Skip to main content
Top
Published in: Acta Diabetologica 5/2016

01-10-2016 | Review Article

The state of the art of islet transplantation and cell therapy in type 1 diabetes

Authors: Silvia Pellegrini, Elisa Cantarelli, Valeria Sordi, Rita Nano, Lorenzo Piemonti

Published in: Acta Diabetologica | Issue 5/2016

Login to get access

Abstract

In patients with type 1 diabetes (T1D), pancreatic β cells are destroyed by a selective autoimmune attack and their replacement with functional insulin-producing cells is the only possible cure for this disease. The field of islet transplantation has evolved significantly from the breakthrough of the Edmonton Protocol in 2000, since significant advances in islet isolation and engraftment, together with improved immunosuppressive strategies, have been reported. The main limitations, however, remain the insufficient supply of human tissue and the need for lifelong immunosuppression therapy. Great effort is then invested in finding innovative sources of insulin-producing β cells. One old alternative with new recent perspectives is the use of non-human donor cells, in particular porcine β cells. Also the field of preexisting β cell expansion has advanced, with the development of new human β cell lines. Yet, large-scale production of human insulin-producing cells from stem cells is the most recent and promising alternative. In particular, the optimization of in vitro strategies to differentiate human embryonic stem cells into mature insulin-secreting β cells has made considerable progress and recently led to the first clinical trial of stem cell treatment for T1D. Finally, the discovery that it is possible to derive human induced pluripotent stem cells from somatic cells has raised the possibility that a sufficient amount of patient-specific β cells can be derived from patients through cell reprogramming and differentiation, suggesting that in the future there might be a cell therapy without immunosuppression.
Literature
4.
go back to reference Saudek CD, Duckworth WC, Giobbie-Hurder A et al (1996) Implantable insulin pump vs multiple-dose insulin for non-insulin-dependent diabetes mellitus: a randomized clinical trial. Department of Veterans Affairs Implantable Insulin Pump Study Group. JAMA 276:1322–1327CrossRefPubMed Saudek CD, Duckworth WC, Giobbie-Hurder A et al (1996) Implantable insulin pump vs multiple-dose insulin for non-insulin-dependent diabetes mellitus: a randomized clinical trial. Department of Veterans Affairs Implantable Insulin Pump Study Group. JAMA 276:1322–1327CrossRefPubMed
6.
go back to reference Venturini M, Angeli E, Maffi P et al (2005) Technique, complications, and therapeutic efficacy of percutaneous transplantation of human pancreatic islet cells in type 1 diabetes: the role of US. Radiology 234:617–624. doi:10.1148/radiol.2342031356 CrossRefPubMed Venturini M, Angeli E, Maffi P et al (2005) Technique, complications, and therapeutic efficacy of percutaneous transplantation of human pancreatic islet cells in type 1 diabetes: the role of US. Radiology 234:617–624. doi:10.​1148/​radiol.​2342031356 CrossRefPubMed
8.
go back to reference Scharp DW, Lacy PE, Santiago JV et al (1990) Insulin independence after islet transplantation into type I diabetic patient. Diabetes 39:515–518CrossRefPubMed Scharp DW, Lacy PE, Santiago JV et al (1990) Insulin independence after islet transplantation into type I diabetic patient. Diabetes 39:515–518CrossRefPubMed
10.
go back to reference Bertuzzi F, Verzaro R, Provenzano V, Ricordi C (2007) Brittle type 1 diabetes mellitus. Curr Med Chem 14:1739–1744CrossRefPubMed Bertuzzi F, Verzaro R, Provenzano V, Ricordi C (2007) Brittle type 1 diabetes mellitus. Curr Med Chem 14:1739–1744CrossRefPubMed
11.
go back to reference Ballinger WF, Lacy PE (1972) Transplantation of intact pancreatic islets in rats. Surgery 72:175–186PubMed Ballinger WF, Lacy PE (1972) Transplantation of intact pancreatic islets in rats. Surgery 72:175–186PubMed
12.
go back to reference Kemp CB, Knight MJ, Scharp DW et al (1973) Effect of transplantation site on the results of pancreatic islet isografts in diabetic rats. Diabetologia 9:486–491CrossRefPubMed Kemp CB, Knight MJ, Scharp DW et al (1973) Effect of transplantation site on the results of pancreatic islet isografts in diabetic rats. Diabetologia 9:486–491CrossRefPubMed
13.
go back to reference Najarian JS, Sutherland DE, Matas AJ et al (1977) Human islet transplantation: a preliminary report. Transplant Proc 9:233–236PubMed Najarian JS, Sutherland DE, Matas AJ et al (1977) Human islet transplantation: a preliminary report. Transplant Proc 9:233–236PubMed
14.
go back to reference Ricordi C, Lacy PE, Finke EH et al (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420CrossRefPubMed Ricordi C, Lacy PE, Finke EH et al (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420CrossRefPubMed
15.
go back to reference Oberholzer J, Triponez F, Mage R et al (2000) Human islet transplantation: lessons from 13 autologous and 13 allogeneic transplantations. Transplantation 69:1115–1123CrossRefPubMed Oberholzer J, Triponez F, Mage R et al (2000) Human islet transplantation: lessons from 13 autologous and 13 allogeneic transplantations. Transplantation 69:1115–1123CrossRefPubMed
16.
go back to reference Secchi A, Socci C, Maffi P et al (1997) Islet transplantation in IDDM patients. Diabetologia 40:225–231CrossRefPubMed Secchi A, Socci C, Maffi P et al (1997) Islet transplantation in IDDM patients. Diabetologia 40:225–231CrossRefPubMed
19.
go back to reference Brennan DC, Kopetskie HA, Sayre PH et al (2015) Long-term follow-up of the edmonton protocol of islet transplantation in the United States. Am J Transplant. doi:10.1111/ajt.13458 PubMed Brennan DC, Kopetskie HA, Sayre PH et al (2015) Long-term follow-up of the edmonton protocol of islet transplantation in the United States. Am J Transplant. doi:10.​1111/​ajt.​13458 PubMed
21.
go back to reference Brooks AM, Walker N, Aldibbiat A et al (2013) Attainment of metabolic goals in the integrated UK islet transplant program with locally isolated and transported preparations. Am J Transplant 13:3236–3243. doi:10.1111/ajt.12469 CrossRefPubMed Brooks AM, Walker N, Aldibbiat A et al (2013) Attainment of metabolic goals in the integrated UK islet transplant program with locally isolated and transported preparations. Am J Transplant 13:3236–3243. doi:10.​1111/​ajt.​12469 CrossRefPubMed
23.
go back to reference Lablanche S, Borot S, Wojtusciszyn A et al (2015) Five-year metabolic, functional, and safety results of patients with type 1 diabetes transplanted with allogenic islets within the Swiss-French GRAGIL network. Diabetes Care 38:1714–1722. doi:10.2337/dc15-0094 CrossRefPubMed Lablanche S, Borot S, Wojtusciszyn A et al (2015) Five-year metabolic, functional, and safety results of patients with type 1 diabetes transplanted with allogenic islets within the Swiss-French GRAGIL network. Diabetes Care 38:1714–1722. doi:10.​2337/​dc15-0094 CrossRefPubMed
27.
go back to reference Posselt AM, Szot GL, Frassetto LA et al (2010) Islet transplantation in type 1 diabetic patients using calcineurin inhibitor-free immunosuppressive protocols based on T-cell adhesion or costimulation blockade. Transplantation 90(12):1595–1601CrossRefPubMedPubMedCentral Posselt AM, Szot GL, Frassetto LA et al (2010) Islet transplantation in type 1 diabetic patients using calcineurin inhibitor-free immunosuppressive protocols based on T-cell adhesion or costimulation blockade. Transplantation 90(12):1595–1601CrossRefPubMedPubMedCentral
28.
go back to reference Nijhoff MF, Engelse MA, Dubbeld J et al (2015) Glycemic stability through islet-after-kidney transplantation using an alemtuzumab-based induction regimen and long-term triple-maintenance immunosuppression. Am J Transplant. doi:10.1111/ajt.13425 PubMed Nijhoff MF, Engelse MA, Dubbeld J et al (2015) Glycemic stability through islet-after-kidney transplantation using an alemtuzumab-based induction regimen and long-term triple-maintenance immunosuppression. Am J Transplant. doi:10.​1111/​ajt.​13425 PubMed
29.
go back to reference Maffi P, Berney T, Nano R et al (2014) Calcineurin inhibitor-free immunosuppressive regimen in type 1 diabetes patients receiving islet transplantation: single-group phase 1/2 trial. Transplantation. doi:10.1097/TP.0000000000000396 PubMed Maffi P, Berney T, Nano R et al (2014) Calcineurin inhibitor-free immunosuppressive regimen in type 1 diabetes patients receiving islet transplantation: single-group phase 1/2 trial. Transplantation. doi:10.​1097/​TP.​0000000000000396​ PubMed
32.
go back to reference Watanabe M, Yamashita K, Suzuki T et al (2013) ASKP1240, a fully human anti-CD40 monoclonal antibody, prolongs pancreatic islet allograft survival in nonhuman primates. Am J Transplant 13:1976–1988. doi:10.1111/ajt.12330 CrossRefPubMed Watanabe M, Yamashita K, Suzuki T et al (2013) ASKP1240, a fully human anti-CD40 monoclonal antibody, prolongs pancreatic islet allograft survival in nonhuman primates. Am J Transplant 13:1976–1988. doi:10.​1111/​ajt.​12330 CrossRefPubMed
35.
go back to reference Klymiuk N, Aigner B, Brem G, Wolf E (2010) Genetic modification of pigs as organ donors for xenotransplantation. Mol Reprod Dev 77:209–221PubMed Klymiuk N, Aigner B, Brem G, Wolf E (2010) Genetic modification of pigs as organ donors for xenotransplantation. Mol Reprod Dev 77:209–221PubMed
36.
go back to reference Groth CG, Korsgren O, Tibell A et al (1994) Transplantation of porcine fetal pancreas to diabetic patients. Lancet (London, England) 344:1402–1404CrossRef Groth CG, Korsgren O, Tibell A et al (1994) Transplantation of porcine fetal pancreas to diabetic patients. Lancet (London, England) 344:1402–1404CrossRef
37.
go back to reference Galili U, Shohet SB, Kobrin E et al (1988) Man, apes, and Old World monkeys differ from other mammals in the expression of alpha-galactosyl epitopes on nucleated cells. J Biol Chem 263:17755–17762PubMed Galili U, Shohet SB, Kobrin E et al (1988) Man, apes, and Old World monkeys differ from other mammals in the expression of alpha-galactosyl epitopes on nucleated cells. J Biol Chem 263:17755–17762PubMed
38.
go back to reference Patience C, Takeuchi Y, Weiss RA (1997) Infection of human cells by an endogenous retrovirus of pigs. Nat Med 3:282–286CrossRefPubMed Patience C, Takeuchi Y, Weiss RA (1997) Infection of human cells by an endogenous retrovirus of pigs. Nat Med 3:282–286CrossRefPubMed
39.
go back to reference Cardona K, Korbutt GS, Milas Z et al (2006) Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med 12:304–306. doi:10.1038/nm1375 CrossRefPubMed Cardona K, Korbutt GS, Milas Z et al (2006) Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med 12:304–306. doi:10.​1038/​nm1375 CrossRefPubMed
40.
go back to reference Hering BJ, Wijkstrom M, Graham ML et al (2006) Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med 12:301–303. doi:10.1038/nm1369 CrossRefPubMed Hering BJ, Wijkstrom M, Graham ML et al (2006) Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med 12:301–303. doi:10.​1038/​nm1369 CrossRefPubMed
41.
go back to reference Shin JS, Kim JM, Kim JS et al (2015) Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am J Transplant 15:2837–2850. doi:10.1111/ajt.13345 CrossRefPubMed Shin JS, Kim JM, Kim JS et al (2015) Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am J Transplant 15:2837–2850. doi:10.​1111/​ajt.​13345 CrossRefPubMed
44.
go back to reference Rayat GR, Rajotte RV, Ao Z, Korbutt GS (2000) Microencapsulation of neonatal porcine islets: protection from human antibody/complement-mediated cytolysis in vitro and long-term reversal of diabetes in nude mice. Transplantation 69:1084–1090CrossRefPubMed Rayat GR, Rajotte RV, Ao Z, Korbutt GS (2000) Microencapsulation of neonatal porcine islets: protection from human antibody/complement-mediated cytolysis in vitro and long-term reversal of diabetes in nude mice. Transplantation 69:1084–1090CrossRefPubMed
45.
go back to reference Dufrane D, Goebbels R-M, Gianello P (2010) Alginate macroencapsulation of pig islets allows correction of streptozotocin-induced diabetes in primates up to 6 months without immunosuppression. Transplantation 90:1054–1062. doi:10.1097/TP.0b013e3181f6e267 CrossRefPubMed Dufrane D, Goebbels R-M, Gianello P (2010) Alginate macroencapsulation of pig islets allows correction of streptozotocin-induced diabetes in primates up to 6 months without immunosuppression. Transplantation 90:1054–1062. doi:10.​1097/​TP.​0b013e3181f6e267​ CrossRefPubMed
47.
49.
go back to reference Teta M, Long SY, Wartschow LM et al (2005) Very slow turnover of beta-cells in aged adult mice. Diabetes 54:2557–2567CrossRefPubMed Teta M, Long SY, Wartschow LM et al (2005) Very slow turnover of beta-cells in aged adult mice. Diabetes 54:2557–2567CrossRefPubMed
51.
go back to reference Levine F, Wang S, Beattie GM et al (1995) Development of a cell line from the human fetal pancreas. Transplant Proc 27:3410PubMed Levine F, Wang S, Beattie GM et al (1995) Development of a cell line from the human fetal pancreas. Transplant Proc 27:3410PubMed
52.
go back to reference De la Tour D, Halvorsen T, Demeterco C et al (2001) Beta-cell differentiation from a human pancreatic cell line in vitro and in vivo. Mol Endocrinol 15:476–483PubMed De la Tour D, Halvorsen T, Demeterco C et al (2001) Beta-cell differentiation from a human pancreatic cell line in vitro and in vivo. Mol Endocrinol 15:476–483PubMed
53.
go back to reference Narushima M, Kobayashi N, Okitsu T et al (2005) A human beta-cell line for transplantation therapy to control type 1 diabetes. Nat Biotechnol 23:1274–1282CrossRefPubMed Narushima M, Kobayashi N, Okitsu T et al (2005) A human beta-cell line for transplantation therapy to control type 1 diabetes. Nat Biotechnol 23:1274–1282CrossRefPubMed
54.
go back to reference Ravassard P, Hazhouz Y, Pechberty S et al (2011) A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. J Clin Invest 121:3589–3597CrossRefPubMedPubMedCentral Ravassard P, Hazhouz Y, Pechberty S et al (2011) A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. J Clin Invest 121:3589–3597CrossRefPubMedPubMedCentral
57.
go back to reference Thomson JA, Itskovitz-Eldor J, Shapiro SS et al (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147CrossRefPubMed Thomson JA, Itskovitz-Eldor J, Shapiro SS et al (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147CrossRefPubMed
58.
go back to reference D’Amour KA, Bang AG, Eliazer S et al (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24:1392–1401. doi:10.1038/nbt1259 CrossRefPubMed D’Amour KA, Bang AG, Eliazer S et al (2006) Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol 24:1392–1401. doi:10.​1038/​nbt1259 CrossRefPubMed
61.
go back to reference Kroon E, Martinson LA, Kadoya K et al (2008) Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26:443–452CrossRefPubMed Kroon E, Martinson LA, Kadoya K et al (2008) Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol 26:443–452CrossRefPubMed
62.
go back to reference Schulz TC, Young HY, Agulnick AD et al (2012) A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE 7:e37004CrossRefPubMedPubMedCentral Schulz TC, Young HY, Agulnick AD et al (2012) A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE 7:e37004CrossRefPubMedPubMedCentral
64.
go back to reference Rezania A, Bruin JE, Xu J et al (2013) Enrichment of human embryonic stem cell-derived NKX6.1-expressing pancreatic progenitor cells accelerates the maturation of insulin-secreting cells in vivo. Stem Cells 31:2432–2442. doi:10.1002/stem.1489 CrossRefPubMed Rezania A, Bruin JE, Xu J et al (2013) Enrichment of human embryonic stem cell-derived NKX6.1-expressing pancreatic progenitor cells accelerates the maturation of insulin-secreting cells in vivo. Stem Cells 31:2432–2442. doi:10.​1002/​stem.​1489 CrossRefPubMed
67.
go back to reference Rezania A, Bruin JE, Arora P et al (2014) Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. doi:10.1038/nbt.3033 PubMed Rezania A, Bruin JE, Arora P et al (2014) Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. doi:10.​1038/​nbt.​3033 PubMed
68.
go back to reference Kelly OG, Chan MY, Martinson LA et al (2011) Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol 29:750–756CrossRefPubMed Kelly OG, Chan MY, Martinson LA et al (2011) Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol 29:750–756CrossRefPubMed
69.
go back to reference Jiang W, Sui X, Zhang D et al (2011) CD24: a novel surface marker for PDX1-positive pancreatic progenitors derived from human embryonic stem cells. Stem Cells 29:609–617CrossRefPubMed Jiang W, Sui X, Zhang D et al (2011) CD24: a novel surface marker for PDX1-positive pancreatic progenitors derived from human embryonic stem cells. Stem Cells 29:609–617CrossRefPubMed
70.
go back to reference Osafune K, Caron L, Borowiak M et al (2008) Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol 26:313–315CrossRefPubMed Osafune K, Caron L, Borowiak M et al (2008) Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol 26:313–315CrossRefPubMed
77.
84.
go back to reference Nakagawa M, Koyanagi M, Tanabe K et al (2008) Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 26:101–106. doi:10.1038/nbt1374 CrossRefPubMed Nakagawa M, Koyanagi M, Tanabe K et al (2008) Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 26:101–106. doi:10.​1038/​nbt1374 CrossRefPubMed
86.
go back to reference Motté E, Szepessy E, Suenens K et al (2014) Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts. Am J Physiol Endocrinol Metab 307:E838–E846. doi:10.1152/ajpendo.00219.2014 CrossRefPubMed Motté E, Szepessy E, Suenens K et al (2014) Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts. Am J Physiol Endocrinol Metab 307:E838–E846. doi:10.​1152/​ajpendo.​00219.​2014 CrossRefPubMed
Metadata
Title
The state of the art of islet transplantation and cell therapy in type 1 diabetes
Authors
Silvia Pellegrini
Elisa Cantarelli
Valeria Sordi
Rita Nano
Lorenzo Piemonti
Publication date
01-10-2016
Publisher
Springer Milan
Published in
Acta Diabetologica / Issue 5/2016
Print ISSN: 0940-5429
Electronic ISSN: 1432-5233
DOI
https://doi.org/10.1007/s00592-016-0847-z

Other articles of this Issue 5/2016

Acta Diabetologica 5/2016 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.