Abstract
Purpose of review
Human islet transplantation has proven to be a highly effective treatment for patients with labile type 1 diabetes mellitus, which can free patients from daily glucose monitoring and insulin injections. However, the shortage of islet donors limits its’ broad application. Porcine islet xenotransplantation presents a solution to the donor shortage and recent advances in genetic modification and immunosuppressive regimens provide renewed enthusiasm for the potential of this treatment.
Recent findings
Advances in genetic editing technology are leading to multigene modified porcine islet donors with alterations in expression of known xenoantigens, modifications of their complement and coagulation systems, and modifications to gain improved immunological compatibility. Recent NHP-based trials of costimulation blockade using CD154 blockade show promising improvements in islet survival, whereas results targeting CD40 are less consistent. Furthermore, trials using IL-6 receptor antagonism have yet to demonstrate improvement in glucose control and suffer from poor graft revascularization.
Summary
This review will detail the current status of islet xenotransplantation as a potential treatment for type I diabetes mellitus, focusing on recent advances in porcine xenogeneic islet production, assessment in nonhuman primate preclinical models, the outcome of human clinical trials and review barriers to translation of xenoislets to the clinic.
Keywords: genetic modification, porcine islet xenotransplantation, type 1 diabetes mellitus
INTRODUCTION
Human pancreatic islet transplantation is an effective treatment for patients with type 1 diabetes mellitus (T1DM) with hypoglycemia unawareness [1]. In 2000, a leap forward in the quest for an effective cell-based β-cell replacement occurred with the development of the Edmonton islet transplantation protocol. Since then, further efforts by international groups have optimized the procedural techniques and immunosuppressive regimens leading to long-term insulin independence for many patients [2-4]. Despite these encouraging results, human islet transplantation is limited by the scarcity of suitable pancreata from deceased donors [5]. The current organ donor supply will never meet the clinical demand, thus alternative islet sources of unlimited supply are being explored, such as bioartificial devices, gene therapy, stem cell-derived islet transplantation and xenotransplantation.
Many aspects of islet xenotransplantation have been recently reviewed as the field reaches toward broad clinical relevance [6-10]. Although most islet transplants occur via infusion into the portal system, Smood et al. [11■] evaluated the optimal site for transplantation, questioning if the well-accepted intraportal route is truly superior to other sites, such as the renal subcapsular space. Kemter et al. [12■] recently reviewed work related to optimal porcine islet isolation and culture and alternate strategies to improve maturation and engraftment. Kemter [13■] also summarized recent advances in the genetic engineering of porcine islet donors and the important distinctions between source of islets, mode of delivery and site of transplantation. In this review, we will outline the rejection processes important in islet xenotransplantation, review optimal targets for porcine genetic modification, recent preclinical nonhuman primate models, human clinical trials, and the persistent challenges of clinical utilization of porcine islet xenotransplantation in the treatment of T1DM.
MAJOR BARRIERS
Instant blood mediated inflammatory reaction
Both human and porcine islet transplantation must overcome the immediate inflammatory response called instant blood-mediated inflammatory reaction (IBMIR) upon intraportal transplantation [14]. Although the cause of IBMIR is not perfectly understood, one theory suggests that in the normal state, only bone marrow-derived blood cells and endothelium are exposed to blood components. Exposure to other cells and to the extracellular matrix is non-physiologic, triggering inflammation, coagulation, and complement. Tissue factor, which is constitutively expressed on many cell types, including vessel adventitia, renal glomeruli and islets, and inducible by the inflammatory response on vessel endothelium, platelets, and neutrophils, has been demonstrated to be integral in the initiation of IBMIR [15,16]. This reaction is not unique to xenotransplantation and can be recapitulated ex-vivo in studies that have been informative about the underlying mechanisms that include activation of immune mediators, the complement and coagulation systems and platelet aggregation [17-19]. IBMIR leads to immediate loss of a significant portion of transplanted islets [20]. This is partially overcome by increasing the number of islets transplanted to sustain an adequate islet mass. IBMIR is particularly pronounced in islet xenotransplantation, in which immediate platelet consumption, activation of complement and a rapid dysregulated release of insulin representing islet injury is seen [21,22]. Efforts to overcome IBMIR include encapsulation strategies, genetic modifications of porcine donors and improved immunosuppressive regimens.
Chronic rejection
Sustaining long-term function of islets in both preclinical pig to non-human primate (NHP) trials and in human alloislet transplantation has proven challenging because of immune responses as well as detrimental effects of necessary immunosuppressive medications. In numerous studies, histologic evaluation of xenoislets has shown T-cell infiltration representing residual rejection not controlled by current immunosuppression regimens [23-26]. In addition to T-cell-mediated rejection, there are additional, poorly understood, nonimmune mechanisms resulting in the late loss of insulin independence seen even in autologous islet transplant recipients, in which alloimmunity and immunosuppression confounders are avoided [27].
PORCINE GENETIC MODIFICATIONS
Genetic modifications to target rejection
The difference in the expression of cell surface carbohydrate epitopes between pigs and humans and the role of human preformed antibodies in hyperacute rejection (HAR) of xenografts allowed for an actionable target for porcine genetic modifications [28,29]. The major carbohydrate antigen targets of human preformed antiporcine antibodies include galactosyl-α1,3-galactose, synthesized by α-1,3-galactosyltransferase, N-acetylneuraminic acid, synthesized by cytidine monophosphate-N-acetylneuraminic acid hydroxylase, and an Sd(a)-like glycan made by β-1,4-N-acetyl-galactosaminyl transferase 2 [13■]. The development of pigs deficient in these carbohydrates occurred initially by single genetic modification, cloning, and natural breeding, but with advancements in gene-editing techniques, such as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9, modifications can now occur through rapid, multiple molecular techniques [10,30]. These initial modifications allowed researchers to largely overcome the first hurdle evident within xenotransplantation of HAR; however, further barriers persisted, despite reducing antibody binding to porcine endothelial cells by more than 90%. In addition to residual antibodies to as yet undefined targets, activation of complement occurs during both HAR and IBMIR and there are species incompatibilities in complement components and natural complement pathway regulatory proteins (CPRPs). Thus, further efforts in porcine genetic modification focused on complement regulators [17,31]. Xenoislets from pigs with the human complement regulators, CD46, CD55, and CD59, have been evaluated in preclinical models and manifest less early islet loss because of IBMIR but inconsistent improvement in long-term islet survival [32-35]. This finding is compatible with the results of islet transplantation using multitransgenic pigs [36].
Genetic modifications to target infection transmission
The theoretical risk of transmission of porcine infectious diseases to humans was a central concern that tempered initial interest in xenotransplantation in the 1990s and remains a concern today. Dedicated efforts to eliminate clinical and subclinical infection of porcine donors with various porcine viruses include delivery via cesarean section and maintenance in designated pathogen-free facilities [37■■]. These steps can effectively reduce transmission of a multitude of viruses; however, porcine endogenous retroviruses (PERVs) are not eliminated as these viruses are integrated into the porcine genome in varying copy number in all strains tested to date. Whether PERVs have a physiologic role in the pig host remains uncertain [38]. Recently, PERV inactivation was achieved through CRISPR/Cas9 mediated editing of multiple genomic sites [39]. Furthermore, it is evident that different porcine tissues and organs harbor variable levels of infectious agents. Crossan et al. [37■■] evaluated neonatal and adult porcine islets in culture and found that even when PBMC tested positive for specific viruses, the islet cells remained free of the pathogen, implying that islet cells may pose a lower infectious risk than other organs.
Genetic modifications of porcine insulin
Both bovine and porcine insulin was historically utilized in the treatment of diabetes prior to the transition to synthetic human insulin [40]. Porcine insulin has a similar structure to human insulin, with only a single amino acid difference (alanine in pigs and threonine in humans at position B30) [41]. Given that porcine insulin is more immunogenic and less effective, efforts are underway to develop genetically modified pigs that express human insulin [42■■,43]. Yang et al. [43] successfully generated pigs that expressed human insulin by utilizing somatic cell nuclear transfer (SCNT) technology with transcription activator-like effector nucleases or CRISPR/Cas9, combined with single-stranded oligonucleotides to modify the pig insulin gene. These pigs expressed human insulin, but porcine C-peptide, which have different amino acid sequences and lengths [44]. More recently, Cho et al. [42■■] used CRISPR/Cas9 to generate insulin-deficient pigs and subsequent SCNT to develop piglets that expressed both human C-peptide and insulin. Of note, the insulin expression levels were low and the piglets died after three days of life, thus further optimization of this strategy is necessary.
PRECLINICAL NON-HUMAN PRIMATE STUDIES
Prior work
Over the past twenty years, many groups have investigated pig islet cell xenotransplantation in NHP, recently reviewed by Kemter and Liu [8,13■]. Table 1 summarizes free, intraportal islet xenotransplantation trials since 2006, with recent studies and novel approaches reviewed below.
Table 1.
Porcine-free, intraportal islet xenotransplantation in nonhuman primates since 2006
| Donor pig | Recipient type | Average number of islets (IEQ/kg) |
Immunosuppression | Maximum graft survival (days) |
References |
|---|---|---|---|---|---|
| SNU miniature pigs Adult |
STZ-induced diabetic rhesus monkeys (n = 9) | 96 090 | ATG, sirolimus, tacrolimus, anti-CD40 mAb, belatacept, CVF, adalimumab | >320 | [45■■] |
| SNU miniature pigs Adult |
STZ-induced diabetic rhesus monkeys (n = 5) | 93 575 | ATG, sirolimus, tacrolimus, anti-CD40 mAb, tocilizumab, CVF, adalimumab | 176 | [50■■] |
| SNU miniature pigs Adult | STZ-induced diabetic rhesus monkeys (n = 5) | 100 000 | ATG, sirolimus, tacrolimus, anti-CD154 mAb, CVF, adalimumab | 603 | [23] |
| GTKO/hCD46/hCD39 or GTKO/CD46/TFPI/CTLA4-Ig or GTKO/CD46/TFPI/CTL4-Ig/CD39 Adult |
STZ induced diabetic cynomolgus monkeys (n = 5) | 85 000 | ATG, MMF, anti-CD154 mAb | 365 | [36] |
| Large white × Landrace GTKO/hCD55/hCD59/HT Neonatal |
Nondiabetic baboons WT n = 4 Genetically modified n = 5 |
7644 (WT) 17889 (genetically modified) |
WT: none Genetically modified: ATG, MMF, tacrolimus |
30 | [34] |
| hCD46 genetically modified adult | STZ-induced diabetic cynomolgus monkeys (n = 9) | 85–100000 | ATG, MMF, anti-CD154 mAb | 396 | [35] |
| Outbred wild type or GTKO adult | STZ induced diabetic cynomolgus monkeys (n = 10) | 40 000 | ATG, tacrolimus, rapamycin, anti-CD154 mAb, MMF | >58 | [71] |
| Duroc or Large White Crossbreeds Neonatal |
Diabetic rhesus macaques s/p pancreatectomy (n = 9) | 50 000 | Anti-CD 154 mAb, basiliximab, sirolimus, belatacept | >260 | [25] |
| Outbred swine and inbred miniature swine Adult |
STZ-induced diabetic cynomolgus monkeys (n = 12) | 25 000 | Basilixumab, FTY720 or tacrolimus, everolimus, anti-CD154 mAb, leflunomide | >187 | [46] |
ATG, Antithymocyte globulin; CVF, cobra venom factor; IEQ, insulin equivalents; mAb, monoclonal antibody; MMF, mycophenolate mofetil; SNU, Seoul National University; STZ, streptozotocin; WT, wild type.
Costimulation blockade
Costimulation blockade through anti-CD40 and anti-CD154 monoclonal antibodies (mAb) is thought to have great potential as an immunosuppressive strategy by interfering with a key signaling pathway required for optimal activation of T cells, B cells, macrophages, and dendritic cells. This immunosuppressive approach has led to recent progress in porcine islet xenotransplantation, as well as xenotransplantation in general. Shin et al. [45■■] completed nine xeno islet transplantations from Seoul National University (SNU) miniature pigs to Streptozotocin (STZ)-induced diabetic rhesus monkeys via the portal system. The immunosuppressive regimen was composed of anti-CD40 mAb in conjunction with belatacept or tacrolimus. This regimen achieved one long-term survivor (>6 months); however, the results were inconsistent, apparently because of a lack of control of both IBMIR and cell-mediated rejection with this immunosuppressive regimen. This contrasts with their experience with anti-CD154 mAb-based immunosuppression in which five STZ-induced diabetic rhesus monkeys underwent islet cell transplantation from SNU miniature pigs [23]. In this study, graft survival with adequate glycemic control was achieved for greater than six months in four of the five monkeys. This is consistent with prior studies using anti-CD154 mAb-based immunosuppressive regimens; however, clinical advancement of CD154 targeting antibodies was halted because of thromboembolic complications [25,46-48]. More recently, a number of companies have developed modified antibodies against CD154 that are designed to eliminate thromboembolic triggering. The Pittsburgh group reviewed their experience with 14 STZ-induced diabetic cynomolgus monkeys, all of which were maintained on an anti-CD154 mAb-based immunosuppressive based regimen and experienced no thromboembolic complications, using only Aspirin 81 mg per day and a low-dose continuous heparin infusion to maintain intravenous catheter patency [49].
IL-6 receptor antagonist
In addition to optimizing immunosuppression, Min et al. [50■■] evaluated the effects of addition of tocilizumab, an IL-6 receptor antagonist. Theoretically, IL-6 has both positive and negative effects with regards to islet xenotransplantation. It is an important mediator in angiogenesis leading to revascularization of the islets but it also plays a role in the deleterious inflammatory response and coagulation dysregulation. Five STZ-induced diabetic rhesus monkeys underwent portal vein islet transplantation with islets derived from SNU miniature pigs. The immunosuppressive regimen included anti-CD40 mAb, sirolimus, tacrolimus, thymoglobulin, cobra venom factor, and adalimumab (TNF-α neutralizing mAb). The monkeys treated with tocilizumab were noted to have decreased levels of CRP production, representing less initial inflammation, but poorer revascularization of the islets. Graft survival was variable, with the longest surviving 176 days with 134 days independent of insulin.
TRANSLATION TO HUMANS
Human islet xenotransplantation
Trials of porcine islet xenotransplantation in humans have varied in their approach and success (Table 2). The first human islet xenotransplantation was conducted by Groth et al. [51] in 1994. Ten patients who had received a kidney allotransplant, on standard immunosuppression, received fetal pig islet cell clusters either under the kidney capsule (n = 2) or via the portal system (n = 8). Each transplant required pancreata from 39 to 100 fetuses, with the average yield per fetus of 10000 islet cell clusters. The patients tolerated the procedures well, but the porcine islets did not improve their glycemic control.
Table 2.
Human clinical trials utilizing porcine islets
| Islet source | Recipient | Islet type | Average number of islets |
Site | Immunosuppression | References |
|---|---|---|---|---|---|---|
| Auckland Island | T1DM (n = 8) | Neonatal Encapsulated |
5000 and 10 000 IEQ/kg | Peritoneum | None | [58] |
| Auckland Island | T1DM (n = 14) | Neonatal Encapsulated |
5000 or 10 000 or 15 000 or 20 000 IEQ/kg | Peritoneum | None | [57] |
| Xeno-1 | T1DM (n = 22) | Neonatal Free |
55 000 IEQ/kg | Intraportal | Cyclosporine, prednisolone, MMF, tacrolimus, sirolimus, OKT-3 | [56] |
| New Zealand bred | T1DM adolescents (n = 23) | Neonatal with Sertoli cells | 13 000–20 000 IEQ/kg (first transplant) | Subcutaneous | None | [54,55] |
| Cross-White Breed | T1DM (n = 1) with kidney transplant (n = 1) | Neonatal Encapsulated |
15 000 IEQ/kg | Peritoneum | None (n = 1) and cyclosporine, prednisone, azathioprine | [52,53] |
| Swedish Landrace | T1DM with kidney transplant (n = 10) | Fetal Free |
200 000–1 million islets | Kidney capsule or intraportal | ATG, 15-deoxyspergualin, cyclosporine, prednisolone, azathioprine | [51] |
ATG, Antithymocyte globulin; IEQ, insulin equivalents; MMF, mycophenolate mofetil; T1DM, type 1 diabetes mellitus.
Subsequently, Elliott et al. [52] completed two intraperitoneal transplants of genetically unmodified neonatal alginate-encapsulated islets in one patient without immunosuppression and one who was on standard immunosuppression because of a prior kidney transplant. Importantly, neither patient showed evidence of PERV transmission and both had short-term improvement in their insulin requirements and glycosylated hemoglobin (HbA1c). The nonimmunosuppressed patient was followed for over nine years and underwent laparoscopic islet harvest, revealing intact nodules with encapsulated islets that were similar in size to initial implantation, but opaque and rigid. When assessed for functionality, the islets were capable of low-level insulin production [53]. Whether the modest improvement in A1c was attributable to the graft or to the improved diabetes care and attention associated with the transplant is unclear.
Valdes-Gonzalez et al. [54] instituted a novel approach, creating a vascularized collagen environment prior to islet xenotransplantation by implanting a subcutaneous collagen-generating steel rod. Twelve adolescent diabetic patients underwent transplant with an average of 17 798 insulin equivalents (IEQ)/kg body weight combined with Sertoli cells, derived from New Zealand 7–10-day-old piglets. Eleven of the patients underwent retransplant 6–9 months later and four underwent a third transplant. Immunohistochemistry on three explanted devices showed the presence of insulin-producing cells and glucagon positive cells, with CD3 T-cell infiltrate. At four years, half of the patients achieved reduction in their insulin needs, and at 5–7 years of follow-up of a similar cohort, the team reported sustained improvement in HbA1c with less chronic complications of diabetes, including neuropathy, retinopathy, and nephropathy [55].
Wang et al. [56] reported 21 patients who underwent islet xenotransplantation utilizing neonatal islets from the xeno-1 pig bred in the Animal House of Central South University of Changsha, China. The islets, 55 000 IEQ/kg body weight, were implanted in the liver via the hepatic artery and patients were immunosuppressed. Patients saw a reduction in their insulin requirements and improvement in HbA1c in the first three months. Although none achieved long-term control, one patient was able to discontinue insulin therapy for seven days. No significant complications developed and there was no evidence of PERV transmission, with up to six years posttransplant follow-up.
In 2011, the group in New Zealand completed a Phase 1/2a xenotransplantation trial of encapsulated neonatal porcine islets transplanted to the peritoneum of 14 nonimmunosuppressed diabetic patients [57]. Patients received 5000, 10 000, 15 000, or 20 000 IEQ/kg; however, there were no apparent outcome differences between the groups. Patients experienced a reduction in unaware hypoglycemic events; however, minimal changes to HbA1c or daily insulin doses were evident at one year. This work led to a trial conducted in Argentina in which eight patients underwent intraperitoneal transplant of either 5000 or 10 000 IEQ/kg of encapsulated neonatal porcine islets [58]. Again, there was no evidence of PERV transmission [59]. Patients experienced fewer episodes of unaware hypoglycemia and improved HbA1c; however, marginal change to their daily insulin requirement.
Persistent challenges
Prior to moving forward with further human clinical trials of unencapsulated porcine islets, it will be necessary to obtain sustained (>6 months), consistent improvement in diabetes management, with either insulin independence or a greatly reduced insulin requirement, in a porcine to NHP model using clinically tolerable immunosuppression [60,61]. Current success in NHP preclinical models has been achieved using immunosuppressive regimens that are not clinically applicable, predominately based upon costimulation and complement blockade. As the ability to gain more extensive genetically modified donor pigs increases, there is hope that the addition of human CPRPs and coagulation regulatory elements will help overcome this barrier. Additionally, the ability to cure clinical diabetes and achieve insulin independence in NHPs under current xenotransplantation techniques has been inconsistent. It is clear that a method to mitigate IBMIR and immediate xenograft damage in a clinically relevant manner is necessary prior to clinical translation, as is effective therapy to combat cellular immunity.
In humans, the number of transplanted islets has been demonstrated to be the greatest predictor of insulin independence after islet transplantation [62]. In the recent Clinical Islet Transplantation Consortium trial, an average transplanted islet mass of more than 16 000 IEQ/kg had been delivered in those achieving insulin independence [63]. There are clear species differences between pigs, NHPs, and humans regarding metabolic demands, islet insulin release, and glycemic control [64]. NHPs have been shown to have a higher daily insulin requirement/kg than humans, related to differences in dietary energy requirements and eating habits, and in most preclinical islet xenotransplantation studies, a high islet mass is utilized, typically 50 000–100 000 IEQ/kg [64]. For allotransplantation in NHP, a mass of 25 000 IEQ/kg is generally curative. Translating this number to a human dose/kg of 50 000 IEQ per 70 kg person would require 10 adult pigs and greater than ninety juvenile pigs per recipient [65]. Although it is anticipated that a smaller islet mass will be required for human islet xenotransplantation, it is still a concern as to the practicality and ability to obtain sufficient numbers of islets for transplant.
These concerns are compounded by other emerging technologies to treat diabetes. In particular, recent advances allow stem cell differentiation into islet-like clusters (ILCs) that are glucose responsive and approach near-normal patterns of insulin secretion [66]. The composition of these ILCs differs from that of normal islets, lacking antigen-presenting cells and endothelium, and in some cases, expansion of other endocrine cell types (such as enterochromaffin cells) [67■,68■]. The impact of these differences remains to be determined but in small and large animal studies, glycemic control can be excellent. Despite these issues, a clear advantage of ILCs is that they can be generated in massive numbers and could be further genetically modified to reduce immunogenicity [69■■,70■].
CONCLUSION
Recent advances in porcine genetic modification and immunosuppressive regimens in preclinical pig-to-NHP islet xenotransplantation studies have led to new insights into the pathogenesis of islet cell rejection and progress toward achieving successful islet xenotransplantation. There are persistent barriers to clinical human islet xenotransplantation and further efforts should remain focused on establishment of an optimal genetically modified porcine islet source and establishment of long-term functioning islets with clinically relevant immunosuppression.
KEY POINTS.
Instant blood-mediated inflammatory reaction and chronic T-cell-mediated rejection persist in both human islet transplantation and porcine islet xenotransplantation.
Genetic modification of porcine islet donors target coagulation and complement regulatory elements, reduction of infectivity, and optimization of insulin type and release.
Recent preclinical pig-to-NHP trials demonstrate improved outcomes with anti-CD154 mAb-based immunosuppression when compared with anti-CD40 mAb-based treatment.
Inconsistent outcomes in NHP preclinical trials, the large number of xenoislets needed for insulin independence and progress made in alternative stem cell-derived β-cell replacement may impede progress toward clinical relevance of porcine xenotransplantation
Financial support and sponsorship
None.
Footnotes
Conflicts of interest
There are no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
■ of special interest
■■ of outstanding interest
- 1.Shapiro AM, Pokrywczynska M, Ricordi C. Clinical pancreatic islet transplantation. Nat Rev Endocrinol 2017; 13:268–277. [DOI] [PubMed] [Google Scholar]
- 2.Shapiro AM, Lakey JR, Ryan EA, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 2000; 343:230–238. [DOI] [PubMed] [Google Scholar]
- 3.Schuetz EC, Anazawa PH, Cross RS, et al. β cell replacement therapy: the next 10 years. Transplantation 2018; 102:215–229. [DOI] [PubMed] [Google Scholar]
- 4.Barton FB, Rickels MR, Alejandro R, et al. Improvement in outcomes of clinical islet transplantation. Diabetes Care 2012; 35:1436–1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Gamble A, Pepper AR, Bruni A, et al. The journey of islet cell transplantation and future development. Islets 2018; 10:80–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Park CG, Bottino R, Hawthorne WJ. Current status of islet xenotransplantation. Int J Surg 2015; 23:261–266. [DOI] [PubMed] [Google Scholar]
- 7.Reichart B, Niemann H, Chavakis T, et al. Xenotransplantation of porcine islet cells as a potential option for the treatment of type 1 diabetes in the future. Horm Metab Res 2015; 47:31–35. [DOI] [PubMed] [Google Scholar]
- 8.Liu Z, Hu W, He T, et al. Pig-to-primate islet xenotransplantation: past, present, and future. Los Angeles, CA: SAGE Publications; 2017. ; 925–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dhanasekaran M, George JJ, Loganathan G, et al. Pig islet xenotransplantation. Curr Opin Organ Transplant 2017; 22:452–462. [DOI] [PubMed] [Google Scholar]
- 10.Hawthorne WJ, Lew AM, Thomas HE. Genetic strategies to bring islet xenotransplantation to the clinic. Curr Opin Organ Transplant 2016; 21:476–483. [DOI] [PubMed] [Google Scholar]
- 11.■.Smood B, Bottino R, Hara H, et al. Is the renal subcapsular space the preferred site for clinical porcine islet xenotransplantation? Review article. Int J Surg 2019; 69:100–107.This review contrasts intraportal islet xenotransplantation with the renal subcapsular site, comparing physiologic differences between the two sites.
- 12.■.Kemter E, Wolf E. Recent progress in porcine islet isolation, culture and engraftment strategies for xenotransplantation. Curr Opin Organ Transplant 2018; 23:633–641.This review highlights progress optimal porcine islet isolation, culture, and engraftment, comparing different methods and types of islets.
- 13.■.Kemter E, Denner J, Wolf E. Will genetic engineering carry xenotransplantation of pig islets to the clinic? Curr Diab Rep 2018; 18:103.This review describes porcine genetic modifications utilized in preclinical trials of islet xenotransplantation and how each affects islet survival.
- 14.Bennet W, Groth CG, Larsson R et al. Isolated human islets trigger an instant blood mediated inflammatory reaction: implications for intraportal islet transplantation as a treatment for patients with type 1 diabetes. Upsala J Med Sci 2000; 105:125–133. [DOI] [PubMed] [Google Scholar]
- 15.Ji M, Yi S, Smith-Hurst H, et al. The importance of tissue factor expression by porcine NICC in triggering IBMIR in the xenograft setting. Transplantation 2011; 91:841–846. [DOI] [PubMed] [Google Scholar]
- 16.Mackman N Role of tissue factor in hemostasis and thrombosis. Blood Cells, Mol, Dis 2006; 36:104–107. [DOI] [PubMed] [Google Scholar]
- 17.Van Der Windt DJ, Bottino R, Casu A, et al. Rapid loss of intraportally transplanted islets: an overview of pathophysiology and preventive strategies. Oxford, UK: Blackwell Publishing Ltd; 2007; 288–297. [DOI] [PubMed] [Google Scholar]
- 18.Johansson H, Lukinius A, Moberg L, et al. Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes 2005; 54:1755–1762. [DOI] [PubMed] [Google Scholar]
- 19.Moberg L, Johansson H, Lukinius A, et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet 2002; 360:2039–2045. [DOI] [PubMed] [Google Scholar]
- 20.Eriksson O, Eich T, Sundin A, et al. Positron emission tomography in clinical islet transplantation. Am J Transplant 2009; 9:2816–2824. [DOI] [PubMed] [Google Scholar]
- 21.Bennet W, Sundberg B, Lundgren T, et al. Damage to porcine islets of Langerhans after exposure to human blood in vitro, or after intraportal transplantation to cynomologus monkeys: protective effects of sCR1 and heparin. Transplantation 2000; 69:711–719. [DOI] [PubMed] [Google Scholar]
- 22.Miyagawa S, Yamamoto A, Matsunami K, et al. Complement regulation in the GalT KO era. Xenotransplantation 2010; 17:11–25. [DOI] [PubMed] [Google Scholar]
- 23.Shin JS, Kim JM, Kim JS, et al. Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets (report). Am J Transplant 2015; 15:2837. [DOI] [PubMed] [Google Scholar]
- 24.Kirchhof N, Shibata S, Wijkstrom M, et al. Reversal of diabetes in nonimmunosuppressed rhesus macaques by intraportal porcine islet xenografts precedes acute cellular rejection. Xenotransplantation 2004; 11:396–407. [DOI] [PubMed] [Google Scholar]
- 25.Cardona K, Gregory SK, Zvonimir M, et al. Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med 2006; 12:304. [DOI] [PubMed] [Google Scholar]
- 26.Shin JS, Kim JS, Kim JM, et al. Minimizing immunosuppression in islet xenotransplantation. Immunotherapy 2014; 6:419–430. [DOI] [PubMed] [Google Scholar]
- 27.Wilson G, Sutton J, Abbott D, et al. Long-term outcomes after total pancreatectomy and islet cell autotransplantation is it a durable operation? Ann Surg 2014; 260:659–667. [DOI] [PubMed] [Google Scholar]
- 28.Good AH, Cooper DK, Malcolm AJ, et al. Identification of carbohydrate structures that bind human antiporcine antibodies: implications for discordant xenografting in humans. Transplant Proc 1992; 24:559. [PubMed] [Google Scholar]
- 29.Galili U, Shohet SB, Kobrin E, et al. Man, apes, and Old World monkeys differ from other mammals in the expression of alpha-galactosyl epitopes on nucleated cells. J Biol Chem 1988; 263:17755–17762. [PubMed] [Google Scholar]
- 30.Phelps CJ, Koike C, Vaught TD, et al. Production of alpha 1,3-galactosyl-transferase-deficient pigs. Science (New York, NY) 2003; 299:411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Diamond EL, Quinn MC, Martin JM, et al. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation 2001; 71:132–142. [DOI] [PubMed] [Google Scholar]
- 32.Nagaraju S, Bottino R, Wijkstrom M, et al. Islet xenotransplantation from genetically engineered pigs. Curr Opin Organ Transplant 2013; 18:695–702. [DOI] [PubMed] [Google Scholar]
- 33.Fodor WL, Williams BL, Matis LA, 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 USA 1994; 91:11153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hawthorne WJ, Salvaris EJ, Phillips P, et al. Control of IBMIR in neonatal porcine islet xenotransplantation in baboons. Am J Transplant 2014; 14:1300–1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Van Der Windt DJ, Bottino R, Casu A, et al. Long-term controlled normoglycemia in diabetic non-human primates after transplantation with hCD46 transgenic porcine islets. Am J Transplant 2009; 9:2716–2726. [DOI] [PubMed] [Google Scholar]
- 36.Bottino R, Wijkstrom M, van Der Windt DJ, et al. Pig-to-monkey islet xenotransplantation using multi-transgenic pigs. Am J Transplant 2014; 14:2275–2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.■■.Crossan C, O’Hara Z, Mourad N, et al. Examining the potential for porcine-derived islet cells to harbour viral pathogens. Xenotransplantation 2018; 25:e12375.This study evaluates for the presence of viral pathogens in porcine islets as compared with PBMC and found that islets were negative for all viruses tested.
- 38.Denner J, Tönjes RR. Infection barriers to successful xenotransplantation focusing on porcine endogenous retroviruses. Clin Microbiol Rev 2012; 25:318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yang L, Guell M, Niu D, et al. Genome-wide inactivation of porcine endogenous retroviruses (PERVs). Science 2015; 350:1101–1104. [DOI] [PubMed] [Google Scholar]
- 40.Clark AJ, Adeniyi-Jones RO, Knight G, et al. Biosynthetic human insulin in the treatment of diabetes: a double-blind crossover trial in established diabetic patients. Lancet (London, Engl) 1982; 2:354–357. [DOI] [PubMed] [Google Scholar]
- 41.Nicol DS, Smith LF. Amino-acid sequence of human insulin. Nature 1960; 187:483. [DOI] [PubMed] [Google Scholar]
- 42.■■.Cho B, Lee EJ, Ahn SM, et al. Production of genetically modified pigs expressing human insulin and C-peptide as a source of islets for xenotransplantation. Transgen Res 2019; 28:549–559.In this study, the investigators generated pigs expressing human insulin and human C-peptide utilizing fibroblasts from proinslin knockout pigs and somatic cell nuclear transfer. Pigs expressing human insulin and c-peptide were created but survived only three days.
- 43.Yang Y, Wang K, Wu H, et al. Genetically humanized pigs exclusively expressing human insulin are generated through custom endonuclease-mediated seamless engineering. J Mol Cell Biol 2016; 8:174–177. [DOI] [PubMed] [Google Scholar]
- 44.Rubenstein AH, Welbourne WP, Mako M, et al. Comparative immunology of bovine, porcine and human proinsulins and C-peptides. Diabetes 1970; 19:546–553. [DOI] [PubMed] [Google Scholar]
- 45.■■.Shin JS, Kim JM, Min BH, et al. Pre-clinical results in pig-to-non-human primate islet xenotransplantation using anti-CD40 antibody (2C10R4)-based immunosuppression. Xenotransplantation 2018; 25:. doi: 10.1111/xen.12356.This study of pig islet xenotransplantation to STZ-induced diabetic rhesus monkeys resulted in modest survival of porcine islets utlizing an anti-CD40 monoclonal antibody-based immunosuppressive regimen, with inconsistent results when compared with monkeys treated with anti-CD154 monoclonal antibody.
- 46.Hering BJ, Wijkstrom M, Graham ML, et al. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med 2006; 12:301–303. [DOI] [PubMed] [Google Scholar]
- 47.Bottino R, Knoll MF, Graeme-Wilson J, et al. Safe use of anti-CD154 monoclonal antibody in pig islet xenotransplantation in monkeys. Xenotransplantation 2017; 24:. doi: 10.1111/xen.12283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ford ML, Larsen CP. Translating costimulation blockade to the clinic: lessons learned from three pathways. Oxford, UK: Blackwell Publishing Ltd; 2009; 294–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bottino R, Knoll MF, Graeme-Wilson J, et al. Safe use of anti-CD154 monoclonal antibody in pig islet xenotransplantation in monkeys. Xenotransplantation 2017; 24:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.■■.Min BH, Shin JS, Kim JM, et al. Delayed revascularization of islets after transplantation by IL-6 blockade in pig to non-human primate islet xenotransplantation model. Xenotransplantation 2018; 25:. doi: 10.1111/xen.12374.Tocilizumab, an IL-6 receptor antagonist was utilized in a pig to monkey model of islet xenotransplantation, resulting in delayed revascularization with minimal effect on islet survival.
- 51.Groth CG, Tibell A, Tollemar J, et al. Transplantation of porcinefetal pancreas to diabetic patients. Lancet 1994; 344:1402–1404. [DOI] [PubMed] [Google Scholar]
- 52.Elliott RB, Escobar L, Garkavenko O, et al. No evidence of infection with porcine endogenous retrovirus in recipients of encapsulated porcine islet xenografts. Cell Transplant 2000; 9:895–901. [DOI] [PubMed] [Google Scholar]
- 53.Elliott R, Escobar L, Tan P, et al. Live encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after xenotransplantation. Xenotransplantation 2007; 14:157–161. [DOI] [PubMed] [Google Scholar]
- 54.Valdés-González RA, Dorantes LM, Garibay GN, et al. Xenotransplantation of porcine neonatal islets of Langerhans and Sertoli cells: a 4-year study. Eur J Endocrinol 2005; 153:419–427. [DOI] [PubMed] [Google Scholar]
- 55.Valdes-Gonzalez R, Rodriguez-Ventura AL, White DJ, et al. Long-term follow-up of patients with type 1 diabetes transplanted with neonatal pig islets. Clin Exp Immunol 2010; 162:537–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang W, Mo Z, Ye B, et al. A clinical trial of xenotransplantation of neonatal pig islets for diabetic patients. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2011; 36:1134–1140. [DOI] [PubMed] [Google Scholar]
- 57.Matsumoto S, Tan P, Baker J, et al. Clinical porcine islet xenotransplantation under comprehensive regulation. Transplant Proc 2014; 46:1992–1995. [DOI] [PubMed] [Google Scholar]
- 58.Matsumoto S, Abalovich A, Wechsler C, et al. Clinical benefit of islet xenotransplantation for the treatment of type 1 diabetes. EBioMedicine 2016; 12:255–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Morozov VA, Wynyard S, Matsumoto S, et al. No PERV transmission during a clinical trial of pig islet cell transplantation. Virus Res 2017; 227:34–40. [DOI] [PubMed] [Google Scholar]
- 60.Hering BJ, Cozzi E, Spizzo T, et al. First update of the International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: executive summary. Xenotransplantation 2016; 23:3–13. [DOI] [PubMed] [Google Scholar]
- 61.Cooper DK, Bottino R, Gianello P, et al. First update of the International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: Chapter 4: preclinical efficacy and complication data required to justify a clinical trial. Xenotransplantation 2016; 23:46–52. [DOI] [PubMed] [Google Scholar]
- 62.Keymeulen B, Gillard P, Mathieu C, et al. Correlation between beta cell mass and glycemic control in type 1 diabetic recipients of islet cell graft. Proc Natl Acad Sci USA 2006; 103:17444–17449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hering BJ, Clarke WR, Bridges ND, et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes care 2016; 39:1230–1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Graham ML, Bellin MD, Papas KK, et al. Species incompatibilities in the pig-to-macaque islet xenotransplant model affect transplant outcome: a comparison with allotransplantation. Xenotransplantation 2011; 18:328–342. [DOI] [PubMed] [Google Scholar]
- 65.Vanderschelden R, Sathialingam M, Alexander M, et al. Cost and scalability analysis of porcine islet isolation for islet transplantation: comparison of juvenile, neonatal and adult pigs. Cell Transplant 2019; 28:967–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bose B, Shenoy SP, Konda S, et al. Human embryonic stem cell differentiation into insulin secreting β-cells for diabetes. Cell Biology Int 2012; 36:1013–1020. [DOI] [PubMed] [Google Scholar]
- 67.■.Loretelli C, Assi E, Seelam AJ, Ben Nasr M, Fiorina P. Cell therapy for type 1 diabetes. Expert Opin Biol Ther 2020; 1–11; doi: 10.1080/14712598.2020.1748596. [published online ahead of print]This review highlights the recent advances in cell-based therapies for type 1 diabetes mellitus including the role of regulatory T cells, hematopoietic stem cells, tolerogenic dendritic cells, embryonic and induced pluripotent stem cells.
- 68.■.Cito M, Pellegrini S, Piemonti L, et al. The potential and challenges of alternative sources of β cells for the cure of type 1 diabetes. Endocr Connect 2018; 7:R114–R125.This review outlines recent advances in alternative sources for β-cell replacement.
- 69.■■.Odorico J, Markmann J, Melton D, et al. Report of the key opinion leaders meeting on stem cell-derived beta cells. Transplantation 2018; 102:1223–1229.This article outlines the current status of stem cell-derived β-cell replacement and the necessary steps to bring this technology to clinical relevance.
- 70.■.Memon B, Abdelalim EM. Stem cell therapy for diabetes: beta cells versus pancreatic progenitors. Cells 2020; 9:283.This review contrasts human pluripotent stem cell-derived β-cells and pancreatic progenitors for islet cell replacement.
- 71.Rood PP, Bottino R, Balamurugan AN, et al. Reduction of early graft loss after intraportal porcine islet transplantation in monkeys. Transplantation 2007; 83:202–210. [DOI] [PubMed] [Google Scholar]
