Journal List > Clin Transplant Res > v.39(1) > 1516090373

Kim and Park: Current status of pancreatic islet xenotransplantation

Abstract

Pancreatic islet transplantation represents the optimal treatment for severe hypoglycemia, a serious complication experienced by patients with long-term type 1 diabetes who are undergoing insulin therapy. However, the limited availability of donor organs restricts its widespread use. Porcine pancreatic islets could offer a viable alternative to address this organ shortage. For successful pancreatic islet xenotransplantation using porcine pancreatic islets, efficacy and safety must first be demonstrated in pig-to-nonhuman primate (NHP) preclinical studies, as outlined in the consensus statement of the International Xenotransplantation Association. Our group has achieved long-term survival of wild-type porcine islet grafts in immunosuppressed NHPs by employing two immunosuppressive protocols: one based on CD40-CD40L blockade and another utilizing clinically available immunosuppressants. A clinical trial for pancreatic islet xenotransplantation, following the latter protocol, has received approval from the Korean Ministry of Food and Drug Safety (MFDS). This review aims to highlight the results of clinical trials involving porcine islet xenotransplantation to date, along with the age-specific and other characteristics of the porcine islets used in these trials and the preclinical NHP studies that support them. It offers insights into the perspectives around the first clinical islet xenotransplantation approved by the Korean MFDS, emphasizing improved long-term graft survival.

HIGHLIGHTS
  • Porcine islet transplantation is the one of the optional treatment for severe hypoglycemia in long-term type 1 diabetes patients on insulin therapy.

  • Porcine pancreatic islets present a promising solution to mitigate the shortage of donor organs.

  • Before using porcine islets for xenotransplantation, its efficacy and safety must be rigorously validated in pig-to-nonhuman primate preclinical studies.

  • Advancing xenotransplantation research in South Korea requires government and private investment, along with regulatory reforms.

INTRODUCTION

Type 1 diabetes is an autoimmune disease characterized by the destruction of insulin-secreting beta cells in the pancreatic islets. This results in a deficiency of endogenous insulin, necessitating conventional insulin treatment [1]. However, patients with type 1 diabetes who retain less than 5% of their beta cell volume require intensive insulin therapy to mitigate complications associated with the disease [2]. The administration of large doses of insulin can lead to life-threatening severe hypoglycemic events [3].
To reduce hypoglycemic events resulting from insulin overdose, allogeneic islet transplantation has been explored [4]. In 2000, successful clinical trials of allogeneic islet transplantation were conducted using the Edmonton protocol [5]. Subsequently, in 2016, phase 3 clinical trial outcomes were published, demonstrating that the procedure could prevent severe hypoglycemia and achieve more stable glycemic control [6]. In 2023, the US Food and Drug Administration approved the use of allogeneic islets as a biological product, thus establishing allogeneic islet transplantation as a standard treatment option for type 1 diabetes, comparable to intensive insulin therapy [7].
However, considering the large number of patients with unstable type 1 diabetes, the issue of donor shortage for allogeneic islet transplantation must be addressed. To overcome this challenge, researchers have investigated the use of porcine islets [8]. The primary benefit of porcine islet xenotransplantation is the potential for an unlimited supply of healthy islets. However, porcine islets elicit a stronger immune response than allogeneic islet transplants. Additionally, xenogeneic islets are classified as medicinal products, meaning that both the production of designated pathogen-free pigs [9] and the isolation of their islets must adhere to pharmaceutical production standards [10]. While the use of designated pathogen-free pigs reduces the risk of zoonosis, concerns regarding porcine endogenous retroviruses (PERVs) remain [11]. Moreover, such studies are subject to ethical considerations and legal regulations [12]. Despite these challenges, porcine islet xenotransplantation is considered a potential solution to address the donor shortage in allogeneic islet transplantation for patients with unstable type 1 diabetes. Clinical trials have been conducted in various countries, including Sweden, China, New Zealand, Russia, and Mexico [1319].

CLINICAL APPLICATION OF PORCINE ISLET XENOTRANSPLANTATION

In 1994, Groth et al. [13] transplanted fetal porcine islet-like cell clusters (ICCs) into 10 patients with type 1 diabetes mellitus. The porcine ICCs were delivered to the liver via the portal vein in eight patients and implanted beneath the kidney capsule in two patients. As the patients were undergoing concurrent kidney transplantation, they were induced with antithymocyte globulin (ATG) or 15-deoxyspergualin and maintained on a regimen of cyclosporine, prednisolone, and azathioprine. Of the patients who received transplants through the portal vein, four exhibited porcine C-peptide positivity in their urine for 200–400 days after transplantation. Biopsies confirmed the presence of insulin- and glucagon-positive cells, although no change in insulin requirements was noted relative to pretransplantation levels. Although no clinical benefits were observed following the transplantation of porcine ICCs, the study was the first to demonstrate that porcine islets could survive in the human body.
In 2005, a study conducted in Mexico involved transplanting macroencapsulated neonatal porcine islets with Sertoli cells into 12 adolescent patients with diabetes [14]. The macrodevice, constructed from surgical-grade stainless steel mesh tubes, was implanted subcutaneously. Over a period of 2 months, vascularized collagen tissue was allowed to infiltrate the device. Subsequently, a polytetrafluoroethylene rod was removed from the macrodevice, and 250,000 neonatal islets along with 30 to 100 Sertoli cells were implanted in its place. Eleven patients received a second transplant 6 to 9 months after the initial procedure. Of the total group, six patients experienced a reduction in insulin requirements, while the other six saw an increase. Notably, two patients achieved insulin independence. However, the clinical trial was met with criticism for being conducted on adolescent patients without a clear regulatory framework.
In 2013, a Chinese research team transplanted neonatal porcine islets into the livers of 10 adult patients with type 1 diabetes through the portal vein [15]. This clinical trial was conducted under the supervision of a Chinese governmental agency. To prevent the instant blood-mediated inflammatory reaction (IBMIR), patients received anticoagulant therapy for 7 days following transplantation. Clinically available immunosuppressants and autologous regulatory T (Treg) cells were employed to control immune responses. Although only a slight reduction in insulin requirements was noted, the procedure did not result in any zoonotic infections, including those from PERVs.
Clinical trials of microencapsulated neonatal porcine islet transplantation have been conducted in New Zealand, Russia, and Argentina from the early 2000s to the present [1619]. These trials have shown a reduction in insulin dose and improved hypoglycemic awareness in most recipients, with some patients achieving long-term insulin independence. Consequently, this approach to xenogeneic islet transplantation is regarded as promising. The results of these clinical trials are summarized in Table 1.
In Korea, a sponsor-initiated trial using the islets of adult Seoul National University (SNU) pigs was approved by the Ministry of Food and Drug Safety in December 2022. However, due to internal issues with the sponsor, the trial has not yet commenced. Once underway, it will be conducted under a clear regulatory framework. Clinical trials require robust regulatory oversight to ensure safety and efficacy, and this trial is designed to adhere to those standards.

AGE-SPECIFIC CHARACTERISTICS OF DONOR PIG ISLETS FOR CLINICAL TRIALS

The age of donor pigs significantly influences the properties of islet products (Table 2) [20]. Researchers have experimented with the isolation and transplantation of pancreatic islets from pigs of various ages [21]. The islet structure in fetal and neonatal pigs is markedly different from that in adult pigs, primarily because the matrix and connective tissue in the pancreas of younger pigs are not fully developed. Consequently, ICCs can be relatively easily isolated from these organs through enzymatic digestion [22]. ICCs are primarily composed of endocrine tissue, which survives during in vitro culture, while the exocrine tissue dies. This process enables the purification of ICCs. However, ICCs require a maturation period after transplantation before they can begin secreting insulin. Therefore, even if porcine ICCs avoid immune rejection and successfully engraft, exogenous insulin therapy is necessary until their maturation is complete.
Isolation methods used for fetal and neonatal pancreases have been adapted for use with young pig pancreases. Typically, approximately 30,000 islet equivalents (IEQ) can be harvested from the pancreas of one young pig. However, Matsumoto et al. [23] reported isolating up to 180,000 IEQ from a single young pig. Islets from young pigs offer several advantages over neonatal islets, including higher islet yield and the capacity for immediate insulin secretion, which is attributed to their maturation during the culture period.
Despite the benefit of immediate insulin secretion, islets from young adult and adult pigs are delicate, which complicates the process of islet isolation. It is widely recognized that the success and yield of islet isolation depend heavily on the breed and age of the pig. Although the SNU miniature pig is used as an adult, it has demonstrated a higher yield in comparison to other breeds, such as Prestige World Genetics miniature pigs and adult market pigs sourced from a local slaughterhouse [24]. Consequently, the choice of pig breed should be made carefully when considering the use of adult pig islets for clinical or research applications.

PRECLINICAL NONHUMAN PRIMATE STUDIES FOR CLINICAL TRIALS OF PORCINE ISLET XENOTRANSPLANTATION

In collaboration with the World Health Organization, the International Xenotransplantation Association (IXA) published a consensus statement in 2009 to guide the safe conduct of clinical trials involving porcine islet cells. This guidance is detailed in “The International Xenotransplantation Association consensus statement on the conditions for undertaking clinical trials of porcine islet products in type 1 diabetes.” One obstacle to the clinical adoption of porcine islet xenotransplantation is the difficulty in meeting success criteria in preclinical studies involving nonhuman primates (NHPs) [25]. The updated IXA consensus statement from 2016 specify requirements for NHP studies. These include maintaining fasting blood glucose levels below 150 mg/dL and nonfasting levels below 200 mg/dL, either without the need for exogenous insulin or with a significant reduction in insulin requirements, in at least four of six consecutive NHPs following porcine islet transplantation with a clinically available immunosuppressive regimen [26]. Also, all cases should have a follow-up period of at least 6 months, with one or two successful cases ideally undergoing follow-up for 12 months.
Various research teams have conducted studies on porcine islet xenotransplantation in preclinical NHP models to adhere to the IXA guidelines. Among these efforts, the present article summarizes the results of groups that achieved long-term graft survival (Table 3) [2743]. In 2015, our group was the first to demonstrate a proof of concept by successfully reversing diabetes in five diabetic NHPs. This was accomplished through the transplantation of wild-type porcine islets, combined with an immunosuppressive regimen that included an anti-CD154 monoclonal antibody (mAb; 5C8)—which is not considered clinically applicable—and low-dose sirolimus, with or without ex vivo expanded Treg cells [27]. Subsequently, we reported similar, albeit less robust, outcomes using an anti-CD40 mAb (2C10R4) [28], which has potential for clinical use. Most recently, we maintained normal glycemic control in three of seven diabetic NHPs and significantly reduced the need for exogenous insulin in five of these seven animals. This was achieved using a clinically applicable immunosuppressive regimen that included ATG, adalimumab, anakinra, tocilizumab, intravenous immunoglobulin, tacrolimus, belimumab, rapamycin, and tofacitinib [29]. While this study could substantially advance the area of islet xenotransplantation clinical trials, the intensity of the immunosuppressive protocol used limits its widespread application. Consequently, additional strategies may be necessary, such as employing genetically engineered pigs or developing novel immunosuppressive drugs with fewer adverse effects.

FUTURE PERSPECTIVES

The production of galactosyltransferase knockout pigs in 2002 was a key milestone in xenotransplantation research, eliminating the α-galactosidase (αGal) antigen and leading to the first major wave of advancements in the field [44]. Since then, the advent of clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR-associated protein 9 (Cas9) technology has enabled the creation of various genetically engineered pigs. These include animals with multiple human gene knockins to mitigate immune responses; those with knock-outs of multiple carbohydrate antigens such as Gal, 5NeuGc, and B4GalNT2; and even PERV-free pigs. This development has prompted a second wave of progress in xenotransplantation [45]. The authors anticipate that the third wave will incorporate innovative immunological therapies. These may include chemical drugs or antibodies that regulate T cell receptor (TCR) signaling, costimulation, and cytokines, as well as cell-based treatments such as Treg cells.
Regarding potential strategies, first, interventions targeting TCR signaling (also known as signal 1) have been utilized to prevent graft rejection. Numerous immunosuppressive drugs have been developed to target TCR signaling pathways as therapeutic interventions [46]. Chemical immunosuppressive drugs, including mammalian target of rapamycin inhibitors (sirolimus and everolimus), calcineurin inhibitors (tacrolimus and cyclosporine), the Janus kinase inhibitor tofacitinib, and purine and pyrimidine synthesis inhibitors (6-mercaptopurine, mycophenolate mofetil, and methotrexate), have been widely used in preclinical NHP studies of porcine islet xenotransplantation to interfere with the signals that activate T cells and promote their proliferation [47]. In addition to methods that inhibit the TCR signaling pathway, approaches that deplete T cells (such as ATG; polyclonal immunoglobulins [Igs] from rabbits immunized with human thymocytes; or alemtuzumab, an anti-CD52 mAb) or induce T cell anergy (such as daclizumab or basiliximab, blocking and internalizing the interleukin [IL]-2 receptor to reduce IL-2–dependent T cell activation and proliferation) are being employed in combination [48]. In preclinical NHP studies of porcine islet xenotransplantation, all groups that demonstrated long-term graft survival used sirolimus/everolimus, mycophenolate mofetil, tacrolimus, or tofacitinib (Table 3). When costimulatory blockades such as anti-CD154, anti-CD40, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) Ig, or anti-LFA-1 were used, long-term graft survival was achieved even with the use of a single signal 1 blocker. However, in immunosuppressive regimens that are clinically available without costimulatory blockade, long-term graft survival was only observed when three signal 1 blockers—sirolimus, tacrolimus, and tofacitinib—were used together [29]. Thus, it appears essential to incorporate costimulatory blockade in preclinical NHP studies of porcine islet xenotransplantation to ensure long-term graft survival.
Second, a costimulatory blockade strategy, such as anti-CD40L, anti-CD40, anti-LFA-1, LFA-3 Ig, or CTLA-4 Ig, can be considered, with a primary focus on inhibiting the CD40-CD40L pathway. Preclinical studies have shown that CD40-CD40L blockade exhibits remarkable efficacy in inducing tolerance across various autoimmune disease models and in promoting the acceptance of skin and organ transplants [49]. In preclinical trials involving NHPs with porcine islet transplants, long-term islet survival was observed using anti-CD154 mAbs [27,3036]. The success of these antibodies is attributed to their critical role in the development of autoimmune diseases and the mediation of graft rejection. These impressive preclinical outcomes have spurred considerable interest in developing anti-CD154 therapies for human diseases, leading to clinical trials of native IgG1 CD154 antibodies in the late 1990s. However, these trials were halted due to fatal thromboembolic events caused by the simultaneous binding of CD154 to Fc receptors on platelets [50]. Recent advancements in immunotherapy have focused on modifying or substituting immunoglobulins to prevent binding to FcγRIIA, thereby avoiding platelet activation. Notable examples include frexalimab and tegoprubart, which are humanized CD154-specific IgG1 antibodies with silencing mutations in the Fc domain. Additionally, dapirolizumab pegol, a polyethylene glycol-conjugated anti-CD154 Fab fragment, and dazodalibep, a nonantibody biological antagonist, are being examined. These CD154 inhibitors have shown a favorable safety profile, with no thromboembolic activity observed in hundreds of patients and healthy controls. Promising results have also emerged from phase 2 clinical trials for conditions like systemic lupus erythematosus, Sjogren syndrome, and multiple sclerosis, with several candidates advancing to pivotal phase 3 trials [51].
The anti-CD40 inhibitor, a component of the CD40-CD40L blockade, has also displayed promising results in both preclinical and clinical trials. In NHP models, an anti-CD40 mAb (chi220; with B cell-depleting effects) significantly prolonged the survival of neonatal porcine islet grafts, with five of six recipients achieving insulin independence. In that study, up to 50,000 IEQ/kg of neonatal porcine islets were transplanted into six pancreatectomized diabetic macaques via the portal vein. The immunosuppression regimen included induction therapy with CD40-mAb and basiliximab, along with maintenance therapy using sirolimus and belatacept [37]. Separately, Shin et al. [28] demonstrated more than 321 days of adult porcine islet graft survival using a combination of ATG, cobra venom factor, antitumor necrosis factor (TNF), anti-CD40 (2C10R4), rapamycin, and tacrolimus. In another report, iscalimab also displayed a favorable safety profile and improvements compared to placebo in non-thymectomized patients with moderate to severe myasthenia gravis [52]. Clinical trials using anti-CD40 antibodies for various autoimmune diseases are ongoing, and the results are eagerly awaited.
Third, a strategy to inhibit cytokines should be considered to prevent the rejection of porcine islets. Both clinical islet allotransplantation and porcine islet xenotransplantation involve infusing islets through the portal vein to allow the transplanted islets to engraft in the liver. Consequently, the islets come into direct contact with the recipient’s blood, resulting in IBMIR and massive early islet loss. IBMIR is a complex phenomenon that triggers the activation of the complement system and coagulation pathway. Platelets rapidly bind to the islets, and neutrophils and monocytes infiltrate the area [53]. During IBMIR, proinflammatory cytokines such as high-mobility group box 1 protein, IL-8, TNF-α, IL-1β, and IL-6 are produced, leading to further islet destruction [54]. Various inhibitors of proinflammatory cytokines are available. For TNF-α, options include etanercept, infliximab, adalimumab, certolizumab pegol, and golimumab [55]. Anakinra, a recombinant form of the human IL-1 receptor, can be used to block both IL-1α and IL-1β, while canakinumab, a human IgG1/κ mAb, specifically targets IL-1β [56]. For IL-6, options include tocilizumab, an antibody that blocks the IL-6 receptor, and siltuximab, an antibody that targets IL-6 itself [57]. The efficacy of TNF-α blockers (infliximab, certolizumab pegol, and golimumab), an IL-1β blocker (canakinumab), and an IL-6 blocker (siltuximab) has not yet been explored in preclinical NHP studies of porcine islet xenotransplantation, and this information is highly anticipated.
Fourth, Treg cells have been investigated in preclinical NHP studies of porcine islet xenotransplantation to induce immune tolerance. Shin et al. [35] utilized autologous Treg cells characterized by CD4(+) CD25(high) CD127(low) markers in combination with immunosuppressive agents such as ATG, cobra venom factor, anti-CD154 mAb, and sirolimus. In their study, two diabetic NHPs exhibited promising results; one achieved 500 days of graft survival and the other 1,000 days, illustrating the potential of Treg cells in promoting immune tolerance [35]. Although polyclonal Treg cells are a promising tool for inducing transplantation tolerance, their lack of specificity requires the administration of high doses. By genetically modifying these cells to express chimeric antigen receptors (CAR) that target mismatched donor human leukocyte antigen molecules, CAR Treg cells could become much more effective in preventing allograft rejection [58]. Researchers are also examining the use of swine leukocyte antigen-specific CAR Treg cells to induce tolerance in xenotransplantation. This strategy is expected to be incorporated into future porcine islet xenotransplantation protocols, offering new potential for improved outcomes [59].

CONCLUSION

The advancements in TCR signaling blockades, CD40-CD40L costimulation blockades, cytokine blockers, and Treg cell therapies offer hope for increased clinical accessibility. These developments have potential applications in the xenotransplantation of porcine organs, positioning them within what could be considered a “third wave” in this field. Xenotransplantation research is a highly active field in South Korea. As part of a national initiative, the government plans to invest approximately 38 billion Korean won into this area of study over the next 4 years. Preclinical studies involving NHPs are underway, with a focus on the transplantation of solid organs such as kidneys, hearts, and livers, as well as cellular tissues including islets, corneas, and skin. To advance xenotransplantation research in South Korea, sustained investment from both the government and the private sector, coupled with regulatory reforms, is crucial.

ARTICLE INFORMATION

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Funding/Support

This research was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant No. HI23C0923). Additionally, this was supported by a research grant from the Korean Society for Transplantation (2025-00-03001-001).

Author Contributions

All the work was done by Jong-Min Kim and Chung-Gyu Park. All authors read and approved the final manuscript.

REFERENCES

1. Toren E, Burnette KS, Banerjee RR, Hunter CS, Tse HM. 2021; Partners in crime: beta-cells and autoimmune responses complicit in type 1 diabetes pathogenesis. Front Immunol. 12:756548. DOI: 10.3389/fimmu.2021.756548. PMID: 34691077. PMCID: PMC8529969.
crossref
2. Korsgren O, Lundgren T, Felldin M, Foss A, Isaksson B, Permert J, et al. 2008; Optimising islet engraftment is critical for successful clinical islet transplantation. Diabetologia. 51:227–32. DOI: 10.1007/s00125-007-0868-9. PMID: 18040664.
crossref
3. Rzepczyk S, Dolińska-Kaczmarek K, Uruska A, Żaba C. 2022; The other face of insulin-overdose and its effects. Toxics. 10:123. DOI: 10.3390/toxics10030123. PMID: 35324747. PMCID: PMC8955302.
crossref
4. Rother KI, Harlan DM. 2004; Challenges facing islet transplantation for the treatment of type 1 diabetes mellitus. J Clin Invest. 114:877–83. DOI: 10.1172/JCI23235. PMID: 15467822. PMCID: PMC518676.
crossref
5. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, et al. 2000; Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 343:230–8. DOI: 10.1056/NEJM200007273430401. PMID: 10911004.
crossref
6. Hering BJ, Clarke WR, Bridges ND, Eggerman TL, Alejandro R, Bellin MD, et al. 2016; Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care. 39:1230–40. DOI: 10.2337/dc15-1988. PMID: 27208344. PMCID: PMC5317236.
crossref
7. Stabler CL, Russ HA. 2023; Regulatory approval of islet transplantation for treatment of type 1 diabetes: implications and what is on the horizon. Mol Ther. 31:3107–8. DOI: 10.1016/j.ymthe.2023.10.011. PMID: 37865099. PMCID: PMC10638039.
crossref
8. Hering BJ, Cooper DK, Cozzi E, Schuurman HJ, Korbutt GS, Denner J, et al. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: executive summary. Xenotransplantation. 16:196–202. DOI: 10.1111/j.1399-3089.2009.00547.x. PMID: 19799759.
9. Schuurman HJ. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: chapter 2: source pigs. Xenotransplantation. 16:215–22.
10. Korbutt GS. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: chapter 3: pig islet product manufacturing and release testing. Xenotransplantation. 16:223–8. DOI: 10.1111/j.1399-3089.2009.00542.x. PMID: 19799762.
crossref
11. Denner J, Schuurman HJ, Patience C. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: chapter 5: strategies to prevent transmission of porcine endogenous retroviruses. Xenotransplantation. 16:239–48. DOI: 10.1111/j.1399-3089.2009.00544.x. PMID: 19799764.
crossref
12. Cozzi E, Tallacchini M, Flanagan EB, Pierson RN 3rd, Sykes M, Vanderpool HY. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: chapter 1: key ethical requirements and progress toward the definition of an international regulatory framework. Xenotransplantation. 16:203–14. DOI: 10.1111/j.1399-3089.2009.00540.x. PMID: 19799760.
crossref
13. Groth CG, Korsgren O, Tibell A, Tollemar J, Möller E, Bolinder J, et al. 1994; Transplantation of porcine fetal pancreas to diabetic patients. Lancet. 344:1402–4. DOI: 10.1016/S0140-6736(94)90570-3. PMID: 7968077.
crossref
14. Valdes-Gonzalez R, Rodriguez-Ventura AL, White DJ, Bracho-Blanchet E, Castillo A, Ramírez-González B, et al. 2010; Long-term follow-up of patients with type 1 diabetes transplanted with neonatal pig islets. Clin Exp Immunol. 162:537–42. DOI: 10.1111/j.1365-2249.2010.04273.x. PMID: 20964645. PMCID: PMC3026557.
crossref
15. Ma X, Li S, Wang J, Xu C, Wang W. 2023; Establishment of a donor pig for xenotransplantation clinical trials based on the principle of Changsha Communiqué. Health Care Sci. 2:129–34. DOI: 10.1002/hcs2.37. PMID: 38938766. PMCID: PMC11080849.
crossref
16. Elliott RB, Escobar L, Tan PL, Muzina M, Zwain S, Buchanan C. 2007; Live encapsulated porcine islets from a type 1 diabetic patient 9.5 yr after xenotransplantation. Xenotransplantation. 14:157–61. DOI: 10.1111/j.1399-3089.2007.00384.x. PMID: 17381690.
crossref
17. Skinner SJ, Tan PL, Garkavenko O, Muzina M, Escobar L, Elliott RB. Cell replacement therapy: the rationale for encapsulated porcine islet transplantation, type 1 diabetes complications. InTech;2011. DOI: 10.5772/24824.
18. Matsumoto S, Tan P, Baker J, Durbin K, Tomiya M, Azuma K, et al. 2014; Clinical porcine islet xenotransplantation under comprehensive regulation. Transplant Proc. 46:1992–5. DOI: 10.1016/j.transproceed.2014.06.008. PMID: 25131091.
crossref
19. Matsumoto S, Abalovich A, Wechsler C, Wynyard S, Elliott RB. 2016; Clinical benefit of islet xenotransplantation for the treatment of type 1 diabetes. EBioMedicine. 12:255–62. DOI: 10.1016/j.ebiom.2016.08.034. PMID: 27592597. PMCID: PMC5078586.
crossref
20. Park CG, Bottino R, Hawthorne WJ. 2015; Current status of islet xenotransplantation. Int J Surg. 23(Pt B):261–6. DOI: 10.1016/j.ijsu.2015.07.703. PMID: 26253846.
crossref
21. Nagaraju S, Bottino R, Wijkstrom M, Trucco M, Cooper DK. 2015; Islet xenotransplantation: what is the optimal age of the islet-source pig? Xenotransplantation. 22:7–19. DOI: 10.1111/xen.12130. PMID: 25130196.
crossref
22. Korbutt GS, Elliott JF, Ao Z, Smith DK, Warnock GL, Rajotte RV. 1996; Large scale isolation, growth, and function of porcine neonatal islet cells. J Clin Invest. 97:2119–29. DOI: 10.1172/JCI118649. PMID: 8621802. PMCID: PMC507287.
crossref
23. Matsumoto S, Tomiya M, Sawamoto O. 2016; Current status and future of clinical islet xenotransplantation. J Diabetes. 8:483–93. DOI: 10.1111/1753-0407.12395. PMID: 26987992.
crossref
24. Kim JH, Kim HI, Lee KW, Yu JE, Kim SH, Park HS, et al. 2007; Influence of strain and age differences on the yields of porcine islet isolation: extremely high islet yields from SPF CMS miniature pigs. Xenotransplantation. 14:60–6. DOI: 10.1111/j.1399-3089.2006.00364.x. PMID: 17214705.
crossref
25. Cooper DK, Casu A. 2009; The International Xenotransplantation Association consensus statement on conditions for undertaking clinical trials of porcine islet products in type 1 diabetes: chapter 4: pre-clinical efficacy and complication data required to justify a clinical trial. Xenotransplantation. 16:229–38. DOI: 10.1111/j.1399-3089.2009.00543.x. PMID: 19799763.
crossref
26. Cooper DK, Bottino R, Gianello P, Graham M, Hawthorne WJ, Kirk AD, et al. 2016; 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: pre-clinical efficacy and complication data required to justify a clinical trial. Xenotransplantation. 23:46–52. DOI: 10.1111/xen.12226. PMID: 26916706.
crossref
27. Shin JS, Kim JM, Kim JS, Min BH, Kim YH, Kim HJ, et al. 2015; Long-term control of diabetes in immunosuppressed nonhuman primates (NHP) by the transplantation of adult porcine islets. Am J Transplant. 15:2837–50. DOI: 10.1111/ajt.13345. PMID: 26096041.
crossref
28. Shin JS, Kim JM, Min BH, Yoon IH, Kim HJ, Kim JS, et al. 2018; Pre-clinical results in pig-to-non-human primate islet xenotransplantation using anti-CD40 antibody (2C10R4)-based immunosuppression. Xenotransplantation. 25:e12356. DOI: 10.1111/xen.12356. PMID: 29057561. PMCID: PMC5809197.
crossref
29. Kim JM, Hong SH, Chung H, Shin JS, Min BH, Kim HJ, et al. 2021; Long-term porcine islet graft survival in diabetic non-human primates treated with clinically available immunosuppressants. Xenotransplantation. 28:e12659. DOI: 10.1111/xen.12659. PMID: 33155753.
crossref
30. Hering BJ, Wijkstrom M, Graham ML, Hårdstedt M, Aasheim TC, Jie T, et al. 2006; Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 12:301–3. DOI: 10.1038/nm1369. PMID: 16491083.
crossref
31. Cardona K, Korbutt GS, Milas Z, Lyon J, Cano J, Jiang W, et al. 2006; Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat Med. 12:304–6. DOI: 10.1038/nm1375. PMID: 16501570.
crossref
32. van der Windt DJ, Bottino R, Casu A, Campanile N, Smetanka C, He J, et al. 2009; Long-term controlled normoglycemia in diabetic non-human primates after transplantation with hCD46 transgenic porcine islets. Am J Transplant. 9:2716–26. DOI: 10.1111/j.1600-6143.2009.02850.x. PMID: 19845582.
crossref
33. Thompson P, Badell IR, Lowe M, Cano J, Song M, Leopardi F, et al. 2011; Islet xenotransplantation using gal-deficient neonatal donors improves engraftment and function. Am J Transplant. 11:2593–602. DOI: 10.1111/j.1600-6143.2011.03720.x. PMID: 21883917. PMCID: PMC3226931.
crossref
34. Bottino R, Wijkstrom M, van der Windt DJ, Hara H, Ezzelarab M, Murase N, et al. 2014; Pig-to-monkey islet xenotransplantation using multi-transgenic pigs. Am J Transplant. 14:2275–87. DOI: 10.1111/ajt.12868. PMID: 25220221. PMCID: PMC4169326.
crossref
35. Shin JS, Min BH, Kim JM, Kim JS, Yoon IH, Kim HJ, et al. 2016; Failure of transplantation tolerance induction by autologous regulatory T cells in the pig-to-non-human primate islet xenotransplantation model. Xenotransplantation. 23:300–9. DOI: 10.1111/xen.12246. PMID: 27387829.
crossref
36. Hawthorne WJ, Salvaris EJ, Chew YV, Burns H, Hawkes J, Barlow H, et al. 2022; Xenotransplantation of genetically modified neonatal pig islets cures diabetes in baboons. Front Immunol. 13:898948. DOI: 10.3389/fimmu.2022.898948. PMID: 35784286. PMCID: PMC9243461.
crossref
37. Thompson P, Cardona K, Russell M, Badell IR, Shaffer V, Korbutt G, et al. 2011; CD40-specific costimulation blockade enhances neonatal porcine islet survival in nonhuman primates. Am J Transplant. 11:947–57. DOI: 10.1111/j.1600-6143.2011.03509.x. PMID: 21521467. PMCID: PMC4845096.
crossref
38. Thompson P, Badell IR, Lowe M, Turner A, Cano J, Avila J, et al. 2012; Alternative immunomodulatory strategies for xenotransplantation: CD40/154 pathway-sparing regimens promote xenograft survival. Am J Transplant. 12:1765–75. DOI: 10.1111/j.1600-6143.2012.04031.x. PMID: 22458586. PMCID: PMC3387302.
crossref
39. Hecht G, Eventov-Friedman S, Rosen C, Shezen E, Tchorsh D, Aronovich A, et al. 2009; Embryonic pig pancreatic tissue for the treatment of diabetes in a nonhuman primate model. Proc Natl Acad Sci U S A. 106:8659–64. DOI: 10.1073/pnas.0812253106. PMID: 19433788. PMCID: PMC2688963.
crossref
40. Sun Y, Ma X, Zhou D, Vacek I, Sun AM. 1996; Normalization of diabetes in spontaneously diabetic cynomologus monkeys by xenografts of microencapsulated porcine islets without immunosuppression. J Clin Invest. 98:1417–22. DOI: 10.1172/JCI118929. PMID: 8823307. PMCID: PMC507568.
crossref
41. Elliott RB, Escobar L, Tan PL, Garkavenko O, Calafiore R, Basta P, et al. 2005; Intraperitoneal alginate-encapsulated neonatal porcine islets in a placebo-controlled study with 16 diabetic cynomolgus primates. Transplant Proc. 37:3505–8. DOI: 10.1016/j.transproceed.2005.09.038. PMID: 16298643.
crossref
42. Dufrane D, Goebbels RM, Gianello P. 2010; Alginate macroencapsulation of pig islets allows correction of streptozotocin-induced diabetes in primates up to 6 months without immunosuppression. Transplantation. 90:1054–62. DOI: 10.1097/TP.0b013e3181f6e267. PMID: 20975626.
crossref
43. Vériter S, Gianello P, Igarashi Y, Beaurin G, Ghyselinck A, Aouassar N, et al. 2014; Improvement of subcutaneous bioartificial pancreas vascularization and function by coencapsulation of pig islets and mesenchymal stem cells in primates. Cell Transplant. 23:1349–64. DOI: 10.3727/096368913X663550. PMID: 23461890.
crossref
44. Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, Im GS, et al. 2002; Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science. 295:1089–92. DOI: 10.1126/science.1068228. PMID: 11778012.
crossref
45. Hawthorne WJ, Lew AM, Thomas HE. 2016; Genetic strategies to bring islet xenotransplantation to the clinic. Curr Opin Organ Transplant. 21:476–83. DOI: 10.1097/MOT.0000000000000353. PMID: 27517501.
crossref
46. Hamawy MM. 2003; Targeting proximal T cell receptor signaling in transplantation. Transplantation. 75:1921–7. DOI: 10.1097/01.TP.0000070168.42915.47. PMID: 12829888.
crossref
47. Pallet N, Fernández-Ramos AA, Loriot MA. 2018; Impact of immunosuppressive drugs on the metabolism of T cells. Int Rev Cell Mol Biol. 341:169–200. DOI: 10.1016/bs.ircmb.2018.05.009. PMID: 30262032.
48. Meneghini M, Bestard O, Grinyo JM. Immunosuppressive drugs modes of action. Best Pract Res Clin Gastroenterol. 2021; 54-55:101757. DOI: 10.1016/j.bpg.2021.101757. PMID: 34874841.
crossref
49. Laman JD, Claassen E, Noelle RJ. 2017; Functions of CD40 and its ligand, gp39 (CD40L). Crit Rev Immunol. 37:371–420. DOI: 10.1615/CritRevImmunol.v37.i2-6.100. PMID: 29773027.
crossref
50. Kawai T, Andrews D, Colvin RB, Sachs DH, Cosimi AB. 2000; Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand. Nat Med. 6:114. DOI: 10.1038/72162.
crossref
51. Laman JD, Molloy M, Noelle RJ. 2024; Switching off autoimmunity. Science. 385:827–9. DOI: 10.1126/science.ade6949. PMID: 39172850.
crossref
52. GomezMancilla B, Meriggioli MN, Genge A, Roubenoff R, Espié P, Dupuy C, et al. 2024; Efficacy and safety of iscalimab, a novel anti-CD40 monoclonal antibody, in moderate-to-severe myasthenia gravis: a phase 2 randomized study. J Clin Neurosci. 119:76–84. DOI: 10.1016/j.jocn.2023.11.013. PMID: 37988976.
crossref
53. Nilsson B, Ekdahl KN, Korsgren O. 2011; Control of instant blood-mediated inflammatory reaction to improve islets of Langerhans engraftment. Curr Opin Organ Transplant. 16:620–6. DOI: 10.1097/MOT.0b013e32834c2393. PMID: 21971510.
crossref
54. Kanak MA, Takita M, Kunnathodi F, Lawrence MC, Levy MF, Naziruddin B. 2014; Inflammatory response in islet transplantation. Int J Endocrinol. 2014:451035. DOI: 10.1155/2014/451035. PMID: 24883060. PMCID: PMC4021753.
crossref
55. Lim H, Lee SH, Lee HT, Lee JU, Son JY, Shin W, et al. 2018; Structural biology of the TNFα antagonists used in the treatment of rheumatoid arthritis. Int J Mol Sci. 19:768. DOI: 10.3390/ijms19030768. PMID: 29518978. PMCID: PMC5877629.
crossref
56. Fabiani C, Vitale A, Emmi G, Lopalco G, Vannozzi L, Guerriero S, et al. 2017; Interleukin (IL)-1 inhibition with anakinra and canakinumab in Behçet's disease-related uveitis: a multicenter retrospective observational study. Clin Rheumatol. 36:191–7. DOI: 10.1007/s10067-016-3506-4. PMID: 27981463.
crossref
57. Speake C, Habib T, Lambert K, Hundhausen C, Lord S, Dufort MJ, et al. 2022; IL-6-targeted therapies to block the cytokine or its receptor drive distinct alterations in T cell function. JCI Insight. 7:e159436. DOI: 10.1172/jci.insight.159436. PMID: 36282595. PMCID: PMC9746808.
crossref
58. Gille I, Claas FH, Haasnoot GW, Heemskerk MH, Heidt S. 2022; Chimeric antigen receptor (CAR) regulatory T-cells in solid organ transplantation. Front Immunol. 13:874157. DOI: 10.3389/fimmu.2022.874157. PMID: 35720402. PMCID: PMC9204347.
crossref
59. Zimmermann K, Noyan F, Hust M, Schwinzer R, Hundrieser J, Hardtke-Wolenski M, et al. 2018; Operational tolerance in xenotransplantation by use of regulatory T cells with a SLA-specific chimeric antigen receptor. Transplantation. 102:S742. DOI: 10.1097/01.tp.0000543734.57229.97.
crossref

Table 1
Clinical application of porcine islet xenotransplantation
Study Country/study type Donor pig Antirejection method Transplantation site Clinical outcome
Groth et al. (1994) [13] Sweden/case series Fetal islet (55 to 77-day-old fetus) ATG or 15-deoxyspergualin, cyclosporine, prednisolone, azathioprine Intraportal, kidney subcapsular space Eight renal transplant recipients with diabetes; porcine C-peptide–positive for 200–400 days
Valdes-Gonzalez et al. (2010) [14] Mexico/trial Neonatal islet (5 to 9-day-old piglet) Islet + Sertoli cells via a collagen tube Subcutaneous device (6 × 0.8 cm stainless steel mesh tube) 23 Patients with type 1 DM; all patients tested positive for porcine C-peptide, and insulin-free status was achieved in 2 patients for several months
Ma et al. (2023) [15] China/trial Neonatal islet Anticoagulant therapy for 7 days; clinically accepted immunosuppressant drugs and autologous Treg Portal vein 10 Patients with type 1 DM; the conditions of these patients improved substantially
Elliott et al. (2007) [16] New Zealand/trial Term gestation piglet via hysterotomy Alginate encapsulation Abdominal cavity 2 Patients with type 1 DM in New Zealand (reduction of insulin dose with C-peptide positivity)
Skinner et al. (2011) [17] Russia/trial Neonatal islet (7 to 21-day-old piglet) Alginate encapsulation Abdominal cavity 8 Patients with type 1 DM in Russia (achieved insulin-free status in 2 cases)
Matsumoto et al. (2014) [18] New Zealand/trial Neonatal islet (7 to 21-day-old piglet) Alginate encapsulation Abdominal cavity 16 Patients with type 1 DM in New Zealand (reduction of hypoglycemic events)
Matsumoto et al. (2016) [19] Argentina/trial Neonatal islet (7 to 21-day-old piglet) Alginate encapsulation Abdominal cavity 21 Patients with type 1 DM in Argentina (reduction of insulin dose and undetected hypoglycemia)

ATG, antithymocyte globulin; DM, diabetes mellitus; Treg, regulatory T.

Table 2
Age-specific characteristics of porcine islets
Characteristic Embryonic and fetal Neonatal and young Young adult and adult
Age Embryonic days 24–100 1–5 days (neonatal)
7–22 days (young)
12–15 weeks (young adult)
>2 years
Islet size (μm) <50 (embryonic)
80 (fetal)
50–150 50–100 (young adult)
100–200 (adult)
Composition of beta cells after in vitro culture Embryonic
10% (fetal)
25%–50% >70%
Islet yield per pancreas (IEQs) Embryonic
–8,000 (fetal)
25,000–30,000 500–1,500 IEQ/g of pancreas (young adult)
1,000–5,000 IEQ/g of pancreas (adult)
Isolation and purification Easy Easy Very difficult (young adult)
Difficult (adult)
Tissue culture Resistant to hypoxia
Nonendocrine cells disappear
Resistant to hypoxia
Nonendocrine cells disappear
Fragile
Difficult to culture
In vivo functioning Delayed >4 to 6 months (embryonic)
Delayed >8 weeks (fetal)
Delayed >4 weeks (neonatal)
Within hours (young)
Within hours
Cost Low Low High
Pigs required per patient 60 Anlages (embryonic)
70–100 (fetal)
Potential ethical issues
7–10 2–4
Clinical trial experience Yes Yes No

IEQ, islet equivalents.

Modified from Park et al. [20] according to the Creative Commons License.

Table 3
Long-term graft survival of porcine islets in nonhuman primates
Study Pig islet source (recipient) Maximum graft survival (day) Immunosuppressive regimen Transplantation site
Hering et al. (2006) [30] WT adult (C) >187 Anti-IL-2R + anti-CD154 + FTY720 + rapamycin Portal vein
Cardona et al. (2006) [31] WT neonatal (R) >260 Anti-IL-2R + anti-CD154 + CTLA-4 Ig + rapamycin Portal vein
van der Windt et al. (2009) [32] hCD46-Tg adult (C) 396 ATG + anti-CD154 + MMF Portal vein
Thompson et al. (2011) [33] GTKO neonatal (R) 249 Anti-CD154 + anti-LFA-1 + CTLA-4 Ig + MMF Portal vein
Bottino et al. (2014) [34] Multi-Tg adult (C) 365 ATG + anti-CD154 + MMF Portal vein
Thompson et al. (2012) [38] WT neonatal (R) 114 Anti-IL-2R + anti-LFA-1 + CTLA-4 Ig + MMF + LFA-3 Ig Portal vein
Thompson et al. (2011) [37] WT neonatal (R) >203 Anti-IL-2R + anti-CD40 + CTLA-4 Ig + rapamycin Portal vein
Shin et al. (2015) [27] WT adult (R) >603 ATG + CVF + anti-TNF + anti-CD154 + rapamycin (+ Treg) Portal vein
Shin et al. (2016) [35] WT adult (R) 1,000 ATG + CVF + anti-TNF + anti-CD154 → anti-CD40 + rapamycin (+ Treg) Portal vein
Shin et al. (2018) [28] WT adult (R) >320 ATG + CVF + anti-TNF + anti-CD40 + rapamycin + tacrolimus Portal vein
Kim et al. (2021) [29] WT adult (R) >222 ATG + anti-TNF, IL-1, IL-6 + IVIg + tacrolimus + belimumab + rapamycin + tofacitinib Portal vein
Hawthorne et al. (2022) [36] GTKO, CD55, and 59 KI (B) 675 Anti-CD2 + tacrolimus + CTLA-4 Ig + Anti-CD154 Portal vein
Hecht et al. (2009) [39] WT fetal (C) 393 ATG + rituximab + FTY720 + anti-CD25 + CTLA-4 Ig + everolimus Omental pouch
Sun et al. (1996) [40] WT adult (C) 804 Alginate-encapsulated Peritoneal cavity
Elliot et al. (2005) [41] WT neonatal (C) >252 Alginate-encapsulated Peritoneal cavity
Dufrane et al. (2010) [42] WT adult (C) 180 Alginate-encapsulated with monolayer device Subcutaneous
Vériter et al. (2014) [43] WT adult (C) 240 Alginate-encapsulated with monolayer device Subcutaneous

WT, wild-type; C, cynomolgus; IL, interleukin; R, rhesus; CTLA-4, cytotoxic T lymphocyte-associated antigen-4; Ig, immunoglobulin; ATG, antithymocyte globulin; MMF, mycophenolate mofetil; GTKO, α1,3-galactosyl transferase-gene knockout; CVF, cobra venom factor; TNF, tumor necrosis factor; Treg, regulatory T; IVIg, intravenous immunoglobulin; B, baboon.

TOOLS
Similar articles