Skip to main content
Top
Published in: Current Diabetes Reports 11/2018

01-11-2018 | Immunology, Transplantation, and Regenerative Medicine (L Piemonti and V Sordi, Section Editors)

Will Genetic Engineering Carry Xenotransplantation of Pig Islets to the Clinic?

Authors: Elisabeth Kemter, Joachim Denner, Eckhard Wolf

Published in: Current Diabetes Reports | Issue 11/2018

Login to get access

Abstract

Purpose of Review

Porcine islets represent a potentially attractive beta-cell source for xenotransplantation into patients with type 1 diabetes, who are not eligible to islet allo-transplantation due to a lack of suitable human donor organs. Recent progress in genetic engineering/gene editing of donor pigs provides new opportunities to overcome rejection of xeno-islets, to improve their engraftment and insulin secretion capacity, and to reduce the risk for transmission of porcine endogenous retroviruses. This review summarizes the current issues and progress in islet xenotransplantation with special emphasis on genetically modified/gene edited donor pigs.

Recent Findings

Attempts to overcome acute rejection of xeno-islets, especially after intraportal transplantation into the liver, include the genetic elimination of specific carbohydrate antigens such as αGal, Neu5Gc, and Sd(a) for which humans and—in part—non-human primates have natural antibodies that bind to these targets leading to activation of complement and coagulation. A complementary approach is the expression of one or more human complement regulatory proteins (hCD46, hCD55, hCD59). Transgenic attempts to overcome cellular rejection of islet xenotransplants include the expression of proteins that inhibit co-stimulation of T cells. Expression of glucagon-like peptide-1 and M3 muscarinic receptors has been shown to increase the insulin secretion of virally transduced porcine islets in vitro and it will be interesting to see the effects of these modifications in transgenic pigs and islet products derived from them. Genome-wide inactivation of porcine endogenous retrovirus (PERV) integrants by mutating their pol genes using CRISPR/Cas9 is a recent approach to reduce the risk for PERV transmission by xeno-islets.

Summary

Genetic engineering/gene editing of xeno-islet donor pigs facilitated major progress towards clinical islet xenotransplantation. The required set of genetic modifications will depend on the source of islets (fetal/neonatal vs. adult), the mode of delivery (encapsulated vs. free), and the transplantation site.
Literature
3.
go back to reference Rickels MR, Stock PG, de Koning EJP, Piemonti L, Pratschke J, Alejandro R, et al. Defining outcomes for beta-cell replacement therapy in the treatment of diabetes: a consensus report on the Igls criteria from the IPITA/EPITA opinion leaders workshop. Transpl Int. 2018;31(4):343–52. https://doi.org/10.1111/tri.13138.CrossRefPubMed Rickels MR, Stock PG, de Koning EJP, Piemonti L, Pratschke J, Alejandro R, et al. Defining outcomes for beta-cell replacement therapy in the treatment of diabetes: a consensus report on the Igls criteria from the IPITA/EPITA opinion leaders workshop. Transpl Int. 2018;31(4):343–52. https://​doi.​org/​10.​1111/​tri.​13138.CrossRefPubMed
10.
go back to reference Steffen A, Kiss T, Schmid J, Schubert U, Heinke S, Lehmann S et al. Production of high-quality islets from goettingen minipigs: Choice of organ preservation solution, donor pool, and optimal cold ischemia time. Xenotransplantation. 2017;24(1). https://doi.org/10.1111/xen.12284.CrossRef Steffen A, Kiss T, Schmid J, Schubert U, Heinke S, Lehmann S et al. Production of high-quality islets from goettingen minipigs: Choice of organ preservation solution, donor pool, and optimal cold ischemia time. Xenotransplantation. 2017;24(1). https://​doi.​org/​10.​1111/​xen.​12284.CrossRef
34.
37.
go back to reference Diamond LE, Quinn CM, Martin MJ, Lawson J, Platt JL, Logan JS. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation. 2001;71(1):132–42.CrossRef Diamond LE, Quinn CM, Martin MJ, Lawson J, Platt JL, Logan JS. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation. 2001;71(1):132–42.CrossRef
38.
go back to reference McKenzie IF, Li YQ, Xing PX, Dinatale I, Koulmanda M, Loveland BE, et al. CD46 protects pig islets from antibody but not cell-mediated destruction in the mouse. Xenotransplantation. 2003;10(6):615–21.CrossRef McKenzie IF, Li YQ, Xing PX, Dinatale I, Koulmanda M, Loveland BE, et al. CD46 protects pig islets from antibody but not cell-mediated destruction in the mouse. Xenotransplantation. 2003;10(6):615–21.CrossRef
40.
go back to reference Cozzi E, White DJ. The generation of transgenic pigs as potential organ donors for humans. Nat Med. 1995;1(9):964–6.CrossRef Cozzi E, White DJ. The generation of transgenic pigs as potential organ donors for humans. Nat Med. 1995;1(9):964–6.CrossRef
42.
go back to reference Mandel TE, Koulmanda M, Cozzi E, Waterworth P, Tolan M, Langford G, et al. Transplantation of normal and DAF-transgenic fetal pig pancreas into cynomolgus monkeys. Transplant Proc. 1997;29(1–2 /01):940.CrossRef Mandel TE, Koulmanda M, Cozzi E, Waterworth P, Tolan M, Langford G, et al. Transplantation of normal and DAF-transgenic fetal pig pancreas into cynomolgus monkeys. Transplant Proc. 1997;29(1–2 /01):940.CrossRef
43.
go back to reference Fodor WL, Williams BL, Matis LA, Madri JA, Rollins SA, Knight JW, et al. Expression of a functional human complement inhibitor in a transgenic pig as a model for the prevention of xenogeneic hyperacute organ rejection. Proc Natl Acad Sci U S A. 1994;91(23):11153–7.CrossRef Fodor WL, Williams BL, Matis LA, Madri JA, Rollins SA, Knight JW, et al. Expression of a functional human complement inhibitor in a transgenic pig as a model for the prevention of xenogeneic hyperacute organ rejection. Proc Natl Acad Sci U S A. 1994;91(23):11153–7.CrossRef
52.
go back to reference • Wolf-van Buerck L, Schuster M, Oduncu FS, Baehr A, Mayr T, Guethoff S, et al. LEA29Y expression in transgenic neonatal porcine islet-like cluster promotes long-lasting xenograft survival in humanized mice without immunosuppressive therapy. Sci Rep. 2017;7(1):3572. https://doi.org/10.1038/s41598-017-03913-4. Local transgene expression of the immunoregulator LEA29Y by the graft induce local immuneregulation and enables free islet transplant survival without systemic immunosuppression. CrossRefPubMedPubMedCentral • Wolf-van Buerck L, Schuster M, Oduncu FS, Baehr A, Mayr T, Guethoff S, et al. LEA29Y expression in transgenic neonatal porcine islet-like cluster promotes long-lasting xenograft survival in humanized mice without immunosuppressive therapy. Sci Rep. 2017;7(1):3572. https://​doi.​org/​10.​1038/​s41598-017-03913-4. Local transgene expression of the immunoregulator LEA29Y by the graft induce local immuneregulation and enables free islet transplant survival without systemic immunosuppression. CrossRefPubMedPubMedCentral
53.
go back to reference Martin C, Plat M, Nerriere-Daguin V, Coulon F, Uzbekova S, Venturi E, et al. Transgenic expression of CTLA4-Ig by fetal pig neurons for xenotransplantation. Transgenic Res. 2005;14(4):373–84.CrossRef Martin C, Plat M, Nerriere-Daguin V, Coulon F, Uzbekova S, Venturi E, et al. Transgenic expression of CTLA4-Ig by fetal pig neurons for xenotransplantation. Transgenic Res. 2005;14(4):373–84.CrossRef
57.
go back to reference Klose R, Kemter E, Bedke T, Bittmann I, Kelsser B, Endres R, et al. Expression of biologically active human TRAIL in transgenic pigs. Transplantation. 2005;80(2):222–30.CrossRef Klose R, Kemter E, Bedke T, Bittmann I, Kelsser B, Endres R, et al. Expression of biologically active human TRAIL in transgenic pigs. Transplantation. 2005;80(2):222–30.CrossRef
59.
go back to reference Buermann A, Petkov S, Petersen B, Hein R, Lucas-Hahn A, Baars W, et al. Pigs expressing the human inhibitory ligand PD-L1 (CD 274) provide a new source of xenogeneic cells and tissues with low immunogenic properties. Xenotransplantation. 2018; https://doi.org/10.1111/xen.12387.CrossRef Buermann A, Petkov S, Petersen B, Hein R, Lucas-Hahn A, Baars W, et al. Pigs expressing the human inhibitory ligand PD-L1 (CD 274) provide a new source of xenogeneic cells and tissues with low immunogenic properties. Xenotransplantation. 2018; https://​doi.​org/​10.​1111/​xen.​12387.CrossRef
66.
go back to reference Lee HS, Lee JG, Yeom HJ, Chung YS, Kang B, Hurh S, et al. The introduction of human heme oxygenase-1 and soluble tumor necrosis factor-alpha receptor type I with human IgG1 fc in porcine islets prolongs islet xenograft survival in humanized mice. Am J Transplant. 2016;16(1):44–57. https://doi.org/10.1111/ajt.13467.CrossRefPubMed Lee HS, Lee JG, Yeom HJ, Chung YS, Kang B, Hurh S, et al. The introduction of human heme oxygenase-1 and soluble tumor necrosis factor-alpha receptor type I with human IgG1 fc in porcine islets prolongs islet xenograft survival in humanized mice. Am J Transplant. 2016;16(1):44–57. https://​doi.​org/​10.​1111/​ajt.​13467.CrossRefPubMed
85.
go back to reference Rayat GR, Rajotte RV, Hering BJ, Binette TM, Korbutt GS. In vitro and in vivo expression of Galalpha-(1,3)gal on porcine islet cells is age dependent. J Endocrinol. 2003;177(1):127–35.CrossRef Rayat GR, Rajotte RV, Hering BJ, Binette TM, Korbutt GS. In vitro and in vivo expression of Galalpha-(1,3)gal on porcine islet cells is age dependent. J Endocrinol. 2003;177(1):127–35.CrossRef
96.
go back to reference • Samy KP, Davis RP, Gao Q, Martin BM, Song M, Cano J, et al. Early barriers to neonatal porcine islet engraftment in a dual transplant model. Am J Transplant. 2018;18(4):998–1006. https://doi.org/10.1111/ajt.14601. This dual islet transplant model provides robust insights into pathomechanism of (xeno)transplant rejection due to properly control experiments comparing modified xenoislet preparations within one transplant recipient. CrossRefPubMed • Samy KP, Davis RP, Gao Q, Martin BM, Song M, Cano J, et al. Early barriers to neonatal porcine islet engraftment in a dual transplant model. Am J Transplant. 2018;18(4):998–1006. https://​doi.​org/​10.​1111/​ajt.​14601. This dual islet transplant model provides robust insights into pathomechanism of (xeno)transplant rejection due to properly control experiments comparing modified xenoislet preparations within one transplant recipient. CrossRefPubMed
99.
go back to reference • Mourad NI, Perota A, Xhema D, Galli C, Gianello P. Transgenic expression of glucagon-like Peptide-1 (GLP-1) and activated muscarinic receptor (M3R) significantly improves pig islet secretory function. Cell Transplant. 2017;26(5):901–11. https://doi.org/10.3727/096368916x693798. Insulin content and secretory function of pig islets can be distinctly increased by genetic modifications. CrossRefPubMedPubMedCentral • Mourad NI, Perota A, Xhema D, Galli C, Gianello P. Transgenic expression of glucagon-like Peptide-1 (GLP-1) and activated muscarinic receptor (M3R) significantly improves pig islet secretory function. Cell Transplant. 2017;26(5):901–11. https://​doi.​org/​10.​3727/​096368916x693798​. Insulin content and secretory function of pig islets can be distinctly increased by genetic modifications. CrossRefPubMedPubMedCentral
105.
go back to reference Egerer S, Fiebig U, Kessler B, Zakhartchenko V, Kurome M, Reichart B, Kupatt C, Klymiuk N, Wolf E, Denner J, Bähr A. Early weaning completely eliminates porcine cytomegalovirus from a newly established pig donor facility for xenotransplantation. Xenotransplantation. 2018;25:e12449. https://doi.org/10.1111/xen.12449.CrossRef Egerer S, Fiebig U, Kessler B, Zakhartchenko V, Kurome M, Reichart B, Kupatt C, Klymiuk N, Wolf E, Denner J, Bähr A. Early weaning completely eliminates porcine cytomegalovirus from a newly established pig donor facility for xenotransplantation. Xenotransplantation. 2018;25:e12449. https://​doi.​org/​10.​1111/​xen.​12449.CrossRef
108.
go back to reference Crossan C, Mourad NI, Smith K, Gianello P, Scobie L. Assessment of porcine endogenous retrovirus transmission across an alginate barrier used for the encapsulation of porcine islets. Xenotransplantation. 2018:e12409. https://doi.org/10.1111/xen.12409. Crossan C, Mourad NI, Smith K, Gianello P, Scobie L. Assessment of porcine endogenous retrovirus transmission across an alginate barrier used for the encapsulation of porcine islets. Xenotransplantation. 2018:e12409. https://​doi.​org/​10.​1111/​xen.​12409.
110.
go back to reference • Denner J, Graham M. Xenotransplantation of islet cells: what can the non-human primate model bring for the evaluation of efficacy and safety? Xenotransplantation. 2015;22(3):231–5. https://doi.org/10.1111/xen.12169. Evidence is provided that non-human primates are of reduced value for efficacy and safety evaluation of islet xenotransplantation. CrossRefPubMed • Denner J, Graham M. Xenotransplantation of islet cells: what can the non-human primate model bring for the evaluation of efficacy and safety? Xenotransplantation. 2015;22(3):231–5. https://​doi.​org/​10.​1111/​xen.​12169. Evidence is provided that non-human primates are of reduced value for efficacy and safety evaluation of islet xenotransplantation. CrossRefPubMed
115.
go back to reference Denner J, Specke V, Thiesen U, Karlas A, Kurth R. Genetic alterations of the long terminal repeat of an ecotropic porcine endogenous retrovirus during passage in human cells. Virology. 2003;314(1):125–33.CrossRef Denner J, Specke V, Thiesen U, Karlas A, Kurth R. Genetic alterations of the long terminal repeat of an ecotropic porcine endogenous retrovirus during passage in human cells. Virology. 2003;314(1):125–33.CrossRef
Metadata
Title
Will Genetic Engineering Carry Xenotransplantation of Pig Islets to the Clinic?
Authors
Elisabeth Kemter
Joachim Denner
Eckhard Wolf
Publication date
01-11-2018
Publisher
Springer US
Published in
Current Diabetes Reports / Issue 11/2018
Print ISSN: 1534-4827
Electronic ISSN: 1539-0829
DOI
https://doi.org/10.1007/s11892-018-1074-5

Other articles of this Issue 11/2018

Current Diabetes Reports 11/2018 Go to the issue

Pathogenesis of Type 1 Diabetes (A Pugliese and SJ Richardson, Section Editors)

The Role of Accessory Cells in Islet Homeostasis

Immunology, Transplantation, and Regenerative Medicine (L Piemonti and V Sordi, Section Editors)

Can We Re-Engineer the Endocrine Pancreas?

Pathogenesis of Type 2 Diabetes and Insulin Resistance (M-E Patti, Section Editor)

Rethinking Bile Acid Metabolism and Signaling for Type 2 Diabetes Treatment

Pathogenesis of Type 1 Diabetes (A Pugliese and SJ Richardson, Section Editors)

Pancreas Pathology During the Natural History of Type 1 Diabetes

Health Care Delivery Systems and Implementation in Diabetes (ME McDonnell and AR Sadhu, Section Editors)

App-Based Insulin Calculators: Current and Future State

Lifestyle Management to Reduce Diabetes/Cardiovascular Risk (B Conway and H Keenan, Section Editors)

Vegetarian Diets and the Risk of Diabetes