication, antiplatelet therapy with low-dose aspirin is added by the second day (124). Again, serially monitoring of the hematocrit and PTT is required for the first few days together with hepatic transaminase levels as indicators of transient ischemic injury to the hepatic parenchyma following intraportal islet infusion (125, 126). Perioperative antibiotic prophylaxis should follow institutional practice for coverage against skin flora until culture results are available from the transplanted islet product.

Peritransplant management for islet allotransplantation. At the time a compatible islet preparation is available and enters culture, induction and maintenance immunosuppression is initiated in the hospital where intensive insulin therapy is maintained targeting normoglycemia. At the time of islet infusion, anti-inflammatory therapy (e.g., etanercept) and anticoagulation are initiated, with unfractionated heparin transitioned to a subcutaneous regimen with antiplatelet therapy using low-dose aspirin added by the second day. [Shown is the B7 protocol from Hering BJ, Clarke WR, Bridges ND, et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 2016;39:1230–1240; other approaches may vary.]
Figure 4.

Peritransplant management for islet allotransplantation. At the time a compatible islet preparation is available and enters culture, induction and maintenance immunosuppression is initiated in the hospital where intensive insulin therapy is maintained targeting normoglycemia. At the time of islet infusion, anti-inflammatory therapy (e.g., etanercept) and anticoagulation are initiated, with unfractionated heparin transitioned to a subcutaneous regimen with antiplatelet therapy using low-dose aspirin added by the second day. [Shown is the B7 protocol from Hering BJ, Clarke WR, Bridges ND, et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 2016;39:1230–1240; other approaches may vary.]

Exogenous insulin is maintained for at least several weeks after the islet infusion to allow time for establishment of arterialization and adequate oxygenation of the intrahepatic islets. On the day of transplantation, it is helpful to transition from subcutaneous to IV insulin delivery that serves to both more effectively target normoglycemia prior to the islet infusion (124) and allow for temporary withdrawal of insulin should hypoglycemia result during the first 6 hours following infusion from islet degranulation (127). Any hypoglycemia can be corrected by continuous infusion of 10% dextrose with care taken not to overcorrect and expose the transplanted islets to hyperglycemia, with removal and reinstitution of IV insulin as soon as stable normoglycemia is restored. Transition back to subcutaneous insulin delivery should occur prior to the first posttransplant meal where bolus insulin should follow the individual’s pretransplant carbohydrate ratio. It remains important that intensive insulin therapy with basal-bolus insulin dosing based on at least four times daily glucose monitoring be continued together with adherence to a carbohydrate-controlled diet to maintain normoglycemia during this critical period of islet engraftment.

Immunosuppression for the alloislet recipient

Most current protocols implement induction immunosuppression with T lymphocyte–depleting agents such as thymoglobulin as discussed above (18, 19, 76) or alemtuzumab, a humanized monoclonal antibody against the cell surface glycoprotein CD52 (58, 128), or T lymphocyte inhibitory agents that block the IL-2 receptor (e.g., basiliximab). T lymphocyte depletion results in more effective immunosuppression than does IL-2 receptor blockade (129, 130), but results in cytokine release for which mitigation strategies such as that described above in the B7 protocol are likely important for optimizing transplanted islet survival and engraftment. In support of this concept, data from the Collaborative Islet Transplant Registry indicate that induction regimens based on a T lymphocyte–depleting agent result in superior rates of long-term insulin independence when compared with use of an IL-2 receptor antagonist, but only when combined with a TNF-α inhibitor (e.g., etanercept) (131). Consistent with this analysis, a single-arm trial adding the TNF-α infliximab to the IL-2 receptor antagonist daclizumab did not result in an improvement in the rate of insulin independence achieved over historical experience using daclizumab alone (132). Nevertheless, IL-2 receptor antagonists are preferentially used at the time of subsequent islet infusions to minimize adverse effects related to overimmunosuppression (19), and when the presence of allergy precludes use of a T lymphocyte–depleting agent (133).

Maintenance immunosuppression for islet transplant recipients remains calcineurin inhibitor based, either as low-dose tacrolimus used in combination with the mammalian target of rapamycin (mTOR) inhibitor sirolimus (also known as rapamycin) (19), or more standard dosing tacrolimus used in combination with mycophenolic acid as for kidney transplantation (58, 76). Substituting the calcineurin inhibitor with cyclosporine and the mTOR inhibitor with everolimus has also demonstrated efficacy in a phase 2 study (134). Glucocorticoids are avoided for recipients of islet transplantation alone; however, maintenance dosing of glucocorticoids (e.g., prednisone at 5 mg daily) that may be in place in support of a prior kidney transplant is generally continued, as this dosing remains physiologic and has not been associated with inducing insulin resistance (135). Antimicrobial prophylaxis against Pneumocystisjiroveci pneumonia is typically achieved with trimethoprim/sulfamethoxazole, and duration should consider recovery of the lymphocyte count when depleting agents are used for induction. In the case of sulfa allergy, atovaquone is preferred over dapsone as a second line agent because dapsone can affect interpretation of the HbA1c (136). Antiviral prophylaxis against cytomegalovirus (CMV) reactivation is required when either the donor or recipient evidences prior exposure by positive CMV antibodies and can be achieved with valganciclovir. When both donor and recipient are CMV negative, antiviral prophylaxis against herpes simplex and varicella zoster virus reactivation can be achieved with acylclovir. Standard immunizations for persons with diabetes should be up to date, including against pneumococcus, as well as diphtheria, tetanus, and pertussis, whereas those against live attenuated viruses such as varicella zoster virus are contraindicated. Annual influenza vaccination should be performed using recommended inactivated and recombinant vaccines, not those using live attenuated virus.

Clinical Benefits of Islet Transplantation

Outcomes following islet autotransplantation

Pain and quality of life

In a subgroup (n = 207) of the largest cohort of 409 autoislet recipients from the University of Minnesota treated between 1977 and 2011, 85% experienced resolution or significant improvement in pain and narcotic use (29). In a subgroup (n = 193) of the 250 patients treated after 2006 reporting on health-related quality of life during 2 years postoperatively, SF-36 measures improved significantly in all dimensions, irrespective of the ability to discontinue narcotic analgesia (29). Insulin independence was associated with greater improvement in dimensions of physical functioning, limitations, and vitality than for those who were insulin-dependent (29). Overall results of TPIAT were judged as good to excellent by 90% of recipients, fair by 8%, and poor by 2% (29). In another series of 85 consecutive patients undergoing total pancreatectomy (50 of whom received islet autotransplantation), >90% required chronic opiate analgesia before surgery, which reduced to ~40% at 1 year and further to ∼15% at 5 years (36), again supporting 85% effectiveness of pancreatectomy in addressing painful pancreatitis. Patient-reported outcomes were available from 50 of the 85 patients, with significant reductions noted in pain severity and frequency, and all viewing the operation as successful, although a few noted experiencing frequent problems (36). Assuming that missing data occurred at random in these larger studies, the results are consistent with earlier (33, 137) and smaller series (138) supporting that TPIAT effectively addresses the primary indication of chronic pain and reduced quality of life in ~80% of recipients.

Insulin independence and glycemic control

In most large series of islet autotransplantation at a median follow-up of 2 years approximately one-third of patients are insulin-independent (Table 1), approximately one-third are managed with daily long-acting insulin alone, and approximately one-third require intensive insulin therapy with a basal-bolus regimen administered by multidose injection or pump delivery (25, 32, 34, 36). Patients with minimal change disease on pancreatic imaging have higher isolated islet yields that are associated with higher likelihood of achieving insulin independence (32, 139). Similarly, preoperative imaging of pancreatic atrophy, calcifications, or ductal dilatation were each associated with lower isolated islet yields (140) and measures of posttransplant islet function (141). An early series from the University of Minnesota reported that about three-fourths of recipients receiving >300,000 autoislets [the number of islets normally found in a healthy pancreas is ~1 million (142, 143)] were insulin-independent at 2 years (137). Subsequently, insulin independence at 2 years was reported in about two-thirds of patients receiving >5000 islet equivalents [IE; standardized to an islet diameter of 150 µm (144)] per kg body weight, compared with about one-third of patients receiving <5000 but >2500 IE/kg, and very few patients receiving <2500 IE/kg (25). During 3 years of follow-up, HbA1c <7.0% was maintained in 94% who received >5000 IE/kg, with this rate declining to 86% and 71% when <5000 but >2500 IE/kg and <2500 IE/kg were used, respectively; overall, HbA1c <7.0% was maintained in 82% of recipients (29). Note, however, that despite these generally excellent outcomes in glycemic control when sufficient islets are used, some autoislet recipients have reported problems with postprandial hypoglycemic episodes (see below).

Pediatric outcomes

When considering children <18 years of age, outcomes for pain resolution and quality-of-life improvement are similar to those reported for adults, with >80% able to attend school or work (145). A study of 19 consecutive children aged 5 to 18 years undergoing TPIAT demonstrated significant improvement in all SF-36 measures (146). Insulin-independence rates are closer to 40% at 2 years in children undergoing TPIAT, with >50% of the youngest recipients between 5 and 12 years old insulin-independent at 1 year (120). In children during 3 years of follow-up, HbA1c <7.0% was maintained in 100% who received >5000 IE/kg, with this rate declining to 96% and 81% when <5000 but >2500 IE/kg and <2500 IE/kg were used, respectively (120).

Younger age and shorter duration of pancreatitis have been associated with higher islet yields, and similar to adults (137, 140, 147), prior pancreatic surgery and more extensive pancreatic fibrosis and atrophy were associated with lower islet yields (148), again arguing for earlier consideration of TPIAT in the course of recurrent acute or chronic pancreatitis (32). A subsequent study of 17 children between 3 and 8 years of age who underwent TPIAT demonstrated pain resolution in all, and two-thirds were insulin-independent with a median 2 years of follow-up (149). However, the durability of insulin independence throughout puberty and adolescence where both hormonal insulin resistance and accelerated growth significantly increase demand for insulin secretion remains uncertain (150), and longer-term follow-up in pediatric patients is warranted.

Long-term outcomes

There is attrition in insulin independence with time; available series with 5-year follow-up have reported rates between 10% and 15% (25, 36), or approximately half the rates initially achieved that continue to depend on the transplanted islet mass (29). Nevertheless, many patients have been reported with insulin independence persisting for >10 years following TPIAT (25, 29, 120, 151). For previously nondiabetic patients undergoing ~70% to 80% pancreatic resection for trauma or benign neoplasms located at the neck of the pancreas and subsequent islet autotransplantation, all achieved long-term normoglycemia, with ~95% insulin-independent at 2 years (90, 152, 153) and >90% remaining so with up to 7.5 years median follow-up (153). Thus, strategies that preserve or enhance the surviving functional islet β-cell mass are critical to ensuring long-term durability of the metabolic benefit afforded by islet autotransplantation.

Outcomes following islet allotransplantation

Early clinical results

The Edmonton protocol established that glucocorticoid-free immunosuppression together with a subsequent islet infusion from a second donor pancreas could reproducibly render a recipient with type 1 diabetes insulin-independent provided >9000 IE/kg were transplanted (11). However, by a median 2 years of follow-up most patients returned to requiring some insulin therapy despite ongoing evidence of islet graft function indicated by restored C-peptide production (154), suggesting establishment of only a marginal mass of surviving islets after transplantation. These disappointing long-term outcomes for insulin independence were confirmed by an international multicenter clinical trial of the Edmonton protocol, which also confirmed a more positive outcome of improved glycemic control without hypoglycemia experienced by >70% of recipients selected on the basis of experiencing refractory, disabling hypoglycemia (155). This multicenter clinical trial also demonstrated a center effect for achieving the primary outcome of insulin independence, where this occurred most frequently at sites with more extensive experience with pancreatic processing for islet isolation and immunosuppression management in transplant recipients. At the same time it became apparent that the withdrawal of immunosuppression for clinically failed islet grafts resulted in a high rate of sensitization to HLAs (156, 157), raising theoretical concern that finding suitable donor organs for possible future transplant indications may be more difficult. Thus, the emphasis in outcomes shifted away from insulin independence using islets procured from multiple donors to the stabilization of metabolic control using the least number of donor pancreases required for durable clinical benefit (158). This would require a focus on the peritransplant period to ensure that a larger number of high-quality islets survived the engraftment process.

Improving outcomes and quality of life

The biggest change came with the introduction of immunosuppressive protocols at the University of Minnesota that incorporated more potent induction agents with a period of islet culture and peritransplant anti-inflammatory, antithrombotic, and intensive insulin therapy, with improved rates of insulin independence occurring more frequently with islets isolated from a single donor (18) and most remaining insulin-independent for >2 years after transplant (134), suggesting that a greater proportion of the transplanted islets survived engraftment. The effect of peritransplant intensive antithrombotic and insulin therapy on increasing the rate of insulin independence achieved with islets transplanted from a single donor pancreas was independently confirmed (124). Importantly, these results were obtained using similar low-dose calcineurin inhibitor and mTOR inhibitor maintenance therapy as in the Edmonton protocol, providing evidence that appropriate dosing of currently available immunotherapy is not toxic to islets nor rate limiting for improving transplanted islet graft survival and functional durability. Although 1-year single donor insulin independence occurred less frequently (13 of 48 subjects) in the multicenter B7 trial (19) than in the pilot study from the University of Minnesota (5 of 8 subjects) (18), a similarly high rate of sustained single donor insulin independence (7 of 11 subjects) was reported by at least one other center participating in the B7 study (159), showing that these results are reproducible but may be center-dependent owing to careful attention paid to less emphasized variables such as maintenance of strict glycemic control during the period of islet engraftment. Examining the transplanted islet mass in these two cohorts further indicates that although not always sufficient, transplantation of >6000 IE/kg recipient body weight is necessary for a chance of obtaining insulin independence from a single donor (18, 159).

In the largest cohort of 677 alloislet recipients reported by the international Collaborative Islet Transplant Registry, when analyzed by year of transplantation as early (1999 to 2002, n = 214), middle (2003 to 2006, n = 255), or recent (2007 to 2010, n = 208), 55% of recipients from the most recent era remained insulin-independent at 2 years (69). Rates of insulin independence at 3 years after transplant improved from 27% to 37% to 44%, with the proportion of recipients receiving sequential islet infusions decreasing from 60% to 65% to <50% in the most recent era (69). Despite higher rates of sustained insulin independence observed with the transplantation of islets from fewer donor pancreases in the most recent era, receiving islets from more than one donor remained associated with a higher likelihood of achieving insulin independence across all eras by multivariate analysis (69). Other factors associated with a higher likelihood of achieving insulin independence in the multivariate analysis included increasing recipient age, islet culture ≥6 hours, islet stimulation index ≥1.5, and induction therapy with T lymphocyte depletion combined with a TNF-α inhibitor (69). Additionally, ever achieving insulin independence was associated with significantly prolonged islet graft function and accompanying metabolic stability regardless of a return to some insulin therapy (69). During 3 to 5 years of follow-up, near-normal glycemic control indicated by HbA1c <6.5% was maintained by ~60% of recipients across all eras. Although >90% of patients were experiencing severe hypoglycemia prior to transplant, >90% remained free of severe hypoglycemia events in all eras through 5 years of follow-up (69). These results support the benefit of islet culture (18, 132), islet quality, and more potent induction regimens (131) on achieving insulin independence, as well as the practice of considering a subsequent islet infusion when necessary to achieve insulin independence to maximize the long-term benefit of metabolic control imparted by a functioning islet graft (160, 161).

In a cohort of 23 alloislet recipients followed for 3 years, diabetes quality of life improved significantly and was mainly influenced by glycemic control and not insulin independence, although the reintroduction of insulin led to some modification of effect (162). Importantly, procedure- or immunosuppression-related adverse events did not negatively impact quality of life (162). A subsequent report of health-related quality of life that included 28 alloislet recipients followed for 3 years did not show significant improvement, although fear of hypoglycemia was markedly reduced during the period of observation to levels below that of disease controls with type 1 diabetes not being considered for islet transplantation (163). In a cohort of 27 alloislet recipients followed for 5 years, improvement was seen for depression and some but not all general health-related quality-of-life measures, with a robust and durable reduction in fear of hypoglycemia for both behavior and anxiety assessments (164). These results show a clear benefit in islet allotransplantation for improving quality of life that has been reduced by problematic hypoglycemia.

Standardizing outcomes

With the emphasis of islet allotransplantation shifting to establishing metabolic control in the absence of hypoglycemia, programs for islet alone transplantation established selection criteria based on patients experiencing severe hypoglycemia accompanied by hypoglycemia unawareness or marked glycemic lability and a primary outcome of achieving an HbA1c <7.0% in the absence of severe hypoglycemia (Table 2). The results of prospective single-arm clinical trials of islet allotransplantation indicate >80% of recipients achieve this primary outcome at 1 year (76, 165), and that this effect can be further sustained at 2 years in >80% of recipients (19). Importantly, in the phase 3 B7 trial only those recipients who lost islet graft function entirely experienced a recurrence of severe hypoglycemia, and so >90% of recipients were protected from experiencing a severe hypoglycemic event during 2 years of follow-up (19). Moreover, in the B7 study that involved the largest consecutive cohort of 48 transplant recipients, there were significant improvements in diabetes distress, fear of hypoglycemia, as well as patient self-assessments of personal well-being (166). Health-related quality-of-life measures improved significantly in five of the eight SF-36 domains, and results were not significantly different between those who achieved or did not achieve insulin independence (166). Thus, although the burden of chronic immunosuppressive drug treatment is often argued as a reason against consideration of islet transplantation as a therapy for type 1 diabetes, these phase 3 study results clearly demonstrate that appropriately selected candidates experience clinically significant and personally meaningful improvement in glycemic control and quality of life.

“…these phase 3 study results clearly demonstrate that appropriately selected candidates experience clinically significant and personally meaningful improvement in glycemic control and quality of life.”

Table 2.

Comparison of Published Consortium Trials With Standardized Outcomes for Islet Allotransplantation

YearsAustralian (76)
United Kingdom (58)
GRAGIL-2 (165)
National Institutes of Health (B7) (19)
2013a2008–20112003–20102008–2012
Number of patients17202448
Duration of islet culture, h12–24<24<436–72
Median islet yield, IE/kg15,3668770971611,972
HbA1c, 1-y median, %6.5b6.36.25.6
HbA1c, 2-y median, %NR6.2NR5.6
Insulin independence, 1-y median, %NR4537.552
Insulin independence, 2-y median, %47154642
YearsAustralian (76)
United Kingdom (58)
GRAGIL-2 (165)
National Institutes of Health (B7) (19)
2013a2008–20112003–20102008–2012
Number of patients17202448
Duration of islet culture, h12–24<24<436–72
Median islet yield, IE/kg15,3668770971611,972
HbA1c, 1-y median, %6.5b6.36.25.6
HbA1c, 2-y median, %NR6.2NR5.6
Insulin independence, 1-y median, %NR4537.552
Insulin independence, 2-y median, %47154642

Abbreviations: IE/kg, IE/kg recipient body weight, whereby an IE approximates a standard islet diameter of 150 µm (144); NR, not reported.

a

Publication year.

b

Mean reported (76).

Table 2.

Comparison of Published Consortium Trials With Standardized Outcomes for Islet Allotransplantation

YearsAustralian (76)
United Kingdom (58)
GRAGIL-2 (165)
National Institutes of Health (B7) (19)
2013a2008–20112003–20102008–2012
Number of patients17202448
Duration of islet culture, h12–24<24<436–72
Median islet yield, IE/kg15,3668770971611,972
HbA1c, 1-y median, %6.5b6.36.25.6
HbA1c, 2-y median, %NR6.2NR5.6
Insulin independence, 1-y median, %NR4537.552
Insulin independence, 2-y median, %47154642
YearsAustralian (76)
United Kingdom (58)
GRAGIL-2 (165)
National Institutes of Health (B7) (19)
2013a2008–20112003–20102008–2012
Number of patients17202448
Duration of islet culture, h12–24<24<436–72
Median islet yield, IE/kg15,3668770971611,972
HbA1c, 1-y median, %6.5b6.36.25.6
HbA1c, 2-y median, %NR6.2NR5.6
Insulin independence, 1-y median, %NR4537.552
Insulin independence, 2-y median, %47154642

Abbreviations: IE/kg, IE/kg recipient body weight, whereby an IE approximates a standard islet diameter of 150 µm (144); NR, not reported.

a

Publication year.

b

Mean reported (76).

These outcomes have recently been further validated against intensive insulin therapy. The multicenter Australian consortium assessed glycemic control and variability in patients with type 1 diabetes and severe hypoglycemia while on multiple daily injections of insulin, again after ~1 year of continuous subcutaneous insulin infusion (pump), and again 1 year following islet allotransplantation (167); continuous subcutaneous insulin infusion did not affect HbA1c but did modestly reduce glucose variability and lowered the rate of severe hypoglycemia events, whereas islet transplantation reduced HbA1c from a median 8.2% to 6.4% that was associated with a further marked reduction in glucose variability and elimination of hypoglycemia. The multicenter TRIMECO study enrolled patients with type 1 diabetes experiencing either severe hypoglycemia or poor glycemic control after kidney transplantation in the first randomized clinical trial to assess a transplant intervention for the treatment of diabetes (20); after 6 months the HbA1c was significantly lower with islet allotransplantation compared with intensive insulin therapy (5.6% vs 8.2%; P < 0.0001), and 23 of 25 subjects were protected from severe hypoglycemia events with islet transplantation compared with only 8 of 22 subjects receiving optimized medical management (P < 0.0001). Importantly, the outcome of achieving an HbA1c <7.0% in the absence of severe hypoglycemia was achieved in 21 of 25 subjects by islet transplantation and 0 of 22 with intensive insulin therapy (P < 0.0001), and there was significantly improved health-related quality of life in the transplant group compared with insulin group (20). Although the duration of comparative assessment in the TRIMECO study was limited to 6 months to allow subjects to cross over and receive the alloislet intervention, these results further substantiate by randomized clinical trial design the 2-year outcomes reported from the single cohort phase 3 B7 trial (19, 166).

Recent criteria have been developed for defining outcomes for β-cell replacement therapy in the treatment of diabetes regardless of the source being an islet or pancreas transplant (102, 103). These Igls criteria are based on the achievement of HbA1c targets, absence of severe hypoglycemia events, reduction in insulin requirements, and restoration of clinically significant C-peptide production, and they provide consistent classification of treatment success as that reported by the phase 3 B7 trial (19). Use of consistent definitions of graft functional and clinical outcomes is critical to informing the comparative effectiveness of existing and future forms of β-cell replacement therapy.

Long-term outcomes and comparison with pancreas transplantation

Longer-term outcomes have also significantly improved for graft function following islet allotransplantation. Although the initial 5-year outcomes data reported that with the Edmonton protocol 10% of alloislet recipients remained insulin-independent (154), other groups have reported 5-year insulin independence using the Edmonton protocol in >20% to 45% of both islet-alone and islet-after-kidney recipients (165, 168). In more recent protocols that depend on induction therapy with T cell depletion and TNF-α inhibition, 5-year insulin independence may be as high as 50% and significantly prolonged when compared with induction therapy based on IL-2 receptor blockade as used in the Edmonton protocol (131). As with TPIAT, patients are now being reported with insulin independence persisting >10 years following islet allotransplantation (169, 170).

As the rate of long-term insulin independence following islet transplantation is approaching the 5-year insulin independence rates of ~50% reported for pancreas transplantation alone and pancreas-after-kidney transplantation (171), consideration should be made for islet transplantation as an alternative to solitary pancreas transplantation because it carries considerably less risk for morbidity by avoiding major abdominal surgery. However, with simultaneous pancreas/kidney transplantation 5-year insulin independence is >70% (171), supporting the primacy of transplanting a pancreas simultaneously with a deceased donor kidney (when a living kidney donor is not available) for long-term durable graft function and resultant glycemic control. The superior long-term functional outcomes associated with simultaneous pancreas/kidney transplantation has been attributed to less risk for immunologic graft loss because transplanting organs from the same donor (and consequently HLA type) enables earlier detection of rejection episodes using biomarkers from both the kidney (serum creatinine) and pancreas (serum amylase and lipase) with treatment directed at one organ resolving subclinical rejection present in the biochemically stable organ. Insulin independence rates are lower with simultaneous islet/kidney transplantation, even when a second islet infusion is included, although reported series have relied on the Edmonton protocol (172). Nevertheless, this approach resulted in significant improvement in HbA1c without hypoglycemia when compared with a control group of type 1 diabetic individuals who had undergone kidney transplant alone, and significantly less morbidity than that seen in a control group of type 1 diabetic individuals who had undergone simultaneous pancreas/kidney transplantation where 40% required relaparotomy (172). Ideally, pancreas and islet transplantation should be offered as complementary, not competing, approaches to providing β-cell replacement to provide the most optimal clinical benefit to risk balance for each potential candidate while maximizing use of the largest number of quality donor pancreases offered for transplantation (173).

Metabolic Benefits and Consequences of Islet Transplantation

Islet autotransplantation following total pancreatectomy

β-Cell secretory capacity

The prevention or amelioration of postpancreatectomy diabetes by islet autotransplantation is dependent on the mass of islet β-cells that survives engraftment (151). As already discussed, most recipients receiving >300,000 autoislets (of the ~1 million islets normally found in a healthy pancreas) (137), or receiving >5000 IE/kg body weight achieve insulin independence, with very few receiving <2500 IE/kg meeting this objective (25). Because the number of islets that survive engraftment are always likely to be less than the number transplanted, the functional β-cell mass of an islet graft can be most accurately determined in vivo from the β-cell secretory capacity. The β-cell secretory capacity is measured by glucose potentiation of insulin or C-peptide release in response to injection of a nonglucose insulin secretagogue such as arginine or glucagon, and it correlates with calculated β-cell mass in animal models of β-cell reduction (174), and with resection (175) and transplantation (176–178) of a hemipancreas in humans. In insulin-independent, normoglycemic autoislet recipients, the β-cell secretory capacity correlates with the number of transplanted islets, an observation now replicated in independent cohorts of 8 and 10 patients (179, 180). In these cohorts of autoislet recipients selected on the basis of achieving optimal functional outcomes and studied between 1 and 8 years after TPIAT, the β-cell secretory capacity was ~35% to 40% of normal, very close to that predicted on the basis of the numbers of islets transplanted (179, 180) (Fig. 5). Whether the presumably lower engrafted β-cell mass for a comparable number of islets transplanted in less successful cases may be caused by nonspecific inflammatory and thrombotic mechanisms remains a focus of investigation (43). In four patients studied before and almost 1 year after TPIAT, the decrease in β-cell secretory capacity was inversely proportional to the number of islets transplanted, meaning that those who received more islets maintained closer to their presurgical insulin secretory reserve (179). Further longitudinal assessment of five patients who each received >300,000 autoislets and achieved insulin independence and normoglycemia demonstrated stable β-cell secretory capacity over 2 to 4 years, including in a patient between 12 and 13 years after TPIAT (151). These results support that islet survival and long-term functional durability may require establishing a functional islet β-cell mass >30% of normal.

Functional islet β-cell mass estimated from the β-cell secretory capacity. β-Cell secretory capacity is measured as the acute insulin response to glucose-potentiated arginine and shown for insulin-independent autoislet and alloislet recipients as the percentage of nondiabetic control values. Although in both groups a functional islet β-cell mass of ~40% to 50% of normal is achieved, when corrected for the number of islet equivalents transplanted (and assuming 1 million islets are present in the controls), there remains an ~25% loss of islet β-cell mass in the alloislet recipients.
Figure 5.

Functional islet β-cell mass estimated from the β-cell secretory capacity. β-Cell secretory capacity is measured as the acute insulin response to glucose-potentiated arginine and shown for insulin-independent autoislet and alloislet recipients as the percentage of nondiabetic control values. Although in both groups a functional islet β-cell mass of ~40% to 50% of normal is achieved, when corrected for the number of islet equivalents transplanted (and assuming 1 million islets are present in the controls), there remains an ~25% loss of islet β-cell mass in the alloislet recipients.

Metabolic testing prior to TPIAT is important to evaluate for pancreatogenic (type 3c) diabetes and assess some measure of functional β-cell capacity for secretion. This is typically done by assessment of HbA1c, fasting glucose, and oral glucose tolerance in response to either a standardized oral glucose tolerance test (OGTT; 75 g of glucose) or mixed-meal tolerance test (6 mL/kg up to 360 mL of Boost® high protein or equivalent containing ~50 g of carbohydrate) (181). Both 1-hour and 2-hour measures of glucose should be assessed postingestion because an elevated 1-hour OGTT glucose may already indicate a reduced β-cell secretory capacity in patients with pancreatic disease (182). Indeed, not only do standard criteria for prediabetes (HbA1c ≥5.8%, fasting glucose ≥104 mg/dL, 2-hour glucose ≥141 mg/dL) predict the need for insulin therapy after TPIAT, but also a 1-hour glucose ≥189 mg/dL (183). Corresponding C-peptide levels, with or without insulin, should be measured during the OGTT (183) or mixed-meal tolerance test (184), with the combination of fasting glucose <100 mg/dL and stimulated C-peptide >4 ng/mL best predicting the likelihood of an islet isolation yield ≥2500 IE/kg (184). These same measures also appear predictive for estimating islet yield from pediatric patients (185). Nevertheless, these oral challenge techniques and even IV glucose tolerance testing that measures first-phase insulin secretion as a surrogate measure of β-cell secretory capacity (186) lack precision to predict islet isolation outcome for any individual (184). Using arginine stimulation testing in deceased organ donors, an acute insulin response ≥55 µU/mL best predicted the likelihood of an islet isolation yield ≥250,000 IE and/or ≥4000 IE/kg (187). Further study is warranted to determine whether glucose potentiation of arginine induced insulin secretion to provide an estimate of functional β-cell mass prior to TPIAT may better predict islet isolation outcomes and postsurgical diabetes risk.

Insulin sensitivity

Insulin sensitivity has variably been measured as modestly impaired or normal in otherwise normoglycemic patients with chronic pancreatitis (188, 189), a finding likely explained by the presence of a chronic disease studied in various phases of evolution. Importantly, insulin sensitivity as measured by the frequently sampled IV glucose tolerance test increased from before to 12 and 18 months following TPIAT during a randomized clinical trial of the dipeptidyl pepdidase-4 inhibitor sitagliptin vs placebo in both study groups (190). Although this well-conducted study did not find any differences in metabolic outcomes with sitaglitpin use in TPIAT recipients, the normal estimates of insulin sensitivity measured during follow-up suggest that any potential decrease in insulin sensitivity due to chronic pancreatitis resolves postoperatively, assuming at least that near-normal glycemic control is maintained as demonstrated in the referenced study (190).

Glucose counterregulation

Hypoglycemia is reported by some insulin-independent recipients of autoislets that in some cases may be severe (191, 192), and suggests the presence of an inherent defect in glucose counterregulation, that is, the physiologic defense against the development of low blood glucose. Normally in response to decreasing plasma glucose, endogenous insulin secretion is turned off and glucagon secretion is activated with the resulting decreased ratio of insulin to glucagon exposed to the liver increasing endogenous glucose production to prevent or correct the development of hypoglycemia. Secondary to this primary islet response against hypoglycemia, brain recognition of hypoglycemia activates sympathoadrenal responses to release epinephrine that becomes critical for increasing endogenous glucose production when the glucagon response is impaired (193) and also generates autonomic symptoms that alert the individual to ingest food. Thus, defects in both islet and sympathoadrenal responses must interact to contribute to the development of hypoglycemia (Fig. 6).

Potential mechanisms explaining the risk for hypoglycemia experienced by recipients of intrahepatic autoislets following total pancreatectomy, as well as the amelioration of hypoglycemia experienced by type 1 diabetic recipients of intrahepatic alloislets. [Includes data from Rickels MR, Liu C, Shlansky-Goldberg RD, et al. Improvement in beta-cell secretory capacity after human islet transplantation according to the B7 protocol. Diabetes 2013; 62:2890-2897; and from Robertson RP, Bogachus LD, Oseid E, et al. Assessment of beta-cell mass and alpha- and beta-cell survival and function by arginine stimulation in human autologous islet recipients. Diabetes 2015;64:565–572.]
Figure 6.

Potential mechanisms explaining the risk for hypoglycemia experienced by recipients of intrahepatic autoislets following total pancreatectomy, as well as the amelioration of hypoglycemia experienced by type 1 diabetic recipients of intrahepatic alloislets. [Includes data from Rickels MR, Liu C, Shlansky-Goldberg RD, et al. Improvement in beta-cell secretory capacity after human islet transplantation according to the B7 protocol. Diabetes 2013; 62:2890-2897; and from Robertson RP, Bogachus LD, Oseid E, et al. Assessment of beta-cell mass and alpha- and beta-cell survival and function by arginine stimulation in human autologous islet recipients. Diabetes 2015;64:565–572.]

In 1992 it was first reported that TPIAT recipients had impaired glucagon responses to insulin-induced hypoglycemia, even though they had normal glucagon responses to IV arginine and a liver biopsy demonstrated α-cells containing glucagon-positive granules (194). This led to a study in total pancreatectomized dogs that compared glucagon secretion during insulin-induced hypoglycemia in groups of animals receiving either intrahepatic autoislets or intraperitoneal autoislets (51). The dogs receiving intraperitoneal islets had greater glucagon responses than did the dogs receiving intrahepatic autoislets, although both groups had similarly reduced glucagon responses to IV arginine (51). In another study of total pancreatectomized dogs receiving either intraomental autoislets or intrasplenic autoislets, mild non–insulin-medicated hypoglycemia resulted in normal inhibition of insulin secretion but impaired glucagon responses in both groups of animals, which again displayed similarly reduced glucagon responses to IV arginine (195). More recently, in a cohort of intrahepatic TPIAT recipients routinely studied by the investigators but not selected for having reported hypoglycemia, it was discovered that upon close questioning 11 out of 14 recipients acknowledged episodes of hypoglycemia during daily living that occurred either after meals and/or during exercise (52). Self-monitoring of blood glucose levels in these 11 documented glucose levels that reached nadirs of ~37 mg/dL and continuous glucose monitoring in five of these recipients demonstrated ~15% of time spent with hypoglycemia (<70 mg/dL) (52). In this study, hyperinsulinemic hypoglycemic clamps demonstrated markedly impaired glucagon responses in recipients of intrahepatic islets only compared with normal glucagon responses present in five recipients who had received autoislets in nonhepatic as well as hepatic sites; again, glucagon responses to IV arginine were similarly reduced in all transplant recipients (52).

One hypothesis to explain post-TPIAT hypoglycemia is that intrahepatic islets fail to detect hypoglycemia owing to the locally higher glucose levels present in the liver. In support of this hypothesis, in a rodent model of intrahepatic islet transplantation only after glycogen depletion did an increase in glucagon occur in response to hypoglycemia, suggesting that an increase in endogenous glucose production and consequent intrahepatic levels may inhibit activation of intrahepatic α-cells, as local glucose levels may be higher than the peripherally measured hypoglycemia (196). However, the intrahepatic islet β-cells exposed to the same local glucose levels do appropriately respond to hypoglycemia with complete suppression of insulin secretion (52), evidencing appropriate sensing of peripheral glycemia. Although the evidence supports that nonhepatic islets secrete more glucagon in response to hypoglycemia than do their intrahepatic counterparts, recipients of nonhepatic islets received a larger total number of islets, and those placed in the pelvic portion of the peritoneal cavity revascularize in the distribution of the systemic rather than portal circulation that would bypass first-past hepatic extraction of glucagon leading to higher peripherally measured levels (197). A difference in venous drainage by transplant site would also explain why impaired glucagon responses were seen in dogs with intraomental or intrasplenic autoislets that are both nonhepatic but still portally drained (195), whereas dogs with intaperitoneal autoislets exhibited normal responses (51). Future prospective studies are needed to compare the glucagon response to hypoglycemia in recipients of comparable numbers of islets transplanted in hepatic and nonhepatic sites, as well as the endogenous glucose production response because the peripherally measured glucagon levels are markers of the α-cell response and not direct measures of the counterregulatory response.

Another hypothesis to explain post-TPIAT hypoglycemia is that alimentary hypoglycemia develops as a consequence of the Roux-en-Y gastrointestinal reconstruction that follows pancreaticoduodenectomy and is similar to other well-recognized forms of “late dumping syndrome” where altered nutrient transit with rapid delivery to the distal small intestine leads to more rapid glucose absorption and exaggerated glucagon-like peptide-1 (GLP-1) and insulin responses (198, 199) with dysregulated islet function that in some individuals leads to late postprandial hypoglycemia (198, 200–202). In support of this hypothesis, the hypoglycemia that is associated with TPIAT occurs postprandially and can be managed with widely varying degrees of success by the ingestion of small frequent meals and avoidance of simple sugars (52, 191).

In a mixed-meal study involving 10 TPIAT patients experiencing postprandial hypoglycemia, glucose levels reached a higher early peak than in controls, and then fell to levels below baseline between 4 and 6 hours postingestion that in five patients included a nadir <70 mg/dL not seen in controls (203). When corrected for the numbers of islets transplanted, the insulin and C-peptide secretory responses were greater than normal, whereas the glucagon response was greater early after the meal and lower later postingestion compared with controls (203). These data were interpreted to suggest that a mismatch between insulin levels and glucose levels may occur in the later hours after TPIAT meals such that endogenous insulin levels induce hypoglycemia. In this study glucagon levels did not increase above normal in the recipients who developed hypoglycemia postprandially. Such a defect in glucagon secretion despite lower late postprandial glucose concentrations is also exhibited by patients with other forms of alimentary hypoglycemia who have intact native islets and may be explained by a combination of the paracrine effect of the increased β-cell response on inhibiting α-cell function (204), with an additional contribution from the glucagonostatic effect of exaggerated GLP-1 release (205, 206), and desensitization of central mechanisms for responding to low blood glucose. In fact, nondiabetic individuals studied before and after Roux-en-Y gastrointestinal reconstruction as part of gastric bypass surgery for obesity develop reduced neurohormonal responses to insulin-induced hypoglycemia, including a marked reduction in glucagon and epinephrine secretion (207). Because exposure to even mild hypoglycemia leads to blunting of subsequent sympathoadrenal responses to hypoglycemia, including sympathetic nervous system augmentation of glucagon release as well as activation of epinephrine secretion (208), it is possible that induction of hypoglycemia-associated autonomic failure is operative after TPIAT and contributes to the impaired glucagon responses to hypoglycemia. Additionally, subtle differences in the endogenous glucose production response during exercise in autoislet recipients might contribute to the development of hypoglycemia (209), particularly if conducted late postprandially that would further contribute to the risk for experiencing hypoglycemia.

Islet allotransplantation for type 1 diabetes

β-Cell secretory capacity

The reversal of diabetes by islet allotransplantation is also dependent on the mass of islet β-cells that survives engraftment (159). As already discussed, most recipients with type 1 diabetes receiving a total of >9000 IE/kg from at least two donor pancreases under the Edmonton protocol achieve insulin independence (11); however, by a median 2-year follow-up, most patients return to requiring some insulin therapy despite ongoing evidence of islet graft function indicated by restored C-peptide production (154), suggesting establishment of only a marginal mass of surviving islets posttransplantation. Indeed, in insulin-independent, normoglycemic alloislet recipients treated with the Edmonton protocol at two institutions, the β-cell secretory capacity was only ~25% of normal despite receiving >600,000 to 900,000 IE (210, 211). Using another glucocorticoid-free regimen with islets transplanted from pooled donors to achieve insulin independence, Keymeulen et al. (212) also demonstrated a β-cell secretory capacity of ~25% of normal in alloislet recipients. Collectively, these results indicate a markedly reduced engrafted islet β-cell mass in alloislet recipients that is just at the margin of what is required to avoid hyperglycemia, and so help to explain the eventual return to insulin therapy experienced by most recipients treated under the Edmonton protocol.

The lower functional islet β-cell mass for the numbers transplanted under the Edmonton protocol supports early loss of transplanted islets before engraftment due to nonspecific inflammatory and thrombotic mechanisms (66, 213) that has been estimated by positron emission tomography/CT study to affect ~25% of the islet product (214). Further early islet loss may be attributed to excessive metabolic demand inducing endoplasmic reticulum stress (215). Importantly, new induction immunosuppression protocols introduced by the University of Minnesota and adopted elsewhere incorporated peritransplant anti-inflammatory, antithrombotic, and intensive insulin therapy, with improved rates of insulin independence occurring more frequently with islets isolated from a single donor (18, 124) and being sustained for a longer duration posttransplant (131, 134), suggesting that a greater proportion of the transplanted islets survived engraftment. The B7 protocol was designed to test this approach using the T lymphocyte–depleting polyclonal antibody thymoglobulin together with a period of islet culture and the TNF-α inhibitor etanercept and pentoxifylline to address inflammation, anticoagulation to address thrombosis, and intensive insulin therapy for at least 2 months to minimize metabolic demand during engraftment.

Following the B7 protocol, the resulting β-cell secretory capacity was ~40% to 50% of normal in 11 consecutively treated patients who received >600,000 IE, with all becoming insulin-independent (159) (Fig. 5). Seven of the patients received islets from a single donor pancreas and four received islets from a second donor pancreas, such that when considering the lower numbers of islets transplanted than with the Edmonton protocol represents an approximately threefold improvement in engraftment efficiency with the B7 protocol with repeat measures of the β-cell secretory capacity stable at 1 year (159). These findings suggest that a reserve capacity for insulin secretion more than necessary to achieve initial insulin independence is required to maintain long-term islet graft function. Indeed, during the second year of follow-up only 1 patient of the 11 with the lowest β-cell secretory capacity returned to low-dose insulin therapy to maintain normoglycemia (216). Why more islets appear to survive in successful cases of islet autotransplantation when compared with allotransplantation (Fig. 5) is unknown, but it may be due to the source being a living rather than deceased donor or some tropic factor derived from the nonislet tissue present in higher amounts in less pure preparations. Similar to autoislets, however, these results from alloislets also support that islet survival and long-term functional durability may require establishing a functional islet β-cell mass >30% of normal.

Importantly, the improved results for engrafted islet β-cell mass with islet allotransplantation were obtained using the same low-dose tacrolimus and sirolimus maintenance immunosuppression as in the Edmonton protocol. Although both tacrolimus and sirolimus have known human islet toxicity at levels supratherapeutic for their desired immunosuppressive effects (217, 218), β-cell secretory capacity is fully normal in uncomplicated whole-pancreas transplant recipients who have received 100% of an islet β-cell mass despite tacrolimus-based immunosuppression (219), and studies using sirolimus at plasma drug concentrations targeted in clinical practice have no deleterious effect on human islets (220). Curiously, in a canine autoislet transplant model, treatment with sirolimus was associated with a 13% reduction in insulin clearance without any change in insulin sensitivity (221), and which might exert a modest benefit in terms of extending the metabolic effect of secreted insulin. These findings support that modern dosing of currently available immunotherapy is not toxic to islets nor rate limiting for improving transplanted islet graft survival and functional durability.

Insulin sensitivity

Insulin sensitivity is impaired in type 1 diabetes (222), a consequence of absent endogenous insulin secretion, exposure to frequent periods of sustained hyperglycemia (223), and impaired sensitivity of lipolysis to inhibition by insulin with accompanying increases in free fatty acids (224, 225) that affects insulin action at both the liver and skeletal muscle (226–228). Intraheptic islet transplantation restores endogenous insulin secretion that not only corrects the hyperglycemia of type 1 diabetes, but also normalizes free fatty acid metabolism that is associated with improvement in insulin sensitivity (229–231). Interestingly, current glucocorticoid-free immunosuppression with tacrolimus and sirolimus has been implicated in the development of insulin resistance in rodents (232, 233), whereas in both a canine islet autotransplant model with or without the calcineurin inhibitor with cyclosporine (221) and a nonhuman primate islet allotransplant model (234) treatment with sirolimus did not impair insulin sensitivity. Moreover, in humans, appropriate dosing of sirolimus for its immunosuppressive effects in nondiabetic subjects demonstrated no effect on hepatic insulin sensitivity and actually enhanced peripheral insulin sensitivity during nutrient stimulation with amino acids (235). In longitudinal assessment of the same 12 individuals with type 1 diabetes from before to after islet allotransplantation, measures of both hepatic and peripheral insulin sensitivity improved to estimates not different than normal (236, 237). Thus, intrahepatic islet transplantation corrects the defects in insulin action present at both the liver and skeletal muscle in type 1 diabetes, and modern dosing of glucocorticoid-free immunosuppression with low-dose tacrolimus and sirolimus does not induce insulin resistance in this population.

Glucose counterregulation

In contrast to reports of hypoglycemia following islet autotransplantation, type 1 diabetic recipients of islet allotransplantation are entirely protected from hypoglycemia for the duration of islet graft function, even when requiring low-dose insulin therapy to maintain normoglycemia (Fig. 6). With the autoimmune destruction of islet β-cells in type 1 diabetes, the near-total loss of endogenous insulin secretion not only eliminates any autoregulatory capability to reduce excessive insulin, but also abolishes the islet α-cell glucagon response to hypoglycemia that requires paracrine activation from neighboring β-cells through an intraislet decrement in insulin secretion (238–240). In type 1 diabetic recipients of intrahepatic islet transplants, there is recovery of the physiologic islet cell responses to insulin-induced hypoglycemia whereby endogenous insulin secretion is appropriately suppressed and glucagon secretion is partially restored (241–243). In earlier cross-sectional studies involving subjects treated according to the Edmonton protocol, the glucagon response to insulin-induced hypoglycemia was either reported as being absent in one study (244) or, when compared with euglycemic control experiments [that control for the inhibitory effect of experimental hyperinsulinemia on glucagon secretion (245)], was reported as partial albeit markedly less than normal (241, 243). Subsequent longitudinal study of the same 12 patients investigated before and 6 months after islet allotransplantation (whereby 11 were treated according to the B7 protocol) demonstrated a significant improvement in the glucagon response to ~40% to 50% of normal that was associated with complete restoration of the endogenous glucose production response to insulin-induced hypoglycemia (242), an effect that remained upon repeat assessment at 18 months (216). The magnitude of the glucagon response was similar to the ~40% to 50% of a normal β-cell secretory capacity reported from this cohort of B7 treated patients (159). These results are consistent with the partial glucagon responses to insulin-induced hypoglycemia observed in type 1 diabetic recipients of pancreatic segments (246), and they support the dependence of the glucagon response on the number of transplanted islets surviving engraftment. It remains possible that the release of glucagon from islets within the liver may create an early increase in endogenous glucose production with subsequent attenuation of the stimulated glucagon levels measured peripherally by locally increased glucose. However, the normal suppression of C-peptide levels during hypoglycemia indicates that intrahepatic islet β-cells do appropriately sense and respond to the degree of peripheral hypoglycemia (241, 242), arguing against islet exposure to locally increased glucose.

“…type 1 diabetic recipients of islet allotransplantation are entirely protected from hypoglycemia for the duration of islet graft function.”

In the absence of intact islet cell responses to hypoglycemia in type 1 diabetes, secondary sympathoadrenal (epinephrine secretion and autonomic symptom generation) responses become critical to increase endogenous glucose production from the liver and alert the individual to ingest food to correct low blood glucose, whereas pituitary–adrenal (GH and cortisol secretion) responses generally operate later in an attempt to maintain increased hepatic glucose production (247). However, both the glycemic threshold (i.e., the glucose level that elicits the response) and magnitude of these hormonal and symptom responses are impaired by recurrent episodes of hypoglycemia that induce brain adaptation to low blood glucose and lead to a syndrome of hypoglycemia-associated autonomic failure, also known as hypoglycemia unawareness (248), that increases the risk of life-threatening hypoglycemia 20-fold in type 1 diabetes (249). By 6 months following islet allotransplantation, there is normalization of the glycemic thresholds for activation of epinephrine, GH, and autonomic symptom responses to insulin-induced hypoglycemia (250), evidencing recovery from hypoglycemia-associated autonomic failure. Although the magnitude of the epinephrine response may be only partially improved after 6 months (242, 250), additional longitudinal follow-up of the same alloislet recipients for 2 years demonstrated essentially no time with hypoglycemia (<70 mg/dL) by continuous glucose monitoring, and after 18 months complete normalization of the epinephrine response to insulin-induced hypoglycemia (216). These findings indicate that >6 months of strict avoidance of hypoglycemia may be required for complete autonomic nervous system function recovery in patients with hypoglycemia unawareness.

Importantly, even in alloislet recipients requiring insulin to support partial graft function there is modest recovery of glucagon and epinephrine secretion with partial restoration of the endogenous glucose production response to insulin-induced hypoglycemia that is associated with clinical protection from hypoglycemia (243). Continuous glucose monitoring studies have demonstrated similar reductions in mean glucose, glucose variability, and serious hypoglycemia (<54 to 60 mg/dL) relative to that in type 1 diabetes for both insulin-independent and insulin-requiring alloislet recipients (251–253). That the islet graft is responsible for these improvements in continuous glucose monitoring metrics of glycemic control is supported by their significant correlation with measures of islet β-cell graft function (254, 255). Thus, even partial improvements in glucose counterregulation and hypoglycemia symptom recognition likely work in concert with reduced glucose variability to provide the robust protection from hypoglycemia afforded by islet transplantation. Such avoidance of hypoglycemia is distinct from the results described above following TPIAT, and from outcomes seen with nontransplant intervention for type 1 diabetes complicated by hypoglycemia unawareness (256).

Secondary complications of diabetes

The effect of islet allotransplantation on secondary complications of diabetes has been assessed by a few nonrandomized studies. Thompson and colleagues (257) conducted a prospective cohort study of islet allotransplantation in 26 nonuremic patients with type 1 diabetes vs prior or ongoing intensive medical therapy focused on improving glycemic, blood pressure (including use of angiotensin system blockade), and serum lipid control in another 18 waitlisted patients. Baseline HbA1c was ~8.0% in both groups, and after a median ~36-month follow-up was significantly lower at 6.6% following islet transplantation vs 7.4% with intensive medical therapy (258) whereas blood pressure and lipids were comparable (257). The primary outcome of change in measured glomerular filtration rate (GFR) was less after islet transplantation, being −1.4 mL/min/y/1.73 m2 vs −3.6 mL/min/y/1.73 m2 following institution of intensive medical therapy; there were no between group differences in urinary albumin excretion or progression over time (259). Retinopathy graded as greater than mild nonproliferative progressed in 0 of 51 eyes following islet transplantation and more often in 10 of 82 eyes during intensive medical therapy; nerve conduction studies remained stable in both groups (258). In a single cohort followed for 5 years following islet allotransplantation, sensory, but not motor, nerve conduction studies improved over time but differences were not evident until after 3 years (260). These findings support a possible effect of islet allotransplantation on slowing the progression of diabetes microvascular complications, and they suggest that future randomized studies will require >3 years of follow-up to confirm differences from intensive medical therapy.

Mechanisms of Islet Graft Dysfunction and Failure

Metabolic exhaustion: endoplasmic reticulum stress, oxidative stress, islet amyloid

Since the first experiments of islet transplantation using animal models, a nonimmunologic mechanism of islet “stress” has been speculated to account for cases of recurrent hyperglycemia in the absence of evidence of immune system activation (2). As discussed for both autoislet and alloislet recipients, a reserve capacity for insulin secretion more than necessary to achieve initial insulin independence (>30% of normal β-cell secretory capacity) is required to maintain long-term islet graft function. This reserve capacity likely protects islets from excessive metabolic demand inducing endoplasmic reticulum stress (215), oxidative stress (261), or amyloid deposition (262), as has been described in preclinical islet transplant models and that may be toxic to islets. Excessive β-cell demand for secretion may be indicated by increased secretion of proinsulin relative to insulin, resulting in an elevated molar ratio of proinsulin to insulin, which occurs with recruitment of immature secretory granules containing an abundance of incompletely processed proinsulin (263). Hyperglycemia per se decreases β-cell insulin content and increases β-cell secretion of proinsulin from isolated human islets (264). Autoislet recipients with hyperglycemia similarly demonstrate decreased fasting insulin and C-peptide with elevations of proinsulin and the proinsulin-to-insulin ratio (265). Importantly, in studies involving insulin-independent and insulin-dependent alloislet recipients where the mean HbA1c was ~6.0%, fasting proinsulin-to-insulin and proinsulin secretory ratios in response to glucose-potentiated arginine were normal (219, 266). In contrast, in a study involving autoislet and alloislet recipients where the mean HbA1c was ~7.1%, fasting proinsulin-to-insulin ratios were elevated (267), consistent with an effect of exposure to chronic hyperglycemia. Thus, the maintenance of near-normal glycemia (HbA1c ≤6.5%) with insulin therapy when appropriate may alleviate excessive metabolic demand on the islet graft as indicated by protection against the secretion of incompletely processed proinsulin, and prevent stress-induced exhaustion of islet graft function.

Additional evidence for the importance of avoiding hyperglycemia in islet recipients comes from the finding of amyloid deposition in intrahepatic islets of three out of four cases with postmortem examination ~2 to 5 years after transplant (268, 269). Islet amyloid is composed of islet amyloid polypeptide (IAPP or amylin) fibrils deposited within and surrounding β-cells where they exhibit direct toxicity (270). IAPP is cosecreted from the β-cell with insulin (271), but normally is inhibited from forming amyloid by appropriate proportions of insulin and other factors in the β-cell (272). During glucose-potentiated arginine testing in insulin-independent alloislet recipients there is disproportionately increased IAPP-to-insulin secretion (273), suggesting that hyperglycemia may disturb regulation of insulin and IAPP within transplanted islets and facilitate the development of islet amyloid. Moreover, incompletely processed pro-IAPP may be more amyloidogenic than IAPP (274), and hyperglycemic islet transplant recipients with disproportionately increased fasting proinsulin also exhibit disproportionately increased fasting levels of the pro-IAPP1–48 intermediary (275). In a nonhuman primate transplant model, intrahepatic islets developed amyloid deposits coincident with animal growth, increasing β-cell secretory demand with the presence of amyloid predating a decline in β-cell secretory capacity and subsequent recurrence of hyperglycemia (262). Thus, avoiding hyperglycemia and reducing metabolic demand through the provision of exogenous insulin when necessary should prevent nonimmunologic islet graft loss and improve long-term functional outcomes for islet recipients (276).

Insulin therapy should also be provided to islet recipients during periods of intercurrent illness that may increase metabolic demand for insulin, as well as in advance of the anticipated development of insulin resistance as occurs with glucocorticoid therapy and pregnancy. Indeed, the administration of glucocorticoids for indications not related to the islet transplant has been shown to result in severe hyperglycemia and consequently islet graft failure in previously insulin-independent autoislet recipients (277). In addition to the marked induction of insulin resistance by glucocorticoids increasing demand for insulin secretion from a reduced functional β-cell mass, the hyperglycemia resulting from an inadequate hyperinsulinemic response occurs together with glucocorticoid-induced elevation of free fatty acids that accelerate β-cell damage through glucolipotoxicity (278). In a canine model of intrahepatic islet autotransplantation, a short course of the glucocorticoid prednisone induced hyperglycemia and accelerated the time to islet graft failure, but importantly these effects were prevented by the administration of insulin for the duration of glucocorticoid exposure (279). Pregnancy increases metabolic demands for insulin through both the development of maternal insulin resistance and the requirement for increased nutritional intake to support maternal and fetal growth. Few reports are available but they support a benefit to preemptive initiation of insulin at the time pregnancy is detected to maintain gestational targets for fasting and postprandial glucose that can result in optimal birth outcomes and prevent deterioration of islet graft function with a return to postgestational insulin independence (280–284). These observations support that appropriately timed administration of exogenous insulin to alleviate transplanted β-cells from increased requirements for secretion during periods of excessive metabolic demand while avoiding glucotoxicity can prevent stress-induced islet graft dysfunction and loss, and it is important to sustaining long-term islet graft function for transplant recipients.

“…reducing metabolic demand through the provision of exogenous insulin when necessary should prevent nonimmunologic islet graft loss…”

Another strategy for alloislet recipients in addition to the provision of insulin therapy to protect marginal islet grafts from further functional deterioration is administering a supplemental islet infusion. Similar to infusing islets from a second or sometimes a third donor pancreas to achieve insulin independence that is associated with more durable long-term islet graft function (69), patients who later return to a requirement for insulin therapy may be considered for a supplemental islet infusion to reestablish a reserve capacity for insulin secretion with the goal of restoring insulin independence and further promoting long-term islet graft function (161, 258). For this strategy to succeed it is important that immunologic mechanisms are not responsible for the islet graft dysfunction. The reported studies to date have administered supplemental islet infusions with adjuvant treatment including the GLP-1 agonist exenatide based on evidence that GLP-1 has antiapoptotic effects on cultured human islets (285, 286). Another study administered exenatide at the time of initial islet infusions that resulted in early insulin independence (287), but subsequently supplemental islet infusions were required to maintain more durable freedom from exogenous insulin (288). Future randomized controlled trials of adjuvant treatment with a GLP-1 agonist at the time of an initial or supplemental islet infusion on surviving functional islet graft mass are needed.

In other studies patients with islet graft dysfunction were treated with exenatide that was associated with a reduction in insulin requirements without any improvement in glycemic control during a 3- to 6-month period (289, 290); however, by 12 months most patients required an increase in exogenous insulin or a supplemental islet infusion to maintain glycemic control (291). More islet recipients experienced nausea and vomiting with exenatide than that reported in studies of patients with type 2 diabetes, and the tolerated doses were lower. In islet recipients GLP-1 acutely increases the proinsulin secretory ratio in response to glucose-potentiated arginine (292), and chronic exenatide leads to disproportionate increases in proinsulin and IAPP secretion relative to insulin following mixed-meal stimulation (291), effects that suggest GLP-1 agonists increase metabolic demand on a marginal islet β-cell mass that may be detrimental compared with use of exogenous insulin on maintaining long-term islet graft function. Similar ineffectiveness has been reported with the dipeptidyl peptidase-4 inhibitor sitagliptin that slows degradation of the endogenous incretin hormones GLP-1 and glucose-dependent insulinotropic polypeptide, where a 6-month treatment in alloislet recipients was associated with a reduction in insulin requirements without any improvement in glycemic control (293). Consistent with these results, a randomized controlled trial of a 12-month treatment with sitagliptin in autoislet recipients did not affect metabolic outcomes (190).

Alloimmune rejection and sensitization

As for all allogeneic forms of transplantation, alloislets are susceptible to both acute and chronic forms of rejection that may be associated with early and late loss of islet graft function. Alloislets are typically not matched to the recipient for HLAs, and with the possibility for the transplantation of islets from more than one donor there is potential for both a larger number of mismatched HLAs as well as repeat HLA mismatches. Pretransplant sensitization to islet donor HLAs from previous exposure to nonself HLAs through pregnancy, red blood cell transfusions, solid organ transplantation, or human tissue grafts can result in acute rejection of an islet transplant (294, 295). Compared with solid organ transplants, isolated islets are more susceptible to complete loss following acute rejection, likely due to their presentation to the recipient’s immune system as a dispersed cell suspension (296). Because there is no established treatment available for acute rejection of an islet transplant, the transplantation of alloislets bearing HLAs reactive against even very low levels of preformed alloantibody in the recipient should be avoided even when T and B lymphocyte crossmatches are negative. In fact, even alloislet recipients previously sensitized to HLAs not present on an islet graft also experience a higher rate of graft loss that may be explained by cross-reactive or epitope-spreading alloantibody (297, 298). Acute cellular rejection may also develop in the absence of alloantibody, and in one case diagnosed clinically 1 month after transplantation, a 6-day course of high-dose glucocorticoid administered under islet protection with a continuous IV insulin infusion rescued islet graft function (299).

Chronic rejection may also occur with the development of de novo alloantibodies specific for donor HLA that has been temporally associated with loss of islet graft function and β-cell secretory capacity (300, 301). Islet graft loss appears more often with the development of de novo alloantibodies against the islet graft occurring in the context of a reduction in immunosuppression (302) rather than when alloantibodies are detected under adequate immunosuppression (157, 303). Although the identification of anti-islet donor HLA antibodies is a negative prognostic marker for future islet functional outcomes (304), whether early detection and subsequent intensification of immunosuppression may prolong islet graft function and survival is not known. In one case where the development of anti-islet donor HLA antibodies coincided with a deterioration in islet graft function 2 years after transplantation, treatment of clinically diagnosed humoral rejection with rituximab and IV immunoglobulins led to improvement in glycemic control and reestablishment of insulin independence (305).

Although sensitization to HLAs with the development of anti-islet donor alloantibodies has been reported in up to 20% of islet recipients receiving ongoing immunosuppressive drug treatment, the risk of sensitization increases with withdrawal of immunosuppression such that most patients discontinuing immunosuppression develop anti-HLA antibodies when assessed by sensitive assays based on flow cytometry methods (156, 157, 306). In islet recipients undergoing immunosuppression withdrawal, increased cytotoxic T lymphocyte avidity for mismatched HLAs may be seen even in the absence of alloantibody (307). In single-center analyses, the risk of sensitization has not been associated with the number of islet donors or number of mismatched HLAs between the islet recipient and donors (156, 157, 306); however, in an analysis conducted by the Collaborative Islet Transplant Registry there was a trend toward lower risk for sensitization when subsequent islet infusions contained repeat HLA class I mismatches (308). Until the consequences of sensitization on outcomes of potentially needed future organ transplants are known, it reasons that both minimizing the number of islet donors used per recipient and, in the absence of donor-specific anti-HLA antibodies (and ideally cytotoxic T lymphocyte responses), repeating HLA mismatches with subsequent islet infusions may decrease the risk of sensitization and requires further investigation.

Autoimmune recurrence in type 1 diabetes

Unique to islet allotransplantation for type 1 diabetes is the possibility for a recurrence of autoimmunity affecting the transplanted islet β-cells that can also result in early or late loss of islet graft function (309). Recurrent autoimmune diabetes was first demonstrated following initially successful transplantation of hemipancreases donated by identical twins that following reversal of diabetes underwent selective β-cell destruction by the twin recipients with type 1 diabetes, thereby recapitulating the development of the autoimmune disease (310). Although standard immunosuppressive therapy for the most part prevents autoimmune recurrence, there are well-documented cases of recurrent autoimmune diabetes affecting standard deceased donor whole-pancreas transplant recipients (311, 312). Patients with type 1 diabetes exhibiting elevated titers of autoantibodies and cellular autoimmunity prior to islet transplantation appear at increased risk for failure to achieve insulin independence and subsequent islet graft loss (313–316). That recurrent autoimmune diabetes can manifest in an islet recipient has been documented in biopsy studies where insulitis containing lymphocytic islet infiltration is seen with the absence of insulin-positive β-cells and presence of glucagon-positive α-cells (317, 318). Such islet graft loss may be heralded by either an increase in titer of an existing autoantibody or the development of a new autoantibody or cellular autoimmune response after transplant (304, 314, 319, 320). Unfortunately, no strategy for intensification of immunosuppression has been tested to determine whether islet graft function and survival may be prolonged after the identification of new autoimmune markers. Nevertheless, current immunosuppression for islet transplantation, as for pancreas transplantation, does abrogate autoimmunity for the vast majority of recipients with type 1 diabetes. Minimization of long-term immunosuppression that may be acceptable without increasing the risk for alloimmune rejection may nonetheless allow for recurrent autoimmunity resulting in chronic islet or pancreas graft loss. Interestingly, increases in both autoantibody and cellular autoimmune responses against type 1 diabetes–associated antigens glutamic acid decarboxylase-65 and islet antigen-2 have been observed during the course of developing de novo alloimmune responses in islet transplant recipients (298, 307), which might accelerate immune-mediated destruction of islet β-cells in the context of alloimmune recognition in patients with type 1 diabetes.

Another possible potentiating factor for the development of recurrent autoimmune diabetes in islet transplant recipients is active CMV infection. Although occurring rarely following islet transplantation as discussed below, CMV infection has been associated with a trend toward shorter islet graft survival (321). Although CMV may directly infect allograft tissues and increase the risk for allograft rejection (322), CMV infection has also been associated with the development of type 1 diabetes (323) and with the recurrence of autoimmune diabetes following pancreas transplantation (324). Autoreactive T cell clones from individuals with type 1 diabetes have been shown to recognize CMV peptide, supporting molecular mimicry as a potential mechanism for the induction of β-cell autoimmunity by CMV (325). Future investigation is required to assess this potential mechanism for islet graft loss and to gain insight into the mechanisms leading to the initial development of type 1 diabetes that may be recapitulated in the context of islet allotransplantation.

Procedural Risks and Complications of Islet Transplantation

Islet autotransplantation

Procedural risks

As already discussed, the main procedural risks of islet autotransplantation at the time of total pancreatectomy are portal vein thrombosis, surgical site bleeding that may be exacerbated by the heparinization required to minimize the risk for portal vein thrombosis, and the introduction of pathogens from a contaminated islet preparation. Minimization of tissue volume through purification of the islet preparation, monitoring of portal venous pressures during islet infusion, and placement of a portion of the islet product in the peritoneal cavity when elevated portal venous pressure is encountered can all reduce the risk of portal vein thrombosis (44). However, because autoislet preparations are kept relatively impure to optimize islet recovery, the tissue volume is higher for the number of islets transplanted and results in greater increases in portal venous pressure with autoislet compared with alloislet transplantation (326), and the risk for portal branch vein thrombosis may be estimated at ~10% (44). When a portal branch vein clot remains present by hospital discharge, long-term anticoagulation is required for 3 to 6 months to reestablish patency of the portal tree and prevent clot propagation. Independent of the potential effects on the portal vasculature, islet engraftment in the liver can result in periportal steatosis due to locally elevated insulin concentrations surrounding the intrahepatic islets that appears as heterogeneous echogenicity when assessed by ultrasound (327); such changes in radiologic liver appearance have not been associated with any adverse consequences for liver or islet graft function.

“Unique to islet allotransplantation for type 1 diabetes is the possibility for a recurrence of autoimmunity…”

Most patients considering TPIAT have undergone endoscopic or surgical interventions with the potential for introducing enteric bacteria into the pancreatic ductal system, which can lead to colonization of the pancreas with microorganisms. Risk for experiencing an infectious complication is minimized by avoiding islet culture and ensuring that perioperative antibiotics cover upper gastrointestinal tract flora while awaiting the results of microbial cultures taken from the transplanted islet product. Although positive microbial cultures may be seen in most cases, the risk of a related infectious complication is reduced to <15% with perioperative antibiotic prophylaxis covering both Gram-positive cocci and enteric Gram-negative bacilli (328–330). Additional infectious complications with pathogens not related to the islet product also occur as expected during recovery from pancreatectomy surgery.

Islet allotransplantation

Procedural risks

The procedural risks for islet allotransplantation are similar to those for islet autotransplantation, although they generally occur at lower frequency owing to the higher purity of the islet product, less invasive procedure for islet delivery, and use of pancreata that have not been previously instrumented. As with intraportal delivery of autoislets, portal pressure changes are related to the transplant tissue volume, but with the higher level of purification the complete islet preparation can invariably be accommodated within the portal circulation under heparinization with the risk for portal branch vein thrombosis <5% (77, 331); maintenance of therapeutic heparinization for the first 48 hours after infusion reduces this risk further (332). In addition to heparinization, use of the bag method for islet infusion instead of a syringe also helps to decrease the incidence of portal branch vein thrombosis (333). With the closed gravity-fed bag system the rate of islet infusion is controlled with natural feedback decreasing the rate with any increase in portal pressure, and so prevents the precipitous development of portal hypertension (333). While baseline portal pressures remain normal after two subsequent islet infusions, a greater increase in portal pressure during subsequent transplants has been reported by some (77) but not all (126) series. Thus, although up to three islet preparations may be safely accommodated by the liver over time (77, 126), the risk for developing portal hypertension or branch vein thrombosis may also increase over time, and this suggests that consideration should be made that even greater purity and lower tissue volume preparations be used with subsequent islet infusions.

Bleeding complications are primarily associated with the percutaneous transhepatic approach to access the portal venous circulation for islet infusion with a risk <10% (331). The primary types of bleeding complications include perihepatic hematomas, which are usually self-limited, and intraabdominal hemorrhage that usually requires transfusion of packed red blood cells and laparoscopy or laparotomy to ensure achievement of hemostasis and evacuate intraabdominal blood. Rarely, injury to a hepatic artery may result in formation of an arteriovenous fistula or arterial pseudoaneurysm that may rupture and necessitate embolization of the feeding artery for bleeding control (334, 335). Other rare complications can include injury to a subcostal blood vessel or puncture of the gallbladder. The use of ultrasound guidance may reduce the number of needle passes required to gain access to the portal vein, helping to minimize the risk for procedural complications. The use of a Gelfoam gelatin sponge as a hemostatic plug instilled in the hepatic catheter track helps to reduce (70), but does not eliminate (71), the risk of experiencing a bleeding complication. Other hemostatic agents may be more effective provided that ablation of the entire intraparenchymal catheter tract is achieved (336, 337). Initial results using Avitene microfibrillar collagen paste suggest that the bleeding risk can be eliminated entirely (338). A very low risk for bleeding may also be achieved using a minilaparotomy approach with direct visualization of the mesenteric vein used for islet infusion to the portal circulation (72); however, this transmesenteric approach introduces risk for other common surgical complications and may require a longer hospitalization (339).

Elevated liver enzymes are commonly encountered during the first several days following intrahepatic islet transplantation with a pattern of transaminases that may reach twofold to fivefold the upper limit of normal, is related to the peri-infusion change in portal venous pressure, and typically peaks after a few days and declines by the second week after infusion (125, 126). This transient response to presumed hepatic ischemia or nonspecific inflammatory injury is not accentuated by subsequent islet infusions, in fact appears greater with initial transplants, and is not associated with any long-term sequelae. Similar to that reported for intrahepatic autoislets, alloislet engraftment in the liver can result in periportal steatosis due to locally elevated insulin concentrations surrounding the intrahepatic islets (340) that appears as heterogeneous echogenicity when assessed by ultrasound (341); such changes in radiologic liver appearance have not been associated with any adverse consequences for liver or islet graft function, although they could indicate islets that are under increased demand for insulin secretion that warrants further investigation.

Immunosuppressive drug risk and toxicity

The requirement for immunosuppressive drug treatment to protect alloislets from alloimmune rejection and recurrent autoimmunity introduces additional risks that may be specific to the individual immunosuppressive drug agents as well as related more generally to immunosuppression. As discussed earlier, allergy or intolerance to one immunosuppressive drug agent can most often be addressed by substitution with an alternative agent from the same or similar drug class. Important to all immunosuppressive drug approaches is the risk for certain immunosuppression-related infections and malignancies (342). Antimicrobial prophylaxis is required against P.jiroveci pneumonia and against reactivation of prior infection from members of the herpes virus family (herpes simplex virus, varicella zoster virus, and CMV, the latter when either the donor or recipient evidences prior exposure by positive CMV antibodies). Transmission of CMV from seropositive donors to seronegative recipients is observed less frequently with alloislets than that reported with solid organ grafts due to the low tissue volume, absence of lymphoid tissue, and low number of passenger leukocytes seen in islet preparations. Nevertheless, CMV viremia has been detected in ∼5% of alloislet recipients despite receiving initial prophylaxis with valganciclovir and can occur late after transplantation with more risk seen in individuals treated with T lymphocyte–depleting compared with IL-2 receptor–blocking antibodies (321, 343). Although symptomatic disease remains rare, monitoring for viremia should extend beyond the period of prophylaxis and any time symptoms such as unexplained fever, diarrhea, myelosuppression, or abnormal liver function tests present that might be compatible with CMV infection. Posttransplantation lymphoproliferative disorders are associated with Epstein-Barr virus (EBV) infection developing under immunosuppression; risk is markedly reduced (<1%) for individuals with evidence of immunity against EBV prior to transplantation (344). The first two cases of posttransplantation lymphoproliferative disorders in islet transplant recipients were recently reported, having presented at 24 and 80 months after transplant (345). Risk for the development of skin cancers, predominantly squamous-cell carcinomas, is increased during exposure to chronic immunosuppression and necessitates sun protection and regular dermatologic monitoring for early detection and ablation of premalignant lesions (346).

An important concern related to immunosuppressive drug toxicity is the risk for impairment in kidney function that has been associated with both calcineurin inhibitors and mTOR inhibitors. When alloislets are transplanted in patients with type 1 diabetes, the use of low-dose tacrolimus in combination with sirolimus is associated with decline in estimated GFR of ~5 mL/min/y/1.73 m2 (155, 347). Importantly, however, the change in kidney function is very heterogeneous with some patients experiencing steeper declines and others improvement in measures of GFR (347). Early reductions in GFR may be related to normalization of glycemia with associated alleviation of glomerular hyperfiltration and the renovascular constricting effects of calcineurin inhibitors that is dose-dependent (348). Some patients taking tacrolimus in combination with sirolimus develop albuminuria (~5%) that may be associated with more accelerated reduction in kidney function, but fortunately is reversible upon discontinuation of the sirolimus and replacement with mycophenolic acid (349). Use of tacrolimus in combination with mycophenolic acid following alloislet transplantation has been associated with a lower decline in measured GFR of 1.4 mL/min/y/1.73 m2 that is also less than the 3.6 mL/min/y/1.73 m2 observed in a control group receiving intensive medical therapy with no between-group differences seen in urinary albumin excretion or progression (259). Thus, careful selection of patients for normal kidney function prior to initiation of immunosuppression and careful monitoring of both kidney function and albuminuria while receiving immunosuppression are critical to appropriately tailor immunosuppressive drug choices and targets to minimize the potential for adverse renal effects (350). Similar consideration is also important for islet-after-kidney transplantation, where there is a similar need to balance immunosuppressive protection of both the islet and kidney grafts with the potential nephrotoxicity of the required immunosuppression that is nevertheless directed by that required for the kidney rather than the islet transplant. More research is required to determine the relative benefit of long-term improvement in glycemic control vs harm of immunosuppressive drug therapy on kidney function in patients with type 1 diabetes.

“The requirement for immunosuppressive drug treatment to protect alloislets from alloimmune rejection and recurrent autoimmunity introduces additional risks…”

Future Developments in Clinical Islet Transplantation

Alternative sources of islet tissue

Porcine islets

The number of islets available for transplantation is a major limitation for both autoislet and alloislet approaches to β-cell replacement therapy. Therefore, the establishment of an unlimited source of islet tissue for transplantation has been a long-sought-after goal. One approach has been the development of xenoislets for transplantation that is most advanced for porcine islets. Porcine islets have the advantages of sourcing from established pathogen-free herds, targeting similar normoglycemia as present in humans, and physiologic capability to handle large demands for insulin secretion. Importantly, porcine IAPP contains amino acid substitutions in the region corresponding to residues 20 to 29 that prevent the formation of fibrils (351, 352). Intrahepatically transplanted porcine islets in an autograft model maintained normoglycemia in young pigs that experienced a 50% increase in body weight during an 18-month period without the development islet amyloidosis (353), and so porcine xenoislets may be less susceptible to metabolic exhaustion of an established transplanted islet β-cell mass over time. Disadvantages include the larger immunologic barrier of xenogeneic than allogeneic tissue that presents an additional risk for hyperacute rejection and requires more intensive immunosuppression (354, 355) and theoretical concern over the potential for transmission of zoonotic infections such as porcine endogenous retroviruses. Novel genome editing approaches may enable the breeding of pigs with significantly lower immunogenicity (356) and retroviral burden (357) that ideally would enable β-cell replacement therapy with less immunosuppression than is presently required for alloislets. Should this promise be realized, there is additional hope that porcine islets would also escape recognition by memory T lymphocytes and evade autoimmune recurrence.

As with humans, intraportal delivery for intrahepatic engraftment has had the most success with diabetes reversal by porcine islets in streptozotocin-induced diabetic nonhuman primates (358). Although genetically engineered porcine islets continue to undergo preclinical evaluation using this model, clinical phase 1/2 studies of alginate microencapsulated porcine islets (Diabecell) transplanted into the peritoneal cavity of patients with type 1 diabetes have been completed in New Zealand (NCT00940173) and Argentina (NCT01739829) (ClinicalTrials.gov; accessed 7 May 2018). Safety data using these nongenetically engineered porcine islets have not detected transmission of either porcine endogenous retroviruses or other porcine microorganisms (359, 360), although efficacy outcomes have not been reported. Whether microencapsulated islets transplanted in sites other than the liver can physiologically regulate glucose homeostasis as effectively as revascularized intrahepatic islets remains to be determined (361).

Stem cell–derived islets

Recent progress in the generation of functional islet β-cells from human stem cell sources has raised hope for the establishment of another unlimited source of islet tissue for transplantation. One approach involves the differentiation of human embryonic stem cells (hESCs) to a pancreatic progenitor stage in vitro that has the potential to further differentiate into functional pancreatic islets in vivo capable of reversing streptozotocin-induced diabetes in immunodeficient mouse models (362, 363). However, this process was also accompanied by the sporadic growth of mesodermal cells reminiscent of the formation of teratomas, and it highlighted the potential risk of transplanting immature stem cell–derived tissue. Such progenitor tissue has also been shown to reverse streptozotocin-induced diabetes in immunodeficient mice when transplanted under the skin using a macroencapsulation device (TheraCyte) (364). Use of a macroencapsulation device allows for removal of the stem cell–derived islet graft for safety monitoring or for cause assessment, and it may allow for immunoisolation of what is essentially an allogenic graft when embryonic stem cells are used (365). Although untested in a large animal model, a combination hESC-derived pancreatic progenitor in a macroencapsulation devise (VC-02) is currently under clinical phase 1/2 evaluation in patients with type 1 diabetes in the United States (NCT03163511, ClinicalTrials.gov; accessed 7 May 2018). Because mature human alloislets survive and function poorly in encapsulation devices (366), these early phase clinical studies should inform whether stem cell–derived islets may be more resistant to hypoxia than isolated human islets or may require alternative encapsulation approaches incorporating oxygen delivery (367).

Another approach to stem cell–derived islets involves the differentiation of hESCs to a pancreatic islet stage in vitro that has the capacity for glucose-dependent insulin secretion before transplantation and can reverse (368) or ameliorate (369) diabetes in immunodeficient mouse models. These stem cell–derived islets are again allogeneic and so will require the development of compatible immunosuppressive or immunoisolation (370) approaches before translation to early phase clinical trials. Stem cell–derived islets have also been generated from patients with type 1 diabetes using inducible pluripotent stem cells (371) and somatic cell nuclear transfer (372). Although the generation of cell therapy using a patient’s own genetic material will avoid alloimmunity upon transplantation, and so may be most attractive for patients with pancreatogenic forms of type 3c diabetes, recurrent autoimmunity will remain a barrier for patients with type 1 diabetes. As these cell products are considered for testing in early phase clinical trials, additional work is also required for understanding their physiology in comparison with isolated human islets as both an in vitro (373) and in vivo reference standard.

Novel approaches to combat engraftment-limiting inflammation and coagulation

As discussed, nonspecific inflammatory and thrombotic mechanisms are activated upon transplanted islet exposure to blood in the portal circulation that effect early islet survival and the efficiency of islet engraftment in the liver. Although the adoption of a number of approaches, including islet culture, peritransplant anti-inflammatory therapy with pentoxifylline and TNF-α inhibitors (e.g., etanercept), and peritransplant anticoagulation therapy with heparin products, has been associated with an improvement in intrahepatic islet engraftment efficiency (159), there remains a loss of transplanted islet mass in the early posttransplant period (Fig. 5). Additional targets for anti-inflammatory therapy based on preclinical studies using mouse models of intrahepatic islet transplantation include the protease inhibitor α-1-antitrypsin (374) and the chemokine receptor for CXCL1 and CXCL8 (CXCR1/2) (375). α-1-Antitrypsin may promote transplanted islet survival in part through induction of IL-1 receptor antagonist production (376), and administration of the IL-1 receptor antagonist anakinra also promotes survival of transplanted human islets, including an additive effect to that also seen with etanercept (377). Inhibition of IL-1β signaling with anakinra protects islet β-cells directly against apoptosis and indirectly against the potentiation of amyloid formation (378).

Preliminary results from early phase clinical trials in humans have reported favorable effects of anakinra (379) and of reparixin, an allosteric inhibitor of CXCR1/2 (375), on functional outcomes following intrahepatic alloislet transplantation in a small number of subjects. CXCL1 is released by islets in response to IL-1β and operates as a chemokine for polymorphonuclear and natural killer cells expressing CXCR1/2 that are attracted to the engraftment site within the liver (375). Reparixin is currently under clinical phase 2/3 evaluation in a North American trial of intrahepatic islet autotransplantation following total pancreatectomy (NCT01967888), as well as phase 3 evaluation in an international trial of intrahepatic islet allotransplantation for type 1 diabetes (NCT01817959) (ClinicalTrials.gov; accessed 7 May 2018). Future comparative studies will be required to determine which approach, or combination of approaches, can most safely and effectively protect transplanted islets from inflammation-mediated destruction during the critical posttransplantation period until engraftment.

Novel approaches to immunosuppression

The dependence of islet allotransplantation on calcineurin inhibitor–based regimens for immunosuppression limits current application for patients with type 1 diabetes because of the risk for renal toxicity and consequent impairment in kidney function. Two novel approaches employing inhibition of T lymphocyte adhesion and costimulation have undergone evaluation as part of calcuneurin inhibitor–free immunosuppression regimens in islet transplantation. The anti–leukocyte functional antigen-1 antibody efalizumab inhibits T lymphocyte activation and trafficking by blocking the costimulatory attachment of the CD11a subunit of leukocyte functional antigen-1 to the intercellular adhesion molecule-1. Belatacept is a fusion protein composed of the Fc fragment of human IgG1 immunoglobulin linked to the extracellular domain of cytotoxic T lymphocyte-associated protein-4, which blocks the costimulatory attachment of CD28 to CD80 and CD86 on antigen-presenting cells that is required for T-cell activation. Efalizumab-based, calcineurin inhibitor–free immunosuppression has been associated with the achievement of insulin-independence in 12 intrahepatic alloislet recipients with eight patients receiving islets from a single donor pancreas and four receiving islets from a second donor pancreas (380, 381), and belatacept-based, calcineurin inhibitor–free immunosuppression has been associated with the achievement of insulin independence in five intrahepatic alloislet recipients with four patients receiving islets from a single donor pancreas and one receiving islets from a second donor pancreas (382). All patients have exhibited remarkably stable kidney function through at least a median 1-year follow-up.

The islet graft functional outcomes for efalizumab- and belatacept-based immunosuppression are similar to what has been reported using the B7 protocol that depends on the calcineurin inhibitor tacrolimus (159), and they support that inhibition of T lymphocyte adhesion and costimulation can provide effective protection against alloimmune and autoimmune responses, as well as likely anti-inflammatory effects that may promote islet engraftment. There have been concerns, however, regarding immunologic safety. Both efalizumab and belatacept have been associated with increased risk of posttransplant lymphoproliferative disorders and progressive multifocal leukoencephalopathy caused by reactivation of JC virus infection. Efalizumab was withdrawn from the market by its sponsor after four cases of progressive multifocal leukoencephalopathy were reported out of >40,000 patients receiving the drug for the treatment of psoriasis, and belatacept has had its prescriptions related to kidney transplantation restricted to patients with evidence of immunity against EBV and so at lower risk for posttransplant lymphoproliferative disorders. The lower doses of these agents studied in islet allotransplantation than used for their labeled indications may allow for an acceptable risk/benefit profile, and a completed phase 2 study of belatacept in intrahepatic alloislet transplantation for type 1 diabetes will provide important safety and efficacy data regarding this approach (NCT00468403; ClinicalTrials.gov; accessed 7 May 2018).

“…elevated markers of β-cell injury at 24 hours after islet transplantation have been associated with worse islet graft functional outcomes within the first 3 months posttransplant…”

Novel assays to monitor β-cell death

The identification of islet β-cell loss following transplantation depends on functional assessment of glycemic control, requirements for exogenous insulin, and measures of insulin secretion (often using C-peptide) that is most accurate when using dynamic evaluation of the β-cell secretory capacity as an estimate of functional islet β-cell mass (102, 103). Implementation of these metabolic methods to replace less sophisticated traditional measures of β-cell dysfunction (such as fasting levels of glucose, C-peptide, and HbA1c) are essential to better detect cellular injury prior to the development of islet graft dysfunction. These more sensitive indicators could lead to interventions that might prevent clinically significant islet graft loss. This may be particularly helpful to either guide immune monitoring of humoral and cellular alloimmune and autoimmune markers, or to interpret the potential significance of a newly detected alloantigen or autoantigen reactivity for a transplanted islet β-cell graft before functional deterioration is evident. Because as much as 25% of the transplanted islet mass may be lost early following intraportal infusion (214), several approaches have been used to identify damaged islets in the circulation during this period, including by insulin mRNA (383, 384), glutamic acid decarboxylase-65 (385), miRNA375 (386), and unmethylated insulin DNA (387, 388). These studies have also been used to detect β-cell death in cultured islets prior to transplantation (385, 387), and so may be useful for investigations aimed at optimizing both islet viability following isolation and survival after infusion.

Importantly, elevated markers of β-cell injury at 24 hours after islet transplantation have been associated with worse islet graft functional outcomes within the first 3 months posttransplant (385, 388), and they can be modulated by anti-inflammatory therapy during the first week posttransplant (386), suggesting possible utility for both predicting early engraftment and assessing interventions aimed at improving islet survival during the engraftment period. Additionally, early persistent biomarker elevation has been associated with worse islet graft function at 3 months (387), and secondary “peaks” occurring later during follow-up may predict subsequent islet graft functional decline (384). In contrast to an increase in biomarker signifying islet β-cell injury and death, another approach monitors transplanted islet-specific exosomes isolated from peripheral blood samples using anti-HLA antibodies specific to the islet donor and where preliminary data suggest a decline in islet-specific exosome signal may be detectable prior to autoimmune recurrence (389). Further validation of these novel assays for monitoring posttransplant islet β-cell loss is required in both autoislet and alloislet settings, as well as for alloislets in conjunction with assays for detecting humoral and cellular alloimmunity and autoimmunity.

Conclusions

In conclusion, procedures for islet transplantation are established for patients requiring total or subtotal pancreatectomy for benign diseases of the pancreas where intraportal delivery of autologous islets may prevent or ameliorate postsurgical diabetes, and for patients with type 1 or pancreatogenic (type 3c) diabetes where intraportal delivery of allogeneic islets may establish on-target glycemic control often without the requirement for insulin therapy. The indication for total pancreatectomy with islet autotransplantation is most often to alleviate pain and improve quality of life for patients with recurrent acute or chronic pancreatitis, whereas for islet allotransplantation the indication is most often to alleviate severe hypoglycemia and improve quality of life for patients with excessive glycemic lability or hypoglycemia unawareness, as well as to improve glycemic control for patients already requiring immunosuppression in support of another transplanted organ (e.g., a kidney transplant). Insulin independence is experienced by about one-third of autoislet recipients, who are dependent on the number and health of the islets isolated from their own pancreas, and by about half of alloislet recipients, with the latter sometimes receiving islets from a second (or third) donor pancreas, and is dependent on the functional islet β-cell mass surviving engraftment. The improvement in outcomes for alloislets has been sufficiently robust to consider islet transplantation as a complementary alternative to solitary whole-pancreas transplantation, whereas insulin independence occurs more frequently when pancreas transplantation is performed simultaneous with kidney transplantation from the same organ donor. Novel approaches are under evaluation to further reduce the detrimental consequences of peritransplant inflammation on the intrahepatic engraftment efficiency of both autoislets and alloislets that may result in insulin independence more often and require less donor pancreases, as well as for calcineurin inhibitor–free immunosuppression that may have less potential for adverse effects on kidney function. Xenogeneic and stem cell–derived sources of islet tissue have entered early phase clinical trials, although much remains to be learned regarding the in vivo physiology and immunogenicity of the various products, as well as their potential compatibility with microencapsulation and macroencapsulation devices being tested for immunoisolation and cell retrieval and monitoring purposes. These advances provide more options for cellular therapy in the treatment of diabetes available now as autologous and allogeneic sourced tissue, and they promise potentially unlimited sources of islet tissue available for future application.

Abbreviations

    Abbreviations
     
  • cGMP

    current good manufacturing practice

  •  
  • cGTP

    current good tissue practice

  •  
  • B7

    Clinical Islet Transplantation Consortium

  •  
  • CMV

    cytomegalovirus

  •  
  • EBV

    Epstein-Barr virus

  •  
  • GFR

    glomerular filtration rate

  •  
  • GLP-1

    glucagon-like peptide-1

  •  
  • hESC

    human embryonic stem cell

  •  
  • HLA

    human leukocyte antigen

  •  
  • IAPP

    islet amyloid polypeptide

  •  
  • IE

    islet equivalent

  •  
  • mTOR

    mammalian target of rapamycin

  •  
  • OGTT

    oral glucose tolerance test

  •  
  • PTT

    partial thromboplastin time

  •  
  • TPIAT

    total pancreatectomy and islet autotransplantation

Acknowledgments

We thank Drs. Chengyang Liu and Ali Naji of the University of Pennsylvania Perelman School of Medicine (Philadelphia, PA) for performing a critical review of the manuscript.

Financial Support: This work was supported by Public Health Service Research Grants R01 DK 091331-7 (to M.R.R.), R01 DK 97830-5 (to M.R.R.), and R01 DK 39994-26 (to R.P.R.).

Disclosure Summary: The authors have nothing to disclose.

References

1.

Ballinger
WF
,
Lacy
PE
.
Transplantation of intact pancreatic islets in rats
.
Surgery
.
1972
;
72
:
175
186
.

2.

Reckard
CR
,
Barker
CF
.
Transplantation of isolated pancreatic-islets across strong and weak histocompatibility barriers
.
Transplant Proc
.
1973
;
5
:
761
763
.

3.

Barker
CF
,
Reckard
CR
,
Ziegler
MM
,
Naji
A
.
Liver as an immunologically privileged site for rat pancreatic-islet allografts
.
Diabetes
.
1975
;
24
:
418
.

4.

Kretschmer
GJ
,
Sutherland
DR
,
Matas
AJ
,
Payne
WD
,
Najarian
JS
.
Autotransplantation of pancreatic fragments to the portal vein and spleen of totally pancreatectomized dogs: a comparative evaluation
.
Ann Surg
.
1978
;
187
(
1
):
79
86
.

5.

Najarian
JS
,
Sutherland
DE
,
Baumgartner
D
,
Burke
B
,
Rynasiewicz
JJ
,
Matas
AJ
,
Goetz
FC
.
Total or near total pancreatectomy and islet autotransplantation for treatment of chronic pancreatitis
.
Ann Surg
.
1980
;
192
(
4
):
526
542
.

6.

Gray
DW
,
McShane
P
,
Grant
A
,
Morris
PJ
.
A method for isolation of islets of Langerhans from the human pancreas
.
Diabetes
.
1984
;
33
(
11
):
1055
1061
.

7.

Warnock
GL
,
Ellis
D
,
Rajotte
RV
,
Dawidson
I
,
Baekkeskov
S
,
Egebjerg
J
.
Studies of the isolation and viability of human islets of Langerhans
.
Transplantation
.
1988
;
45
(
5
):
957
963
.

8.

Ricordi
C
,
Lacy
PE
,
Finke
EH
,
Olack
BJ
,
Scharp
DW
.
Automated method for isolation of human pancreatic islets
.
Diabetes
.
1988
;
37
(
4
):
413
420
.

9.

Scharp
DW
,
Lacy
PE
,
Santiago
JV
,
McCullough
CS
,
Weide
LG
,
Falqui
L
,
Marchetti
P
,
Gingerich
RL
,
Jaffe
AS
,
Cryer
PE
,
Anderson
CB
,
Flye
MW
.
Insulin independence after islet transplantation into type I diabetic patient
.
Diabetes
.
1990
;
39
(
4
):
515
518
.

10.

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
.

11.

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
.

12.

Bertuzzi
F
,
Grohovaz
F
,
Maffi
P
,
Caumo
A
,
Aldrighetti
L
,
Nano
R
,
Hengster
P
,
Calori
G
,
Di Carlo
V
,
Bonifacio
E
,
Secchi
A
.
Succesful transplantation of human islets in recipients bearing a kidney graft
.
Diabetologia
.
2002
;
45
(
1
):
77
84
.

13.

Lehmann
R
,
Weber
M
,
Berthold
P
,
Zullig
R
,
Pfammatter
T
,
Moritz
W
,
Madler
K
,
Donath
M
,
Ambuhl
P
,
Demartines
N
,
Clavien
PA
,
Andreia Spinas
G
.
Successful simultaneous islet-kidney transplantation using a steroid-free immunosuppression: two-year follow-up
.
Am J Transplant
.
2004
;
4
(
7
):
1117
1123
.

14.

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
;
GRAGIL group
.
Sequential kidney/islet transplantation: efficacy and safety assessment of a steroid-free immunosuppression protocol
.
Am J Transplant
.
2006
;
6
(
5 Pt 1
):
1049
1058
.

15.

Kaufman
DB
,
Baker
MS
,
Chen
X
,
Leventhal
JR
,
Stuart
FP
.
Sequential kidney/islet transplantation using prednisone-free immunosuppression
.
Am J Transplant
.
2002
;
2
(
7
):
674
677
.

16.

Markmann
JF
,
Deng
S
,
Huang
X
,
Desai
NM
,
Velidedeoglu
EH
,
Lui
C
,
Frank
A
,
Markmann
E
,
Palanjian
M
,
Brayman
K
,
Wolf
B
,
Bell
E
,
Vitamaniuk
M
,
Doliba
N
,
Matschinsky
F
,
Barker
CF
,
Naji
A
.
Insulin independence following isolated islet transplantation and single islet infusions
.
Ann Surg
.
2003
;
237
(
6
):
741
749
.

17.

Hering
BJ
,
Kandaswamy
R
,
Harmon
JV
,
Ansite
JD
,
Clemmings
SM
,
Sakai
T
,
Paraskevas
S
,
Eckman
PM
,
Sageshima
J
,
Nakano
M
,
Sawada
T
,
Matsumoto
I
,
Zhang
HJ
,
Sutherland
DE
,
Bluestone
JA
.
Transplantation of cultured islets from two-layer preserved pancreases in type 1 diabetes with anti-CD3 antibody
.
Am J Transplant
.
2004
;
4
(
3
):
390
401
.

18.

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
.

19.

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
.

20.

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
;
TRIMECO trial investigators
.
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
.

21.

Larsen
JL
.
Pancreas transplantation: Indications and consequences
.
Endocr Rev
.
2004
;
25
(
6
):
919
946
.

22.

White
SA
,
Sutton
CD
,
Weymss-Holden
S
,
Berry
DP
,
Pollard
C
,
Rees
Y
,
Dennison
AR
.
The feasibility of spleen-preserving pancreatectomy for end-stage chronic pancreatitis
.
Am J Surg
.
2000
;
179
(
4
):
294
297
.

23.

Bucher
P
,
Mathe
Z
,
Morel
P
,
Bosco
D
,
Andres
A
,
Kurfuest
M
,
Friedrich
O
,
Raemsch-Guenther
N
,
Buhler
LH
,
Berney
T
.
Assessment of a novel two-component enzyme preparation for human islet isolation and transplantation
.
Transplantation
.
2005
;
79
(
1
):
91
97
.

24.

Szot
GL
,
Lee
MR
,
Tavakol
MM
,
Lang
J
,
Dekovic
F
,
Kerlan
RK
,
Stock
PG
,
Posselt
AM
.
Successful clinical islet isolation using a GMP-manufactured collagenase and neutral protease
.
Transplantation
.
2009
;
88
(
6
):
753
756
.

25.

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
.

26.

Bottino
R
,
Bertera
S
,
Grupillo
M
,
Melvin
PR
,
Humar
A
,
Mazariegos
G
,
Moser
AJ
,
Walsh
RM
,
Fung
J
,
Gelrud
A
,
Slivka
A
,
Soltys
K
,
Wijkstrom
M
,
Trucco
M
.
Isolation of human islets for autologous islet transplantation in children and adolescents with chronic pancreatitis
.
J Transplant
.
2012
;
2012
:
642787
.

27.

Pinkse
GG
,
Bouwman
WP
,
Jiawan-Lalai
R
,
Terpstra
OT
,
Bruijn
JA
,
de Heer
E
.
Integrin signaling via RGD peptides and anti-beta1 antibodies confers resistance to apoptosis in islets of Langerhans
.
Diabetes
.
2006
;
55
(
2
):
312
317
.

28.

Wang
H
,
Desai
KD
,
Dong
H
,
Owzarski
S
,
Romagnuolo
J
,
Morgan
KA
,
Adams
DB
.
Prior surgery determines islet yield and insulin requirement in patients with chronic pancreatitis
.
Transplantation
.
2013
;
95
(
8
):
1051
1057
.

29.

Sutherland
DER
,
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
.

30.

Rabkin
JM
,
Olyaei
AJ
,
Orloff
SL
,
Geisler
SM
,
Wahoff
DC
,
Hering
BJ
,
Sutherland
DE
.
Distant processing of pancreas islets for autotransplantation following total pancreatectomy
.
Am J Surg
.
1999
;
177
(
5
):
423
427
.

31.

Tai
DS
,
Shen
N
,
Szot
GL
,
Posselt
A
,
Feduska
NJ
,
Habashy
A
,
Clerkin
B
,
Core
E
,
Busuttil
RW
,
Hines
OJ
,
Reber
HA
,
Lipshutz
GS
.
Autologous islet transplantation with remote islet isolation after pancreas resection for chronic pancreatitis
.
JAMA Surg
.
2015
;
150
(
2
):
118
124
.

32.

Johnston
PC
,
Lin
YK
,
Walsh
RM
,
Bottino
R
,
Stevens
TK
,
Trucco
M
,
Bena
J
,
Faiman
C
,
Hatipoglu
BA
.
Factors associated with islet yield and insulin independence after total pancreatectomy and islet cell autotransplantation in patients with chronic pancreatitis utilizing off-site islet isolation: Cleveland Clinic experience
.
J Clin Endocrinol Metab
.
2015
;
100
(
5
):
1765
1770
.

33.

Rodriguez Rilo
HL
,
Ahmad
SA
,
D’Alessio
D
,
Iwanaga
Y
,
Kim
J
,
Choe
KA
,
Moulton
JS
,
Martin
J
,
Pennington
LJ
,
Soldano
DA
,
Biliter
J
,
Martin
SP
,
Ulrich
CD
,
Somogyi
L
,
Welge
J
,
Matthews
JB
,
Lowy
AM
.
Total pancreatectomy and autologous islet cell transplantation as a means to treat severe chronic pancreatitis
.
J Gastrointest Surg
.
2003
;
7
(
8
):
978
989
.

34.

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
.

35.

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
.

36.

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
.

37.

Robertson
RP
.
Total pancreatectomy and islet autotransplantation for chronic pancreatitis: breaking down barriers
.
J Clin Endocrinol Metab
.
2015
;
100
(
5
):
1762
1763
.

38.

Griffith
RC
,
Scharp
DW
,
Hartman
BK
,
Ballinger
WF
,
Lacy
PE
.
Morphologic study of intrahepatic portal-vein islet isografts
.
Diabetes
.
1977
;
26
(
3
):
201
214
.

39.

Andersson
A
,
Korsgren
O
,
Jansson
L
.
Intraportally transplanted pancreatic-islets revascularized from hepatic arterial system
.
Diabetes
.
1989
;
38
(
Suppl 1
):
192
195
.

40.

Gardemann
A
,
Jungermann
K
,
Grosse
V
,
Cossel
L
,
Wohlrab
F
,
Hahn
HJ
,
Blech
W
,
Hildebrandt
W
.
Intraportal transplantation of pancreatic-islets into livers of diabetic rats–reinnervation of islets and regulation of insulin-secretion by the hepatic sympathetic-nerves
.
Diabetes
.
1994
;
43
(
11
):
1345
1352
.

41.

Meier
JJ
,
Hong-McAtee
I
,
Galasso
R
,
Veldhuis
JD
,
Moran
A
,
Hering
BJ
,
Butler
PC
.
Intrahepatic transplanted islets in humans secrete insulin in a coordinate pulsatile manner directly into the liver
.
Diabetes
.
2006
;
55
(
8
):
2324
2332
.

42.

Kaufman
DB
,
Morel
P
,
Field
MJ
,
Munn
SR
,
Sutherland
DE
.
Purified canine islet autografts—functional outcome as influenced by islet number and implantation site
.
Transplantation
.
1990
;
50
(
3
):
385
391
.

43.

Naziruddin
B
,
Iwahashi
S
,
Kanak
MA
,
Takita
M
,
Itoh
T
,
Levy
MF
.
Evidence for instant blood-mediated inflammatory reaction in clinical autologous islet transplantation
.
Am J Transplant
.
2014
;
14
(
2
):
428
437
.

44.

Matsumoto
S
,
Takita
M
,
Shimoda
M
,
Sugimoto
K
,
Itoh
T
,
Chujo
D
,
SoRelle
JA
,
Tamura
Y
,
Rahman
AM
,
Onaca
N
,
Naziruddin
B
,
Levy
MF
.
Impact of tissue volume and purification on clinical autologous islet transplantation for the treatment of chronic pancreatitis
.
Cell Transplant
.
2012
;
21
(
4
):
625
632
.

45.

Wilhelm
JJ
,
Bellin
MD
,
Dunn
TB
,
Balamurugan
AN
,
Pruett
TL
,
Radosevich
DM
,
Chinnakotla
S
,
Schwarzenberg
SJ
,
Freeman
ML
,
Hering
BJ
,
Sutherland
DE
,
Beilman
GJ
.
Proposed thresholds for pancreatic tissue volume for safe intraportal islet autotransplantation after total pancreatectomy
.
Am J Transplant
.
2013
;
13
(
12
):
3183
3191
.

46.

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
.

47.

Rafael
E
,
Tibell
A
,
Ryden
M
,
Lundgren
T
,
Savendahl
L
,
Borgstrom
B
,
Arnelo
U
,
Isaksson
B
,
Nilsson
B
,
Korsgren
O
,
Permert
J
.
Intramuscular autotransplantation of pancreatic islets in a 7-year-old child: a 2-year follow-up
.
Am J Transplant
.
2008
;
8
(
2
):
458
462
.

48.

Bertuzzi
F
,
Colussi
G
,
Lauterio
A
,
De Carlis
L
.
Intramuscular islet allotransplantation in type 1 diabetes mellitus
.
Eur Rev Med Pharmacol Sci
.
2018
;
22
:
1731
1736
.

49.

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 [published correction appears in Diabetes. 2014;63:377]
.
Diabetes
.
2013
;
62
(
10
):
3523
3531
.

50.

Cantarelli
E
,
Citro
A
,
Pellegrini
S
,
Mercalli
A
,
Melzi
R
,
Dugnani
E
,
Jofra
T
,
Fousteri
G
,
Mondino
A
,
Piemonti
L
.
Transplant site influences the immune response after islet transplantation: bone marrow versus liver
.
Transplantation
.
2017
;
101
(
5
):
1046
1055
.

51.

Gupta
V
,
Wahoff
DC
,
Rooney
DP
,
Poitout
V
,
Sutherland
DE
,
Kendall
DM
,
Robertson
RP
.
The defective glucagon response from transplanted intrahepatic pancreatic islets during hypoglycemia is transplantation site-determined
.
Diabetes
.
1997
;
46
(
1
):
28
33
.

52.

Bellin
MD
,
Parazzoli
S
,
Oseid
E
,
Bogachus
LD
,
Schuetz
C
,
Patti
ME
,
Dunn
T
,
Pruett
T
,
Balamurugan
AN
,
Hering
B
,
Beilman
G
,
Sutherland
DE
,
Robertson
RP
.
Defective glucagon secretion during hypoglycemia after intrahepatic but not nonhepatic islet autotransplantation
.
Am J Transplant
.
2014
;
14
(
8
):
1880
1886
.

53.

Lee
TC
,
Barshes
NR
,
Brunicardi
FC
,
Alejandro
R
,
Ricordi
C
,
Nguyen
L
,
Goss
JA
.
Procurement of the human pancreas for pancreatic islet transplantation
.
Transplantation
.
2004
;
78
(
3
):
481
483
.

54.

Ihm
SH
,
Matsumoto
I
,
Sawada
T
,
Nakano
M
,
Zhang
HJ
,
Ansite
JD
,
Sutherland
DE
,
Hering
BJ
.
Effect of donor age on function of isolated human islets
.
Diabetes
.
2006
;
55
(
5
):
1361
1368
.

55.

Niclauss
N
,
Bosco
D
,
Morel
P
,
Demuylder-Mischler
S
,
Brault
C
,
Milliat-Guittard
L
,
Colin
C
,
Parnaud
G
,
Muller
YD
,
Giovannoni
L
,
Meier
R
,
Toso
C
,
Badet
L
,
Benhamou
PY
,
Berney
T
.
Influence of donor age on islet isolation and transplantation outcome
.
Transplantation
.
2011
;
91
(
3
):
360
366
.

56.

Hering
BJ
.
Achieving and maintaining insulin independence in human islet transplant recipients
.
Transplantation
.
2005
;
79
(
10
):
1296
1297
.

57.

Lakey
JR
,
Warnock
GL
,
Rajotte
RV
,
Suarez-Alamazor
ME
,
Ao
Z
,
Shapiro
AM
,
Kneteman
NM
.
Variables in organ donors that affect the recovery of human islets of Langerhans
.
Transplantation
.
1996
;
61
(
7
):
1047
1053
.

58.

Brooks
AM
,
Walker
N
,
Aldibbiat
A
,
Hughes
S
,
Jones
G
,
de Havilland
J
,
Choudhary
P
,
Huang
GC
,
Parrott
N
,
Mcgowan
NW
,
Casey
J
,
Mumford
L
,
Barker
P
,
Burling
K
,
Hovorka
R
,
Walker
M
,
Smith
RM
,
Forbes
S
,
Rutter
MK
,
Amiel
S
,
Rosenthal
MJ
,
Johnson
P
,
Shaw
JA
.
Attainment of metabolic goals in the integrated UK islet transplant program with locally isolated and transported preparations
.
Am J Transplant
.
2013
;
13
(
12
):
3236
3243
.

59.

Balamurugan
AN
,
Naziruddin
B
,
Lockridge
A
,
Tiwari
M
,
Loganathan
G
,
Takita
M
,
Matsumoto
S
,
Papas
K
,
Trieger
M
,
Rainis
H
,
Kin
T
,
Kay
TW
,
Wease
S
,
Messinger
S
,
Ricordi
C
,
Alejandro
R
,
Markmann
J
,
Kerr-Conti
J
,
Rickels
MR
,
Liu
C
,
Zhang
X
,
Witkowski
P
,
Posselt
A
,
Maffi
P
,
Secchi
A
,
Berney
T
,
O’Connell
PJ
,
Hering
BJ
,
Barton
FB
.
Islet product characteristics and factors related to successful human islet transplantation from the Collaborative Islet Transplant Registry (CITR) 1999–2010
.
Am J Transplant
.
2014
;
14
(
11
):
2595
2606
.

60.

Markmann
JF
,
Deng
SP
,
Desai
NM
,
Huang
XL
,
Velidedeoglu
E
,
Frank
A
,
Liu
CY
,
Brayman
KL
,
Lian
MM
,
Wolf
B
,
Bell
E
,
Vitamaniuk
M
,
Doliba
N
,
Matschensky
F
,
Markmann
E
,
Barker
CF
,
Naji
A
.
The use of non-heart-beating donors for isolated pancreatic islet transplantation
.
Transplantation
.
2003
;
75
(
9
):
1423
1429
.

61.

Saito
T
,
Gotoh
M
,
Satomi
S
,
Uemoto
S
,
Kenmochi
T
,
Itoh
T
,
Kuroda
Y
,
Yasunami
Y
,
Matsumoto
S
,
Teraoka
S
;
Working Members of the Japanese Pancreas and Islet Transplantation Association
.
Islet transplantation using donors after cardiac death: report of the Japan Islet Transplantation Registry
.
Transplantation
.
2010
;
90
(
7
):
740
747
.

62.

Berney
T
,
Boffa
C
,
Augustine
T
,
Badet
L
,
de Koning
E
,
Pratschke
J
,
Socci
C
,
Friend
P
.
Utilization of organs from donors after circulatory death for vascularized pancreas and islet of Langerhans transplantation: recommendations from an expert group
.
Transpl Int
.
2016
;
29
(
7
):
798
806
.

63.

Ricordi
C
,
Goldstein
JS
,
Balamurugan
AN
,
Szot
GL
,
Kin
T
,
Liu
C
,
Czarniecki
CW
,
Barbaro
B
,
Bridges
ND
,
Cano
J
,
Clarke
WR
,
Eggerman
TL
,
Hunsicker
LG
,
Kaufman
DB
,
Khan
A
,
Lafontant
DE
,
Linetsky
E
,
Luo
X
,
Markmann
JF
,
Naji
A
,
Korsgren
O
,
Oberholzer
J
,
Turgeon
NA
,
Brandhorst
D
,
Chen
X
,
Friberg
AS
,
Lei
J
,
Wang
LJ
,
Wilhelm
JJ
,
Willits
J
,
Zhang
X
,
Hering
BJ
,
Posselt
AM
,
Stock
PG
,
Shapiro
AM
,
Chen
X
.
National Institutes of Health–sponsored Clinical Islet Transplantation Consortium phase 3 trial: manufacture of a complex cellular product at eight processing facilities
.
[published correction appears in Diabetes. 2017;66(9):2531]
Diabetes
.
2016
;
65
(
11
):
3418
3428
.

64.

Markmann
JF
,
Tomaszewski
J
,
Posselt
AM
,
Levy
MM
,
Woehrle
M
,
Barker
CF
,
Naji
A
.
The effect of islet cell-culture in vitro at 24°C on graft-survival and MHC antigen expression
.
Transplantation
.
1990
;
49
(
2
):
272
277
.

65.

Piemonti
L
,
Leone
BE
,
Nano
R
,
Saccani
A
,
Monti
P
,
Maffi
P
,
Bianchi
G
,
Sica
A
,
Peri
G
,
Melzi
R
,
Aldrighetti
L
,
Secchi
A
,
Di Carlo
V
,
Allavena
P
,
Bertuzzi
F
.
Human pancreatic islets produce and secrete MCP-1/CCL2: relevance in human islet transplantation
.
Diabetes
.
2002
;
51
(
1
):
55
65
.

66.

Moberg
L
,
Johansson
H
,
Lukinius
A
,
Berne
C
,
Foss
A
,
Kallen
R
,
Ostraat
O
,
Salmela
K
,
Tibell
A
,
Tufveson
G
,
Elgue
G
,
Nilsson
EK
,
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
.

67.

Bertuzzi
F
,
Marzorati
S
,
Maffi
P
,
Piemonti
L
,
Melzi
R
,
de Taddeo
F
,
Valtolina
V
,
D’Angelo
A
,
di Carlo
V
,
Bonifacio
E
,
Secchi
A
.
Tissue factor and CCL2/monocyte chemoattractant protein-1 released by human islets affect islet engraftment in type 1 diabetic recipients
.
J Clin Endocrinol Metab
.
2004
;
89
(
11
):
5724
5728
.

68.

Marzorati
S
,
Antonioli
B
,
Nano
R
,
Maffi
P
,
Piemonti
L
,
Giliola
C
,
Secchi
A
,
Lakey
JR
,
Bertuzzi
F
.
Culture medium modulates proinflammatory conditions of human pancreatic islets before transplantation
.
Am J Transplant
.
2006
;
6
(
11
):
2791
2795
.

69.

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
XR
,
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
.

70.

Goss
JA
,
Soltes
G
,
Goodpastor
SE
,
Barth
M
,
Lam
R
,
Brunicardi
FC
,
Froud
T
,
Alejandro
R
,
Ricordi
C
.
Pancreatic islet transplantation: the radiographic approach
.
Transplantation
.
2003
;
76
(
1
):
199
203
.

71.

Owen
RJ
,
Ryan
EA
,
O’Kelly
K
,
Lakey
JR
,
McCarthy
MC
,
Paty
BW
,
Bigam
DL
,
Kneteman
NM
,
Korbutt
GS
,
Rajotte
RV
,
Shapiro
AM
.
Percutaneous transhepatic pancreatic islet cell transplantation in type 1 diabetes mellitus: radiologic aspects
.
Radiology
.
2003
;
229
(
1
):
165
170
.

72.

Gaber
AO
,
Chamsuddin
A
,
Fraga
D
,
Fisher
J
,
Lo
A
.
Insulin independence achieved using the transmesenteric approach to the portal vein for islet transplantation
.
Transplantation
.
2004
;
77
(
2
):
309
311
.

73.

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
:
1761
1766
.

74.

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
.

75.

Kessler
L
,
Bucher
P
,
Milliat-Guittard
L
,
Benhamou
PY
,
Berney
T
,
Penfornis
A
,
Badet
L
,
Thivolet
C
,
Bayle
F
,
Oberholzer
J
,
Renoult
E
,
Brun
MJ
,
Rifle
G
,
Atlan
C
,
Colin
C
,
Morel
P
,
Group
G
.
Influence of islet transportation on pancreatic islet allotransplantation in type 1 diabetic patients within the Swiss-French GRAGIL network
.
Transplantation
.
2004
;
77
(
8
):
1301
1304
.

76.

O’Connell
PJ
,
Holmes-Walker
DJ
,
Goodman
D
,
Hawthorne
WJ
,
Loudovaris
T
,
Gunton
JE
,
Thomas
HE
,
Grey
ST
,
Drogemuller
CJ
,
Ward
GM
,
Torpy
DJ
,
Coates
PT
,
Kay
TW
.
Multicenter Australian trial of islet transplantation: improving accessibility and outcomes
.
Am J Transplant
.
2013
;
13
(
7
):
1850
1858
.

77.

Casey
JJ
,
Lakey
JRT
,
Ryan
EA
,
Paty
BW
,
Owen
R
,
O’Kelly
K
,
Nanji
S
,
Rajotte
RV
,
Korbutt
GS
,
Bigam
D
,
Kneteman
NN
,
Shapiro
AMJ
.
Portal venous pressure changes after sequential clinical islet transplantation
.
Transplantation
.
2002
;
74
:
913
915
.

78.

Toso
C
,
Isse
K
,
Demetris
AJ
,
Dinyari
P
,
Koh
A
,
Imes
S
,
Kin
T
,
Emamaullee
J
,
Senior
P
,
Shapiro
AM
.
Histologic graft assessment after clinical islet transplantation
.
Transplantation
.
2009
;
88
(
11
):
1286
1293
.

79.

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
.

80.

Sjoberg
RJ
,
Kidd
GS
.
Pancreatic diabetes mellitus
.
Diabetes Care
.
1989
;
12
(
10
):
715
724
.

81.

Gudipaty
LR
,
Rickels
MR
. Pancreatogenic (type 3c) diabetes.
Pancreapedia: Exocrine Pancreas Knowledge Base
.
American Pancreatic Association
;
2015
. doi: 10.3998/panc.2015.35.

82.

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
.

83.

Whitcomb
DC
,
Gorry
MC
,
Preston
RA
,
Furey
W
,
Sossenheimer
MJ
,
Ulrich
CD
,
Martin
SP
,
Gates
LK
,
Amann
ST
,
Toskes
PP
,
Liddle
R
,
McGrath
K
,
Uomo
G
,
Post
JC
,
Ehrlich
GD
.
Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene
.
Nat Genet
.
1996
;
14
(
2
):
141
145
.

84.

Schneider
A
,
LaRusch
J
,
Sun
XM
,
Aloe
A
,
Lamb
J
,
Hawes
R
,
Cotton
P
,
Brand
RE
,
Anderson
MA
,
Money
ME
,
Banks
PA
,
Lewis
MD
,
Baillie
J
,
Sherman
S
,
DiSario
J
,
Burton
FR
,
Gardner
TB
,
Amann
ST
,
Gelrud
A
,
George
R
,
Rockacy
MJ
,
Kassabian
S
,
Martinson
J
,
Slivka
A
,
Yadav
D
,
Oruc
N
,
Barmada
MM
,
Frizzell
R
,
Whitcomb
DC
.
Combined bicarbonate conductance-impairing variants in CFTR and SPINK1 variants are associated with chronic pancreatitis in patients without cystic fibrosis
.
Gastroenterology
.
2011
;
140
(
1
):
162
171
.

85.

Whitcomb
DC
.
Genetic risk factors for pancreatic disorders
.
Gastroenterology
.
2013
;
144
(
6
):
1292
1302
.

86.

Cahen
DL
,
Gouma
DJ
,
Nio
Y
,
Rauws
EA
,
Boermeester
MA
,
Busch
OR
,
Stoker
J
,
Lameris
JS
,
Dijkgraaf
MG
,
Huibregtse
K
,
Bruno
MJ
.
Endoscopic versus surgical drainage of the pancreatic duct in chronic pancreatitis
.
N Engl J Med
.
2007
;
356
(
7
):
676
684
.

87.

Moran
RA
,
Klapheke
R
,
John
GK
,
Devlin
S
,
Warren
D
,
Desai
N
,
Sun
Z
,
Walsh
C
,
Kalyani
RR
,
Hall
E
,
Stein
EM
,
Kalloo
AN
,
Zaheer
A
,
Hirose
K
,
Makary
MA
,
Singh
VK
.
Prevalence and predictors of pain and opioid analgesic use following total pancreatectomy with islet autotransplantation for pancreatitis
.
Pancreatology
.
2017
;
17
(
5
):
732
737
.

88.

Oberholzer
J
,
Triponez
F
,
Mage
R
,
Andereggen
E
,
Bühler
L
,
Crétin
N
,
Fournier
B
,
Goumaz
C
,
Lou
J
,
Philippe
J
,
Morel
P
.
Human islet transplantation: lessons from 13 autologous and 13 allogeneic transplantations
.
Transplantation
.
2000
;
69
:
1115
1123
.

89.

Lee
BW
,
Jee
JH
,
Heo
JS
,
Choi
SH
,
Jang
KT
,
Noh
JH
,
Jeong
IK
,
Oh
SH
,
Ahn
YR
,
Chae
HY
,
Min
YK
,
Chung
JH
,
Lee
MK
,
Lee
MS
,
Kim
KW
.
The favorable outcome of human islet transplantation in Korea: experiences of 10 autologous transplantations
.
Transplantation
.
2005
;
79
(
11
):
1568
1574
.

90.

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
.

91.

Oberholzer
J
,
Mathe
Z
,
Bucher
P
,
Triponez
F
,
Bosco
D
,
Fournier
B
,
Majno
P
,
Philippe
J
,
Morel
P
.
Islet autotransplantation after left pancreatectomy for non-enucleable insulinoma
.
Am J Transplant
.
2003
;
3
(
10
):
1302
1307
.

92.

Jindal
RM
,
Ricordi
C
,
Shriver
CD
.
Autologous pancreatic islet transplantation for severe trauma
.
N Engl J Med
.
2010
;
362
(
16
):
1550
.

93.

Khan
A
,
Jindal
RM
,
Shriver
C
,
Guy
SR
,
Vertrees
AE
,
Wang
X
,
Xu
X
,
Szust
J
,
Ricordi
C
.
Remote processing of pancreas can restore normal glucose homeostasis in autologous islet transplantation after traumatic whipple pancreatectomy: technical considerations
.
Cell Transplant
.
2012
;
21
(
6
):
1261
1267
.

94.

Gala-Lopez
BL
,
Semlacher
E
,
Manouchehri
N
,
Kin
T
,
Shapiro
AM
.
Autologous islet transplantation after total pancreatectomy for renal cell carcinoma metastases
.
Am J Transplant
.
2013
;
13
(
9
):
2487
2491
.

95.

Förster
S
,
Liu
X
,
Adam
U
,
Schareck
WD
,
Hopt
UT
.
Islet autotransplantation combined with pancreatectomy for treatment of pancreatic adenocarcinoma: a case report
.
Transplant Proc
.
2004
;
36
(
4
):
1125
1126
.

96.

Alsaif
F
,
Molinari
M
,
Al-Masloom
A
,
Lakey
JR
,
Kin
T
,
Shapiro
AM
.
Pancreatic islet autotransplantation with completion pancreatectomy in the management of uncontrolled pancreatic fistula after whipple resection for ampullary adenocarcinoma
.
Pancreas
.
2006
;
32
(
4
):
430
431
.

97.

Iyegha
UP
,
Asghar
JA
,
Beilman
GJ
.
Total pancreatectomy and islet auto-transplantation as treatment for ampullary adenocarcinoma in the setting of pancreatic ductal disruption secondary to acute necrotizing pancreatitis. A case report
.
JOP
.
2012
;
13
:
239
242
.

98.

Savari
O
,
Golab
K
,
Solomina
J
,
Konsur
E
,
Cieply
K
,
Ramachandran
S
,
Tekin
Z
,
Schenck
L
,
Zhang
SS
,
Tibudan
M
,
Posner
MC
,
Witkowski
P
.
Total pancreatectomy with islet autotransplantation for the ampullary cancer. A case report [published online ahead of print 21 November 2017]
.
J Gastrointest Cancer
doi: 10.1007/s12029-017-0029-4.

99.

Gerich
JE
,
Langlois
M
,
Noacco
C
,
Karam
JH
,
Forsham
PH
.
Lack of glucagon response to hypoglycemia in diabetes—evidence for an intrinsic pancreatic alpha cell defect
.
Science
.
1973
;
182
(
4108
):
171
173
.

100.

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
.

101.

Bergenstal
RM
,
Garg
S
,
Weinzimer
SA
,
Buckingham
BA
,
Bode
BW
,
Tamborlane
WV
,
Kaufman
FR
.
Safety of a hybrid closed-loop insulin delivery system in patients with type 1 diabetes
.
JAMA
.
2016
;
316
(
13
):
1407
1408
.

102.

Rickels
MR
,
Stock
PG
,
de Koning
EJP
,
Piemonti
L
,
Pratschke
J
,
Alejandro
R
,
Bellin
MD
,
Berney
T
,
Choudhary
P
,
Johnson
PR
,
Kandaswamy
R
,
Kay
TW
,
Keymeulen
B
,
Kudva
YC
,
Latres
E
,
Langer
RM
,
Lehmann
R
,
Ludwig
B
,
Markmann
JF
,
Marinac
M
,
Odorico
JS
,
Pattou
F
,
Senior
PA
,
Shaw
JA
,
Vantyghem
MC
,
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
.
Transpl Int
.
2018
;
31
(
4
):
343
352
.

103.

Rickels
MR
,
Stock
PG
,
de Koning
EJ
,
Piemonti
L
,
Pratschke
J
,
Alejandro
R
,
Bellin
MD
,
Berney
T
,
Choudhary
P
,
Johnson
PR
,
Kandaswamy
R
,
Kay
TW
,
Keymeulen
B
,
Kudva
YC
,
Latres
E
,
Langer
RM
,
Lehmann
R
,
Ludwig
B
,
Markmann
JF
,
Marinac
M
,
Odorico
JS
,
Pattou
F
,
Senior
PA
,
Shaw
JA
,
Vantyghem
MC
,
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
;
102
(
9
):
1479
1486
.

104.

Gillard
P
,
Ling
Z
,
Lannoo
M
,
Maes
B
,
Maleux
G
,
Pipeleers
D
,
Keymeulen
B
,
Mathieu
C
.
β-Cell transplantation restores metabolic control and quality of life in a patient with subcutaneous insulin resistance
.
Diabetes Care
.
2004
;
27
(
9
):
2243
2244
.

105.

Leone
JP
,
Kendall
DM
,
Reinsmoen
N
,
Hering
BJ
,
Sutherland
DE
.
Immediate insulin-independence after retransplantation of islets prepared from an allograft pancreatectomy in a type 1 diabetic patient
.
Transplant Proc
.
1998
;
30
(
2
):
319
.

106.

Nijhoff
MF
,
Dubbeld
J
,
van Erkel
AR
,
van der Boog
PJM
,
Rabelink
TJ
,
Engelse
MA
,
de Koning
EJ
.
Islet alloautotransplantation: allogeneic pancreas transplantation followed by transplant pancreatectomy and islet transplantation
.
Am J Transplant
.
2018
;
18
(
4
):
1016
1019
.

107.

Kessler
L
,
Bakopoulou
S
,
Kessler
R
,
Massard
G
,
Santelmo
N
,
Greget
M
,
Moreau
F
,
Helms
O
,
Bosco
D
,
Gasche-Soccal
P
,
Morel
P
,
Wolf
P
,
Berney
T
;
GRAGIL group
.
Combined pancreatic islet-lung transplantation: a novel approach to the treatment of end-stage cystic fibrosis
.
Am J Transplant
.
2010
;
10
(
7
):
1707
1712
.

108.

Klee
P
,
Dirlewanger
M
,
Lavallard
V
,
McLin
VA
,
Mornand
A
,
Pernin
N
,
Petit
LM
,
Soccal
PM
,
Wildhaber
BE
,
Zumsteg
U
,
Blouin
JL
,
Berney
T
,
Schwitzgebel
VM
.
Combined pancreatic islet-lung-liver transplantation in a pediatric patient with cystic fibrosis-related diabetes
.
Horm Res Paediatr
.
2018
;
90
(
4
):
270
274
.

109.

Spijker
HS
,
Wolffenbuttel
BH
,
van der Bij
W
,
Engelse
MA
,
Rabelink
TJ
,
de Koning
EJ
.
Islet-after-lung transplantation in a patient with cystic fibrosis-related diabetes
.
Diabetes Care
.
2014
;
37
(
7
):
e159
e160
.

110.

Hackman
KL
,
Bailey
MJ
,
Snell
GI
,
Bach
LA
.
Diabetes is a major risk factor for mortality after lung transplantation
.
Am J Transplant
.
2014
;
14
(
2
):
438
445
.

111.

Biarnes
M
,
Montolio
M
,
Nacher
V
,
Raurell
M
,
Soler
J
,
Montanya
E
.
β-Cell death and mass in syngeneically transplanted islets exposed to short- and long-term hyperglycemia
.
Diabetes
.
2002
;
51
(
1
):
66
72
.

112.

Juang
JH
,
Bonner-Weir
S
,
Wu
YJ
,
Weir
GC
.
Beneficial influence of glycemic control upon the growth and function of transplanted islets
.
Diabetes
.
1994
;
43
(
11
):
1334
1339
.

113.

Davalli
AM
,
Ricordi
C
,
Socci
C
,
Braghi
S
,
Bertuzzi
F
,
Fattor
B
,
Di
CV
,
Pontiroli
AE
,
Pozza
G
.
Abnormal sensitivity to glucose of human islets cultured in a high glucose medium: partial reversibility after an additional culture in a normal glucose medium
.
J Clin Endocrinol Metab
.
1991
;
72
(
1
):
202
208
.

114.

Dubois
S
,
Madec
AM
,
Mesnier
A
,
Armanet
M
,
Chikh
K
,
Berney
T
,
Thivolet
C
.
Glucose inhibits angiogenesis of isolated human pancreatic islets
.
J Mol Endocrinol
.
2010
;
45
(
2
):
99
105
.

115.

Yumiba
T
,
Miyata
M
,
Izukura
M
,
Tanaka
Y
,
Dousei
T
,
Yamaguchi
T
,
Matsuda
H
.
Islet rest protects against exhaustion of insulin production in transplanted islets
.
Transplant Proc
.
1992
;
24
:
995
996
.

116.

Merino
JF
,
Nacher
V
,
Raurell
M
,
Biarnes
M
,
Soler
J
,
Montanya
E
.
Optimal insulin treatment in syngeneic islet transplantation
.
Cell Transplant
.
2000
;
9
(
1
):
11
18
.

117.

Struyvenberg
MR
,
Martin
CR
,
Freedman
SD
.
Practical guide to exocrine pancreatic insufficiency—breaking the myths
.
BMC Med
.
2017
;
15
(
1
):
29
.

118.

Grau
T
,
Bonet
A
,
Rubio
M
,
Mateo
D
,
Farre
M
,
Acosta
JA
,
Blesa
A
,
Montejo
JC
,
de Lorenzo
AG
,
Mesejo
A
;
Working Group on Nutrition and Metabolism of the Spanish Society of Critical Care
.
Liver dysfunction associated with artificial nutrition in critically ill patients
.
Crit Care
.
2007
;
11
(
1
):
R10
.

119.

Rubin
LG
,
Schaffner
W
.
Clinical practice. Care of the asplenic patient
.
N Engl J Med
.
2014
;
371
(
4
):
349
356
.

120.

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
.

121.

Makhlouf
L
,
Duvivier-Kali
VF
,
Bonner-Weir
S
,
Dieperink
H
,
Weir
GC
,
Sayegh
MH
.
Importance of hyperglycemia on the primary function of allogeneic islet transplants
.
Transplantation
.
2003
;
76
(
4
):
657
664
.

122.

Jackson
AM
,
Connolly
JE
,
Matsumoto
S
,
Noguchi
H
,
Onaca
N
,
Levy
MF
,
Naziruddin
B
.
Evidence for induced expression of HLA class II on human islets: possible mechanism for HLA sensitization in transplant recipients
.
Transplantation
.
2009
;
87
(
4
):
500
506
.

123.

Strieter
RM
,
Remick
DG
,
Ward
PA
,
Spengler
RN
,
Lynch
JP
,
Larrick
J
,
Kunkel
SL
.
Cellular and molecular regulation of tumor necrosis factor-alpha production by pentoxifylline
.
Biochem Biophys Res Commun
.
1988
;
155
(
3
):
1230
1236
.

124.

Koh
A
,
Senior
P
,
Salam
A
,
Kin
T
,
Imes
S
,
Dinyari
P
,
Malcolm
A
,
Toso
C
,
Nilsson
B
,
Korsgren
O
,
Shapiro
AM
.
Insulin-heparin infusions peritransplant substantially improve single-donor clinical islet transplant success
.
Transplantation
.
2010
;
89
(
4
):
465
471
.

125.

Rafael
E
,
Ryan
EA
,
Paty
BW
,
Oberholzer
J
,
Imes
S
,
Senior
P
,
McDonald
C
,
Lakey
JR
,
Shapiro
AM
.
Changes in liver enzymes after clinical islet transplantation
.
Transplantation
.
2003
;
76
(
9
):
1280
1284
.

126.

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
.

127.

Faradji
RN
,
Monroy
K
,
Cure
P
,
Froud
T
,
Baidal
D
,
Pileggi
A
,
Messinger
S
,
Ricordi
C
,
Alejandro
R
.
C-peptide and glucose values in the peritransplant period after intraportal islet infusions in type 1 diabetes
.
Transplant Proc
.
2005
;
37
(
8
):
3433
3434
.

128.

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
.
2016
;
16
(
1
):
246
253
.

129.

Brennan
DC
,
Schnitzler
MA
.
Long-term results of rabbit antithymocyte globulin and basiliximab induction
.
N Engl J Med
.
2008
;
359
(
16
):
1736
1738
.

130.

Group
CSC
,
Haynes
R
,
Harden
P
,
Judge
P
,
Blackwell
L
,
Emberson
J
,
Landray
MJ
,
Baigent
C
,
Friend
PJ
.
Alemtuzumab-based induction treatment versus basiliximab-based induction treatment in kidney transplantation (the 3C Study): a randomised trial
.
Lancet
.
2014
;
384
(
9955
):
1684
1690
.

131.

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
.

132.

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
.

133.

Soleimanpour
SA
,
Sekiguchi
DR
,
Larosa
DF
,
Prak
ETL
,
Naji
A
,
Rickels
MR
.
Hypersensitivity to rabbit antithymocyte globulin in an islet transplant recipient: a case report
.
Transplant Proc
.
2011
;
43
(
9
):
3302
3306
.

134.

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
.

135.

Midtvedt
K
,
Hjelmesaeth
J
,
Hartmann
A
,
Lund
K
,
Paulsen
D
,
Egeland
T
,
Jenssen
T
.
Insulin resistance after renal transplantation: the effect of steroid dose reduction and withdrawal
.
J Am Soc Nephrol
.
2004
;
15
(
12
):
3233
3239
.

136.

Froud
T
,
Faradji
RN
,
Gorn
L
,
Monroy
K
,
Paz
C
,
Baidal
DA
,
Ponte
G
,
Cure
P
,
Poggioli
R
,
Pileggi
A
,
Ricordi
C
,
Alejandro
R
.
Dapsone-induced artifactual A1c reduction in islet transplant recipients
.
Transplantation
.
2007
;
83
(
6
):
824
825
.

137.

Wahoff
DC
,
Papalois
BE
,
Najarian
JS
,
Kendall
DM
,
Farney
AC
,
Leone
JP
,
Jessurun
J
,
Dunn
DL
,
Robertson
RP
,
Sutherland
DE
.
Autologous islet transplantation to prevent diabetes after pancreatic resection. Ann Surg. 1995;222(4):562–575
.

138.

Walsh
RM
,
Saavedra
JR
,
Lentz
G
,
Guerron
AD
,
Scheman
J
,
Stevens
T
,
Trucco
M
,
Bottino
R
,
Hatipoglu
B
.
Improved quality of life following total pancreatectomy and auto-islet transplantation for chronic pancreatitis
.
J Gastrointest Surg
.
2012
;
16
(
8
):
1469
1477
.

139.

Takita
M
,
Naziruddin
B
,
Matsumoto
S
,
Noguchi
H
,
Shimoda
M
,
Chujo
D
,
Itoh
T
,
Sugimoto
K
,
Onaca
N
,
Lamont
J
,
Lara
LF
,
Levy
MF
.
Implication of pancreatic image findings in total pancreatectomy with islet autotransplantation for chronic pancreatitis
.
Pancreas
.
2011
;
40
(
1
):
103
108
.

140.

Khan
KM
,
Desai
CS
,
Kalb
B
,
Patel
C
,
Grigsby
BM
,
Jie
T
,
Gruessner
RW
,
Rodriguez-Rilo
H
.
MRI prediction of islet yield for autologous transplantation after total pancreatectomy for chronic pancreatitis
.
Dig Dis Sci
.
2013
;
58
(
4
):
1116
1124
.

141.

Young
MC
,
Theis
JR
,
Hodges
JS
,
Dunn
TB
,
Pruett
TL
,
Chinnakotla
S
,
Walker
SP
,
Freeman
ML
,
Trikudanathan
G
,
Arain
M
,
Robertson
PR
,
Wilhelm
JJ
,
Schwarzenberg
SJ
,
Bland
B
,
Beilman
GJ
,
Bellin
MD
.
Preoperative computerized tomography and magnetic resonance imaging of the pancreas predicts pancreatic mass and functional outcomes after total pancreatectomy and islet autotransplant
.
Pancreas
.
2016
;
45
(
7
):
961
966
.

142.

Ogilvie
RF
.
A quantitative estimation of the pancreatic islet tissue
.
Q J Med
.
1937
;
6
:
287
300
.

143.

Volk
BW
,
Wellmann
KF
. Quantitative studies of the islets of nondiabetic patients. In:
Volk
BW
,
Wellmann
KF
, eds.
The Diabetic Pancreas
.
New York, NY
:
Plenum
;
1977
:
121
128
.

144.

Ricordi
C
,
Gray
DW
,
Hering
BJ
,
Kaufman
DB
,
Warnock
GL
,
Kneteman
NM
,
Lake
SP
,
London
NJ
,
Socci
C
,
Alejandro
R
,
Zeng Y, Scharp DW, Viviani G, Falqui L, Tzakis A, Bretzel RG, Federlin K, Pozza G, James RF, Rajotte RV, Di Carlo V, Morris PJ, Sutherland DE, Starzl TE, Mintz DH, Lacy PE
.
Islet isolation assessment in man and large animals
.
Acta Diabetol Lat
.
1990
;
27
(
3
):
185
195
.

145.

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
.

146.

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
.

147.

Kobayashi
T
,
Manivel
JC
,
Carlson
AM
,
Bellin
MD
,
Moran
A
,
Freeman
ML
,
Bielman
GJ
,
Hering
BJ
,
Dunn
T
,
Sutherland
DE
.
Correlation of histopathology, islet yield, and islet graft function after islet autotransplantation in chronic pancreatitis
.
Pancreas
.
2011
;
40
(
2
):
193
199
.

148.

Kobayashi
T
,
Manivel
JC
,
Bellin
MD
,
Carlson
AM
,
Moran
A
,
Freeman
ML
,
Hering
BJ
,
Sutherland
DE
.
Correlation of pancreatic histopathologic findings and islet yield in children with chronic pancreatitis undergoing total pancreatectomy and islet autotransplantation
.
Pancreas
.
2010
;
39
(
1
):
57
63
.

149.

Bellin
MD
,
Forlenza
GP
,
Majumder
K
,
Berger
M
,
Freeman
ML
,
Beilman
GJ
,
Dunn
TB
,
Pruett
TL
,
Murati
M
,
Wilhelm
JJ
,
Cook
M
,
Sutherland
DE
,
Schwarzenberg
SJ
,
Chinnakotla
S
.
Total pancreatectomy with islet autotransplantation resolves pain in young children with severe chronic pancreatitis
.
J Pediatr Gastroenterol Nutr
.
2017
;
64
(
3
):
440
445
.

150.

Wahoff
DC
,
Paplois
BE
,
Najarian
JS
,
Farney
AC
,
Leonard
AS
,
Kendall
DM
,
Roberston
RR
,
Sutherland
DE
.
Islet autotransplantation after total pancreatectomy in a child
.
J Pediatr Surg
.
1996
;
31
(
1
):
132
135
.

151.

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
.

152.

Jung
HS
,
Choi
SH
,
Kim
SJ
,
Choi
DW
,
Heo
JS
,
Lee
KT
,
Lee
JK
,
Jang
KT
,
Lee
BW
,
Jee
JH
,
Noh
JH
,
Jeong
IK
,
Yang
TY
,
Oh
SH
,
Ahn
YR
,
Kim
YS
,
No
H
,
Lee
MK
,
Kim
KW
.
Delayed improvement of insulin secretion after autologous islet transplantation in partially pancreatectomized patients
.
Metabolism
.
2009
;
58
(
11
):
1629
1635
.

153.

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
.

154.

Ryan
EA
,
Paty
BW
,
Senior
PA
,
Bigam
D
,
Alfadhli
E
,
Kneteman
NM
,
Lakey
JR
,
Shapir
AM
.
Five-year follow-up after clinical islet transplantation
.
Diabetes
.
2005
;
54
(
7
):
2060
2069
.

155.

Shapiro
AM
,
Ricordi
C
,
Hering
BJ
,
Auchincloss
H
,
Lindblad
R
,
Robertson
P
,
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
.

156.

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
.

157.

Cardani
R
,
Pileggi
A
,
Ricordi
C
,
Gomez
C
,
Baidal
DA
,
Ponte
GG
,
Mineo
D
,
Faradji
RN
,
Froud
T
,
Ciancio
G
,
Esquenazi
V
,
Burke
GW
III
,
Selvaggi
G
,
Miller
J
,
Kenyon
NS
,
Alejandro
R
.
Allosensitization of islet allograft recipients
.
Transplantation
.
2007
;
84
(
11
):
1413
1427
.

158.

Lehmann
R
,
Spinas
GA
,
Moritz
W
,
Weber
M
.
Has time come for new goals in human islet transplantation
?
Am J Transplant
.
2008
;
8
(
6
):
1096
1100
.

159.

Rickels
MR
,
Liu
C
,
Shlansky-Goldberg
RD
,
Soleimanpour
SA
,
Vivek
K
,
Kamoun
M
,
Min
Z
,
Markmann
E
,
Palangian
M
,
Dalton-Bakes
C
,
Fuller
C
,
Chiou
AJ
,
Barker
CF
,
Luning Prak
ET
,
Naji
A
.
Improvement in β-cell secretory capacity after human islet transplantation according to the B7 protocol
.
Diabetes
.
2013
;
62
(
8
):
2890
2897
.

160.

Borot
S
,
Niclauss
N
,
Wojtusciszyn
A
,
Brault
C
,
Demuylder-Mischler
S
,
Muller
Y
,
Giovannoni
L
,
Parnaud
G
,
Meier
R
,
Badet
L
,
Bayle
F
,
Frimat
L
,
Kessler
L
,
Morelon
E
,
Penfornis
A
,
Thivolet
C
,
Toso
C
,
Morel
P
,
Bosco
D
,
Colin
C
,
Benhamou
PY
,
Berney
T
,
Network
G
.
Impact of the number of infusions on 2-year results of islet-after-kidney transplantation in the GRAGIL network
.
Transplantation
.
2011
;
92
:
1031
1038
.

161.

Faradji
RN
,
Tharavanij
T
,
Messinger
S
,
Froud
T
,
Pileggi
A
,
Monroy
K
,
Mineo
D
,
Baidal
DA
,
Cure
P
,
Ponte
G
,
Mendez
AJ
,
Selvaggi
G
,
Ricordi
C
,
Alejandro
R
.
Long-term insulin independence and improvement in insulin secretion after supplemental islet infusion under exenatide and etanercept
.
Transplantation
.
2008
;
86
(
12
):
1658
1665
.

162.

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
.

163.

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
.

164.

Radosevich
DM
,
Jevne
R
,
Bellin
M
,
Kandaswamy
R
,
Sutherland
DE
,
Hering
BJ
.
Comprehensive health assessment and five-year follow-up of allogeneic islet transplant recipients
.
Clin Transplant
.
2013
;
27
(
6
):
E715
E724
.

165.

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
,
Network
G
.
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
.

166.

Foster
ED
,
Bridges
ND
,
Feurer
ID
,
Eggerman
TL
,
Hunsicker
LG
,
Alejandro
R
;
Clinical Islet Transplantation Consortium
.
Improved health-related quality of life in a phase 3 islet transplantation trial in type 1 diabetes complicated by severe hypoglycemia
.
Diabetes Care
.
2018
;
41
(
5
):
1001
1008
.

167.

Holmes-Walker
DJ
,
Gunton
JE
,
Hawthorne
W
,
Payk
M
,
Anderson
P
,
Donath
S
,
Loudovaris
T
,
Ward
GM
,
Kay
TW
,
O’Connell
PJ
.
Islet transplantation provides superior glycemic control with less hypoglycemia compared with continuous subcutaneous insulin infusion or multiple daily insulin injections
.
Transplantation
.
2017
;
101
(
6
):
1268
1275
.

168.

Benomar
K
,
Chetboun
M
,
Espiard
S
,
Jannin
A
,
Le Mapihan
K
,
Gmyr
V
,
Caiazzo
R
,
Torres
F
,
Raverdy
V
,
Bonner
C
,
D’Herbomez
M
,
Pigny
P
,
Noel
C
,
Kerr-Conte
J
,
Pattou
F
,
Vantyghem
MC
.
Purity of islet preparations and 5-year metabolic outcome of allogenic islet transplantation
.
Am J Transplant
.
2018
;
18
(
4
):
945
951
.

169.

Berney
T
,
Ferrari-Lacraz
S
,
Buhler
L
,
Oberholzer
J
,
Marangon
N
,
Philippe
J
,
Villard
J
,
Morel
P
.
Long-term insulin-independence after allogeneic islet transplantation for type 1 diabetes: over the 10-year mark
.
Am J Transplant
.
2009
;
9
(
2
):
419
423
.

170.

Brennan
DC
,
Kopetskie
HA
,
Sayre
PH
,
Alejandro
R
,
Cagliero
E
,
Shapiro
AM
,
Goldstein
JS
,
DesMarais
MR
,
Booher
S
,
Bianchine
PJ
.
Long-term follow-up of the Edmonton protocol of islet transplantation in the United States
.
Am J Transplant
.
2016
;
16
(
2
):
509
517
.

171.

Redfield
RR
,
Rickels
MR
,
Naji
A
,
Odorico
JS
.
Pancreas transplantation in the modern era
.
Gastroenterol Clin North Am
.
2016
;
45
(
1
):
145
166
.

172.

Gerber
PA
,
Pavlicek
V
,
Zuellig
R
,
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
.

173.

Ludwig
B
,
Ludwig
S
,
Steffen
A
,
Saeger
HD
,
Bornstein
SR
.
Islet versus pancreas transplantation in type 1 diabetes: competitive or complementary
?
Curr Diab Rep
.
2010
;
10
(
6
):
506
511
.

174.

Larsen
MO
,
Rolin
B
,
Sturis
J
,
Wilken
M
,
Carr
RD
,
Porksen
N
,
Gotfredsen
CF
.
Measurements of insulin responses as predictive markers of pancreatic beta-cell mass in normal and β-cell-reduced lean and obese Gottingen minipigs in vivo
.
Am J Physiol Endocrinol Metab
.
2006
;
290
(
4
):
E670
E677
.

175.

Seaquist
ER
,
Robertson
RP
.
Effects of hemipancreatectomy on pancreatic alpha and beta cell function in healthy human donors
.
J Clin Invest
.
1992
;
89
(
6
):
1761
1766
.

176.

Christiansen
E
,
Tibell
A
,
Volund
A
,
Rasmussen
K
,
Groth
CG
,
Holst
JJ
,
Pedersen
O
,
Christensen
NJ
,
Madsbad
S
.
Pancreatic endocrine function in recipients of segmental and whole pancreas transplantation
.
J Clin Endocrinol Metab
.
1996
;
81
:
3972
3979
.

177.

Robertson
RP
,
Sutherland
DE
,
Lanz
KJ
.
Normoglycemia and preserved insulin secretory reserve in diabetic patients 10–18 years after pancreas transplantation
.
Diabetes
.
1999
;
48
(
9
):
1737
1740
.

178.

Robertson
RP
.
Consequences on β-cell function and reserve after long-term pancreas transplantation
.
Diabetes
.
2004
;
53
(
3
):
633
644
.

179.

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
.

180.

Robertson
RP
,
Bogachus
LD
,
Oseid
E
,
Parazzoli
S
,
Patti
ME
,
Rickels
MR
,
Schuetz
C
,
Dunn
T
,
Pruett
T
,
Balamurugan
AN
,
Sutherland
DE
,
Beilman
G
,
Bellin
MD
.
Assessment of β-cell mass and α- and β-cell survival and function by arginine stimulation in human autologous islet recipients
.
Diabetes
.
2015
;
64
(
2
):
565
572
.

181.

Rickels
MR
,
Bellin
M
,
Toledo
FG
,
Robertson
RP
,
Andersen
DK
,
Chari
ST
,
Brand
R
,
Frulloni
L
,
Anderson
MA
,
Whitcomb
DC
.
Detection, evaluation and treatment of diabetes mellitus in chronic pancreatitis. Recommendations from PancreasFest 2012
.
Pancreatology
.
2013
;
13
(
4
):
336
342
.

182.

Sheikh
S
,
Gudipaty
L
,
De Leon
DD
,
Hadjiliadis
D
,
Kubrak
C
,
Rosenfeld
NK
,
Nyirjesy
SC
,
Peleckis
AJ
,
Malik
S
,
Stefanovski
D
,
Cuchel
M
,
Rubenstein
RC
,
Kelly
A
,
Rickels
MR
.
Reduced β-cell secretory capacity in pancreatic-insufficient, but not pancreatic-sufficient, cystic fibrosis despite normal glucose tolerance
.
Diabetes
.
2017
;
66
(
1
):
134
144
.

183.

Quartuccio
M
,
Hall
E
,
Singh
V
,
Makary
MA
,
Hirose
K
,
Desai
N
,
Walsh
C
,
Warren
D
,
Sun
Z
,
Stein
E
,
Kalyani
RR
.
Glycemic predictors of insulin independence after total pancreatectomy with islet autotransplantation
.
J Clin Endocrinol Metab
.
2017
;
102
:
801
809
.

184.

Lundberg
R
,
Beilman
GJ
,
Dunn
TB
,
Pruett
TL
,
Chinnakotla
SC
,
Radosevich
DM
,
Robertson
RP
,
Ptacek
P
,
Balamurugan
AN
,
Wilhelm
JJ
,
Hering
BJ
,
Sutherland
DE
,
Moran
A
,
Bellin
MD
.
Metabolic assessment prior to total pancreatectomy and islet autotransplant: utility, limitations and potential
.
Am J Transplant
.
2013
;
13
(
10
):
2664
2671
.

185.

Bellin
MD
,
Blondet
JJ
,
Beilman
GJ
,
Dunn
TB
,
Balamurugan
AN
,
Thomas
W
,
Sutherland
DE
,
Moran
A
.
Predicting islet yield in pediatric patients undergoing pancreatectomy and autoislet transplantation for chronic pancreatitis
.
Pediatr Diabetes
.
2010
;
11
(
4
):
227
234
.

186.

Robertson
RP
.
AIRarg and AIRgluc as predictors of insulin secretory reserve
.
Transplant Proc
.
2004
;
36
(
4
):
1040
1041
.

187.

Hubert
T
,
Strecker
G
,
Gmyr
V
,
Arnalsteen
L
,
Garrigue
D
,
Ezzouaoui
R
,
Caiazzo
R
,
Dezfoulian
G
,
Averland
B
,
Vandewalle
B
,
Vantyghem
MC
,
Kerr-Conte
J
,
Pattou
F
.
Acute insulin response to arginine in deceased donors predicts the outcome of human islet isolation
.
Am J Transplant
.
2008
;
8
(
4
):
872
876
.

188.

Lundberg
R
,
Beilman
GJ
,
Dunn
TB
,
Pruett
TL
,
Freeman
ML
,
Ptacek
PE
,
Berry
KL
,
Robertson
RP
,
Moran
A
,
Bellin
MD
.
Early alterations in glycemic control and pancreatic endocrine function in nondiabetic patients with chronic pancreatitis
.
Pancreas
.
2016
;
45
(
4
):
565
571
.

189.

Yasuda
H
,
Harano
Y
,
Ohgaku
S
,
Kosugi
K
,
Suzuki
M
,
Hidaka
H
,
Kashiwagi
A
,
Shigeta
Y
.
Insulin sensitivity in pancreatitis, liver-diseases, steroid treatment and hyperthyroidism assessed by glucose, insulin and somatostatin infusion
.
Horm Metab Res
.
1984
;
16
(
1
):
3
6
.

190.

Bellin
MD
,
Beilman
GJ
,
Dunn
TB
,
Pruett
TL
,
Sutherland
DE
,
Chinnakotla
S
,
Hodges
JS
,
Lane
A
,
Ptacek
P
,
Berry
KL
,
Hering
BJ
,
Moran
A
.
Sitagliptin treatment after total pancreatectomy with islet autotransplantation: a randomized, placebo-controlled study
.
Am J Transplant
.
2017
;
17
(
2
):
443
450
.

191.

Lin
YK
,
Faiman
C
,
Walsh
RM
,
Stevens
T
,
Bottino
R
,
Hatipoglu
BA
.
Spontaneous hypoglycemia after islet autotransplantation for chronic pancreatitis
.
J Clin Endocrinol Metab
.
2016
;
101
(
10
):
3669
3675
.

192.

Redel
JM
,
Nathan
JD
,
Abu-El-Haija
M
,
Lin
TK
,
Palermo
JJ
,
Elder
DA
.
Severe fasting hypoglycemia in a child after total pancreatectomy with islet autotransplantation
.
Pediatr Transplant
.
2018
;
22
(
1
):
13080
.

193.

Rizza
RA
,
Cryer
PE
,
Gerich
JE
.
Role of glucagon, catecholamines, and growth-hormone in human glucose counter-regulation—effects of somatostatin and combined α-adrenergic and β- adrenergic-blockade on plasma-glucose recovery and glucose flux rates after insulin-induced hypoglycemia
.
J Clin Invest
.
1979
;
64
:
62
71
.

194.

American Diabetes Association
.
6. Glycemic targets
.
Diabetes Care
.
2015
;
38
(
Suppl 1
):
S33
S40
.

195.

Gustavson
SM
,
Rajotte
RV
,
Hunkeler
D
,
Lakey
JR
,
Edgerton
DS
,
Neal
DW
,
Snead
WL
,
Penaloza
AR
,
Cherrington
AD
.
Islet auto-transplantation into an omental or splenic site results in a normal beta cell but abnormal alpha cell response to mild non-insulin-induced hypoglycemia
.
Am J Transplant
.
2005
;
5
(
10
):
2368
2377
.

196.

Zhou
H
,
Zhang
T
,
Bogdani
M
,
Oseid
E
,
Parazzoli
S
,
Vantyghem
MC
,
Harmon
J
,
Slucca
M
,
Robertson
RP
.
Intrahepatic glucose flux as a mechanism for defective intrahepatic islet α-cell response to hypoglycemia
.
Diabetes
.
2008
;
57
(
6
):
1567
1574
.

197.

Jaspan
JB
,
Ruddick
J
,
Rayfield
E
.
Transhepatic glucagon gradients in man—evidence for glucagon extraction by human-liver
.
J Clin Endocrinol Metab
.
1984
;
58
(
2
):
287
292
.

198.

Salehi
M
,
Prigeon
RL
,
D’Alessio
DA
.
Gastric bypass surgery enhances glucagon-like peptide 1-stimulated postprandial insulin secretion in humans
.
Diabetes
.
2011
;
60
(
9
):
2308
2314
.

199.

Bradley
D
,
Conte
C
,
Mittendorfer
B
,
Eagon
JC
,
Varela
JE
,
Fabbrini
E
,
Gastaldelli
A
,
Chambers
KT
,
Su
X
,
Okunade
A
,
Patterson
BW
,
Klein
S
.
Gastric bypass and banding equally improve insulin sensitivity and β cell function
.
J Clin Invest
.
2012
;
122
(
12
):
4667
4674
.

200.

Goldfine
AB
,
Mun
EC
,
Devine
E
,
Bernier
R
,
Baz-Hecht
M
,
Jones
DB
,
Schneider
BE
,
Holst
JJ
,
Patti
ME
.
Patients with neuroglycopenia after gastric bypass surgery have exaggerated incretin and insulin secretory responses to a mixed meal
.
J Clin Endocrinol Metab
.
2007
;
92
(
12
):
4678
4685
.

201.

Salehi
M
,
Gastaldelli
A
,
D’Alessio
DA
.
Blockade of glucagon-like peptide 1 receptor corrects postprandial hypoglycemia after gastric bypass. Gastroenterology. 2014;146(3):669–680.e2
.

202.

Salehi
M
,
Gastaldelli
A
,
D’Alessio
DA
.
Altered islet function and insulin clearance cause hyperinsulinemia in gastric bypass patients with symptoms of postprandial hypoglycemia
.
J Clin Endocrinol Metab
.
2014
;
99
(
6
):
2008
2017
.

203.

Bogachus
LD
,
Bellin
MD
,
Vella
A
,
Robertson
RP
.
Deficient glucagon response to hypoglycemia during a mixed meal in total pancreatectomy/islet autotransplantation recipients
.
J Clin Endocrinol Metab
.
2018
;
103
(
4
):
1522
1529
.

204.

Maruyama
H
,
Hisatomi
A
,
Orci
L
,
Grodsky
GM
,
Unger
RH
.
Insulin within islets is a physiologic glucagon-release inhibitor
.
J Clin Invest
.
1984
;
74
(
6
):
2296
2299
.

205.

Toft-Nielsen
M
,
Madsbad
S
,
Holst
JJ
.
Exaggerated secretion of glucagon-like peptide-1 (GLP-1) could cause reactive hypoglycaemia
.
Diabetologia
.
1998
;
41
(
10
):
1180
1186
.

206.

Rickels
MR
,
Naji
A
.
Reactive hypoglycaemia following GLP-1 infusion in pancreas transplant recipients
.
Diabetes Obes Metab
.
2010
;
12
(
8
):
731
733
.

207.

Abrahamsson
N
,
Borjesson
JL
,
Sundbom
M
,
Wiklund
U
,
Karlsson
FA
,
Eriksson
JW
.
Gastric bypass reduces symptoms and hormonal responses in hypoglycemia
.
Diabetes
.
2016
;
65
(
9
):
2667
2675
.

208.

Heller
SR
,
Cryer
PE
.
Reduced neuroendocrine and symptomatic responses to subsequent hypoglycemia after 1 episode of hypoglycemia in nondiabetic humans
.
Diabetes
.
1991
;
40
(
2
):
223
226
.

209.

Bogachus
LD
,
Oseid
E
,
Bellin
M
,
Vella
A
,
Robertson
RP
.
Deficient endogenous glucose production during exercise after total pancreatectomy/islet autotransplantation
.
J Clin Endocrinol Metab
.
2017
;
102
(
9
):
3288
3295
.

210.

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
.

211.

Paty
BW
,
Imes
S
,
Ryan
EA
,
Senior
PA
,
Robertson
RP
,
Shapiro
AM
.
Insulin secretory reserve as a measure of functional islet mass following islet allotransplantation in type 1 diabetes [abstract]
.
Diabetes
.
2004
;
53
(
Suppl 2
):
A35
A36
.

212.

Keymeulen
B
,
Gillard
P
,
Mathieu
C
,
Movahedi
B
,
Maleux
G
,
Delvaux
G
,
Ysebaert
D
,
Roep
B
,
Vandemeulebroucke
E
,
Marichal
M
,
’t Veld
PI
,
Bogdani
M
,
Hendrieckx
C
,
Gorus
F
,
Ling
ZD
,
van Rood
J
,
Pipeleers
D
.
Correlation between β cell mass and glycemic control in type 1 diabetic recipients of islet cell graft
.
Proc Natl Acad Sci USA
.
2006
;
103
(
46
):
17444
17449
.

213.

Johansson
H
,
Lukinius
A
,
Moberg
L
,
Lundgren
T
,
Berne
C
,
Foss
A
,
Felldin
M
,
Kallen
R
,
Salmela
K
,
Tibe
A
,
Tufveson
G
,
Ekdahl
KN
,
Elgue
G
,
Korsgren
O
,
Nilsson
B
.
Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation
.
Diabetes
.
2005
;
54
(
6
):
1755
1762
.

214.

Eriksson
O
,
Eich
T
,
Sundin
A
,
Tibell
A
,
Tufveson
G
,
Andersson
H
,
Felldin
M
,
Foss
A
,
Kyllonen
L
,
Langstrom
B
,
Nilsson
B
,
Korsgren
O
,
Lundgren
T
.
Positron emission tomography in clinical islet transplantation
.
Am J Transplant
.
2009
;
9
(
12
):
2816
2824
.

215.

Negi
S
,
Park
SH
,
Jetha
A
,
Aikin
R
,
Trernblay
M
,
Paraskevas
S
.
Evidence of endoplasmic reticulum stress mediating cell death in transplanted human islets
.
Cell Transplant
.
2012
;
21
(
5
):
889
900
.

216.

Rickels
MR
,
Peleckis
AJ
,
Markmann
E
,
Dalton-Bakes
C
,
Kong
SM
,
Teff
KL
,
Naji
A
.
Long-term improvement in glucose control and counterregulation by islet transplantation for type 1 diabetes
.
J Clin Endocrinol Metab
.
2016
;
101
(
11
):
4421
4430
.

217.

Drachenberg
CB
,
Klassen
DK
,
Weir
MR
,
Wiland
A
,
Fink
JC
,
Bartlett
ST
,
Cangro
CB
,
Blahut
S
,
Papadimitriou
JC
.
Islet cell damage associated with tacrolimus and cyclosporine: morphological features in pancreas allograft biopsies and clinical correlation
.
Transplantation
.
1999
;
68
(
3
):
396
402
.

218.

Bell
E
,
Cao
X
,
Moibi
JA
,
Greene
SR
,
Young
R
,
Trucco
M
,
Gao
Z
,
Matschinsky
FM
,
Deng
S
,
Markman
JF
,
Naji
A
,
Wolf
BA
.
Rapamycin has a deleterious effect on MIN-6 cells and rat and human islets
.
Diabetes
.
2003
;
52
(
11
):
2731
2739
.

219.

Rickels
MR
,
Mueller
R
,
Teff
KL
,
Naji
A
.
β-Cell secretory capacity and demand in recipients of islet, pancreas, and kidney transplants
.
J Clin Endocrinol Metab
.
2010
;
95
(
3
):
1238
1246
.

220.

Marcelli-Tourvieille
S
,
Hubert
T
,
Moerman
E
,
Gmyr
V
,
Kerr-Conte
J
,
Nunes
B
,
Dherbomez
M
,
Vandewalle
B
,
Pattou
F
,
Vantyghem
MC
.
In vivo and in vitro effect of sirolimus on insulin secretion
.
Transplantation
.
2007
;
83
(
5
):
532
538
.

221.

Kneteman
NM
,
Lakey
JR
,
Wagner
T
,
Finegood
D
.
The metabolic impact of rapamycin (sirolimus) in chronic canine islet graft recipients
.
Transplantation
.
1996
;
61
(
8
):
1206
1210
.

222.

Greenbaum
CJ
.
Insulin resistance in type 1 diabetes
.
Diabetes Metab Res Rev
.
2002
;
18
(
3
):
192
200
.

223.

Yki-Järvinen
H
,
Helve
E
,
Koivisto
VA
.
Hyperglycemia decreases glucose-uptake in type-I diabetes
.
Diabetes
.
1987
;
36
(
8
):
892
896
.

224.

Zuniga-Guajardo
S
,
Zinman
B
.
The metabolic response to the eugylcemic insulin clamp in type-I diabetes and normal humans
.
Metabolism
.
1985
;
34
(
10
):
926
930
.

225.

Trevisan
R
,
Nosadini
R
,
Avogaro
A
,
Lippe
G
,
Duner
E
,
Fioretto
P
,
Deana
R
,
Tessari
P
,
Tiengo
A
,
Velussi
M
,
Cernigoi
A
,
Delprato
S
,
Crepaldi
G
.
Type-I diabetes is characterized by insulin resistance not only with regard to glucose, but also to lipid and amino-acid-metabolism
.
J Clin Endocrinol Metab
.
1986
;
62
(
6
):
1155
1162
.

226.

DeFronzo
RA
,
Hendler
R
,
Simonson
D
.
Insulin resistance is a prominent feature of insulin-dependent diabetes
.
Diabetes
.
1982
;
31
(
9
):
795
801
.

227.

Kacerovsky
M
,
Jones
J
,
Schmid
AI
,
Barosa
C
,
Lettner
A
,
Kacerovsky-Bielesz
G
,
Szendroedi
J
,
Chmelik
M
,
Nowotny
P
,
Chandramouli
V
,
Wolzt
M
,
Roden
M
.
Postprandial and fasting hepatic glucose fluxes in long-standing type 1 diabetes
.
Diabetes
.
2011
;
60
(
6
):
1752
1758
.

228.

Bergman
BC
,
Howard
D
,
Schauer
IE
,
Maahs
DM
,
Snell-Bergeon
JK
,
Eckel
RH
,
Perreault
L
,
Rewers
M
.
Features of hepatic and skeletal muscle insulin resistance unique to type 1 diabetes
.
J Clin Endocrinol Metab
.
2012
;
97
(
5
):
1663
1672
.

229.

Rickels
MR
,
Naji
A
,
Teff
KL
.
Insulin sensitivity, glucose effectiveness, and free fatty acid dynamics after human islet transplantation for type 1 diabetes
.
J Clin Endocrinol Metab
.
2006
;
91
(
6
):
2138
2144
.

230.

Hirsch
D
,
Odorico
J
,
Radke
N
,
Hanson
M
,
Danobeitia
JS
,
Hullett
D
,
Alejandro
R
,
Ricordi
C
,
Fernandez
LA
.
Correction of insulin sensitivity and glucose disposal after pancreatic islet transplantation: preliminary results
.
Diabetes Obes Metab
.
2010
;
12
(
11
):
994
1003
.

231.

Vethakkan
SR
,
Walters
JM
,
Gooley
JL
,
Boston
RC
,
Kay
TW
,
Goodman
DJ
,
Jenkins
AJ
,
Ward
GM
.
Normalized NEFA dynamics during an OGTT after islet transplantation
.
Transplantation
.
2012
;
94
(
7
):
e49
e51
.

232.

Lopez-Talavera
JC
,
Garcia-Ocana
A
,
Sipula
I
,
Takane
KK
,
Cozar-Castellano
I
,
Stewart
AF
.
Hepatocyte growth factor gene therapy for pancreatic islets in diabetes: reducing the minimal islet transplant mass required in a glucocorticoid-free rat model of allogeneic portal vein islet transplantation
.
Endocrinology
.
2004
;
145
(
2
):
467
474
.

233.

Larsen
JL
,
Bennett
RG
,
Burkman
T
,
Ramirez
AL
,
Yamamoto
S
,
Gulizia
J
,
Radio
S
,
Hamel
FG
.
Tacrolimus and sirolimus cause insulin resistance in normal Sprague Dawley rats
.
Transplantation
.
2006
;
82
(
4
):
466
470
.

234.

Soleimanpour
SA
,
Hirshberg
B
,
Bunnell
DJ
,
Sumner
AE
,
Ader
M
,
Remaley
AT
,
Rother
KI
,
Rickels
MR
,
Harlan
DM
.
Metabolic function of a suboptimal transplanted islet mass in nonhuman primates on rapamycin monotherapy
.
Cell Transplant
.
2012
;
21
(
6
):
1297
1304
.

235.

Krebs
M
,
Brunmair
B
,
Brehm
A
,
Artwohl
M
,
Szendroedi
J
,
Nowotny
P
,
Roth
E
,
Fuernsinn
C
,
Promintzer
M
,
Anderwald
C
,
Bischof
M
,
Roden
M
.
The mammalian target of rapamycin pathway regulates nutrient-sensitive glucose uptake in man
.
Diabetes
.
2007
;
56
(
6
):
1600
1607
.

236.

Rickels
MR
,
Kong
SM
,
Fuller
C
,
Dalton-Bakes
C
,
Ferguson
JF
,
Reilly
MP
,
Teff
KL
,
Naji
A
.
Improvement in insulin sensitivity after human islet transplantation for type 1 diabetes
.
J Clin Endocrinol Metab
.
2013
;
98
(
11
):
E1780
E1785
.

237.

Rickels
MR
,
Kong
SM
,
Fuller
C
,
Dalton-Bakes
C
,
Ferguson
JF
,
Reilly
MP
,
Teff
KL
,
Naji
A
.
Insulin sensitivity index in type 1 diabetes and following human islet transplantation: comparison of the minimal model to euglycemic clamp measures
.
Am J Physiol Endocrinol Metab
.
2014
;
306
(
10
):
E1217
E1224
.

238.

Zhou
HR
,
Tran
POT
,
Yang
SL
,
Zhang
T
,
Leroy
E
,
Oseid
E
,
Robertson
RP
.
Regulation of α-cell function by the β-cell during hypoglycemia in Wistar rats: the “switch-off” hypothesis
.
Diabetes
.
2004
;
53
(
6
):
1482
1487
.

239.

Hope
KM
,
Tran
POT
,
Zhou
HR
,
Oseid
E
,
Leroy
E
,
Robertson
RP
.
Regulation of α-cell function by the β-cell in isolated human and rat islets deprived of glucose: the “switch-off” hypothesis
.
Diabetes
.
2004
;
53
(
6
):
1488
1495
.

240.

Cooperberg
BA
,
Cryer
PE
.
Insulin reciprocally regulates glucagon secretion in humans
.
Diabetes
.
2010
;
59
(
11
):
2936
2940
.

241.

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
.

242.

Rickels
MR
,
Fuller
C
,
Dalton-Bakes
C
,
Markmann
E
,
Palanjian
M
,
Cullison
K
,
Tiao
J
,
Kapoor
S
,
Liu
CY
,
Naji
A
,
Teff
KL
.
Restoration of glucose counterregulation by islet transplantation in long-standing type 1 diabetes
.
Diabetes
.
2015
;
64
(
5
):
1713
1718
.

243.

Ang
M
,
Meyer
C
,
Brendel
MD
,
Bretzel
RG
,
Linn
T
.
Magnitude and mechanisms of glucose counterregulation following islet transplantation in patients with type 1 diabetes suffering from severe hypoglycaemic episodes
.
Diabetologia
.
2014
;
57
(
3
):
623
632
.

244.

Paty
BW
,
Ryan
EA
,
Shapiro
AMJ
,
Lakey
JRT
,
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
.

245.

Diamond
MP
,
Hallarman
L
,
Starickzych
K
,
Jones
TW
,
Connollyhoward
M
,
Tamborlane
WV
,
Sherwin
RS
.
Suppression of counterregulatory hormone response to hypoglycemia by insulin per se
.
J Clin Endocrinol Metab
.
1991
;
72
(
6
):
1388
1390
.

246.

Battezzati
A
,
Luzi
L
,
Perseghin
G
,
Bianchi
E
,
Spotti
D
,
Secchi
A
,
Vergani
S
,
DiCarlo
V
,
Pozza
G
.
Persistence of counter-regulatory abnormalities in insulin-dependent diabetes-mellitus after pancreas transplantation
.
Eur J Clin Invest
.
1994
;
24
(
11
):
751
758
.

247.

Cryer
PE
,
Davis
SN
,
Shamoon
H
.
Hypoglycemia in diabetes
.
Diabetes Care
.
2003
;
26
(
6
):
1902
1912
.

248.

Cryer
PE
.
Mechanisms of hypoglycemia-associated autonomic failure in diabetes
.
N Engl J Med
.
2013
;
369
(
4
):
362
372
.

249.

Pedersen-Bjergaard
U
,
Pramming
S
,
Heller
SR
,
Wallace
TM
,
Rasmussen
AK
,
Jorgensen
HV
,
Matthews
DR
,
Hougaard
P
,
Thorsteinsson
B
.
Severe hypoglycaemia in 1076 adult patients with type 1 diabetes: influence of risk markers and selection
.
Diabetes Metab Res Rev
.
2004
;
20
(
6
):
479
486
.

250.

Rickels
MR
,
Schutta
MH
,
Mueller
R
,
Kapoor
S
,
Markmann
JF
,
Naji
A
,
Teff
KL
.
Glycemic thresholds for activation of counterregulatory hormone and symptom responses in islet transplant recipients
.
J Clin Endocrinol Metab
.
2007
;
92
(
3
):
873
879
.

251.

Kessler
L
,
Passemard
R
,
Oberholzer
J
,
Benhamou
PY
,
Bucher
P
,
Toso
C
,
Meyer
P
,
Penfornis
A
,
Badet
L
,
Wolf
P
,
Colin
C
,
Morel
P
,
Pinget
M
.
Reduction of blood glucose variability in type 1 diabetic patients treated by pancreatic islet transplantation
.
Diabetes Care
.
2002
;
25
(
12
):
2256
2262
.

252.

Paty
BW
,
Senior
PA
,
Lakey
JR
,
Shapiro
AM
,
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 Technol Ther
.
2006
;
8
(
2
):
165
173
.

253.

Gorn
L
,
Faradji
RN
,
Messinger
S
,
Monroy
K
,
Baidal
DA
,
Froud
T
,
Mastrototaro
J
,
Ricordi
C
,
Alejandro
R
.
Impact of islet transplantation on glycemic control as evidenced by a continuous glucose monitoring system
.
J Diabetes Sci Technol
.
2008
;
2
(
2
):
221
228
.

254.

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 (β-score greater than 7) is required to abrogate hyperglycemia, whereas a minimal function is necessary to suppress severe hypoglycemia (β-score greater than 3)
.
J Clin Endocrinol Metab
.
2012
;
97
(
11
):
E2078
E2083
.

255.

Brooks
AM
,
Oram
R
,
Home
P
,
Steen
N
,
Shaw
JAM
.
Demonstration of an intrinsic relationship between endogenous C-peptide concentration and determinants of glycemic control in type 1 diabetes following islet transplantation
.
Diabetes Care
.
2015
;
38
(
1
):
105
112
.

256.

Rickels
MR
,
Peleckis
AJ
,
Dalton-Bakes
C
,
Naji
JR
,
Ran
NA
,
Nguyen
HL
,
O’Brien
S
,
Chen
S
,
Lee
I
,
Schutta
MH
.
Continuous glucose monitoring for hypoglycemia avoidance and glucose counterregulation in long-standing type 1 diabetes
.
J Clin Endocrinol Metab
.
2018
;
103
(
1
):
105
114
.

257.

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
.

258.

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
.

259.

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
.

260.

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
e142
.

261.

Robertson
RP
.
Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes
.
J Biol Chem
.
2004
;
279
(
41
):
42351
42354
.

262.

Liu
C
,
Koeberlein
B
,
Feldman
MD
,
Mueller
R
,
Wang
Z
,
Li
Y
,
Lane
K
,
Hoyt
CC
,
Tomaszewski
JE
,
Naji
A
,
Rickels
MR
.
Accumulation of intrahepatic islet amyloid in a nonhuman primate transplant model
.
Endocrinology
.
2012
;
153
(
4
):
1673
1683
.

263.

Seaquist
ER
,
Kahn
SE
,
Clark
PM
,
Hales
CN
,
Porte
D
,
Robertson
RP
.
Hyperproinsulinemia is associated with increased beta cell demand after hemipancreatectomy in humans
.
J Clin Invest
.
1996
;
97
(
2
):
455
460
.

264.

Hostens
K
,
Ling
ZD
,
Van Schravendijk
C
,
Pipeleers
D
.
Prolonged exposure of human β-cells to high glucose increases their release of proinsulin during acute stimulation with glucose or arginine
.
J Clin Endocrinol Metab
.
1999
;
84
(
4
):
1386
1390
.

265.

White
SA
,
Koppiker
NP
,
Burden
AC
,
Davies
MJ
,
Davies
JE
,
Clayton
HA
,
Swift
SM
,
Hales
CN
,
Dennison
AR
,
London
NJ
.
Insulin deficiency and increased intact and 32/33 split proinsulin secretion following human islet autotransplantation
.
Transplant Proc
.
1998
;
30
(
2
):
627
628
.

266.

McDonald
CG
,
Ryan
EA
,
Paty
BW
,
Senior
PA
,
Marshall
SM
,
Lakey
JR
,
Shapiro
AM
.
Cross-sectional and prospective association between proinsulin secretion and graft function after clinical islet transplantation
.
Transplantation
.
2004
;
78
(
6
):
934
937
.

267.

Klimek
AM
,
Soukhatcheva
G
,
Thompson
DM
,
Warnock
GL
,
Salehi
M
,
Rilo
H
,
D’Alessio
D
,
Meneilly
GS
,
Panagiotopoulos
C
,
Verchere
CB
.
Impaired proinsulin processing is a characteristic of transplanted islets
.
Am J Transplant
.
2009
;
9
(
9
):
2119
2125
.

268.

Westermark
GT
,
Westermark
P
,
Berne
C
,
Korsgren
O
.
Widespread amyloid deposition in transplanted human pancreatic islets
.
N Engl J Med
.
2008
;
359
(
9
):
977
979
.

269.

Westermark
GT
,
Davalli
AM
,
Secchi
A
,
Folli
F
,
Kin
T
,
Toso
C
,
Shapiro
AM
,
Korsgren
O
,
Tufveson
G
,
Andersson
A
,
Westermark
P
.
Further evidence for amyloid deposition in clinical pancreatic islet grafts
.
Transplantation
.
2012
;
93
(
2
):
219
223
.

270.

Ritzel
RA
,
Meier
JJ
,
Lin
CY
,
Veldhuis
JD
,
Butler
PC
.
Human islet amyloid polypeptide oligomers disrupt cell coupling, induce apoptosis, and impair insulin secretion in isolated human islets
.
Diabetes
.
2007
;
56
(
1
):
65
71
.

271.

Kahn
SE
,
Dalessio
DA
,
Schwartz
MW
,
Fujimoto
WY
,
Ensinck
JW
,
Taborsky
GJ
,
Porte
D
.
Evidence of cosecretion of islet amyloid polypeptide and insulin by β-Cells
.
Diabetes
.
1990
;
39
(
5
):
634
638
.

272.

Westermark
P
,
Li
ZC
,
Westermark
GT
,
Leckstrom
A
,
Steiner
DF
.
Effects of beta cell granule components on human islet amyloid polypeptide fibril formation
.
FEBS Lett
.
1996
;
379
(
3
):
203
206
.

273.

Rickels
MR
,
Collins
HW
,
Naji
A
.
Amyloid and transplanted islets
.
N Engl J Med
.
2008
;
359
(
25
):
2729
2731
.

274.

Paulsson
JF
,
Westermark
GT
.
Aberrant processing of human proislet amyloid polypeptide results in increased amyloid formation
.
Diabetes
.
2005
;
54
(
7
):
2117
2125
.

275.

Courtade
JA
,
Klimek-Abercrombie
AM
,
Chen
YC
,
Patel
N
,
Lu
PY
,
Speake
C
,
Orban
PC
,
Najafian
B
,
Meneilly
G
,
Greenbaum
CJ
,
Warnock
GL
,
Panagiotopoulos
C
,
Verchere
CB
.
Measurement of pro-islet amyloid polypeptide (1–48) in diabetes and islet transplants
.
J Clin Endocrinol Metab
.
2017
;
102
(
7
):
2595
2603
.

276.

Orr
C
,
Stratton
J
,
Rao
I
,
Al-Sayed
M
,
Smith
C
,
El-Shahawy
M
,
Dafoe
D
,
Mullen
Y
,
Al-Abdullah
I
,
Kandeel
F
.
Quantifying insulin therapy requirements to preserve islet graft function following islet transplantation
.
Cell Transplant
.
2016
;
25
(
1
):
83
95
.

277.

Ngo
A
,
Sutherland
DE
,
Beilman
GJ
,
Bellin
MD
.
Deterioration of glycemic control after corticosteroid administration in islet autotransplant recipients: a cautionary tale
.
Acta Diabetol
.
2014
;
51
(
1
):
141
145
.

278.

Poitout
V
,
Robertson
RP
.
Glucolipotoxicity: fuel excess and β-cell dysfunction
.
Endocr Rev
.
2008
;
29
(
3
):
351
366
.

279.

Rodriguez Rilo
HL
,
Carroll
PB
,
Zeng
YJ
,
Fontes
P
,
Demetris
J
,
Ricordi
C
.
Acceleration of chronic failure of intrahepatic canine islet autografts by a short course of prednisone
.
Transplantation
.
1994
;
57
(
2
):
181
186
.

280.

Wahoff
DC
,
Leone
JP
,
Farney
AC
,
Teuscher
AU
,
Sutherland
DE
.
Pregnancy after total pancreatectomy and autologous islet transplantation
.
Surgery
.
1995
;
117
(
3
):
353
354
.

281.

Jung
HS
,
Choi
SH
,
Noh
JH
,
Ohi
SH
,
Ahn
YR
,
Lee
MK
,
Kim
KW
.
Healthy twin birth after autologous islet transplantation in a pancreatectomized patient due to a benign tumor
.
Transplant Proc
.
2007
;
39
(
5
):
1723
1725
.

282.

Schive
SW
,
Scholz
H
,
Sahraoui
A
,
Kloster-Jensen
K
,
Hafsahl
G
,
Korsgren
O
,
Foss
A
,
Jenssen
TG
.
Graft function 1 year after pregnancy in an islet-transplanted patient
.
Transpl Int
.
2015
;
28
(
10
):
1235
1239
.

283.

Assalino
M
,
Podetta
M
,
Demuylder-Mischler
S
,
Francini
K
,
Pernin
N
,
Randin
JP
,
Bosco
D
,
Andres
A
,
Berney
T
.
Successful pregnancy and delivery after simultaneous islet-kidney transplantation
.
Am J Transplant
.
2018
;
18
(
8
):
2075
2078
.

284.

Rickels
MR
,
Markmann
E
,
Naji
A
.
Successful pregnancies after islet transplantation for type 1 diabetes
.
Am J Transplant
.
2019
;
19
(
1
):
298
299
.

285.

Farilla
L
,
Bulotta
A
,
Hirshberg
B
,
Calzi
SL
,
Khoury
N
,
Noushmehr
H
,
Bertolotto
C
,
Di Mario
U
,
Harlan
DM
,
Perfetti
R
.
Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets
.
Endocrinology
.
2003
;
144
(
12
):
5149
5158
.

286.

Toso
C
,
McCall
M
,
Emamaullee
J
,
Merani
S
,
Davis
J
,
Edgar
R
,
Pawlick
R
,
Kin
T
,
Knudsen
LB
,
Shapiro
AM
.
Liraglutide, a long-acting human glucagon-like peptide 1 analogue, improves human islet survival in culture
.
Transpl Int
.
2010
;
23
(
3
):
259
265
.

287.

Gangemi
A
,
Salehi
P
,
Hatipoglu
B
,
Martellotto
J
,
Barbaro
B
,
Kuechle
JB
,
Qi
M
,
Wang
Y
,
Pallan
P
,
Owens
C
,
Bui
J
,
West
D
,
Kaplan
B
,
Benedetti
E
,
Oberholzer
J
.
Islet transplantation for brittle type 1 diabetes: the UIC protocol
.
Am J Transplant
.
2008
;
8
(
6
):
1250
1261
.

288.

Qi
M
,
Kinzer
K
,
Danielson
KK
,
Martellotto
J
,
Barbaro
B
,
Wang
Y
,
Bui
JT
,
Gaba
RC
,
Knuttinen
G
,
Garcia-Roca
R
,
Tzvetanov
I
,
Heitman
A
,
Davis
M
,
McGarrigle
JJ
,
Benedetti
E
,
Oberholzer
J
.
Five-year follow-up of patients with type 1 diabetes transplanted with allogeneic islets: the UIC experience
.
Acta Diabetol
.
2014
;
51
(
5
):
833
843
.

289.

Al Ghofaili
K
,
Fung
M
,
Ao
ZL
,
Meloche
M
,
Shapiro
RJ
,
Warnock
GL
,
Elahi
D
,
Meneilly
GS
,
Thompson
DM
.
Effect of exenatide on beta cell function after islet transplantation in type 1 diabetes
.
Transplantation
.
2007
;
83
(
1
):
24
28
.

290.

Froud
T
,
Faradji
RN
,
Pileggi
A
,
Messinger
S
,
Baidal
DA
,
Ponte
GM
,
Cure
PE
,
Monroy
K
,
Mendez
A
,
Selvaggi
G
,
Ricordi
C
,
Alejandro
R
.
The use of exenatide in islet transplant recipients with chronic allograft dysfunction: safety, efficacy, and metabolic effects
.
Transplantation
.
2008
;
86
(
1
):
36
45
.

291.

Faradji
RN
,
Froud
T
,
Messinger
S
,
Monroy
K
,
Pileggi
A
,
Mineo
D
,
Tharavanij
T
,
Mendez
AJ
,
Ricordi
C
,
Alejandro
R
.
Long-term metabolic and hormonal effects of exenatide on islet transplant recipients with allograft dysfunction
.
Cell Transplant
.
2009
;
18
(
10–11
):
1247
1259
.

292.

Rickels
MR
,
Mueller
R
,
Markman
JF
,
Naji
A
.
Effect of GLP-1 on β- and α-cell function in isolated islet and whole pancreas transplant recipients
.
J Clin Endocrinol Metab
.
2009
;
94
:
181
189
.

293.

Senior
PA
,
Koh
A
,
Yau
J
,
Imes
S
,
Dinyari
P
,
Malcolm
AJ
,
Light
P
,
Shapiro
AM
.
Sitagliptin plus pantoprazole can restore but not maintain insulin independence after clinical islet transplantation: results of a pilot study
.
Diabet Med
.
2017
;
34
(
2
):
204
212
.

294.

Mohanakumar
T
,
Narayanan
K
,
Desai
N
,
Ramachandran
S
,
Shenoy
S
,
Jendrisak
M
,
Susskind
BM
,
Olack
B
,
Benshoff
N
,
Phelan
DL
,
Brennan
DC
,
Fernandez
LA
,
Odorico
JS
,
Polonsky
KS
.
A significant role for histocompatibility in human islet transplantation
.
Transplantation
.
2006
;
82
(
2
):
180
187
.

295.

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
.

296.

Naji
A
,
Barker
CF
,
Silvers
WK
.
Relative vulnerability of isolated pancreatic-islets, parathyroid, and skin allografts to cellular and humoral immunity
.
Transplant Proc
.
1979
;
11
(
1
):
560
562
.

297.

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
.

298.

Brooks
AM
,
Carter
V
,
Liew
A
,
Marshall
H
,
Aldibbiat
A
,
Sheerin
NS
,
Manas
DM
,
White
SA
,
Shaw
JA
.
De novo donor-specific hla antibodies are associated with rapid loss of graft function following islet transplantation in type 1 diabetes
.
Am J Transplant
.
2015
;
15
(
12
):
3239
3246
.

299.

Moreau
F
,
Toti
F
,
Bayle
F
,
Berney
T
,
Egelhofer
H
,
Chastre
M
,
Richard
MJ
,
Greget
M
,
Masson
D
,
Zobairi
F
,
Benhamou
PY
,
Kessler
L
;
GRAGIL group
.
Rescue of a pancreatic islet graft after steroid therapy
.
Transplantation
.
2012
;
93
(
3
):
e10
e11
.

300.

Olack
BJ
,
Swanson
CJ
,
Flavin
KS
,
Phelan
D
,
Brennan
DC
,
White
NH
,
Lacy
PE
,
Scharp
DW
,
Poindexter
N
,
Mohanakumar
T
.
Sensitization to HLA antigens in islet recipients with failing transplants
.
Transplant Proc
.
1997
;
29
(
4
):
2268
2269
.

301.

Rickels
MR
,
Kamoun
M
,
Kearns
J
,
Markmann
JF
,
Naji
A
.
Evidence for allograft rejection in an islet transplant recipient and effect on beta-cell secretory capacity
.
J Clin Endocrinol Metab
.
2007
;
92
(
7
):
2410
2414
.

302.

Rickels
MR
,
Kearns
J
,
Markmann
E
,
Palanjian
M
,
Markmann
JF
,
Naji
A
,
Kamoun
M
. HLA sensitization in islet transplantation. In:
Cecka
JM
,
Terasaki
PI
, eds.
Clinical Transplants 2006
.
Los Angeles, CA
:
UCLA Tissue Typing Laboratory
;
2007
:
413
420
.

303.

Ferrari-Lacraz
S
,
Berney
T
,
Morel
P
,
Marangon
N
,
Hadaya
K
,
Demuylder-Mischler
S
,
Pongratz
G
,
Pernin
N
,
Villard
J
.
Low risk of anti-human leukocyte antigen antibody sensitization after combined kidney and islet transplantation
.
Transplantation
.
2008
;
86
(
2
):
357
359
.

304.

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
.

305.

Kessler
L
,
Parissiadis
A
,
Bayle
F
,
Moreau
F
,
Pinget
M
,
Froelich
N
,
Cazenave
JP
,
Berney
T
,
Benhamou
PY
,
Hanau
D
,
Group
GS
.
Evidence for humoral rejection of a pancreatic islet graft and rescue with rituximab and IV immunoglobulin therapy
.
Am J Transplant
.
2009
;
9
(
8
):
1961
1966
.

306.

Hilbrands
R
,
Gillard
P
,
Van der Torren
CR
,
Ling
Z
,
Verheyden
S
,
Jacobs-Tulleneers-Thevissen
D
,
Roep
BO
,
Claas
FH
,
Demanet
C
,
Gorus
FK
,
Pipeleers
D
,
Keymeulen
B
.
Predictive factors of allosensitization after immunosuppressant withdrawal in recipients of long-term cultured islet cell grafts
.
Transplantation
.
2013
;
96
(
2
):
162
169
.

307.

Huurman
VA
,
van der Torren
CR
,
Gillard
P
,
Hilbrands
R
,
van der Meer-Prins
EP
,
Duinkerken
G
,
Gorus
FK
,
Claas
FH
,
Keymeulen
B
,
Roelen
DL
,
Pipeleers
DG
,
Roep
BO
.
Immune responses against islet allografts during tapering of immunosuppression—a pilot study in 5 subjects
.
Clin Exp Immunol
.
2012
;
169
(
2
):
190
198
.

308.

Naziruddin
B
,
Wease
S
,
Stablein
D
,
Barton
FB
,
Berney
T
,
Rickels
MR
,
Alejandro
R
.
HLA class i sensitization in islet transplant recipients: report from the Collaborative Islet Transplant Registry
.
Cell Transplant
.
2012
;
21
(
5
):
901
908
.

309.

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
.

310.

Sibley
RK
,
Sutherland
DE
,
Goetz
F
,
Michael
AF
.
Recurrent diabetes mellitus in the pancreas iso- and allograft. A light and electron microscopic and immunohistochemical analysis of four cases
.
Lab Invest
.
1985
;
53
:
132
144
.

311.

Tydén
G
,
Reinholt
FP
,
Sundkvist
G
,
Bolinder
J
.
Recurrence of autoimmune diabetes mellitus in recipients of cadaveric pancreatic grafts
.
N Engl J Med
.
1996
;
335
(
12
):
860
863
.

312.

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
ZB
,
Mendez
A
,
Chen
LD
,
Sageshima
J
,
Ruiz
P
,
Ciancio
G
,
Ricordi
C
,
Reijonen
H
,
Nepom
GT
,
Burke
GW
,
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
.

313.

Jaeger
C
,
Brendel
MD
,
Hering
BJ
,
Eckhard
M
,
Bretzel
RG
.
Progressive islet graft failure occurs significantly earlier in autoantibody-positive than in autoantibody-negative IDDM recipients of intrahepatic islet allografts
.
Diabetes
.
1997
;
46
(
11
):
1907
1910
.

314.

Bosi
E
,
Braghi
S
,
Maffi
P
,
Scirpoli
M
,
Bertuzzi
F
,
Pozza
G
,
Secchi
A
,
Bonifacio
E
.
Autoantibody response to islet transplantation in type 1 diabetes
.
Diabetes
.
2001
;
50
(
11
):
2464
2471
.

315.

Huurman
VA
,
Hilbrands
R
,
Pinkse
GG
,
Gillard
P
,
Duinkerken
G
,
van de Linde
P
,
van der Meer-Prins
PM
,
Versteeg-van der Voort Maarschalk
MF
,
Verbeeck
K
,
Alizadeh
BZ
,
Mathieu
C
,
Gorus
FK
,
Roelen
DL
,
Claas
FH
,
Keymeulen
B
,
Pipeleers
DG
,
Roep
BO
.
Cellular islet autoimmunity associates with clinical outcome of islet cell transplantation
.
PLoS One
.
2008
;
3
(
6
):
e2435
.

316.

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
.

317.

Stegall
MD
,
Lafferty
KJ
,
Kam
I
,
Gill
RG
.
Evidence of recurrent autoimmunity in human allogeneic islet transplantation
.
Transplantation
.
1996
;
61
(
8
):
1272
1274
.

318.

Autoimmunity after islet-cell allotransplantation
.
N Engl J Med
.
2006
;
355
(
13
):
1397
1399
.

319.

Demeester
S
,
Balke
EM
,
Van der Auwera
BJ
,
Gillard
P
,
Hilbrands
R
,
Lee
D
,
Van de Velde
U
,
Ling
Z
,
Roep
BO
,
Pipeleers
DG
,
Gorus
FK
,
Keymeulen
B
.
HLA-A*24 carrier status and autoantibody surges posttransplantation associate with poor functional outcome in recipients of an islet allograft
.
Diabetes Care
.
2016
;
39
(
6
):
1060
1064
.

320.

Chujo
D
,
Foucat
E
,
Takita
M
,
Itoh
T
,
Sugimoto
K
,
Shimoda
M
,
Yagi
K
,
Yamagishi
M
,
Tamura
Y
,
Yu
L
,
Naziruddin
B
,
Levy
MF
,
Ueno
H
,
Matsumoto
S
.
Emergence of a broad repertoire of GAD65-specific T-cells in type 1 diabetes patients with graft dysfunction after allogeneic islet transplantation
.
Cell Transplant
.
2012
;
21
(
12
):
2783
2795
.

321.

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
.

322.

Smelt
MJ
,
Faas
MM
,
de Haan
BJ
,
Draijer
C
,
Hugenholtz
GC
,
de Haan
A
,
Engelse
MA
,
de Koning
EJ
,
de Vos
P
.
Susceptibility of human pancreatic β cells for cytomegalovirus infection and the effects on cellular immunogenicity
.
Pancreas
.
2012
;
41
(
1
):
39
49
.

323.

Pak
CY
,
Eun
HM
,
McArthur
RG
,
Yoon
JW
.
Association of cytomegalovirus infection with autoimmune type 1 diabetes
.
Lancet
.
1988
;
2
(
8601
):
1
4
.

324.

Zanone
MM
,
Favaro
E
,
Quadri
R
,
Miceli
I
,
Giaretta
F
,
Romagnoli
R
,
David
E
,
Perin
PC
,
Salizzoni
M
,
Camussi
G
.
Association of cytomegalovirus infections with recurrence of humoral and cellular autoimmunity to islet autoantigens and of type 1 diabetes in a pancreas transplanted patient
.
Transpl Int
.
2010
;
23
(
3
):
333
337
.

325.

Roep
BO
,
Hiemstra
HS
,
Schloot
NC
,
De Vries
RR
,
Chaudhuri
A
,
Behan
PO
,
Drijfhout
JW
.
Molecular mimicry in type 1 diabetes: immune cross-reactivity between islet autoantigen and human cytomegalovirus but not Coxsackie virus
.
Ann NY Acad Sci
.
2002
;
958
(
1
):
163
165
.

326.

Kawahara
T
,
Kin
T
,
Shapiro
AM
.
A comparison of islet autotransplantation with allotransplantation and factors elevating acute portal pressure in clinical islet transplantation
.
J Hepatobiliary Pancreat Sci
.
2012
;
19
(
3
):
281
288
.

327.

Ong
SL
,
Pollard
C
,
Rees
Y
,
Garcea
G
,
Webb
M
,
Illouz
S
,
Berry
DP
,
Dennison
AR
.
Ultrasound changes within the liver after total pancreatectomy and intrahepatic islet cell autotransplantation
.
Transplantation
.
2008
;
85
(
12
):
1773
1777
.

328.

Wray
CJ
,
Ahmad
SA
,
Lowy
AM
,
D’Alessio
DA
,
Gelrud
A
,
Choe
KA
,
Soldano
DA
,
Matthews
JB
,
Rodriguez-Rilo
HL
.
Clinical significance of bacterial cultures from 28 autologous islet cell transplant solutions
.
Pancreatology
.
2005
;
5
(
6
):
562
569
.

329.

Johnson
CN
,
Morgan
KA
,
Owczarski
SM
,
Wang
H
,
Fried
J
,
Adams
DB
.
Autotransplantation of culture-positive islet product: is dirty always bad
?
HPB
.
2014
;
16
(
7
):
665
669
.

330.

Berger
MG
,
Majumder
K
,
Hodges
JS
,
Bellin
MD
,
Schwarzenberg
SJ
,
Gupta
S
,
Dunn
TB
,
Beilman
GJ
,
Pruett
TL
,
Freeman
ML
,
Wilhelm
JJ
,
Sutherland
DE
,
Chinnakotla
S
.
Microbial contamination of transplant solutions during pancreatic islet autotransplants is not associated with clinical infection in a pediatric population
.
Pancreatology
.
2016
;
16
(
4
):
555
562
.

331.

Bucher
P
,
Mathe
Z
,
Bosco
D
,
Becker
C
,
Kessler
L
,
Greget
M
,
Benhamou
PY
,
Andres
A
,
Oberholzer
J
,
Buhler
L
,
Morel
P
,
Berney
T
.
Morbidity associated with intraportal islet transplantation
.
Transplant Proc
.
2004
;
36
(
4
):
1119
1120
.

332.

Kawahara
T
,
Kin
T
,
Kashkoush
S
,
Gala-Lopez
B
,
Bigam
DL
,
Kneteman
NM
,
Koh
A
,
Senior
PA
,
Shapiro
AM
.
Portal vein thrombosis is a potentially preventable complication in clinical islet transplantation
.
Am J Transplant
.
2011
;
11
(
12
):
2700
2707
.

333.

Baidal
DA
,
Froud
T
,
Ferreira
JV
,
Khan
A
,
Alejandro
R
,
Ricordi
C
.
The bag method for islet cell infusion
.
Cell Transplant
.
2003
;
12
(
7
):
809
813
.

334.

Onaca
N
,
Naziruddin
B
,
Randall
HB
,
Meler
JD
,
Sanchez
EQ
,
Matsumoto
S
,
Noguchi
H
,
Jackson
A
,
Diamond
NG
,
Klintmalm
GB
,
Levy
MF
.
False aneurysm of a hepatic artery branch complicating intrahepatic islet transplantation
.
Transpl Int
.
2009
;
22
(
6
):
663
666
.

335.

Bozkurt
NC
,
Leao Peixoto
EM
,
Froud
T
,
Herrada
E
,
Corrales
A
,
Ricordi
C
,
Alejandro
R
.
Hepatic hematoma after islet cell transplantation
.
Transplantation
.
2013
;
95
(
12
):
e73
e76
.

336.

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
.

337.

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
.

338.

Gaba
RC
,
Kobayashi
KR
,
Bui
JT
,
Lokken
RP
,
Lipnik
AJ
,
Ray
CE
Jr
,
Oberholzer
J
.
Liver track embolization after islet cell transplant: comparison of two techniques
.
AJR Am J Roentgenol
.
2017
;
208
(
5
):
1134
1140
.

339.

Caiazzo
R
,
Vantyghem
MC
,
Raverdi
V
,
Bonner
C
,
Gmyr
V
,
Defrance
F
,
Leroy
C
,
Sergent
G
,
Hubert
T
,
Ernst
O
,
Noel
C
,
Kerr-Conte
J
,
Pattou
F
.
Impact of procedure-related complications on long-term islet transplantation outcome
.
Transplantation
.
2015
;
99
(
5
):
979
984
.

340.

Markmann
JF
,
Rosen
M
,
Siegelman
ES
,
Soulen
MC
,
Deng
SP
,
Barker
CF
,
Naji
A
.
Magnetic resonance-defined periportal steatosis following intraportal islet transplantation—a functional footprint of islet graft survival
?
Diabetes
.
2003
;
52
(
7
):
1591
1594
.

341.

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
.

342.

Fishman
JA
.
Infection in solid-organ transplant recipients
.
N Engl J Med
.
2007
;
357
(
25
):
2601
2614
.

343.

Cure
P
,
Pileggi
A
,
Faradji
RN
,
Baidal
DA
,
Froud
T
,
Selvaggi
G
,
Ricordi
C
,
Alejandro
R
.
Cytomegalovirus infection in a recipient of solitary allogeneic islets
.
Am J Transplant
.
2006
;
6
(
5 Pt 1
):
1089
1090
.

344.

Dierickx
D
,
Habermann
TM
.
Post-transplantation lymphoproliferative disorders in adults
.
N Engl J Med
.
2018
;
378
(
6
):
549
562
.

345.

Peters
A
,
Olateju
T
,
Deschenes
J
,
Shankarnarayan
SH
,
Chua
N
,
Shapiro
AM
,
Senior
P
.
Posttransplant lymphoproliferative disorder after clinical islet transplantation: report of the first two cases
.
Am J Transplant
.
2017
;
17
(
9
):
2474
2480
.

346.

Euvrard
S
,
Kanitakis
J
,
Claudy
A
.
Skin cancers after organ transplantation
.
N Engl J Med
.
2003
;
348
(
17
):
1681
1691
.

347.

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
.

348.

Naesens
M
,
Kuypers
DR
,
Sarwal
M
.
Calcineurin inhibitor nephrotoxicity
.
Clin J Am Soc Nephrol
.
2009
;
4
:
481
508
.

349.

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
.

350.

Leitão
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
.

351.

Potter
KJ
,
Abedini
A
,
Marek
P
,
Klimek
AM
,
Butterworth
S
,
Driscoll
M
,
Baker
R
,
Nilsson
MR
,
Warnock
GL
,
Oberholzer
J
,
Bertera
S
,
Trucco
M
,
Korbutt
GS
,
Fraser
PE
,
Raleigh
DP
,
Verchere
CB
.
Islet amyloid deposition limits the viability of human islet grafts but not porcine islet grafts
.
Proc Natl Acad Sci USA
.
2010
;
107
(
9
):
4305
4310
.

352.

Zhang
X
,
Cheng
B
,
Gong
H
,
Li
C
,
Chen
H
,
Zheng
L
,
Huang
K
.
Porcine islet amyloid polypeptide fragments are refractory to amyloid formation
.
FEBS Lett
.
2011
;
585
(
1
):
71
77
.

353.

Emamaullee
JA
,
Merani
S
,
Toso
C
,
Kin
T
,
Al-Saif
F
,
Truong
W
,
Pawlick
R
,
Davis
J
,
Edgar
R
,
Lock
J
,
Bonner-Weir
S
,
Knudsen
LB
,
Shapiro
AM
.
Porcine marginal mass islet autografts resist metabolic failure over time and are enhanced by early treatment with liraglutide
.
Endocrinology
.
2009
;
150
(
5
):
2145
2152
.

354.

Hering
BJ
,
Wijkstrom
M
,
Graham
ML
,
Harstedt
M
,
Aasheim
TC
,
Jie
T
,
Ansite
JD
,
Nakano
M
,
Cheng
J
,
Li
W
,
Moran
K
,
Christians
U
,
Finnegan
C
,
Mills
CD
,
Sutherland
DE
,
Bansal-Pakala
P
,
Murtaugh
MP
,
Kirchhof
N
,
Schuurman
HJ
.
Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates
.
Nat Med
.
2006
;
12
(
3
):
301
303
.

355.

Cardona
K
,
Korbutt
GS
,
Milas
Z
,
Lyon
J
,
Cano
J
,
Jiang
W
,
Bello-Laborn
H
,
Hacquoil
B
,
Strobert
E
,
Gangappa
S
,
Weber
CJ
,
Pearson
TC
,
Rajotte
RV
,
Larsen
CP
.
Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways
.
Nat Med
.
2006
;
12
(
3
):
304
306
.

356.

van der Windt
DJ
,
Bottino
R
,
Casu
A
,
Campanile
N
,
Smetanka
C
,
He
J
,
Murase
N
,
Hara
H
,
Ball
S
,
Loveland
BE
,
Ayares
D
,
Lakkis
FG
,
Cooper
DK
,
Trucco
M
.
Long-term controlled normoglycemia in diabetic non-human primates after transplantation with hCD46 transgenic porcine islets
.
Am J Transplant
.
2009
;
9
(
12
):
2716
2726
.

357.

Denner
J
.
Paving the path toward porcine organs for transplantation
.
N Engl J Med
.
2017
;
377
(
19
):
1891
1893
.

358.

Bartlett
ST
,
Markmann
JF
,
Johnson
P
,
Korsgren
O
,
Hering
BJ
,
Scharp
D
,
Kay
TW
,
Bromberg
J
,
Odorico
JS
,
Weir
GC
,
Bridges
N
,
Kandaswamy
R
,
Stock
P
,
Friend
P
,
Gotoh
M
,
Cooper
DK
,
Park
CG
,
O’Connell
P
,
Stabler
C
,
Matsumoto
S
,
Ludwig
B
,
Choudhary
P
,
Kovatchev
B
,
Rickels
MR
,
Sykes
M
,
Wood
K
,
Kraemer
K
,
Hwa
A
,
Stanley
E
,
Ricordi
C
,
Zimmerman
M
,
Greenstein
J
,
Montanya
E
,
Otonkoski
T
.
Report from IPITA-TTS opinion leaders meeting on the future of beta-cell replacement
.
Transplantation
.
2016
;
100
(
Suppl 2
):
S1
S44
.

359.

Wynyard
S
,
Nathu
D
,
Garkavenko
O
,
Denner
J
,
Elliott
R
.
Microbiological safety of the first clinical pig islet xenotransplantation trial in New Zealand
.
Xenotransplantation
.
2014
;
21
(
4
):
309
323
.

360.

Morozov
VA
,
Wynyard
S
,
Matsumoto
S
,
Abalovich
A
,
Denner
J
,
Elliott
R
.
No PERV transmission during a clinical trial of pig islet cell transplantation
.
Virus Res
.
2017
;
227
:
34
40
.

361.

Basta
G
,
Montanucci
P
,
Luca
G
,
Boselli
C
,
Noya
G
,
Barbaro
B
,
Qi
M
,
Kinzer
KP
,
Oberholzer
J
,
Calafiore
R
.
Long-term metabolic and immunological follow-up of nonimmunosuppressed patients with type 1 diabetes treated with microencapsulated islet allografts: four cases
.
Diabetes Care
.
2011
;
34
(
11
):
2406
2409
.

362.

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
.

363.

Rezania
A
,
Bruin
JE
,
Riedel
MJ
,
Mojibian
M
,
Asadi
A
,
Xu
J
,
Gauvin
R
,
Narayan
K
,
Karanu
F
,
O’Neil
JJ
,
Ao
Z
,
Warnock
GL
,
Kieffer
TJ
.
Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice
.
Diabetes
.
2012
;
61
(
8
):
2016
2029
.

364.

Bruin
JE
,
Rezania
A
,
Xu
J
,
Narayan
K
,
Fox
JK
,
O’Neil
JJ
,
Kieffer
TJ
.
Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice
.
Diabetologia
.
2013
;
56
(
9
):
1987
1998
.

365.

van der Torren
CR
,
Zaldumbide
A
,
Duinkerken
G
,
Brand-Schaaf
SH
,
Peakman
M
,
Stange
G
,
Martinson
L
,
Kroon
E
,
Brandon
EP
,
Pipeleers
D
,
Roep
BO
.
Immunogenicity of human embryonic stem cell-derived beta cells
.
Diabetologia
.
2017
;
60
(
1
):
126
133
.

366.

Carlsson
PO
,
Espes
D
,
Sedigh
A
,
Rotem
A
,
Zimerman
B
,
Grinberg
H
,
Goldman
T
,
Barkai
U
,
Avni
Y
,
Westermark
GT
,
Carlbom
L
,
Ahlstrom
H
,
Eriksson
O
,
Olerud
J
,
Korsgren
O
.
Transplantation of macroencapsulated human islets within the bioartificial pancreas βAir to patients with type 1 diabetes mellitus
.
Am J Transplant
.
2018
;
18
(
7
):
1735
1744
.

367.

Ludwig
B
,
Reichel
A
,
Steffen
A
,
Zimerman
B
,
Schally
AV
,
Block
NL
,
Colton
CK
,
Ludwig
S
,
Kersting
S
,
Bonifacio
E
,
Solimena
M
,
Gendler
Z
,
Rotem
A
,
Barkai
U
,
Bornstein
SR
.
Transplantation of human islets without immunosuppression
.
Proc Natl Acad Sci USA
.
2013
;
110
(
47
):
19054
19058
.

368.

Rezania
A
,
Bruin
JE
,
Arora
P
,
Rubin
A
,
Batushansky
I
,
Asadi
A
,
O’Dwyer
S
,
Quiskamp
N
,
Mojibian
M
,
Albrecht
T
,
Yang
YHC
,
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
.

369.

Pagliuca
FW
,
Millman
JR
,
Gurtler
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
.

370.

Vegas
AJ
,
Veiseh
O
,
Gurtler
M
,
Millman
JR
,
Pagliuca
FW
,
Bader
AR
,
Doloff
JC
,
Li
J
,
Chen
M
,
Olejnik
K
,
Tam
HH
,
Jhunjhunwala
S
,
Langan
E
,
Aresta-Dasilva
S
,
Gandham
S
,
McGarrigle
JJ
,
Bochenek
MA
,
Hollister-Lock
J
,
Oberholzer
J
,
Greiner
DL
,
Weir
GC
,
Melton
DA
,
Langer
R
,
Anderson
DG
.
Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice [published correction appears in Nat Med. 2016;22(4):446]
.
Nat Med
.
2016
;
22
(
3
):
306
311
.

371.

Millman
JR
,
Xie
C
,
Van Dervort
A
,
Gurtler
M
,
Pagliuca
FW
,
Melton
DA
.
Generation of stem cell-derived β-cells from patients with type 1 diabetes [published correction appears in Nat Commun. 2016;7:12379]
.
Nat Commun
.
2016
;
7
(
1
):
11463
.

372.

Sui
L
,
Danzl
N
,
Campbell
SR
,
Viola
R
,
Williams
D
,
Xing
Y
,
Wang
Y
,
Phillips
N
,
Poffenberger
G
,
Johannesson
B
,
Oberholzer
J
,
Powers
AC
,
Leibel
RL
,
Chen
X
,
Sykes
M
,
Egli
D
.
β-Cell replacement in mice using human type 1 diabetes nuclear transfer embryonic stem cells
.
Diabetes
.
2018
;
67
(
1
):
26
35
.

373.

Motté
E
,
Szepessy
E
,
Suenens
K
,
Stange
G
,
Bomans
M
,
Jacobs-Tulleneers-Thevissen
D
,
Ling
Z
,
Kroon
E
,
Pipeleers
D
;
Beta Cell Therapy Consortium EU-FP7
.
Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts
.
Am J Physiol Endocrinol Metab
.
2014
;
307
(
9
):
E838
E846
.

374.

Wang
J
,
Sun
Z
,
Gou
W
,
Adams
DB
,
Cui
W
,
Morgan
KA
,
Strange
C
,
Wang
H
.
α-1 Antitrypsin enhances islet engraftment by suppression of instant blood-mediated inflammatory reaction
.
Diabetes
.
2017
;
66
(
4
):
970
980
.

375.

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
.

376.

Abecassis
A
,
Schuster
R
,
Shahaf
G
,
Ozeri
E
,
Green
R
,
Ochayon
DE
,
Rider
P
,
Lewis
EC
.
α1-Antitrypsin increases interleukin-1 receptor antagonist production during pancreatic islet graft transplantation
.
Cell Mol Immunol
.
2014
;
11
(
4
):
377
386
.

377.

McCall
M
,
Pawlick
R
,
Kin
T
,
Shapiro
AM
.
Anakinra potentiates the protective effects of etanercept in transplantation of marginal mass human islets in immunodeficient mice
.
Am J Transplant
.
2012
;
12
(
2
):
322
329
.

378.

Park
YJ
,
Warnock
GL
,
Ao
Z
,
Safikhan
N
,
Meloche
M
,
Asadi
A
,
Kieffer
TJ
,
Marzban
L
.
Dual role of interleukin-1β in islet amyloid formation and its β-cell toxicity: implications for type 2 diabetes and islet transplantation
.
Diabetes Obes Metab
.
2017
;
19
(
5
):
682
694
.

379.

Matsumoto
S
,
Takita
M
,
Chaussabel
D
,
Noguchi
H
,
Shimoda
M
,
Sugimoto
K
,
Itoh
T
,
Chujo
D
,
SoRelle
J
,
Onaca
N
,
Naziruddin
B
,
Levy
MF
.
Improving efficacy of clinical islet transplantation with iodixanol-based islet purification, thymoglobulin induction, and blockage of IL-1β and TNF-α
.
Cell Transplant
.
2011
;
20
(
10
):
1641
1647
.

380.

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
.

381.

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
.

382.

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
.

383.

Ritz-Laser
B
,
Oberholzer
J
,
Toso
C
,
Brulhart
MC
,
Zakrzewska
K
,
Ris
F
,
Bucher
P
,
Morel
P
,
Philippe
J
.
Molecular detection of circulating beta-cells after islet transplantation
.
Diabetes
.
2002
;
51
(
3
):
557
561
.

384.

Berney
T
,
Mamin
A
,
James Shapiro
AM
,
Ritz-Laser
B
,
Brulhart
MC
,
Toso
C
,
Demuylder-Mischler
S
,
Armanet
M
,
Baertschiger
R
,
Wojtusciszyn
A
,
Benhamou
PY
,
Bosco
D
,
Morel
P
,
Philippe
J
.
Detection of insulin mRNA in the peripheral blood after human islet transplantion predicts deterioration of metabolic control
.
Am J Transplant
.
2006
;
6
(
7
):
1704
1711
.

385.

Ling
Z
,
De Pauw
P
,
Jacobs-Tulleneers-Thevissen
D
,
Mao
R
,
Gillard
P
,
Hampe
CS
,
Martens
GA
,
In’t Veld
P
,
Lernmark
A
,
Keymeulen
B
,
Gorus
F
,
Pipeleers
D
.
Plasma GAD65, a marker for early β-cell loss after intraportal islet cell transplantation in diabetic patients
.
J Clin Endocrinol Metab
.
2015
;
100
(
6
):
2314
2321
.

386.

Kanak
MA
,
Takita
M
,
Shahbazov
R
,
Lawrence
MC
,
Chung
WY
,
Dennison
AR
,
Levy
MF
,
Naziruddin
B
.
Evaluation of microRNA375 as a novel biomarker for graft damage in clinical islet transplantation
.
Transplantation
.
2015
;
99
(
8
):
1568
1573
.

387.

Bellin
MD
,
Clark
P
,
Usmani-Brown
S
,
Dunn
TB
,
Beilman
GJ
,
Chinnakotla
S
,
Pruett
TL
,
Ptacek
P
,
Hering
BJ
,
Wang
Z
,
Gilmore
T
,
Wilhelm
JJ
,
Hodges
JS
,
Moran
A
,
Herold
KC
.
Unmethylated insulin DNA is elevated after total pancreatectomy with islet autotransplantation: assessment of a novel beta cell marker
.
Am J Transplant
.
2017
;
17
(
4
):
1112
1118
.

388.

Gala-Lopez
BL
,
Neiman
D
,
Kin
T
,
O’Gorman
D
,
Pepper
AR
,
Malcolm
AJ
,
Pianzin
S
,
Senior
PA
,
Campbell
P
,
Glaser
B
,
Dor
Y
,
Shemer
R
,
Shapiro
AMJ
.
Beta cell death by cell-free DNA and outcome after clinical islet transplantation
.
Transplantation
.
2018
;
102
(
6
):
978
985
.

389.

Vallabhajosyula
P
,
Korutla
L
,
Habertheuer
A
,
Yu
M
,
Rostami
S
,
Yuan
CX
,
Reddy
S
,
Liu
C
,
Korutla
V
,
Koeberlein
B
,
Trofe-Clark
J
,
Rickels
MR
,
Naji
A
.
Tissue-specific exosome biomarkers for noninvasively monitoring immunologic rejection of transplanted tissue
.
J Clin Invest
.
2017
;
127
(
4
):
1375
1391
.