1
|
Lansberry TR, Stabler CL. Immunoprotection of cellular transplants for autoimmune type 1 diabetes through local drug delivery. Adv Drug Deliv Rev 2024; 206:115179. [PMID: 38286164 PMCID: PMC11140763 DOI: 10.1016/j.addr.2024.115179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune condition that results in the destruction of insulin-secreting β cells of the islets of Langerhans. Allogeneic islet transplantation could be a successful treatment for T1DM; however, it is limited by the need for effective, permanent immunosuppression to prevent graft rejection. Upon transplantation, islets are rejected through non-specific, alloantigen specific, and recurring autoimmune pathways. Immunosuppressive agents used for islet transplantation are generally successful in inhibiting alloantigen rejection, but they are suboptimal in hindering non-specific and autoimmune pathways. In this review, we summarize the challenges with cellular immunological rejection and therapeutics used for islet transplantation. We highlight agents that target these three immune rejection pathways and how to package them for controlled, local delivery via biomaterials. Exploring macro-, micro-, and nano-scale immunomodulatory biomaterial platforms, we summarize their advantages, challenges, and future directions. We hypothesize that understanding their key features will help identify effective platforms to prevent islet graft rejection. Outcomes can further be translated to other cellular therapies beyond T1DM.
Collapse
Affiliation(s)
- T R Lansberry
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - C L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Immunology and Pathology, College of Medicine, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
Collapse
|
2
|
Szempruch KR, Walter K, Ebert N, Bridgens K, Desai CS. Pharmacological management of patients undergoing total pancreatectomy with auto-islet transplantation. Pancreatology 2022; 22:656-664. [PMID: 35490122 DOI: 10.1016/j.pan.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/17/2022] [Accepted: 04/15/2022] [Indexed: 12/11/2022]
Abstract
Chronic pancreatitis results in permanent parenchymal destruction of the pancreas gland leading to anatomical and physiological consequences for patients. Surgical management varies, and some patients require total pancreatectomy with autologous islet cell transplantation (TPIAT). Patients undergoing TPIAT require complex and diligent management after surgery. This encompasses the management of glucose control (endocrine function of the pancreas) and supplementing loss of exocrine function of the pancreas with digestive enzymes. Other areas of management include optimizing pain relief while reducing narcotic usage, providing antimicrobial prophylaxis, and reducing loss of islet cells by improving its integrity through anticoagulation and use of anti-inflammatory agents. Each aspect of care is unique to this population. However, comprehensive reviews on its pharmacological management are scarce. This review will discuss the available literature to date surrounding all aspects of pharmacological management of patients undergoing TPIAT.
Collapse
Affiliation(s)
- Kristen R Szempruch
- Pharmacy Department, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Krysta Walter
- Pharmacy Department, Michigan Medicine, Ann Arbor, MI, USA
| | - Natassha Ebert
- Pharmacy Department, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Kathryn Bridgens
- Department of Nutrition and Food Services, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Chirag S Desai
- Department of Surgery, Transplant, University of North Carolina Medical Center, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Lemos JRN, Baidal DA, Poggioli R, Fuenmayor V, Chavez C, Alvarez A, Linetsky E, Mauvais-Jarvis F, Ricordi C, Alejandro R. Prolonged Islet Allograft Function is Associated With Female Sex in Patients After Islet Transplantation. J Clin Endocrinol Metab 2022; 107:e973-e979. [PMID: 34727179 PMCID: PMC8852206 DOI: 10.1210/clinem/dgab787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Islet transplantation (ITx) has proved to be effective in preventing severe hypoglycemia and improving metabolic control in selected subjects with type 1 diabetes. Long-term graft function remains a challenge. Estrogens have been shown to protect β cells from metabolic stresses and improve revascularization of transplanted human islets in the mouse. We aimed to evaluate the influence of sex in allograft survival of ITx recipients. METHODS We analyzed a retrospective cohort of ITx recipients (n = 56) followed-up for up to 20 years. Allograft failure was defined as a stimulated C-peptide <0.3 ng/mL during a mixed-meal tolerance test. Subjects were divided into recipients of at least 1 female donor (group 1) and recipients of male donors only (group 2). RESULTS Group 1 subjects (n = 25) were aged 41.5 ± 8.4 years and group 2 subjects (n = 22) 45.9 ± 7.3 years (P = 0.062). Female recipient frequency was 44.8% (n = 13) in group 1 and 55.2% (n = 16) in group 2 (P = 0.145). Group 2 developed graft failure earlier than group 1 (680 [286-1624] vs 1906 [756-3256] days, P = 0.038). We performed additional analyses on female recipients only from each group (group 1, n = 16; group 2, n = 20). Female recipients in group 1 exhibited prolonged allograft function compared with group 2, after adjustment for confounders (odds ratio, 28.6; 95% CI, 1.3-619.1; P < 0.05). CONCLUSION Recipients of islets from at least 1 female donor exhibited prolonged graft survival compared with recipients of islets from exclusively male donors. In addition, female recipients exhibited prolonged survival compared with male recipients following ITx of at least 1 female donor.
Collapse
Affiliation(s)
- Joana R N Lemos
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David A Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Tulane Center of Excellence in Sex Based Biology & Medicine, New Orleans, LA 70112, USA
| | - Raffaella Poggioli
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Virginia Fuenmayor
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carmen Chavez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ana Alvarez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Elina Linetsky
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Franck Mauvais-Jarvis
- Tulane Center of Excellence in Sex Based Biology & Medicine, New Orleans, LA 70112, USA
- Diabetes Discovery Research & Sex-Based Medicine Laboratory, New Orleans, LA 70112, USA
- Section of Endocrinology and Metabolism, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Cellular Transplantation, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
4
|
Walker S, Appari M, Forbes S. Considerations and challenges of islet transplantation and future therapies on the horizon. Am J Physiol Endocrinol Metab 2022; 322:E109-E117. [PMID: 34927459 DOI: 10.1152/ajpendo.00310.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Islet transplantation is a treatment for selected adults with type 1 diabetes and severe hypoglycemia. Islets from two or more donor pancreases, a scarce resource, are usually required to impact glycemic control, but the treatment falls short of a cure. Islets are avascular when transplanted into the hypoxic liver environment and subjected to inflammatory insults, immune attack, and toxicity from systemic immunosuppression. The Collaborative Islet Transplant Registry, with outcome data on over 1,000 islet transplant recipients, has demonstrated that larger islet numbers transplanted and older age of recipients are associated with better outcomes. Induction with T-cell depleting agents and the TNF-α inhibitor etanercept and maintenance systemic immunosuppression with mTOR inhibitors in combination with calcineurin inhibitors also appear advantageous, but concerns remain over immunosuppressive toxicity. We discuss strategies and therapeutics that address specific challenges of islet transplantation, many of which are at the preclinical stage of development. On the horizon are adjuvant cell therapies with mesenchymal stromal cells and regulatory T cells that have been used in preclinical models and in humans in other contexts; such a strategy may enable reductions in immunosuppression in the early peri-transplant period when the islets are vulnerable to apoptosis. Human embryonic stem cell-derived islets are in early-phase clinical trials and hold the promise of an inexhaustible supply of insulin-producing cells; effective encapsulation of such cells or, silencing of the human leukocyte antigen (HLA) complex would eliminate the need for immunosuppression, enabling this therapy to be used in all those with type 1 diabetes.
Collapse
Affiliation(s)
- Sophie Walker
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Mahesh Appari
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Shareen Forbes
- BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Transplant Unit, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
- Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Rios P, Baidal D, Lemos J, Camhi SS, Infante M, Padilla N, Gil AMA, Fuenmayor V, Ambut J, Qasmi FA, Mantero AM, Cayetano SM, Ruiz P, Ricordi C, Alejandro R. Long-term Persistence of Allosensitization After Islet Allograft Failure. Transplantation 2021; 105:2490-2498. [PMID: 33481552 PMCID: PMC8289921 DOI: 10.1097/tp.0000000000003635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Allosensitization has been reported after discontinuation of immunosuppression following graft failure in islet transplantation (ITx) recipients, though duration of its persistence is unknown. METHODS We evaluated 35 patients with type 1 diabetes who received ITx, including 17 who developed graft failure (ITx alone, n = 13; ITx plus bone marrow-derived hematopoietic stem cells, n = 4) and 18 with persistent graft function. Panel-reactive antibody (PRA) was measured yearly for the duration of graft function within 1 y after graft failure at enrollment and yearly thereafter. RESULTS In ITx alone graft failure patients, 61% (8/13) were PRA-positive at 6 y postgraft failure, and 46% (6/13) developed donor-specific anti-HLA antibodies (DSA to 2 ± 1 donors) during follow-up. The degree of sensitization was variable (cPRA ranging between 22% and 100% after graft failure). Allosensitization persisted for 7-15 y. Three subjects (3/13) were not allosensitized. In ITx plus bone marrow-derived hematopoietic stem cell recipients, cPRA-positivity (88%-98%) and DSA positivity persisted for 15 y in 75% (3/4) of subjects. CONCLUSIONS Allosensitization was minimal while subjects remained on immunosuppression, but after discontinuation of immunosuppressive therapy, the majority of subjects (77%) became allosensitized with persistence of PRA positivity for up to 15 y. Persistence of allosensitization in this patient population is of clinical importance as it may result in longer transplant waiting list times for identification of a suitable donor in the case of requiring a subsequent transplant.
Collapse
Affiliation(s)
- Paola Rios
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - David Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Joana Lemos
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Stephanie S. Camhi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Marco Infante
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Nathalia Padilla
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Ana M. Alvarez Gil
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Virginia Fuenmayor
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Jonathan Ambut
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Fatima A. Qasmi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro M. Mantero
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL
| | - Shari Messinger Cayetano
- Division of Biostatistics, Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL
| | - Phillip Ruiz
- Department of Surgery and Pathology, University of Miami Miller School of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
- Division of Cellular Transplantation, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
6
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Tahbaz M, Yoshihara E. Immune Protection of Stem Cell-Derived Islet Cell Therapy for Treating Diabetes. Front Endocrinol (Lausanne) 2021; 12:716625. [PMID: 34447354 PMCID: PMC8382875 DOI: 10.3389/fendo.2021.716625] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Insulin injection is currently the main therapy for type 1 diabetes (T1D) or late stage of severe type 2 diabetes (T2D). Human pancreatic islet transplantation confers a significant improvement in glycemic control and prevents life-threatening severe hypoglycemia in T1D patients. However, the shortage of cadaveric human islets limits their therapeutic potential. In addition, chronic immunosuppression, which is required to avoid rejection of transplanted islets, is associated with severe complications, such as an increased risk of malignancies and infections. Thus, there is a significant need for novel approaches to the large-scale generation of functional human islets protected from autoimmune rejection in order to ensure durable graft acceptance without immunosuppression. An important step in addressing this need is to strengthen our understanding of transplant immune tolerance mechanisms for both graft rejection and autoimmune rejection. Engineering of functional human pancreatic islets that can avoid attacks from host immune cells would provide an alternative safe resource for transplantation therapy. Human pluripotent stem cells (hPSCs) offer a potentially limitless supply of cells because of their self-renewal ability and pluripotency. Therefore, studying immune tolerance induction in hPSC-derived human pancreatic islets will directly contribute toward the goal of generating a functional cure for insulin-dependent diabetes. In this review, we will discuss the current progress in the immune protection of stem cell-derived islet cell therapy for treating diabetes.
Collapse
Affiliation(s)
- Meghan Tahbaz
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
- *Correspondence: Eiji Yoshihara,
| |
Collapse
|
8
|
Maffi P, Lundgren T, Tufveson G, Rafael E, Shaw JAM, Liew A, Saudek F, Witkowski P, Golab K, Bertuzzi F, Gustafsson B, Daffonchio L, Ruffini PA, Piemonti L. Targeting CXCR1/2 Does Not Improve Insulin Secretion After Pancreatic Islet Transplantation: A Phase 3, Double-Blind, Randomized, Placebo-Controlled Trial in Type 1 Diabetes. Diabetes Care 2020; 43:710-718. [PMID: 32019854 PMCID: PMC7876579 DOI: 10.2337/dc19-1480] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/11/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Reparixin is an inhibitor of CXCR1/2 chemokine receptor shown to be an effective anti-inflammatory adjuvant in a pilot clinical trial in allotransplant recipients. RESEARCH DESIGN AND METHODS A phase 3, multicenter, randomized, double-blind, parallel-assignment study (NCT01817959) was conducted in recipients of islet allotransplants randomized (2:1) to reparixin or placebo in addition to immunosuppression. Primary outcome was the area under the curve (AUC) for C-peptide during the mixed-meal tolerance test at day 75 ± 5 after the first and day 365 ± 14 after the last transplant. Secondary end points included insulin independence and standard measures of glycemic control. RESULTS The intention-to-treat analysis did not show a significant difference in C-peptide AUC at both day 75 (27 on reparixin vs. 18 on placebo, P = 0.99) and day 365 (24 on reparixin vs. 15 on placebo, P = 0.71). There was no statistically significant difference between treatment groups at any time point for any secondary variable. Analysis of patient subsets showed a trend for a higher percentage of subjects retaining insulin independence for 1 year after a single islet infusion in patients receiving reparixin as compared with patients receiving placebo (26.7% vs. 0%, P = 0.09) when antithymocyte globulin was used as induction immunosuppression. CONCLUSIONS In this first double-blind randomized trial, islet transplantation data obtained with reparixin do not support a role of CXCR1/2 inhibition in preventing islet inflammation-mediated damage.
Collapse
Affiliation(s)
- Paola Maffi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Torbjörn Lundgren
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska, Sweden
| | | | | | - James A M Shaw
- Institute of Cellular Medicine, Newcastle University, and Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, U.K
| | - Aaron Liew
- Institute of Cellular Medicine, Newcastle University, and Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, U.K
| | - Frantisek Saudek
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Piotr Witkowski
- Transplantation Institute, University of Chicago Medicine, Chicago, IL
| | - Karolina Golab
- Transplantation Institute, University of Chicago Medicine, Chicago, IL
| | | | | | - Luisa Daffonchio
- Research and Development Department, Dompé farmaceutici S.p.A., Milan, Italy
| | | | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
9
|
Szempruch KR, Banerjee O, McCall RC, Desai CS. Use of anti-inflammatory agents in clinical islet cell transplants: A qualitative systematic analysis. Islets 2019; 11:65-75. [PMID: 31149871 PMCID: PMC6548473 DOI: 10.1080/19382014.2019.1601543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Immunologic and non-immunologic loss of islet cells upon their transplantation into the liver leads to suboptimal outcomes. Anti-inflammatory agents are used during autologous and allogeneic transplantation. The aim of this qualitative systematic literature review is to evaluate their clinical use and safety. Electronic databases Embase, PubMed, Cumulative Index for Nursing and Allied Health Literature, ClinicalTrials.gov, and EU Clinical Trials Register were searched. Of the 216 unique citations, 10 with tumor necrosis factor (TNF) blockers [etanercept (ETA) or infliximab] and 3 with both TNF blockers and an interluekin-1 receptor antagonist [anakinra (ANA)]) were included. Of these, 12 were in allogeneic and one in autologous transplant. Insulin independence with decreased islet cells and number of transfusions were reported with their use. One infection was reported in a group receiving ETA. Analysis suggested that the use of ETA ± ANA have the potential to improve outcomes in islet cell transplant.
Collapse
Affiliation(s)
| | - Oyshik Banerjee
- Department of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C. McCall
- Health Sciences Library University of North Carolina, Chapel Hill, USA
| | - Chirag S. Desai
- Department of Surgery, Abdominal Transplant, University of North Carolina, Chapel Hill, NC, USA
- CONTACT Chirag S. Desai Department of Surgery, Abdominal Transplant, University of North Carolina Medical Center, 4021 Burnett-Womack CB 7211, Chapel Hill, NC 27599
| |
Collapse
|
10
|
Long-term drug modification to the surface of mesenchymal stem cells by the avidin-biotin complex method. Sci Rep 2017; 7:16953. [PMID: 29208980 PMCID: PMC5717103 DOI: 10.1038/s41598-017-17166-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 11/22/2017] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have various functions, making a significant contribution to tissue repair. On the other hand, the viability and function of MSCs are not lasting after an in vivo transplant, and the therapeutic effects of MSCs are limited. Although various chemical modification methods have been applied to MSCs to improve their viability and function, most of conventional drug modification methods are short-term and unstable and cause cytotoxicity. In this study, we developed a method for long-term drug modification to C3H10T1/2 cells, murine mesenchymal stem cells, without any damage, using the avidin-biotin complex method (ABC method). The modification of NanoLuc luciferase (Nluc), a reporter protein, to C3H10T1/2 cells by the ABC method lasted for at least 14 days in vitro without major effects on the cellular characteristics (cell viability, cell proliferation, migration ability, and differentiation ability). Moreover, in vivo, the surface Nluc modification to C3H10T1/2 cells by the ABC method lasted for at least 7 days. Therefore, these results indicate that the ABC method may be useful for long-term surface modification of drugs and for effective MSC-based therapy.
Collapse
|
11
|
Forbes S, Oram RA, Smith A, Lam A, Olateju T, Imes S, Malcolm AJ, Shapiro AMJ, Senior PA. Validation of the BETA-2 Score: An Improved Tool to Estimate Beta Cell Function After Clinical Islet Transplantation Using a Single Fasting Blood Sample. Am J Transplant 2016; 16:2704-13. [PMID: 27017888 PMCID: PMC5074289 DOI: 10.1111/ajt.13807] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/01/2016] [Accepted: 03/19/2016] [Indexed: 01/25/2023]
Abstract
The beta score, a composite measure of beta cell function after islet transplantation, has limited sensitivity because of its categorical nature and requires a mixed-meal tolerance test (MMTT). We developed a novel score based on a single fasting blood sample. The BETA-2 score used stepwise forward linear regression incorporating glucose (in millimoles per liter), C-peptide (in nanomoles per liter), hemoglobin A1c (as a percentage) and insulin dose (U/kg per day) as continuous variables from the original beta score data set (n = 183 MMTTs). Primary and secondary analyses assessed the score's ability to detect glucose intolerance (90-min MMTT glucose ≥8 mmol/L) and insulin independence, respectively. A validation cohort of islet transplant recipients (n = 114 MMTTs) examined 12 mo after transplantation was used to compare the score's ability to detect these outcomes. The BETA-2 score was expressed as follows (range 0-42): [Formula: see text] A score <20 and ≥15 detected glucose intolerance and insulin independence, respectively, with >82% sensitivity and specificity. The BETA-2 score demonstrated greater discrimination than the beta score for these outcomes (p < 0.05). Using a fasting blood sample, the BETA-2 score estimates graft function as a continuous variable and shows greater discrimination of glucose intolerance and insulin independence after transplantation versus the beta score, allowing frequent assessments of graft function. Studies examining its utility to track long-term graft function are required.
Collapse
Affiliation(s)
- S Forbes
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - R A Oram
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - A Smith
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
- Department of Surgery, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - A Lam
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - T Olateju
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - S Imes
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
- Department of Surgery, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - A J Malcolm
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
- Department of Surgery, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - A M J Shapiro
- Department of Surgery, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| | - P A Senior
- Department of Medicine, Clinical Islet Transplant Program, University of Alberta & Alberta Health Services, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
G-CSF and Exenatide Might Be Associated with Increased Long-Term Survival of Allogeneic Pancreatic Islet Grafts. PLoS One 2016; 11:e0157245. [PMID: 27285580 PMCID: PMC4902232 DOI: 10.1371/journal.pone.0157245] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/26/2016] [Indexed: 12/16/2022] Open
Abstract
Background Allogeneic human islet transplantation is an effective therapy for the treatment of patients with Type 1 Diabetes (T1D). The low number of islet transplants performed worldwide and the different transplantation protocols used limit the identification of the most effective therapeutic options to improve the efficacy of this approach. Methods We present a retrospective analysis on the data collected from 44 patients with T1D who underwent islet transplantation at our institute between 2000 and 2007. Several variables were included: recipient demographics and immunological characteristics, donor and transplant characteristics, induction protocols, and additional medical treatment received. Immunosuppression was induced with anti-CD25 (Daclizumab), alone or in association with anti-tumor necrosis factor alpha (TNF-α) treatments (Etanercept or Infliximab), or with anti-CD52 (Alemtuzumab) in association with anti-TNF-α treatments (Etanercept or Infliximab). Subsets of patients were treated with Filgrastim for moderate/severe neutropenia and/or Exenatide for post prandial hyperglycemia. Results The analysis performed indicates a negative association between graft survival (c-peptide level ≥ 0.3 ng/ml) and islet infusion volume, with the caveat that, the progressive reduction of infusion volumes over the years has been paralleled by improved immunosuppressive protocols. A positive association is instead suggested between graft survival and administration of Exenatide and Filgrastim, alone or in combination. Conclusion This retrospective analysis may be of assistance to further improve long-term outcomes of protocols for transplant of islets and other organs.
Collapse
|
13
|
Nijhoff MF, Engelse MA, Dubbeld J, Braat AE, Ringers J, Roelen DL, van Erkel AR, Spijker HS, Bouwsma H, van der Boog PJM, de Fijter JW, Rabelink TJ, de Koning EJP. Glycemic Stability Through Islet-After-Kidney Transplantation Using an Alemtuzumab-Based Induction Regimen and Long-Term Triple-Maintenance Immunosuppression. Am J Transplant 2016; 16:246-53. [PMID: 26288226 DOI: 10.1111/ajt.13425] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 06/01/2015] [Accepted: 06/21/2015] [Indexed: 01/25/2023]
Abstract
Pancreatic islet transplantation is performed in a select group of patients with type 1 diabetes mellitus. Immunosuppressive regimens play an important role in long-term islet function. We aimed to investigate the efficacy of islet transplantation in patients with type 1 diabetes and a previous kidney transplantation using an alemtuzumab-based induction regimen and triple maintenance immunosuppression. Patients with type 1 diabetes, who had received a kidney transplant previously, were treated with alemtuzumab as induction therapy for their first islet transplantation and basiliximab induction therapy for subsequent islet transplantations. Maintenance immunosuppression consisted of triple immunosuppression (tacrolimus, mycophenolate mofetil, and prednisolone). Thirteen patients (age 50.9 ± 9.2 years, duration of diabetes 35 ± 9 years) received a total of 22 islet transplantations. One- and 2-year insulin independence was 62% and 42%, respectively; graft function was 100% and 92%, respectively. HbA1c dropped from 57.2 ± 13.1 (7.4 ± 1.2%) to 44.5 ± 11.8 mmol/molHb (6.2 ± 0.9%) (p = 0.003) after 2 years. Six of 13 patients suffered from severe hypoglycemia before islet transplantation. After transplantation, severe hypoglycemia was restricted to the only patient who lost graft function. Creatinine clearance was unchanged. Islet-after-kidney transplantation in patients with type 1 diabetes using an alemtuzumab-based induction regimen leads to considerable islet allograft function and improvement in glycemic control.
Collapse
Affiliation(s)
- M F Nijhoff
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Centre, Leiden, the Netherlands.,Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - M A Engelse
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - J Dubbeld
- Departments of Surgery, Leiden University Medical Centre, Leiden, the Netherlands
| | - A E Braat
- Departments of Surgery, Leiden University Medical Centre, Leiden, the Netherlands
| | - J Ringers
- Departments of Surgery, Leiden University Medical Centre, Leiden, the Netherlands
| | - D L Roelen
- Immunohematology, Leiden University Medical Centre, Leiden, the Netherlands
| | - A R van Erkel
- Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - H S Spijker
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - H Bouwsma
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - P J M van der Boog
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - J W de Fijter
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - T J Rabelink
- Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| | - E J P de Koning
- Department of Medicine, Division of Endocrinology and Metabolism, Leiden University Medical Centre, Leiden, the Netherlands.,Division of Nephrology and Transplantation, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
14
|
Pancreas Islet Transplantation for Patients With Type 1 Diabetes Mellitus: A Clinical Evidence Review. ONTARIO HEALTH TECHNOLOGY ASSESSMENT SERIES 2015; 15:1-84. [PMID: 26644812 PMCID: PMC4664938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
BACKGROUND Type 1 diabetes mellitus is caused by the autoimmune destruction of pancreatic beta (β) cells, resulting in severe insulin deficiency. Islet transplantation is a β-cell replacement therapeutic option that aims to restore glycemic control in patients with type 1 diabetes. The objective of this study was to determine the clinical effectiveness of islet transplantation in patients with type 1 diabetes, with or without kidney disease. METHODS We conducted a systematic review of the literature on islet transplantation for type 1 diabetes, including relevant health technology assessments, systematic reviews, meta-analyses, and observational studies. We used a two-step process: first, we searched for systematic reviews and health technology assessments; second, we searched primary studies to update the chosen health technology assessment. The Assessment of Multiple Systematic Reviews measurement tool was used to examine the methodological quality of the systematic reviews and health technology assessments. We assessed the quality of the body of evidence and the risk of bias according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) Working Group criteria. RESULTS Our searched yielded 1,354 citations. One health technology assessment, 11 additional observational studies to update the health technology assessment, one registry report, and four guidelines were included; the observational studies examined islet transplantation alone, islet-after-kidney transplantation, and simultaneous islet-kidney transplantation. In general, low to very low quality of evidence exists for islet transplantation in patients with type 1 diabetes with difficult-to-control blood glucose levels, with or without kidney disease, for these outcomes: health-related quality of life, secondary complications of diabetes, glycemic control, and adverse events. However, high quality of evidence exists for the specific glycemic control outcome of insulin independence compared with intensive insulin therapy. For patients without kidney disease, islet transplantation improves glycemic control and diabetic complications for patients with type 1 diabetes when compared with intensive insulin therapy. However, results for health-related quality of life outcomes were mixed, and adverse events were increased compared with intensive insulin therapy. For patients with type 1 diabetes with kidney disease, islet-after-kidney transplantation or simultaneous islet-kidney transplantation also improved glycemic control and secondary diabetic complications, although the evidence was more limited for this patient group. Compared with intensive insulin therapy, adverse events for islet-after-kidney transplantation or simultaneous islet-kidney transplantation were increased, but were in general less severe than with whole pancreas transplantation. CONCLUSIONS For patients with type 1 diabetes with difficult-to-control blood glucose levels, islet transplantation may be a beneficial β-cell replacement therapy to improve glycemic control and secondary complications of diabetes. However, there is uncertainty in the estimates of effectiveness because of the generally low to very low quality of evidence for all outcomes of interest.
Collapse
|
15
|
Papas KK, Bellin MD, Sutherland DER, Suszynski TM, Kitzmann JP, Avgoustiniatos ES, Gruessner AC, Mueller KR, Beilman GJ, Balamurugan AN, Loganathan G, Colton CK, Koulmanda M, Weir GC, Wilhelm JJ, Qian D, Niland JC, Hering BJ. Islet Oxygen Consumption Rate (OCR) Dose Predicts Insulin Independence in Clinical Islet Autotransplantation. PLoS One 2015; 10:e0134428. [PMID: 26258815 PMCID: PMC4530873 DOI: 10.1371/journal.pone.0134428] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 07/10/2015] [Indexed: 12/05/2022] Open
Abstract
Background Reliable in vitro islet quality assessment assays that can be performed routinely, prospectively, and are able to predict clinical transplant outcomes are needed. In this paper we present data on the utility of an assay based on cellular oxygen consumption rate (OCR) in predicting clinical islet autotransplant (IAT) insulin independence (II). IAT is an attractive model for evaluating characterization assays regarding their utility in predicting II due to an absence of confounding factors such as immune rejection and immunosuppressant toxicity. Methods Membrane integrity staining (FDA/PI), OCR normalized to DNA (OCR/DNA), islet equivalent (IE) and OCR (viable IE) normalized to recipient body weight (IE dose and OCR dose), and OCR/DNA normalized to islet size index (ISI) were used to characterize autoislet preparations (n = 35). Correlation between pre-IAT islet product characteristics and II was determined using receiver operating characteristic analysis. Results Preparations that resulted in II had significantly higher OCR dose and IE dose (p<0.001). These islet characterization methods were highly correlated with II at 6–12 months post-IAT (area-under-the-curve (AUC) = 0.94 for IE dose and 0.96 for OCR dose). FDA/PI (AUC = 0.49) and OCR/DNA (AUC = 0.58) did not correlate with II. OCR/DNA/ISI may have some utility in predicting outcome (AUC = 0.72). Conclusions Commonly used assays to determine whether a clinical islet preparation is of high quality prior to transplantation are greatly lacking in sensitivity and specificity. While IE dose is highly predictive, it does not take into account islet cell quality. OCR dose, which takes into consideration both islet cell quality and quantity, may enable a more accurate and prospective evaluation of clinical islet preparations.
Collapse
Affiliation(s)
- Klearchos K. Papas
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, Arizona, United States of America
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Melena D. Bellin
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - David E. R. Sutherland
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Thomas M. Suszynski
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Jennifer P. Kitzmann
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, Arizona, United States of America
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Efstathios S. Avgoustiniatos
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Angelika C. Gruessner
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, Arizona, United States of America
| | - Kathryn R. Mueller
- Institute for Cellular Transplantation, Department of Surgery, University of Arizona, Tucson, Arizona, United States of America
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Gregory J. Beilman
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Appakalai N. Balamurugan
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Gopalakrishnan Loganathan
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Clark K. Colton
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Maria Koulmanda
- The Transplant Institute, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gordon C. Weir
- Joslin Diabetes Center, Boston, Massachusetts, United States of America
| | - Josh J. Wilhelm
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| | - Dajun Qian
- Information Science, City of Hope, Duarte, California, United States of America
| | - Joyce C. Niland
- Information Science, City of Hope, Duarte, California, United States of America
| | - Bernhard J. Hering
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, United States of America
- Schulze Diabetes Institute, Minneapolis, Minnesota, United States of America
| |
Collapse
|
16
|
Ling Z, De Pauw P, Jacobs-Tulleneers-Thevissen D, Mao R, Gillard P, Hampe CS, Martens GA, In't Veld P, Lernmark Å, 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; 100:2314-21. [PMID: 25816051 PMCID: PMC5393519 DOI: 10.1210/jc.2015-1216] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT AND OBJECTIVE Intraportal islet transplantation can restore insulin production in type 1 diabetes patients, but its effect is subject to several interfering processes. To assess the influence of β-cell loss before and during engraftment, we searched for a real-time marker of β-cell destruction. Previous studies showed that 65-kDa isoform of glutamate decarboxylase (GAD65) is discharged by chemically damaged rat β-cells. We therefore examined the utility of the GAD65 assay to detect and quantify destruction of human β-cells in vitro and in vivo. DESIGN AND PARTICIPANTS A time-resolved fluorescence immunoassay was used to measure GAD65 discharge from β-cells after administration of toxins or after intraportal transplantation. The study in patients involved type 1 diabetes recipients of 56 implants. RESULTS GAD65 was discharged from cultured human β-cells between 4 and 24 hours after acute insult and proportional to the number of dying cells. It was also detected in plasma during the first 24 hours after intraportal transplantation of human islet cell grafts. Diabetic nude rat recipients without hyperglycemic correction exhibited higher plasma GAD65 levels than those with normalization. In type 1 diabetes recipients of grafts with 2-5 × 10(6) β-cells per kilogram of body weight, five of six with plasma GAD65 greater than 1 ng/mL failed to increase plasma C-peptide by greater than 0.5 ng/mL at posttransplant month 2, whereas five of six with undetectable plasma GAD 65 and 15 of 19 with intermediate levels did result in such increase. CONCLUSION Plasma GAD65 qualifies as a marker for early β-cell loss after intraportal transplantation. Further studies are needed to extend its clinical utility.
Collapse
Affiliation(s)
- Zhidong Ling
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Pieter De Pauw
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Daniel Jacobs-Tulleneers-Thevissen
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Rui Mao
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Pieter Gillard
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Christiane S Hampe
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Geert A Martens
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Peter In't Veld
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Åke Lernmark
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Bart Keymeulen
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Frans Gorus
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Daniel Pipeleers
- Diabetes Research Center and Universitair Ziekenhuis Brussel (Z.L., P.D.P., D.J.-T.-T., R.M., G.A.M., P.I.V., B.K., F.G., D.P.), Brussels Free University-VUB, B-1090 Brussels, Belgium; Department of Endocrinology (P.G.), Universitair Ziekenhuis Gasthuisberg, Katholieke Universiteit Leuven-KUL, B-3000 Leuven, Belgium; Department of Medicine (C.S.H.), University of Washington, Seattle, Washington 98109; and Department of Clinical Sciences (Å.L.), Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| |
Collapse
|
17
|
Lau J, Vasylovska S, Kozlova EN, Carlsson PO. Surface coating of pancreatic islets with neural crest stem cells improves engraftment and function after intraportal transplantation. Cell Transplant 2015; 24:2263-72. [PMID: 25581301 DOI: 10.3727/096368915x686184] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to develop techniques for surface coating of islets with neural crest stem cells (NCSCs) in order to enable cotransplantation to the clinically used liver site and then investigate engraftment and function intraportally of such bioengineered islets. Mouse islets were coated during incubation with enhanced green fluorescent protein (EGFP)-expressing mouse NCSCs and transplanted into the portal vein to cure diabetic mice. An intravenous glucose tolerance test was performed at 1 month posttransplantation. Islet grafts were retrieved and evaluated for vascular density, nerves, and glial cells. NCSCs expressed a vast number of key angiogenic and neurotrophic factors. Mice transplanted with NCSC-bioengineered islets responded better to the glucose load than recipient mice with control islets. NCSCs remained present in the vicinity or had often migrated into the NCSC-coated islets, and an improved islet graft reinnervation and revascularization was observed. Transplanted NCSCs differentiated into both glial and neural cells in the islet grafts. We conclude that bioengineering of islets with NCSCs for intraportal transplantation provides a possibility to improve islet engraftment and function. Pending successful establishment of protocols for expansion of NCSCs from, for example, human skin or bone marrow, this strategy may be applied to clinical islet transplantation.
Collapse
Affiliation(s)
- Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
18
|
Ahearn AJ, Parekh JR, Posselt AM. Islet transplantation for Type 1 diabetes: where are we now? Expert Rev Clin Immunol 2014; 11:59-68. [DOI: 10.1586/1744666x.2015.978291] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Kanak MA, Takita M, Kunnathodi F, Lawrence MC, Levy MF, Naziruddin B. Inflammatory response in islet transplantation. Int J Endocrinol 2014; 2014:451035. [PMID: 24883060 PMCID: PMC4021753 DOI: 10.1155/2014/451035] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/04/2014] [Accepted: 02/12/2014] [Indexed: 12/23/2022] Open
Abstract
Islet cell transplantation is a promising beta cell replacement therapy for patients with brittle type 1 diabetes as well as refractory chronic pancreatitis. Despite the vast advancements made in this field, challenges still remain in achieving high frequency and long-term successful transplant outcomes. Here we review recent advances in understanding the role of inflammation in islet transplantation and development of strategies to prevent damage to islets from inflammation. The inflammatory response associated with islets has been recognized as the primary cause of early damage to islets and graft loss after transplantation. Details on cell signaling pathways in islets triggered by cytokines and harmful inflammatory events during pancreas procurement, pancreas preservation, islet isolation, and islet infusion are presented. Robust control of pre- and peritransplant islet inflammation could improve posttransplant islet survival and in turn enhance the benefits of islet cell transplantation for patients who are insulin dependent. We discuss several potent anti-inflammatory strategies that show promise for improving islet engraftment. Further understanding of molecular mechanisms involved in the inflammatory response will provide the basis for developing potent therapeutic strategies for enhancing the quality and success of islet transplantation.
Collapse
Affiliation(s)
- Mazhar A. Kanak
- Institute for Biomedical Studies, Baylor University, Waco, TX 76712, USA
| | - Morihito Takita
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | - Faisal Kunnathodi
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | | | - Marlon F. Levy
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| | - Bashoo Naziruddin
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| |
Collapse
|
20
|
Implementation of a simplified method of islet isolation for allogeneic islet transplantation in cynomolgus monkeys. Pancreas 2014; 43:226-35. [PMID: 24518500 DOI: 10.1097/01.mpa.0000437322.44975.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES The present study describes a simple and cost-effective islet isolation procedure. Using this method, allogeneic islets reverse diabetes in cynomolgus monkeys. METHODS Pancreatic tissue from 11 cynomolgus monkeys were digested, collected, and purified using a simplified method. Islet quantification, purity, viability, and glucose static incubation were conducted immediately after isolation. Five streptozotocin-induced monkeys with diabetes were transplanted intrahepatically, and liver biopsies from 3 of these monkeys were taken at different time points for histologic study. RESULTS The mean (SD) of viability, purity, and static glucose incubation stimulation index were 94.4% (2.3%), 91.8% (3.4%), and 2.6 (1.7), respectively. Monkeys who received a mean (SD) dose of 19,968 (2273) islet equivalent per kilogram (n = 4) from 2 to 3 donors who achieved prolonged normoglycemia (57-232 days), whereas the single monkey who received an islet dose of 8000 islet equivalent per kilogram did not experience diabetes reversal. Immunohistochemical assessment of the liver biopsies taken from the monkeys with normoglycemia revealed an insulin- and glucagon-positive islet graft for up to 6 months with minimal peri-islet inflammatory infiltration. CONCLUSIONS This study demonstrates that cynomolgus monkey islets can be successfully and efficiently harvested using a simple isolation method, and these islets can restore normoglycemia in monkeys with diabetes.
Collapse
|
21
|
Azzi J, Geara AS, El-Sayegh S, Abdi R. Immunological aspects of pancreatic islet cell transplantation. Expert Rev Clin Immunol 2014; 6:111-24. [DOI: 10.1586/eci.09.67] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
22
|
Pepper AR, Gala-Lopez B, Ziff O, Shapiro AMJ. Current status of clinical islet transplantation. World J Transplant 2013; 3:48-53. [PMID: 24392308 PMCID: PMC3879523 DOI: 10.5500/wjt.v3.i4.48] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 08/29/2013] [Indexed: 02/05/2023] Open
Abstract
Islet transplantation (IT) is today a well-established treatment modality for selected patients with type 1 diabetes mellitus (T1DM). After the success of the University of Alberta group with a modified approach to the immune protection of islets, the international experience grew along with the numbers of transplants in highly specialized centers. Yet, long-term analysis of those initial results from the Edmonton group indicated that insulin-independence was not durable and most patients return to modest amounts of insulin around the fifth year, without recurrent hypoglycemia events. Many phenomena have been identified as limiting factor for the islet engraftment and survival, and today all efforts are aimed to improve the quality of islets and their engrafting process, as well as more optimized immunosuppression to facilitate tolerance and ultimately, better long term survival. This brief overview presents recent progress in IT. A concise historical perspective is provided, along with the latest efforts to improve islet engraftment, immune protection and ultimately, prolonged graft survival. It is apparent that as the community continues to work together further optimizing IT, it is hopeful a cure for T1DM will soon be achievable.
Collapse
|
23
|
Efficacy of DHMEQ, a NF-κB inhibitor, in islet transplantation: II. Induction DHMEQ treatment ameliorates subsequent alloimmune responses and permits long-term islet allograft acceptance. Transplantation 2013; 96:454-62. [PMID: 23860082 DOI: 10.1097/tp.0b013e31829b077f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Long-term graft deterioration remains a major obstacle in the success of pancreatic islet transplantation (PITx). Antigen-independent inflammatory and innate immune responses strengthen subsequent antigen-dependent immunity; further, activation of nuclear factor (NF)-κB plays a key role during these responses. In this study, we tested our hypothesis that, by the inhibition of NF-κB activation, the suppression of these early responses after PITx could facilitate graft acceptance. METHODS Full major histocompatibility complex (MHC)-mismatched BALB/c (H-2) mice islets were transplanted into streptozotocin-induced diabetic C57BL/6 (B6: H-2) mice. The NF-κB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) was administered for either 3 or 14 days after PITx. To some PITx recipients, tacrolimus was also administered. Islet allograft survival, alloimmune responses, and in vitro effects of DHMEQ on dendritic cells (DCs) were assessed. RESULTS With a vehicle treatment, 600 islet allografts were promptly rejected after PITx. In contrast, 3-day treatment with DHMEQ, followed by 2-week treatment with tacrolimus, allowed permanent acceptance of islet allografts. The endogenous danger-signaling molecule high mobility group complex 1 (HMGB1) was elevated in sera shortly after PITx, whereas DHMEQ administration abolished this elevation. DHMEQ suppressed HMGB1-driven cellular activation and proinflammatory cytokine secretion in mouse bone marrow-derived DCs and significantly reduced the capacity of DCs to prime allogeneic T-cell proliferation in vitro. Finally, the DHMEQ plus tacrolimus regimen reverted the diabetic state with only 300 islet allografts. CONCLUSIONS Inhibition of NF-κB activation by DHMEQ shortly after PITx suppresses HMGB1, which activates DCs and strengthens the magnitude of alloimmune responses; this permits long-term islet allograft acceptance, even in case of fewer islet allografts.
Collapse
|
24
|
Abstract
Islet transplantation is today an accepted modality for treating selected patients with frequent hypoglycemic events or severe glycemic lability. Despite tremendous progress in islet isolation, culture, and preservation, clinical use is still restricted to a limited subset, and lifelong immunosuppression is required. Issues surrounding limited islet revascularization and immune destruction remain. One of the major challenges is to prevent alloreactivity and recurrence of autoimmunity against β-cells. These two hurdles can be effectively reduced by immunosuppressive therapy combining induction and maintenance treatments. The introduction of highly potent and selective biologic agents has significantly reduced the frequency of acute rejection and has prolonged graft survival, while minimizing the complications of this therapeutic scheme. This review will address the most important biological agents used in islet transplantation. We provide a historical perspective of their introduction into clinical practice and their role in current clinical protocols, aiming at improved engraftment efficiency, increased long-term survival, and better overall results of clinical islet transplantation.
Collapse
Affiliation(s)
- Boris Gala-Lopez
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | | | | |
Collapse
|
25
|
Hu M, Wu J, Zhang GY, Wang YM, Watson D, Yi S, Hawthorne WJ, O'connell PJ, Alexander SI. Selective Depletion of Alloreactive T Cells Leads to Long-Term Islet Allograft Survival across a Major Histocompatibility Complex Mismatch in Diabetic Mice. Cell Transplant 2013; 22:1929-41. [DOI: 10.3727/096368912x658025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Islet cell transplantation as a therapy for type 1 diabetes has been limited by progressive graft loss. Significant immunosuppression including T-cell ablation has been used in an attempt to limit islet rejection. Here, we show that CD3+ lymphocytes depleted of alloreactive T cells selected from a mixed lymphocyte reaction (MLR), where responder BALB/c splenocytes stained with carboxyfluorescein succinimidyl ester (CFSE) were stimulated with irradiated C57BL/6 splenocytes for 5 days, infused into diabetic immunodeficient mice are capable of restoring a broad T-cell repertoire and specifically do not reject islet transplants from the strain (C57BL/6) used in the original depletion. These mice demonstrate reconstitution with CD4+ and CD8+ T cells, the capacity to reject third-party grafts (CBA), and restoration of interferon-γ (IFN-γ) responses to third-party alloantigens. Over time, both forkhead box P3-positive (Foxp3+) T regulatory cells (Tregs) and γδ T cells expand, suggesting a role for peripheral tolerance, in addition to the initial depletion of alloreactive T cells, in long-term islet graft survival. Our results suggest that immune restoration with CD3+ lymphocytes where alloreactive T cells are removed can restore cognate immunity without islet allograft loss and recurrence of diabetes.
Collapse
Affiliation(s)
- M. Hu
- Centre for Kidney Research, the Children's Hospital at Westmead, the University of Sydney, NSW, Australia
- Centre for Transplant and Renal Research, Westmead Millennium Institute, the University of Sydney, NSW, Australia
| | - J. Wu
- Centre for Transplant and Renal Research, Westmead Millennium Institute, the University of Sydney, NSW, Australia
| | - G. Y. Zhang
- Centre for Kidney Research, the Children's Hospital at Westmead, the University of Sydney, NSW, Australia
| | - Y. M. Wang
- Centre for Kidney Research, the Children's Hospital at Westmead, the University of Sydney, NSW, Australia
| | - D. Watson
- Centre for Kidney Research, the Children's Hospital at Westmead, the University of Sydney, NSW, Australia
- Centre for Medical Bioscience, University of Wollongong, NSW, Australia
| | - S. Yi
- Centre for Transplant and Renal Research, Westmead Millennium Institute, the University of Sydney, NSW, Australia
| | - W. J. Hawthorne
- Centre for Transplant and Renal Research, Westmead Millennium Institute, the University of Sydney, NSW, Australia
| | - P. J. O'connell
- Centre for Transplant and Renal Research, Westmead Millennium Institute, the University of Sydney, NSW, Australia
| | - S. I. Alexander
- Centre for Kidney Research, the Children's Hospital at Westmead, the University of Sydney, NSW, Australia
| |
Collapse
|
26
|
Marek-Trzonkowska N, Myśliwec M, Siebert J, Trzonkowski P. Clinical application of regulatory T cells in type 1 diabetes. Pediatr Diabetes 2013; 14:322-32. [PMID: 23627860 DOI: 10.1111/pedi.12029] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 01/28/2013] [Accepted: 01/31/2013] [Indexed: 12/21/2022] Open
Abstract
Regulatory T cells (Tregs) are responsible for the maintenance of peripheral tolerance. Animal studies have shown that administration of Tregs can prevent type 1 diabetes (DM1). Several clinical trials attempted to induce Tregs with various agents, and thus provide long-term tolerance of β cells in DM1. Nevertheless, most of these studies have focused on clinical parameters (e.g. C-peptide) and not Treg numbers nor their function after treatment. Therefore, it is not possible to conclude if the majority of these therapies failed because the drugs did not induce Tregs, or if they failed despite Treg expansion. The current knowledge regarding Tregs, along with our experience in Treg therapy of patients with graft versus host disease, prompted us to use ex vivo expanded Tregs in 10 children with recent-onset DM1. No adverse effects in the treated individuals were observed. There was a significant increase in Treg number in peripheral blood immediately after the treatment administration, while the first clinical differences between treated and control patients were observed 4 months after Treg injection. Treated individuals had higher C-peptide levels and lower insulin requirements than non-treated children. Eleven months after diagnosis of DM1, there are still 2 individuals who are independent of exogenous insulin. These results indicate that autologous Tregs are a safe and well-tolerated therapy in children with DM1, which can inhibit or delay the destruction of pancreatic β cells. Additionally, Tregs can be a useful tool for local protection of transplanted pancreatic islets. Isolation and expansion of antigen-specific Tregs is one of the directions for future studies on cellular therapy of DM1.
Collapse
|
27
|
Watanabe M, Yamashita K, Suzuki T, Kamachi H, Kuraya D, Koshizuka Y, Ogura M, Yoshida T, Aoyagi T, Fukumori D, Shimamura T, Okimura K, Maeta K, Miura T, Sakai F, Todo S. ASKP1240, a fully human anti-CD40 monoclonal antibody, prolongs pancreatic islet allograft survival in nonhuman primates. Am J Transplant 2013; 13:1976-88. [PMID: 23841873 DOI: 10.1111/ajt.12330] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 05/08/2013] [Accepted: 05/10/2013] [Indexed: 01/25/2023]
Abstract
A strategy for inhibiting CD40 has been considered as an alternative approach for immunosuppression because of undesirable effects of anti-CD154 monoclonal antibodies (mAbs). Previously, we demonstrated that ASKP1240, which is a fully human anti-CD40 mAb, significantly prolonged kidney and liver allograft survival in cynomolgus monkeys without causing thromboembolic complications. Herein, we evaluated the effect of ASKP1240 on pancreatic islet transplantation (PITx) in cynomolgus monkeys. Diabetes was induced by total pancreatectomy, and islet allografts were transplanted into the liver. Following PITx (8201-12 438 IEQ/kg), blood glucose levels normalized promptly in all animals. Control islet allografts were rejected within 9 days (n = 3), whereas ASKP1240 (10 mg/kg) given on postoperative days 0, 4, 7, 11 and 14 (induction treatment, n = 5) significantly prolonged graft survival time (GST) to >15, >23, 210, 250 and >608 days, respectively. When ASKP1240 (5 mg/kg) was administered weekly thereafter up to post-PITx 6 months (maintenance treatment, n = 4), GST was markedly prolonged to >96, >115, 523 and >607 days. During the ASKP1240 treatment period, both anti-donor cellular responses and development of anti-donor antibodies were abolished, and no serious adverse events were noted. ASKP1240 appears to be a promising candidate for immunosuppression in clinical PITx.
Collapse
Affiliation(s)
- M Watanabe
- First Department of Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
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; 13:1850-8. [PMID: 23668890 DOI: 10.1111/ajt.12250] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/14/2013] [Accepted: 03/14/2013] [Indexed: 01/25/2023]
Abstract
Whilst initial rates of insulin independence following islet transplantation are encouraging, long-term function using the Edmonton Protocol remains a concern. The aim of this single-arm, multicenter study was to evaluate an immunosuppressive protocol of initial antithymocyte globulin (ATG), tacrolimus and mycophenolate mofetil (MMF) followed by switching to sirolimus and MMF. Islets were cultured for 24 h prior to transplantation. The primary end-point was an HbA1c of <7% and cessation of severe hypoglycemia. Seventeen recipients were followed for ≥ 12 months. Nine islet preparations were transported interstate for transplantation. Similar outcomes were achieved at all three centers. Fourteen of the 17 (82%) recipients achieved the primary end-point. Nine (53%) recipients achieved insulin independence for a median of 26 months (range 7-39 months) and 6 (35%) remain insulin independent. All recipients were C-peptide positive for at least 3 months. All subjects with unstimulated C-peptide >0.2 nmol/L had cessation of severe hypoglycemia. Nine of the 17 recipients tolerated switching from tacrolimus to sirolimus with similar graft outcomes. There was a small but significant reduction in renal function in the first 12 months. The combination of islet culture, ATG, tacrolimus and MMF is a viable alternative for islet transplantation.
Collapse
Affiliation(s)
- P J O'Connell
- National Pancreas Transplant Unit, University of Sydney at Westmead Hospital, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ten Years' Evaluation of Diet, Anthropometry, and Physical Exercise Adherence After Islet Allotransplantation. Transplant Proc 2013; 45:2025-8. [DOI: 10.1016/j.transproceed.2013.01.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/15/2013] [Indexed: 12/29/2022]
|
30
|
Takita M, Matsumoto S, Shimoda M, Chujo D, Itoh T, Sorelle JA, Purcell K, Onaca N, Naziruddin B, Levy MF. Safety and tolerability of the T-cell depletion protocol coupled with anakinra and etanercept for clinical islet cell transplantation. Clin Transplant 2013; 26:E471-84. [PMID: 23061757 DOI: 10.1111/ctr.12011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Islet cell transplantation (ICT) is a promising approach to cure patients with type 1 diabetes. We have implemented a new immunosuppression protocol with antithymoglobulin plus anti-inflammatory agents of anakinra and eternacept for induction and tacrolimus plus mycophenolate mofetil for maintenance [T-cell depletion with anti-inflammatory (TCD-AI) protocol], resulting in successful single-donor ICT. METHODS Eight islet recipients with type 1 diabetes reported adverse events (AEs) monthly. AEs were compared between three groups: first infusion with the TCD-AI protocol (TCD-AI-1st) and first and second infusion with the Edmonton-type protocol (Edmonton-1st and Edmonton-2nd). RESULTS The incidence of symptomatic AEs within the initial three months in the TCD-AI-1st group was less than in the Edmonton-1st and Edmonton-2nd groups, with a marginally significant difference (mean ± SE: 5.5 ± 0.3, 7.5 ± 0.5, and 8.3 ± 1.3, respectively; p = 0.07). A significant reduction in liver enzyme elevation after ICT was found in the TCD-AI-1st group compared with the Edmonton-1st and Edmonton-2nd groups (p < 0.05). Because of AEs, all patients in the Edmonton protocol eventually converted to the TCD-AI protocol, whereas all patients tolerated the TCD-AI protocol. CONCLUSIONS TCD-AI protocol can be tolerated for successful ICT, although this study includes small cohort, and large population trial should be taken.
Collapse
Affiliation(s)
- Morihito Takita
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lowe MC, Badell IR, Turner AP, Thompson PW, Leopardi FV, Strobert EA, Larsen CP, Kirk AD. Belatacept and sirolimus prolong nonhuman primate islet allograft survival: adverse consequences of concomitant alefacept therapy. Am J Transplant 2013; 13:312-9. [PMID: 23279640 PMCID: PMC3558637 DOI: 10.1111/j.1600-6143.2012.04341.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/02/2012] [Accepted: 10/15/2012] [Indexed: 01/25/2023]
Abstract
Calcineurin inhibitors (CNI) and steroids are known to promote insulin resistance, and their avoidance after islet transplantation is preferred from a metabolic standpoint. Belatacept, a B7-specific mediator of costimulation blockade (CoB), is clinically indicated as a CNI alternative in renal transplantation, and we have endeavored to develop a clinically translatable, belatacept-based regimen that could obviate the need for both CNIs and steroids. Based on the known synergy between CoB and mTOR inhibition, we studied rhesus monkeys undergoing MHC-mismatched islet allotransplants treated with belatacept and the mTOR inhibitor, sirolimus. To extend prior work on CoB-resistant rejection, some animals also received CD2 blockade with alefacept (LFA3-Ig). Nine rhesus macaques were rendered diabetic with streptozotocin and underwent islet allotransplantation. All received belatacept and sirolimus; six also received alefacept. Belatacept and sirolimus significantly prolonged rejection-free graft survival (median 225 days compared to 8 days in controls receiving basiliximab and sirolimus; p = 0.022). The addition of alefacept provided no additional survival benefit, but was associated with Cytomegalovirus reactivation in four of six animals. No recipients produced donor-specific alloantibodies. The combination of belatacept and sirolimus successfully prevents islet allograft survival in rhesus monkeys, but induction with alefacept provides no survival benefit and increases the risk of viral reactivation.
Collapse
Affiliation(s)
- MC Lowe
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - IR Badell
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - AP Turner
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - PW Thompson
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - FV Leopardi
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - EA Strobert
- Yerkes National Primate Research Center, Atlanta, GA 30322
| | - CP Larsen
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| | - AD Kirk
- Emory Transplant Center, Emory University, Atlanta, GA 30322
| |
Collapse
|
32
|
Piemonti L, Pileggi A. 25 YEARS OF THE RICORDI AUTOMATED METHOD FOR ISLET ISOLATION. CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2013; 1:e128. [PMID: 30505878 PMCID: PMC6267808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The year 2013 marks the 25th anniversary of the Automated Method for islet isolation. The dissociation chamber at the core of the Automated Method was developed by Dr. Camillo Ricordi in 1988 to enhance the disassembling of the pancreatic tissue via a combined enzymatic and mechanical digestion while preserving endocrine cell cluster integrity. This method has ever since become the gold standard for human and large animal pancreas processing, contributing to the success and increasing number of clinical trials of islet transplantation worldwide. Herein we offer an attempt to a comprehensive, yet unavoidably incomplete, historical review of the progress in the field of islet cell transplantation to restore beta-cell function in patients with diabetes.
Collapse
Affiliation(s)
- Lorenzo Piemonti
- Beta Cell Biology Unit, Diabetes Research Institute (OSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | - Antonello Pileggi
- Cell Transplant Center and Diabetes Research Institute, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology of the University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| |
Collapse
|
33
|
Ishii S, Saito T, Ise K, Yamashita M, Sato Y, Saito T, Tsukada M, Oshibe I, Kenjo A, Kimura T, Anazawa T, Suzuki S, Gotoh M. Preservation of pancreatic islets in cold UW solution before transplantation. Islets 2012; 4:32-9. [PMID: 22504908 DOI: 10.4161/isl.18607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Culture of islets prior to transplantation needs to be revisited for maintaining functional islet capacity. This study was conducted to compare cold UW (University of Wisconsin) preservation with conventional culture based on insulin secretory capacity in vitro and in vivo. Islets isolated from Wistar rats were either cultured for 24 h at 37°C in RPMI1640 medium or DMEM containing various concentrations of glucose or preserved for the same period in UW solution or in DMEM solution at 4°C. The islet yield in UW group, but not in other groups, was maintained as comparable with that of fresh islets. Insulin secretory capacity in response to glucose was maintained only in the islets of UW group, but not in other groups. SCID mice given 300 IEQ islets of UW group showed gradual restoration of normoglycemia as found in the mice given freshly isolated islets. Meanwhile, those mice given cultured islets for 24 h at 37°C in RPMI1640 medium showed rapid decrease of blood glucose levels on day 1 followed by relatively elevated levels on day 2, suggesting unstable insulin secretory capacity of islets. Morphological staining with anti-HMGB1 (high mobility group B1) antibody revealed central damage of islets in all culture groups regardless of glucose concentration and in islets of cold DMEM group, whereas those in the UW group were quite intact. These results suggest that cold preservation in UW solution is simple and beneficial in protecting islets morphologically and functionally before transplantation.
Collapse
Affiliation(s)
- Show Ishii
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Takuro Saito
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Kazuya Ise
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Michitoshi Yamashita
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Yoshihiro Sato
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Takaharu Saito
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Manabu Tsukada
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Ikuro Oshibe
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Akira Kenjo
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Takashi Kimura
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Takayuki Anazawa
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| | - Shigeya Suzuki
- Research and Development Division; Kikkoman Co., Ltd; Noda City, Chiba, Japan
| | - Mitsukazu Gotoh
- Department of Surgery; Fukushima Medical University; Fukushima City, Fukushima, Japan
| |
Collapse
|
34
|
Islet transplantation: factors in short-term islet survival. Arch Immunol Ther Exp (Warsz) 2011; 59:421-9. [PMID: 21984594 DOI: 10.1007/s00005-011-0143-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 05/25/2011] [Indexed: 12/20/2022]
Abstract
Islet transplantation has the potential to cure type 1 diabetes. In recent years, the proportion of patients achieving initial insulin independence has improved, but longer term outcomes remain poor compared to those for whole pancreas transplants. This review article will discuss factors affecting islet yield and viability leading up to transplantation and in the immediate post-transplant period.
Collapse
|
35
|
Maffi P, Scavini M, Socci C, Piemonti L, Caldara R, Gremizzi C, Melzi R, Nano R, Orsenigo E, Venturini M, Staudacher C, Del Maschio A, Secchi A. Risks and benefits of transplantation in the cure of type 1 diabetes: whole pancreas versus islet transplantation. A single center study. Rev Diabet Stud 2011; 8:44-50. [PMID: 21720672 DOI: 10.1900/rds.2011.8.44] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Pancreas and islet transplantation are the only available options to replace beta-cell function in patients with type 1 diabetes. Great variability in terms of rate of success for both approaches is reported in the literature and it is difficult to compare the respective risks and benefits. OBJECTIVES The aim of this study was to analyze risks and benefits of pancreas transplantation alone (PTA) and islet transplantation alone (ITA) by making use of the long-term experience of a single center where both transplantations are performed. We focused on the risks and benefits of both procedures, with the objective of better defining indications and providing evidence to support the decision-making process. The outcomes of 33 PTA and 33 ITA were analyzed, and pancreas and islet function (i.e., insulin independence), perioperative events, and long-term adverse events were recorded. RESULTS We observed a higher rate of insulin independence in PTA (75%) versus ITA (59%), with the longer insulin independence among PTA patients receiving tacrolimus. The occurrence of adverse events was higher for PTA patients in terms of hospitalization length and frequency, re-intervention for surgical and immunological acute complications, CMV reactivation, and other infections. CONCLUSIONS In conclusion, these results support the practice of listing patients for PTA when the metabolic control and the progression of chronic complications require a rapid normalization of glucose levels, with the exception of patients with cardiovascular disease, because of the high surgical risks. ITA is indicated when replacement of beta-cell mass is needed in patients with a high surgical risk.
Collapse
Affiliation(s)
- Paola Maffi
- Diabetes Research Institute, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Toso C, Pawlick R, Lacotte S, Edgar R, Davis J, McCall M, Morel P, Mentha G, Berney T, Shapiro AMJ. Detecting rejection after mouse Islet transplantation utilizing islet protein-stimulated ELISPOT. Cell Transplant 2010; 20:955-62. [PMID: 21054945 DOI: 10.3727/096368910x539137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Improved posttransplant monitoring and on-time detection of rejection could improve islet transplantation outcome. The present study explored the possibility of detecting harmful events after mouse islet transplantation measuring the immune responsiveness against islet extracts. Mouse islet transplantations were performed using various donor/recipient combinations, exploring autoimmune (NOD/SCID to NOD, n = 6) and alloimmune events (C57BL/6 to BALB/c, n = 20), a combination of both (C57BL/6 to NOD, n = 8), the absence of both (BALB/c to BALB/c, n = 21), or naive, nontransplanted control mice (n = 14). The immune reactivity was measured by ELISPOT, looking at the ex vivo release of IFN-γ from splenocytes stimulated by islet donor extracts (sonicated islets). The immune reactivity was not altered in the syngeneic and autoimmune models, demonstrating similar levels as nontransplanted controls (p = 0.46 and p = 0.6). Conversely, the occurrence of an allogeneic rejection alone or in combination to autoimmunity was associated to an increase in the level of immune reactivity (p = 0.023 and p = 0.003 vs. respective controls). The observed increase was transient and lost in the postrejection period or after treatment with CTLA4-Ig. Overall, allogeneic rejection was associated to a transient increase in the reactivity of splenocytes against islet proteins. Such a strategy has the potential to improve islet graft monitoring in human and should be further explored.
Collapse
Affiliation(s)
- Christian Toso
- Transplant Unit, Department of Surgery, University of Geneva Hospitals, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Successes and disappointments with clinical islet transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:749-69. [PMID: 20217523 DOI: 10.1007/978-90-481-3271-3_33] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transplantation of pancreatic islets is considered a therapeutic option for patients with type 1 diabetes mellitus who have life-threatening hypoglycaemic episodes. After the procedure, a decrease in the frequency and severity of hypoglycaemic episodes and sustained graft function as indicated by detectable levels of C-peptide can be seen in the majority of patients. However, true insulin independence, if achieved, usually lasts for at most a few years. Apart from the low insulin independence rates, reasons for concern regarding this procedure are the side effects of the immunosuppressive therapy, allo-immunization, and the high costs. Moreover, whether islet transplantation prevents the progression of diabetic micro- and macrovascular complications is largely unknown. Areas of current research include the development of less toxic immunosuppressive regimens, the control of the inflammatory reaction immediately after transplantation, the identification of the optimal anatomical site for islet infusion, and the possibility to encapsulate transplanted islets to protect them from the allo-immune response. At present, pancreatic islet transplantation is still an experimental procedure, which is only indicated for a highly selected group of type 1 diabetic patients with life-threatening hypoglycaemic episodes.
Collapse
|
38
|
Gagliani N, Ferraro A, Roncarolo MG, Battaglia M. Autoimmune diabetic patients undergoing allogeneic islet transplantation: are we ready for a regulatory T-cell therapy? Immunol Lett 2009; 127:1-7. [DOI: 10.1016/j.imlet.2009.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 07/14/2009] [Accepted: 07/20/2009] [Indexed: 11/30/2022]
|
39
|
Mineo D, Pileggi A, Alejandro R, Ricordi C. Point: steady progress and current challenges in clinical islet transplantation. Diabetes Care 2009; 32:1563-9. [PMID: 19638527 PMCID: PMC2713646 DOI: 10.2337/dc09-0490] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Davide Mineo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- Department of Internal Medicine and University Policlinic, Tor Vergata University, Rome, Italy
| | - Antonello Pileggi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- DeWitt-Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Rodolfo Alejandro
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, Florida
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- DeWitt-Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, Florida
- Department of Biomedical Engineering, University of Miami Miller School of Medicine, Miami, Florida
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
| |
Collapse
|