This website uses only technical or equivalent cookies.
For more information click here.

Transplant Immunology

Special Issue 3 - December 2023 - Transplant Immunology

Cardiac xenotransplantation – from bench to bedside

Authors

Key words: xenotransplantation, heart, pig, nonhuman primate, terminal heart failure
Submission Date: 2023-06-22
Publication Date: 2023-10-04

Abstract

Heart transplantation is widely regarded as the most effective and durable treatment option for patients with advanced/terminal cardiac failure. However, the need for organs by far surmounts the donations and the corresponding long waiting lists carry a considerable mortality. Currently, mechanical assist devices are the major alternative, but they come with a high complication rate and only moderate improvements in patients’ quality of life.

An emerging and promising solution is xenotransplantation of genetically multi-modified (GM) pig hearts. Recent studies have shown long-term success in baboons with both heterotopic (non-life-supporting) abdominal and life-supporting orthotopic porcine heart transplants. Notably, in a recent case of compassionate use, a pig heart with ten genetic modifications was implanted into a terminally ill patient, who survived for two months.

This review examines the concept of pig heart xenotransplantation, focusing on challenges such as immune rejection, coagulation dysregulation, and detrimental overgrowth of the transplanted heart; GM strategies and the non-nephrotoxic immunosuppression are discussed to minimize or prevent these issues. This review covers also microbiological (virologic) safety considerations and further requirements for initiating clinical studies.

Abbreviations

αGal: galactose-α(1,3)-galactose

ADCC: antibody-dependent cellular cytotoxicity

APC: antigen-presenting cell

ATMP: advanced therapy medicinal product

B2M: β2-microglobulin

B4GALNT2: β-1,4-N-acetyl-galactosaminyl transferase

BCL2: BCL2 apoptosis regulator

CD39: ectonucleoside triphosphate diphosphohydrolase 1

CD46: membrane cofactor protein (MCP)

CD47: leukocyte surface antigen CD47

CD55: complement decay-accelerating factor (DAF)

CD59: membrane inhibitor of reactive lysis (MIRL)

CMAH: cytidine monophosphate-N-acetylneuraminic acid hydroxylase

CPRP: complement pathway regulatory protein

CRISPR/Cas9: clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9

CTLA4-Ig: cytotoxic T lymphocyte-associated antigen-4-immunoglobulin (abatacept)

DCD: donation after circulatory death

DPF: designated pathogen-free

EMA: European Medicines Agency

EPCR: endothelial protein C receptor (CD201)

ETV2: ETS variant transcription factor 2

FDA: US Food and Drug Administration

GGTA1: α-1,3-galactosyltransferase

GHR: growth hormone receptor

GM: genetically (multi)modified

hiPSC: human induced pluripotent stem cell

HLA: human leukocyte antigen

HMOX1: haeme oxygenase 1

HTx: heart transplantation

ICU: intensive care unit

IGF1R: insulin-like growth factor 1 receptor

IL6R: interleukin 6 receptor

KDR: kinase insert domain receptor

KO: knockout

LEA29Y: affinity optimized CTLA4-Ig (belatacept)

mAb: monoclonal antibody

MMF: mycophenolate mofetil

mTOR: mechanistic target of rapamycin

MYF5/6: myogenic factor 5/6

MYOD: myogenic differentiation

Neu5Gc: N-glycolylneuraminic acid

NHP: nonhuman primate

NK cell: natural killer cell

PCMV: porcine cytomegalovirus

PCR: polymerase chain reaction

PCXD: perioperative xenograft dysfunction

PD-L1: programmed death-ligand 1

PEI: Paul-Ehrlich-Institute

PERV: porcine endogenous retrovirus

PRA: panel reactive antibodies

Sd(a): blood group antigen

SIRPα: signal regulatory protein alpha

SLA: swine leukocyte antigen

TBM: thrombomodulin (CD141)

TF: tissue factor

TFPI: tissue factor pathway inhibitor

tg: transgenic

TKO: triple-knockout

TM: thrombotic microangiopathy

TNFAIP3: TNF alpha induced protein 3 (A20)

TP53: tumour protein P53

INTRODUCTION

Current medical treatments for advanced heart failure are highly effective (reviewed in Bauersachs, 2021 1). However, when all options fail, heart transplantation (HTx) remains the gold standard for patients with end-stage heart disease, offering a high probability level of an extended life time in good general condition. The world-wide shortage of donated human organs results in waiting lists (Tab. I) with annual numbers being twice as high when compared to those of actually done transplants. Long waiting lists carry a considerable mortality; furthermore, additional patients need to be withdrawn due to severe side effects, which make future transplantations impossible.

Taking extended risks in donor selection is one alternative solution 2, donations after circulatory death (DCD) are another 3-5; DCD is, however, not permitted in Germany.

Currently, mechanic assist devices are the major alternative, but they come with a high complication rate and only moderate improvements in patients’ quality of life. The one- and five-year survival rates are 83 and 52 percent, respectively, and, when compared with autologous transplantations, are significantly worse after the longer observation time. The hospital readmission rates, mainly due to infections and bleeding events, are high after three and twelve postoperative months (36 and 68 percent, respectively); withdrawal of care is the main cause of death 6.

Significant progress has been made in pig-to-primate cardiac xenotransplantation, using GM donor pigs, improved preservation techniques, optimized transplantation models, and effective immunosuppressive regimens 7-10, demonstrating the potential for clinical application.

In January 2022, the first compassionate use xenotransplantation of a 10xGM pig heart into a patient with terminal heart failure was done at Maryland University, Baltimore 11. The patient died after 2 months due to multiple pre-operative risk factors and portentous complications such as intra-operative acute aortic dissection involving the renal and mesenteric artery branches and their end-organs; a porcine cytomegalovirus (PCMV) infection caused endothelial damage of the transplant and may also have contributed to the patient’s demise after two post-operative months 11. However, the ability to sustain normal heart function for more than 45 days is widely regarded as a crucial demonstration that clinical cardiac xenotransplantation is feasible.

In this overview, we provide a summary of the background and outline additional steps that we believe are necessary to achieve consistent long-term success and finally translate xenogeneic heart transplantation into clinical reality.

GENETIC MODIFICATION OF SOURCE PIGS TO ALLEVIATE THE PATHOBIOLOGY OF PIG HEART XENOTRANSPLANTATION

Xenogeneic rejection reactions

The intricate nature of organ xenotransplantation’s pathobiology surpasses that of allotransplantation, as the innate immune responses assume a more significant role. The factors contributing to xenograft destruction have been comprehensively reviewed by Cooper et al. 12 in this issue. In brief: during infancy, both humans and nonhuman primates (NHPs) develop antibodies that cross-react with carbohydrate antigens found on the surfaces of genetically unmodified pig cells. Hence, when a wild-type pig organ is transplanted into a human or baboon, these antibodies swiftly bind to the graft’s vascular endothelial cells. In a next quick step, the complement cascade is activated, leukocytes are attracted and infiltrate the porcine heart through (antibody) Fc-receptor-mediated and Fc-independent mechanisms. Consequently, the graft is typically rejected within minutes to hours. This antibody dependent “hyperacute rejection” is characterized by histopathological features such as venous thrombosis, loss of vascular integrity, interstitial haemorrhage, oedema, and infiltration of innate immune cells.

Hyperacute (and later acute vascular) rejections of pig organs in humans or NHPs primarily occur due to (preformed) antibodies targeting galactose α-(1,3)-galactose (αGal). Humans possess additional natural antibodies against N-glycolylneuraminic acid (Neu5Gc) and a glycan resembling the human Sd(a) blood group antigen (often referred to as β4Gal). In contrast, NHPs only exhibit anti-αGal and anti-Sd(a) antibodies (reviewed in Byrne et al., 2018; Sykes and Sachs, 2019) 13,14.

To eliminate the αGal, Neu5Gc, and Sd(a) epitopes as target antigens for xenograft rejection in humans, pigs with inactivated α-1,3-galactosyltransferase (GGTA1) 15, cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) 16,17, and β-1,4-N-acetyl-galactosaminyl transferase 2 (B4GALNT2)/B4GALNT2-like (B4GALNT2L) 18 genes – so-called triple-knockout (TKO) pigs – were generated. However, complement activation can also occur through pathways unrelated to antibody binding, such as ischaemia-reperfusion injury. Therefore, transgenic pigs expressing human complement pathway regulatory (inhibitory) proteins (CPRPs), namely CD46 19, CD55 20, and CD59 21 have been generated; organs derived from animals with transgenic expression of one or more human CPRPs exhibit a significant level of safeguarding against human/NHP complement-mediated injury. In combination with the TKO animals, cell injury is remarkably diminished in these “humanized” porcine organs (reviewed in Galli, 2023 22).

Reducing additional detrimental targets diminishes antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells: since swine leukocyte antigen (SLA)-I has limited ability to bind inhibitory NK cell receptors, human/NHP NK cells directly exhibit cytotoxicity against porcine cells. To counter this, transgenic pigs expressing human leukocyte antigen (HLA)-E/β2-microglobulin (B2M) have been generated as a strategy 23.

Furthermore, porcine cells activate macrophages since porcine CD47 fails to bind the “don’t eat me” signal regulatory protein alpha (SIRPα) on human macrophages. Consequently, transgenic pigs expressing human CD47 have been developed 24.

The activation of human/NHP T cells against porcine xenotransplants occurs either directly through porcine antigen-presenting cells (APCs) presenting porcine peptides or indirectly via human/NHP counterparts. These processes involve several co-stimulatory and co-inhibitory signals; direct T cell activation can be reduced by eliminating or downregulating SLA molecules (as reviewed in Ladowski et al., 2021 25) or by blocking the CD40-CD40L (CD154) costimulatory signal with antibodies (see below). Additionally, transgenic pigs expressing CTLA4-Ig, or its high-affinity derivative LEA29Y 26,27, have been developed to block the CD28-CD80/CD86 co-stimulatory pathway. In another approach, membrane-bound human PD-L1 is expressed on pig cells to activate the inhibitory PD1 receptor on infiltrating human/NHP leukocytes 28. To deplete and inhibit T and NK cells, transgenic pigs have been generated which express a monoclonal anti-human CD2 antibody construct 29.

For LEA29Y, PD-L1 and CD2 transgenic pigs, effective local expression is being investigated as a means to attenuate immune reactions and thus to reduce or even replace the need for systemic immunosuppression.

Xenogeneic coagulation disorders

Coagulation pathway dysregulation is another aspect of the pathobiology associated with pig organ xenotransplantation (as reviewed in Cowan, Robson, 2015; Pierson et al., 2020 30,31). Contributing factors include the immune responses mentioned earlier, which propagate inflammation, vascular damage, and therefore a procoagulant state of the porcine endothelium; molecular incompatibilities between porcine and human/NHP coagulation regulators are a major cause. Although the measures employed to prevent hyperacute xenograft rejection helped to avert systemic life-threatening consumptive coagulopathy, pig hearts, transplanted long-term into baboons abdominally, displayed signs of thrombotic microangiopathy (TM) – even under clinically approved anticoagulation therapy. In contradistinction, transgenic expression of human thrombomodulin (TBM) in donor pigs (e.g., 32-34) successfully prevented TM by overcoming the inability of porcine TBM – in complex with human/NHP thrombin – to activate human/NHP protein C. Activation of this anticoagulation pathway is necessary to prevent the formation of detrimental fibrin clots in the capillary system of the donor organ and the above mentioned TM.

Although the porcine endothelial protein C receptor (EPCR; supporting the activation of protein C) seems to be compatible with the human/NHP protein C pathway 35, transgenic pigs expressing human EPCR have been created to achieve higher EPCR levels and thereby enhance protective thromboregulation.

Other genetic modifications also aim at addressing coagulation dysregulation: Expression of human tissue factor pathway inhibitor (TFPI) to prevent activation of TF-factor VIIa complexes; expression of human ectonucleoside triphosphate diphosphohydrolase 1 (CD39) to inhibit platelet aggregation and consecutive thrombus formation (reviewed in Cowan, Robson, 2015; Pierson et al., 2020 30,31); and siRNA-mediated suppression of porcine TF expression 36.

Additionally, transgenic pigs expressing anti-inflammatory proteins such as human TNF-alpha-induced protein 3 (TNFAIP3 alias A20 37) or human haeme oxygenase 1 (HMOX1 38) have been generated, aiming to prevent or reduce inflammation that may not be adequately controlled by other genetic modifications.

Lethal overgrowth of the donor heart

Detrimental intrinsic (genetically determined) overgrowth of the porcine organ (the heart of an outgrown German Landrace pig weighs one kg) has consistently been observed in preclinical studies of cardiac xenotransplantation 7. In the preclinical setting, overgrowth is retarded with rapamycin, a blocker of the activation of mTOR (mechanistic target of rapamycin), which is part of the signalling cascade of many growth stimulating hormones. To address this issue clinically, one approach involves the generation of donor pigs with loss-of-function mutations in the growth hormone receptor (GHR) gene 39,40. GHR inactivation resulted in approximately 50% reduced body and organ weights. A comprehensive proteome analysis of GHR-deficient pig hearts revealed no major other molecular abnormalities 40; GHR deficiency, along with other genetic modifications, has contributed to the prolonged survival of orthotopic porcine cardiac xenografts beyond 6 months 9,41; in the Maryland case, GHR deficiency was one of the 10 GM of the donor organ. However, GHR deficiency in donor pigs may have side effects, such as marked obesity 39, transient juvenile hypoglycaemia 42, altered liver metabolism 43, and structural/proteomic alterations of the anterior pituitary gland 44. Thus, it is preferably to use a genetic background of source pigs that fits the size of humans.

TOWARDS PARTIALLY HUMANIZED PIG HEARTS: EXOGENESIS

To enhance compatibility between pig hearts and humans, an additional technique called exogenesis has been proposed 45. This approach aims to achieve complete humanization of heart structures, specifically the endothelium (Fig. 1). Exogenesis involves the creation of interspecies chimeras using genetically engineered pig embryos (reviewed in Garry DJ, Garry MG, 2019 46). These embryos lack the ability to generate or sustain certain cell lineages, creating a niche for the introduction of donor human cells or even parts of organs. However, the formation of whole organ chimeras between pigs and humans faces fundamental barriers due to their evolutionary distance. Several mechanisms contribute to this xenogeneic barrier (reviewed in Zheng et al., 2021 47). Apoptosis and cell competition can eliminate donor cells, ligand-receptor incompatibilities between species pose challenges, differences in developmental timing present obstacles, and mismatches in cell adhesion molecules hinder the formation of adequate junctions between human and pig cells. To overcome these barriers, researchers have explored various genetic manipulations in pig embryos. For instance, overexpressing the antiapoptotic factor BCL2 in human induced pluripotent stem cells (hiPSCs) and engrafting them into pig blastocysts lacking ETV2, a master regulator of haematoendothelial lineages, has facilitated the generation of pig embryos with human endothelium 45. Knocking out the kinase insert domain receptor (KDR) gene is another approach to generate porcine embryos with impaired vasculogenesis 48. The development of GM pig hearts with humanized endothelium is of particular significance, as the endothelium is the primary target for rejection mechanisms following xenogeneic heart transplantation.

Furthermore, porcine embryos with a human myogenic lineage have been generated by complementing defective pig blastocysts lacking myogenic factor 5 (MYF5), myogenic differentiation (MYOD), and myogenic factor 6 (MYF6) with TP53-null hiPSCs 49. Inactivation of TP53 in the donor cells allows adaptation to the low TP53 expression in porcine host embryos, resulting in increased chimerism. Strategies to enhance the inter-species chimera competency of human stem cells, such as naïve vs. primed vs intermediate hiPSCs, have been extensively reviewed 47. Additional efforts to improve interspecies chimerism focus on modifying the host embryo, such as inactivating the insulin-like growth factor 1 receptor (IGF1R) gene 50. Systematic analysis of early porcine embryo and heart development is expected to uncover new strategies for enhancing the efficiency of porcine blastocyst complementation with human stem cells. This may involve humanizing essential ligands, receptors, or adhesion molecules in the porcine host embryos 51.

HETEROTOPIC AND ORTHOTOPIC HEART TRANSPLANTATION IN THE PIG-TO-NHP MODEL

Previous comprehensive reviews have examined the early results after pig heart transplantation in NHPs (1968-2013) 52. The baboon (Papio anubis or hamadryas) has been the most commonly used recipient species for preclinical porcine cardiac xenotransplantation, and three transplantation models have been established for these animals (as reviewed in Mohiuddin et al., 2015 53).

In the abdominal heterotopic cardiac xenotransplantation technique (i), the porcine ascending aorta (with both coronary arteries) is connected to the recipient abdominal aorta, the porcine pulmonary artery (with the coronary venous efflux) to the recipient inferior vena cava (Fig. 2A). Upon release of the aortic clamp, the transplanted heart is perfused and starts pumping. As there is no systemic venous (caval) return, the transplant beats empty; the recipient survives on its own organ, which is left untouched in the chest. This easy to accomplish transplantation model is primarily used to assess the effectiveness of immunosuppressive regimens and new combinations of genetic modifications. With appropriate immunosuppressive therapy, pig hearts lacking αGal, expressing hCD46 and hTBM have survived for up to 945 days (median 298 days 32).

In the intrathoracic heterotopic cardiac xenotransplantation technique (ii), the xenograft is placed within the right thoracic cavity and to the right of the recipient heart, resulting in compression of parts of the upper and middle lobes of the right lung 54-56. Four anastomoses are necessary: The connections between the respective left and right atria to achieve physiologically appropriate bi-atrial “inflow”, as well as end-to-side “outflow” connections between both ascending aortae and pulmonary artery trunks; the latter requires an extension with an interposition Dacron or Gore-Tex graft (Fig. 2B). In this “piggyback” position, the xeno-heart can fully or partially support the recipient’s organ, and carries under clinical conditions on average 73% of the total cardiac output 57. The intrathoracic heterotopic cardiac technique has been considered as a potential scenario for an early clinical translation, as the recipient’s native heart can provide the live-saving minimal cardiac output in case a xenograft fails 58. Re-transplantation may then be considered, utilizing either a human or again a porcine organ.

Long-term results after allo-procedures were good, although post-operative anticoagulation is mandatory to avoid thrombus formation within the recipient left ventricle with consecutive systemic emboli 59. Our group in Munich carried out pig-to-baboon intrathoracic heterotopic heart experiments between 2009 and 2013. Short-term results (recipient survival, initial xenograft function) were excellent, but long-term results were limited due to the toxic immunosuppressive therapy at that time (e.g. co-stimulation blockade was not available) and no control of the donor organ overgrowth 60.

The most rigorous preclinical model is the orthotopic cardiac xenotransplantation technique (iii), where the baboon’s own organ is replaced with a GM pig heart using a surgical procedure identical to that of cardiac allotransplantation (Fig. 2C 61). This model extensively proves the life-supporting function of a xenograft heart, and achieving consistent success in good general condition is considered a prerequisite before entering clinical studies 62. Non-ischaemic preservation of the heart using ex vivo perfusion (63; XVIVO, Gothenburg, Sweden), non-nephrotoxic immunosuppression 32, and post-implantation growth control of the xenograft heart 7 are necessary. Table II summarises the results of orthotopic pig-to-baboon heart transplantation experiments.

LIMITATIONS OF THE PIG-TO-NHP CARDIAC XENOTRANSPLANTATION MODELS, THE ALTERNATIVE DECEDENT EXPERIENCE

While inactivation of GGTA1 along with expression of hCD46 and hTBM has proven sufficient to achieve long-term survival in the preclinical orthotopic NHP model, the combination of inactivation of GGTA1, CMAH, and B4GALNT2/B4GALNT2L (TKO) plus transgenic expression of one or several complement pathway regulatory proteins and human TBM is our preferred minimal set of genetic modifications for clinical cardiac xenotransplantation studies. Testing this combination in baboons is complicated by a significant difference in the innate immune response between humans and NHPs. In contrast to humans, all Old-World monkeys, including baboons, express Neu5Gc, as do pigs. When Neu5Gc is deleted in TKO pigs, it appears that another xenoantigen (sometimes known as the ‘4th xenoantigen’, presumable a glycan) is exposed. The structure and identity of the ‘4th xenoantigen’ remains unknown, but most NHPs express natural antibodies reacting with CMAH-KO or TKO cells 18. Binding of these antibodies to TKO pig grafts is associated with a high level of complement-dependent cytotoxicity 70-72 and reduced graft survival in heart 9 and kidney 73 xenotransplantation models. This – in the end clinically irrelevant – phenomenon has proven to be a major obstacle (for e.g. regulatory authorities) in predicting how a TKO pig organ would work in a human recipient 73,74. Additional inactivation of the CMAH gene reduces the antigenicity in a future clinical setting and should therefore be included (but should be avoided in pre-clinical trials 18,72; reviewed in Cooper et al., 2019 75).

In that respect, is the recent so-called “decedent model” of interest, meaning the transplantation of clinical-grade GM porcine organs into brain dead persons with still functioning organs. At New York University, Montgomery et al. 76 attached GGTA1-KO kidneys to groin vessels in two such cases. During the short observation time of 54 h, there were no signs of (hyperacute) rejection and the transplanted organs were passing urine normally. No serum creatinine increases were observed, the recipient’s own kidneys remained however untouched and were working.

In the University of Alabama case, J.E. Locke and her group 77 transplanted two 10xGM kidneys (GGTA1-KO, CMAH-KO, B4GALNT2-KO, GHR-KO; hCD46-tg, hCD55-tg; hTBM-tg, hEPCR-tg, hCD47-tg, hHOMX1-tg; United Therapeutics (UT)/Revivicor, Blacksburg, Virginia, USA) into a nephrectomised decedent. During the 74-h follow-up time, again, there were no signs of rejection; however, the creatinine clearance did not recover. At autopsy, there were histologic features of thrombotic microangiopathy and tubular necrosis.

In June, July 2022 and again at New York University, two orthotopic heart transplantations were done and allowed to beat for 72 hours; again, 10xGM pigs (UT/Revivicor) served as donors and no signs of rejection were recorded 78,79. Although both xenografts beated normally after starting reperfusion with the recipients‘ blood, cardiac function decreased in the further follow-up in one case, attributed to a size mismatch between the donor pig (organ too small) and the recipient.

Taken together, such short-term experiments are probably of limited scientific value, but the unstable condition of a brain-dead recipient does not permit easily longer observation times 80. For more reliable information, xenotransplantations must be done in living patients.

PREREQUISITES FOR CONSISTENT SUCCESS IN (PRE-) CLINICAL CARDIAC XENOTRANSPLANTATIONS

Stable genetic modifications of the source pigs

These aspects have already been discussed in chapter “Genetic modification of source pigs to alleviate the pathobiology of pig heart xenotransplantation”.

Non-ischaemic perfusion preservation of the donor heart

For over 20 years, the preclinical results after orthotopic xenogeneic heart transplantations were unpredictable with a 40-60% peri-operative mortality, inconsistent, despite the use of clinically approved cold static preservation techniques (reviewed in Shu et al., 2022 81). This phenomenon was termed “Peri-operative Cardiac Xenograft Dysfunction” (PCXD) and was thought to be due to ischaemia/reperfusion injury 53,82; when compared to humans, porcine hearts are obviously less resistant against ischaemia. Since December 2015, PCXD has been consistently prevented by a continuous (non-ischaemic) perfusion of the grafts with an 8°C hyperoncotic, oxygenated cardioplegic solution containing erythrocytes, nutrition and hormones 63,83; the perfusion was intermittently continued even during implantation. This perfusion preservation technique was also employed in the clinical Maryland case 11.

Development of a non-nephrotoxic immunosuppressive regimen with CD40-CD154 co-stimulation blockade

Initial pig-to-baboon cardiac xenotransplantation studies used conventional immunosuppressive regimens including cyclophosphamide, cyclosporine A or tacrolimus, mycophenolate mofetil, and corticosteroids without success. Since 2000, co-stimulation blockade – first with anti-CD154 mAb – was applied in abdominal heterotopic heart transplantation experiments (84; reviewed in Bühler et al., 2000 85). After anti-CD154 mAb was found to be thrombogenic in humans, the chimeric anti-CD40 2C10 mAb-based regimen was introduced and has since then contributed to the longest reported cardiac xenograft survivals after heterotopic 32 and orthotopic transplantations in baboons 7,9.

The humanized anti-CD40 antibody KPL-404 (Kiniksa Pharmaceuticals, Lexington, Massachusetts, USA) was given in the recent Maryland case 11.

The following immunosuppressive drugs complemented the treatment: for induction, cortisone, ATG and anti-CD20 antibody; for maintenance, cortisone tapered down, MMF and/or rapamycin (also necessary for graft overgrowth control).

Post-implantation growth control of the xeno-heart

Domestic pig breeds used for xenotransplantation experiments, such as German Landrace or Large White, weigh outgrown 200-300 kg; heart sizes increase proportionally and reach then approximately one kg. This size mismatch is of great importance for both preclinical experiments as well as clinical applications.

For many years, it was believed that after xenogeneic transplantations, the grafts would adapt to the growth regulation of the recipient under the influence of extrinsic (recipient dependent) factors such as hormones and growth factors (reviewed in Lui and Baron, 2011; Penzo-Méndez and Stanger, 2015 86,87). However, Längin et al. 7 could demonstrate more recently that the donor organ growth is regulated intrinsically, which means genetically, or in other words: the xeno-heart in the primate body increases its size as if it still would be within the porcine chest (this “overgrowth” was also observed in xenogeneic kidney transplantation experiments 88,89). After heterotopic thoracic xeno-heart transplantation 60, this phenomenon caused a reduction in pulmonary function, and in the orthotopic model diastolic pump failure and subsequent congestive liver damage 7.

In the preclinical setting, cardiac overgrowth was successfully prevented by decreasing the blood pressure (baboons have a higher blood pressure than pigs), early weaning from cortisone, and treatment with sirolimus (or the i.v. donation of the prodrug temsirolimus), which inhibits activation of the mechanistic target of rapamycin (mTOR) and thereby cardiomyocyte hypertrophy.

An alternative would be use of GHR-KO pigs as described before; GHR-KO was included in the 10xGM pig (United Therapeutics/Revivicor, Blacksburg, VA) used as donor for the already mentioned Maryland case.

In future clinical applications, however, smaller donor animal breeds will be preferred, such as the Auckland Island pigs from New Zealand (NZeno, Auckland), which weigh outgrown between 70 and 90 kg, i.e. perfect for adult humans. In the meantime, a small porcine endogenous retrovirus-C (PERV-C) free herd has been established in a farm near Munich. GM Yucatan minipigs were used by Ma et al., 2022 90.

SAFETY OF XENOTRANSPLANTATION, AVOIDING POTENTIAL INFECTIOUS COMPLICATIONS

The microbiologic and virologic safety profile of porcine xenotransplants is high, since GM donor pigs must be maintained in designated pathogen-free (DPF) barrier facilities ensuring the absence of zoonotic pathogens (reviewed in Fishman, 2018;2020 91,92). In addition, highly sensitive and specific assays have been established for specific pathogens which must be absent from the donor pigs 93. Some of them like the porcine cytomegalovirus (PCMV) had a significant negative effect on cardiac xenograft survival times in preclinical transplantation experiments 94 and may have contributed to the Maryland heart xenograft recipient’s demise 11. It is thus mandatory to use only PCMV negative donor pigs confirmed by negative PCR assays and, more importantly, by absent serological antibodies 95,96. Since there exist no drugs or vaccines, “early weaning” is the only solution at the present time, as it prevents the nasal transmission from the mother to the piglets while sucking milk.

Of special importance are the porcine endogenous retroviruses (PERVs) since they are integrated in the porcine genome and will remain there: PERV-A and PERV-B are present in all pigs, whereas PERV-C is in the genome of most, but not all animals. PERV-A and PERV-B are polytropic and can infect human cells only in vitro, whereas PERV-C is ecotropic and infects only pig cells 97. When compared with PERV-A, recombinants between PERV-A and –C carry higher replication rates 98,99 and must be avoided.

PERV-A, -B and -A/C have been shown to infect human tumour and immortalized cell cultures, but rarely primary cells. To date, PERV transmission has not been detected in numerous preclinical xenotransplantation nor in infection experiments in different species 100 – nor in clinical trials with encapsulated porcine islets in diabetic patients 101,102.

Virologists are now of the opinion, that PERVs don’t pose the tremendous risk, as more than 25 years ago researchers thought it would be 103. Since, however, a PERV infection cannot be ruled out for sure, several strategies have been proposed to prevent an (unlikely) PERV transmission:

  • selection of pigs with low expression of PERVs and therefore a low probability to release infectious particles;
  • selection of PERV-C-negative animals to prevent PERV-A/C recombination (our strategy, which is accepted by both FDA and EMA);
  • inhibition of PERV expression by RNA interference 97;
  • inactivation of PERVs using CRISPR/Cas9 technology 104; this strategy may however be associated with off-target effects 105,106; and
  • use of antiviral drugs.

WHAT EXPERIMENTAL RESULTS WOULD JUSTIFY A FORMAL CLINICAL TRIAL?

In 2000, the ad hoc Xenotransplantation Advisory Committee of the International Society for Heart and Lung Transplantation recommended that consistent survival of NHPs supported by pig orthotopic heart transplants for 3 months would be sufficient to warrant moving to a clinical trial 62. That recommendation was made at a time when even 3-month survivals had been unobtainable.

The state of the science has changed dramatically since those pioneering days, and consequently the experimental evidence needs to be stronger, and has indeed already been achieved 7-9. We therefore suggest that consistent survival of up to 6 months, in the absence of features of irreversible rejection or infection, would be sufficient to warrant moving towards a clinical trial in carefully selected patients. Achieving survival for longer durations, with one or two recipients being followed for nine or even 12 months would be reassuring.

However, we would emphasize that any clinical trials should be carried out by teams with experience of both clinical orthotopic heart transplantation and the preclinical setting of a pig-to-NHP model.

REGULATORY ASPECTS

National regulatory bodies (FDA, EMA; in Germany the Paul-Ehrlich-Institute, PEI) have the authority to determine what experimental benchmarks in preclinical studies are appropriate, what kind of (microbiologic/virologic) safeguards are necessary, before approving clinical trials of xenotransplantation. The necessary regulatory framework in the United States has been summarized in a recent letter 107. Within Europe, guidelines on Advanced Therapy Medicinal Products (ATMP; EC/1394/2007) must be adhered. In addition, national laws, such as the AMG (Arzneimittelgesetz, law for medicinal products, which includes bioengineering) in Germany, will be additionally implemented.

It should be remembered that, to date, no NHP has survived longer than 9 months after orthotopic pig heart transplantation 7,9. Consequently, regulatory authorities like the FDA or EMA may decide that (compassionate use) pig heart transplantations should initially be offered as a bridge-to-allotransplantation; in other words: after several months, a human heart-transplantation would be done if clinically indicated.

“Compassionate use” means in this context that a specific patient has no realistic chance to receive an allograft during lifetime. After an in-depth informed consent, the decision for a cardiac xenotransplant is made. Since under these circumstances the whole responsibility remains within the treating team, no permission from regulatory authorities is necessary.

This is in contradistinction to small pivotal or pilot studies, which need such a permission and which is only granted after a thorough (and lengthy) investigation (in that case primarily, the high level exclusion of the transmission of pathogenic microorganisms).

The FDA recognizes the need for xenotransplantation and accepts that:

  1. pigs with multiple genetic modifications would be required;
  2. cloned pigs could be used for the initial studies; and
  3. complete inactivation of porcine endogenous retroviruses is not required.

There is the necessity for biobanking in order to be prepared for the emergence of unknown micro-organisms.

FIRST PATIENT SELECTION

The patients for the first clinical cardiac xenotransplantation trials must be carefully selected to justify these interventions and ensure very likely favourable outcomes. Initial candidates could be intensive care (ICU) patients who are poor candidates for mechanical circulatory support, such as those with hypertrophic cardiomyopathy, prior mechanical valve replacements, deteriorated aortic bio-protheses, post-infarction ventricular septal defects. These high-risk patients become increasingly unstable due to inotrope requirements and arrhythmias; secondary liver and kidney damages must be considered reversible and pulmonary hypertension medically treatable (reviewed in Reichart et al., 2021 58; Table III).

Paediatric patients with complex congenital heart diseases are of special interest, particularly those with single right ventricular pathophysiology. Although palliative surgical techniques (Norwood, Fontan) provide adequate results in some patients, survival and quality of life are limited, particularly in patients with high-risk anatomic lesions or complex arrhythmias. In contradistinction, these high-risk patients do well after allotransplantation 109, but have a high mortality while waiting for a (usually small) heart. In single-ventricle patients, mechanical circulatory devices are associated with little success 110. In addition, total implantable systems are impossible in those below a bodyweight of 40 kg. In these patients, paracorporeal pump chambers are implanted 111.

HOW DO WE PREDICT THE FUTURE OF CARDIAC XENOTRANSPLANTATION DURING THE NEXT 5-10 YEARS?

For humans with advanced/terminal myocardial disease, allografts will always be preferable. Due to the long waiting lists, and as an alternative, we suggest that bridging with a pig heart xenograft will be introduced into the clinic within the next two years, possibly initially again on an individual compassionate basis, but preferably as part of a formal clinical trial. We expect that trials in both infant and adult patients will be approved. With successful longer-term experience, we predict that cardiac xenotransplantation as destination therapy will soon be an accepted treatment form.

We firmly anticipate that advances will be made in the field of xenotransplantation during the next decade that will surpass those of mechanical assist devices, stem cell technology, and regenerative medicine.

Conflict of interest statement

BR and EW are cofounders of XTransplant GmbH, Starnberg, Germany.

Funding

This work was supported by the Deutsche Forschungsgemeinschaft (DFG; CRC/TR 127 to BR and EW) and by the Swiss National Science Foundation (SNSF; Sinergia grant CRSII5_198577/1 to EW).

Author contributions

EW, BR: drafted the initial manuscript; EW, MS, BR: reviewed and edited the draft. All authors approved the final manuscript.

Ethical consideration

Not applicable.

Figures and tables

Figure 1. Principle of exogenesis to generate partially humanized pig hearts.

Figure 2. Models of pig-to-baboon cardiac xenotransplantation: A) heterotopic abdominal; B) heterotopic thoracic, and C) orthotopic techniques; D: donor; R: recipient (modified with permission from Mohiuddin et al., 2015) 53.

Region Year Size of waiting list Deaths on waiting list
Eurotransplant 2022 1044 116
France 2017 874 43
Spain 2021 495 12
Italy 2022 706 71*
Poland 2017 691 79
United Kingdom 2022 320 22
Austria 2021 112 3
Portugal 2021 93 7
Sweden 2021 90 1
Finland 2021 82 1
Czech Republic 2021 56 20
Denmark 2021 46 1
Ireland 2021 9 1
Source: https://www.statista.com/statistics/; *number in 2021.
Table I. Active waiting lists for a heart transplant in Europe.
Genetically modified donor pigs Organ preservation ISR Growth controla Survival time [days]b Ref.
Knockouts Transgenes
GGTA1 CMAH B4GALNT2(L) GHR hCD46 hCD55 hTBM hEPCR hCD47 hHMOX1
X Static A - < 1(2); 5(2); 9 65
X Static A - < 1(5); 4; 5(3); 9 66
X Static B - 39 67
X Static C - 9; 25 68
X Static C - 0; 2(2); 34; 40; 57 68
X X Static C - 0; 1; 2; 14 68
X X X Static D - 1(3); 3; 30 7
X X X Perfusion D - 4c; 18; 27; 40 7
X X X Perfusion D + 90d (2); 182; 195 7
X X X Perfusion D + 15e; 27e; 90d (2) 8
X X X Blood cardioplegia E - < 1; 4; 29 9
X X X Perfusion E 57 9
X X X Perfusion E - 6 9
X X X X X Perfusion E - < 1 9
X X X Perfusion E - 8 9
X X X X X X X X Perfusion E - 84; 95 9
X X X X X X X Perfusion E - 182f; 264 9
X X Static (UW) F + < 1 10 , 69
X X Static (del Nido) F + 90 10 , 69
X X X Static (del Nido) F + < 1; 241 10 , 69
A: Treatment with mTOR inhibitor temsirolimus or rapamycin; B: Numbers of animals with the same survival time are given in brackets; C: Technical failure; D: Terminated according to experimental protocol; E: Donor infected with porcine cytomegalovirus (PCMV); F: Elective euthanasia. Immunosuppressive regimens (ISR): A: cyclosporine A (CsA), corticosteroids (Cs), cyclophosphamide (CyP); B: CsA, Cs, CyP, mycophenolate mofetil (MMF); C: anti-thymocyte globulin (ATG), rituximab, tacrolimus, sirolimus, TPC (αGal polymer) or GAS914 some recipients; D: induction: anti-CD20 monoclonal antibody (mAb), anti-CD40 mAb or PASylated anti-CD154 Fab, ATG; maintenance: MMF, anti-CD40 mAb or PASylated anti-CD154 Fab, methylprednisolone (MP; tapered down); anti-inflammatory therapy: interleukin-1 receptor (IL1R) blocker, IL6R blocker, tumour necrosis factor alpha (TNFA) inhibitor; E: induction: anti-CD20 mAb, anti-CD40 mAb, ATG, cobra venom factor (CVF) or C1 esterase inhibitor; maintenance: MMF, anti-CD40 mAb; anti-inflammatory therapy: IL6R blocker, TNFA inhibitor. F: induction: ATG, anti-CD20 mAb, C1-esterase inhibitor; maintenance: anti-CD40 mAb, rapamycin, methylprednisolone; adjunctive medication: etanercept (TNFA antagonist), ganciclovir, famotidine, aspirin, bactrim, erythropoietin, triiodothyronine.
Table II. Orthotopic xenogeneic heart transplantation experiments in baboons (from Reichart et al., mod.) 64.
1. Relative or absolute contraindications to mechanical circulatory support, e.g. restrictive or hypertrophic cardiomyopathy presence of a dysfunctional mechanical valve prosthesis or degenerated bio-prosthesis atrial or ventricular septal defect
2. High titres of broadly panel-reactive anti-HLA antibodies (high PRA) that do not cross-react with swine leukocyte antigens (SLA) of the donor animal
3. Chronic rejection after cardiac allotransplantation
4. Infants and children with complex congenital heart disease, e.g., after atrial correction of a transposition of the great arteries, single ventricle circulation after right ventricular Fontan (Norwood III) procedures
*Based on Chaban et al., 2023 108.
Table III. Potential indications for the initial clinical trials of pig heart transplantation*.

References

  1. Bauersachs J. Heart failure drug treatment: the fantastic four. Eur Heart J 2021;42:681-683. https://doi.org/10.1093/eurheartj/ehaa1012
  2. Ruck JM, Zhou AL, Zeiser LB, et al. Trends and three-year outcomes of hepatitis C virus-viremic donor heart transplant for hepatitis C virus-seronegative recipients. JTCVS Open 2022;12:269-279. https://doi.org/10.1016/j.xjon.2022.10.007
  3. Ayer A, Schroder JN, Casalinova S, et al. The future of heart procurement with donation after circulatory death: current practice and opportunities for advancement. J Heart Lung Transplant 2022;41:1385-1390. https://doi.org/10.1016/j.healun.2022.06.016
  4. Messer S, Rushton S, Simmonds L, et al. A national pilot of donation after circulatory death (DCD) heart transplantation within the United Kingdom. J Heart Lung Transplant 2023. https://doi.org/10.1016/j.healun.2023.03.006 [Epub Ahead of Print]
  5. Joshi Y, Scheuer S, Chew H, et al. Heart transplantation from DCD donors in Australia: lessons learned from the first 74 cases. Transplantation 2023;107:361-371. https://doi.org/10.1097/tp.0000000000004294
  6. Yuzefpolskaya M, Schroeder SE, Houston BA, et al. The Society of Thoracic Surgeons Intermacs 2022 Annual Report: Focus on the 2018 Heart Transplant Allocation System. Ann Thorac Surg 2023;115:311-327. https://doi.org/10.1016/j.athoracsur.2022.11.023
  7. Längin M, Mayr T, Reichart B, et al. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 2018;564:430-433. https://doi.org/10.1038/s41586-018-0765-z
  8. Reichart B, Längin M, Radan J, et al. Pig-to-non-human primate heart transplantation: the final step toward clinical xenotransplantation? J Heart Lung Transplant 2020;39:751-757. https://doi.org/10.1016/j.healun.2020.05.004
  9. Mohiuddin MM, Goerlich CE, Singh AK, et al. Progressive genetic modifications of porcine cardiac xenografts extend survival to 9 months. Xenotransplantation 2022;E12744. https://doi.org/10.1111/xen.12744
  10. Litovsky SH, Foote JB, Jagdale A, et al. Cardiac and pulmonary histopathology in baboons following genetically-engineered pig orthotopic heart transplantation. Ann Transplant 2022;27:E935338. https://doi.org/10.12659/aot.935338
  11. Griffith BP, Goerlich CE, Singh AK, et al. Genetically modified porcine-to-human cardiac xenotransplantation. N Engl J Med 2022;387:35-44. https://doi.org/10.1056/NEJMoa2201422
  12. Cooper DKC, Wang L, Kinoshita K, et al. Immunobiological barriers to pig organ xenotransplantation. EJT 2023;1:168-185. https://doi.org/10.57603/EJT-266
  13. Byrne G, Ahmad-Villiers S, Du Z, et al. B4GALNT2 and xenotransplantation: a newly appreciated xenogeneic antigen. Xenotransplantation 2018;25:E12394. https://doi.org/10.1111/xen.12394
  14. Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2019;4:eaau6298. https://doi.org/10.1126/sciimmunol.aau6298
  15. Phelps CJ, Koike C, Vaught TD, et al. Production of alpha 1,3-galactosyltransferase-deficient pigs. Science 2003;299:411-414. https://doi.org/10.1126/science.1078942
  16. Kwon DN, Lee K, Kang MJ, et al. Production of biallelic CMP-Neu5Ac hydroxylase knock-out pigs. Sci Rep 2013;3:1981. https://doi.org/10.1038/srep01981
  17. Lutz AJ, Li P, Estrada JL, et al. Double knockout pigs deficient in N-glycolylneuraminic acid and galactose alpha-1,3-galactose reduce the humoral barrier to xenotransplantation. Xenotransplantation 2013;20:27-35. https://doi.org/10.1111/xen.12019
  18. Estrada JL, Martens G, Li P, et al. Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/beta4GalNT2 genes. Xenotransplantation 2015;22:194-202. https://doi.org/10.1111/xen.12161
  19. Diamond LE, Quinn CM, Martin MJ, et al. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation 2001;71:132-142.
  20. Cozzi E, White DJ. The generation of transgenic pigs as potential organ donors for humans. Nat Med 1995;1:964-966. https://doi.org/10.1038/nm0995-964
  21. 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 U S A 1994;91:11153-11157.
  22. Galli C. Animal engineering for xenotransplantation: genome editing and cloning in pigs for xenotransplantation. EJT 2023;1:186-195. https://doi.org/10.57603/EJT-307
  23. Weiss EH, Lilienfeld BG, Müller S, et al. HLA-E/human beta2-microglobulin transgenic pigs: protection against xenogeneic human anti-pig natural killer cell cytotoxicity. Transplantation 2009;87:35-43. https://doi.org/10.1097/TP.0b013e318191c784
  24. Tena A, Kurtz J, Leonard DA, et al. Transgenic expression of human CD47 markedly increases engraftment in a murine model of pig-to-human hematopoietic cell transplantation. Am J Transplant 2014;14:2713-22. https://doi.org/10.1111/ajt.12918
  25. Ladowski JM, Hara H, Cooper DKC. The role of SLAs in xenotransplantation. Transplantation 2021;105:300-307. https://doi.org/10.1097/tp.0000000000003303
  26. Klymiuk N, van Buerck L, Bahr A, et al. Xenografted islet cell clusters from INSLEA29Y transgenic pigs rescue diabetes and prevent immune rejection in humanized mice. Diabetes 2012;61:1527-1532. https://doi.org/10.2337/db11-1325
  27. Bähr A, Kaser T, Kemter E, et al. Ubiquitous LEA29Y expression blocks T Cell co-stimulation but permits sexual reproduction in genetically modified pigs. PLoS One 2016;11:E0155676. https://doi.org/10.1371/journal.pone.0155676
  28. Buermann A, Petkov S, Petersen B, 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;25:E12387. https://doi.org/10.1111/xen.12387
  29. Nottle MB, Salvaris EJ, Fisicaro N, et al. Targeted insertion of an anti-CD2 monoclonal antibody transgene into the GGTA1 locus in pigs using FokI-dCas9. Sci Rep 2017;7:8383. https://doi.org/10.1038/s41598-017-09030-6
  30. Cowan PJ, Robson SC. Progress towards overcoming coagulopathy and hemostatic dysfunction associated with xenotransplantation. Int J Surg 2015;23:296-300. https://doi.org/10.1016/j.ijsu.2015.07.682
  31. Pierson RN, 3rd, Fishman JA, Lewis GD, et al. Progress toward cardiac xenotransplantation. Circulation 2020;142:1389-1398. https://doi.org/10.1161/circulationaha.120.048186
  32. Mohiuddin MM, Singh AK, Corcoran PC, et al. Chimeric 2C10R4 anti-CD40 antibody therapy is critical for long-term survival of GTKO.hCD46.hTBM pig-to-primate cardiac xenograft. Nat Commun 2016;7:11138. https://doi.org/10.1038/ncomms11138
  33. Iwase H, Ekser B, Satyananda V, et al. Pig-to-baboon heterotopic heart transplantation – exploratory preliminary experience with pigs transgenic for human thrombomodulin and comparison of three costimulation blockade-based regimens. Xenotransplantation 2015;22:211-220. https://doi.org/10.1111/xen.12167
  34. Wuensch A, Baehr A, Bongoni AK, et al. Regulatory sequences of the porcine THBD gene facilitate endothelial-specific expression of bioactive human thrombomodulin in single- and multitransgenic pigs. Transplantation 2014;97:138-147. https://doi.org/10.1097/TP.0b013e3182a95cbc
  35. Salvaris EJ, Moran CJ, Roussel JC, et al. Pig endothelial protein C receptor is functionally compatible with the human protein C pathway. Xenotransplantation 2020;27:E12557. https://doi.org/10.1111/xen.12557
  36. Ahrens HE, Petersen B, Herrmann D, et al. siRNA mediated knockdown of tissue factor expression in pigs for xenotransplantation. Am J Transplant 2015;15:1407-1414. https://doi.org/10.1111/ajt.13120
  37. Oropeza M, Petersen B, Carnwath JW, et al. Transgenic expression of the human A20 gene in cloned pigs provides protection against apoptotic and inflammatory stimuli. Xenotransplantation 2009;16:522-534. https://doi.org/10.1111/j.1399-3089.2009.00556.x
  38. Petersen B, Ramackers W, Lucas-Hahn A, et al. Transgenic expression of human heme oxygenase-1 in pigs confers resistance against xenograft rejection during ex vivo perfusion of porcine kidneys. Xenotransplantation 2011;18:355-368. https://doi.org/10.1111/j.1399-3089.2011.00674.x
  39. Hinrichs A, Kessler B, Kurome M, et al. Growth hormone receptor-deficient pigs resemble the pathophysiology of human Laron syndrome and reveal altered activation of signaling cascades in the liver. Mol Metab 2018;11:113-128. https://doi.org/10.1016/j.molmet.2018.03.006
  40. Hinrichs A, Riedel EO, Klymiuk N, et al. Growth hormone receptor knockout to reduce the size of donor pigs for preclinical xenotransplantation studies. Xenotransplantation 2021;28:E12664. https://doi.org/10.1111/xen.12664
  41. Goerlich CE, Griffith B, Hanna P, et al. The growth of xenotransplanted hearts can be reduced with growth hormone receptor knockout pig donors. J Thorac Cardiovasc Surg 2023;165:E69-E81. https://doi.org/10.1016/j.jtcvs.2021.07.051
  42. Hinrichs A, Renner S, Bidlingmaier M, et al. MECHANISMS IN ENDOCRINOLOGY: transient juvenile hypoglycemia in growth hormone receptor deficiency – mechanistic insights from Laron syndrome and tailored animal models. Eur J Endocrinol 2021;185:R35-R47. https://doi.org/10.1530/eje-21-0013
  43. Riedel EO, Hinrichs A, Kemter E, et al. Functional changes of the liver in the absence of growth hormone (GH) action – proteomic and metabolomic insights from a GH receptor deficient pig model. Mol Metab 2020;36:100978. https://doi.org/10.1016/j.molmet.2020.100978
  44. Shashikadze B, Franzmeier S, Hofmann I, et al. Structural and proteomic repercussions of growth hormone receptor deficiency on the pituitary gland: lessons from a translational pig model. J Neuroendocrinol 2023;E13277. https://doi.org/10.1111/jne.13277
  45. Das S, Koyano-Nakagawa N, Gafni O, et al. Generation of human endothelium in pig embryos deficient in ETV2. Nat Biotechnol 2020;38:297-302. https://doi.org/10.1038/s41587-019-0373-y
  46. Garry DJ, Garry MG. Interspecies chimeras and the generation of humanized organs. Circ Res 2019;124:23-25. https://doi.org/10.1161/circresaha.118.314189
  47. Zheng C, Ballard EB, Wu J. The road to generating transplantable organs: from blastocyst complementation to interspecies chimeras. Development 2021;148:dev195792. https://doi.org/10.1242/dev.195792
  48. Matsunari H, Watanabe M, Hasegawa K, et al. Compensation of disabled organogeneses in genetically modified pig fetuses by blastocyst complementation. Stem Cell Reports 2020;14:21-33. https://doi.org/10.1016/j.stemcr.2019.11.008
  49. Maeng G, Das S, Greising SM, et al. Humanized skeletal muscle in MYF5/MYOD/MYF6-null pig embryos. Nat Biomed Eng 2021;5:805-814. https://doi.org/10.1038/s41551-021-00693-1
  50. Nishimura T, Suchy FP, Bhadury J, et al. Generation of functional organs using a cell-competitive niche in intra- and Inter-species rodent chimeras. Cell Stem Cell 2021;28:141-149.E3. https://doi.org/10.1016/j.stem.2020.11.019
  51. Wolf E, Reichart B, Moretti A, et al. Designer pigs for xenogeneic heart transplantation and beyond. Dis Model Mech 2023;16. https://doi.org/10.1242/dmm.050177
  52. Cooper DK, Satyananda V, Ekser B, et al. Progress in pig-to-non-human primate transplantation models (1998-2013): a comprehensive review of the literature. Xenotransplantation 2014;21:397-419. https://doi.org/10.1111/xen.12127
  53. Mohiuddin MM, Reichart B, Byrne GW, et al. Current status of pig heart xenotransplantation. Int J Surg 2015;23:234-239. https://doi.org/10.1016/j.ijsu.2015.08.038
  54. Barnard CN, Losman JG, Curcio CA, et al. The advantage of heterotopic cardiac transplantation over orthotopic cardiac transplantation in the management of severe acute rejection. J Thorac Cardiovasc Surg 1977;74:918-924.
  55. Novitzky D, Cooper DK, Barnard CN. The surgical technique of heterotopic heart transplantation. Ann Thorac Surg 1983;36:476-482. https://doi.org/10.1016/s0003-4975(10)60492-4
  56. Cooper DK, Novitzky D, Becerra E, et al. Are there indications for heterotopic heart transplantation in 1986? A 2- to 11-year follow-up of 49 consecutive patients undergoing heterotopic heart transplantation. Thorac Cardiovasc Surg 1986;34:300-304. https://doi.org/10.1055/s-2007-1022159
  57. Hildebrandt A, Reichenspurner H, Gordon GD, et al. Heterotopic heart transplantation: mid-term hemodynamic and echocardiographic analysis – the concern of arteriovenous-valve incompetence. J Heart Transplant 1990;9:675-681; discussion 682
  58. Reichart B, Längin M, Denner J, et al. Pathways to clinical cardiac xenotransplantation. Transplantation 2021;105:1930-1943. https://doi.org/10.1097/tp.0000000000003588
  59. Reichenspurner H, Odell JA, Cooper DK, et al. Twenty years of heart transplantation at Groote Schuur Hospital. J Heart Transplant 1987;6:317-323.
  60. Abicht JM, Mayr T, Reichart B, et al. Pre-clinical heterotopic intrathoracic heart xenotransplantation: a possibly useful clinical technique. Xenotransplantation 2015;22:427-442. https://doi.org/10.1111/xen.12213
  61. Lower RR, Shumway NE. Studies on orthotopic homotransplantation of the canine heart. Surg Forum 1960;11:18-19.
  62. Cooper DK, Keogh AM, Brink J, et al. Report of the xenotransplantation Advisory Committee of the International Society for Heart and Lung Transplantation: the present status of xenotransplantation and its potential role in the treatment of end-stage cardiac and pulmonary diseases. J Heart Lung Transplant 2000;19:1125-1165. https://doi.org/10.1016/s1053-2498(00)00224-2
  63. Steen S, Paskevicius A, Liao Q, et al. Safe orthotopic transplantation of hearts harvested 24 hours after brain death and preserved for 24 hours. Scand Cardiovasc J 2016;50:193-200. https://doi.org/10.3109/14017431.2016.1154598
  64. Reichart B, Cooper DKC, Längin M, et al. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023;118:3499-3516. https://doi.org/10.1093/cvr/cvac180
  65. Waterworth PD, Dunning J, Tolan M, et al. Life-supporting pig-to-baboon heart xenotransplantation. J Heart Lung Transplant 1998;17:1201-1207.
  66. Schmoeckel M, Bhatti FN, Zaidi A, et al. Orthotopic heart transplantation in a transgenic pig-to-primate model. Transplantation 1998;65:1570-1577. https://doi.org/10.1097/00007890-199806270-00006
  67. Vial CM, Ostlie DJ, Bhatti FN, et al. Life supporting function for over one month of a transgenic porcine heart in a baboon. J Heart Lung Transplant 2000;19:224-229. https://doi.org/10.1016/s1053-2498(99)00099-6
  68. Byrne GW, Du Z, Sun Z, et al. Changes in cardiac gene expression after pig-to-primate orthotopic xenotransplantation. Xenotransplantation 2011;18:14-27. https://doi.org/10.1111/j.1399-3089.2010.00620.x
  69. Cleveland DC, Jagdale A, Carlo WF, et al. The genetically engineered heart as a bridge to allotransplantation in infants just around the corner? Ann Thorac Surg 2022;114:536-544. https://doi.org/10.1016/j.athoracsur.2021.05.025
  70. Yamamoto T, Iwase H, Patel D, et al. Old World Monkeys are less than ideal transplantation models for testing pig organs lacking three carbohydrate antigens (Triple-Knockout). Sci Rep 2020;10:9771. https://doi.org/10.1038/s41598-020-66311-3
  71. Yamamoto T, Hara H, Iwase H, et al. The final obstacle to successful pre-clinical xenotransplantation? Xenotransplantation 2020;27:E12596. https://doi.org/10.1111/xen.12596
  72. Cui Y, Yamamoto T, Raza SS, et al. Evidence for GTKO/4GalNT2KO pigs as the preferred organ-source for old world nonhuman primates as a preclinical model of xenotransplantation. Transplant Direct 2020;6:E590. https://doi.org/10.1097/txd.0000000000001038
  73. Iwase H, Jagdale A, Yamamoto T, et al. Evidence suggesting that deletion of expression of N-glycolylneuraminic acid (Neu5Gc) in the organ-source pig is associated with increased antibody-mediated rejection of kidney transplants in baboons. Xenotransplantation 2021;28:E12700. https://doi.org/10.1111/xen.12700
  74. Foote JB, Jagdale A, Yamamoto T, et al. Histopathology of pig kidney grafts with/without expression of the carbohydrate Neu5Gc in immunosuppressed baboons. Xenotransplantation 2021;28:E12715. https://doi.org/10.1111/xen.12715
  75. Cooper DKC, Hara H, Iwase H, et al. Justification of specific genetic modifications in pigs for clinical organ xenotransplantation. Xenotransplantation 2019;26:E12516. https://doi.org/10.1111/xen.12516
  76. Montgomery RA, Stern JM, Lonze BE, et al. Results of two cases of pig-to-human kidney xenotransplantation. N Engl J Med 2022;386:1889-1898. https://doi.org/10.1056/NEJMoa2120238
  77. Porrett PM, Orandi BJ, Kumar V, et al. First clinical-grade porcine kidney xenotransplant using a human decedent model. Am J Transplant 2022;22:1037-1053. https://doi.org/10.1111/ajt.16930
  78. Patel NG, Langmaid V. Genetically modified pig heart transplanted into deceased recipient, researchers say. Vol. 2023, CNN, 2022.
  79. Moazami N, Stern JM, Khalil K, et al. Pig-to-human heart xenotransplantation in two recently deceased human recipients. Nat Med 2023;Jul 24. https://doi.org/10.1038/s41591-023-02471-9 [Epub Ahead of Print]
  80. Novitzky D. Electrocardiographic, hemodynamic and endocrine changes occurring during experimental brain death in the Chacma baboon. Heart Transpl 1984;4:63-69.
  81. Shu S, Ren J, Song J. Cardiac xenotransplantation: a promising way to treat advanced heart failure. Heart Fail Rev 2022;27:71-91. https://doi.org/10.1007/s10741-020-09989-x
  82. Byrne GW, McGregor CG. Cardiac xenotransplantation: progress and challenges. Curr Opin Organ Transplant 2012;17:148-154. https://doi.org/10.1097/MOT.0b013e3283509120
  83. Längin M, Reichart B, Steen S, et al. Cold non‐ischemic heart preservation with continuous perfusion prevents early graft failure in orthotopic pig‐to‐baboon xenotransplantation. Xenotransplantation 2021;28:E12636.
  84. Bühler L, Basker M, Alwayn IP, et al. Coagulation and thrombotic disorders associated with pig organ and hematopoietic cell transplantation in nonhuman primates. Transplantation 2000;70:1323-1331. https://doi.org/10.1097/00007890-200011150-00010
  85. Samy KP, Butler JR, Li P, et al. The role of costimulation blockade in solid organ and islet xenotransplantation. J Immunol Res 2017;2017:8415205. https://doi.org/10.1155/2017/8415205
  86. Lui JC, Baron J. Mechanisms limiting body growth in mammals. Endocr Rev 2011;32:422-440. https://doi.org/10.1210/er.2011-0001
  87. Penzo-Méndez AI, Stanger BZ. Organ-size regulation in mammals. Cold Spring Harb Perspect Biol 2015;7:A019240 https://doi.org/10.1101/cshperspect.a019240
  88. Soin B, Ostlie D, Cozzi E, et al. Growth of porcine kidneys in their native and xenograft environment. Xenotransplantation 2000;7:96-100. https://doi.org/10.1034/j.1399-3089.2000.00046.x
  89. Tanabe T, Watanabe H, Shah JA, et al. Role of intrinsic (graft) versus extrinsic (host) factors in the growth of transplanted organs following allogeneic and xenogeneic transplantation. Am J Transplant 2017;17:1778-1790. https://doi.org/10.1111/ajt.14210
  90. Ma D, Hirose T, Lassiter G, et al. Kidney transplantation from triple-knockout pigs expressing multiple human proteins in cynomolgus macaques. Am J Transplant 2022;22:46-57. https://doi.org/10.1111/ajt.16780
  91. Fishman JA. Infectious disease risks in xenotransplantation. Am J Transplant 2018;18:1857-1864. https://doi.org/10.1111/ajt.14725
  92. Fishman JA. Prevention of infection in xenotransplantation: designated pathogen-free swine in the safety equation. Xenotransplantation 2020;27:E12595. https://doi.org/10.1111/xen.12595
  93. Denner J. Sensitive detection systems for infectious agents in xenotransplantation. Xenotransplantation 2020;E12594. https://doi.org/10.1111/xen.12594
  94. Denner J, Längin M, Reichart B, et al. Impact of porcine cytomegalovirus on long-term orthotopic cardiac xenotransplant survival. Sci Rep 2020;10:17531. https://doi.org/10.1038/s41598-020-73150-9
  95. Halecker S, Hansen S, Krabben L, et al. How, where and when to screen for porcine cytomegalovirus (PCMV) in donor pigs for xenotransplantation. Sci Rep 2022;12:21545. https://doi.org/10.1038/s41598-022-25624-1
  96. Fischer N, Gulich B, Keßler B, et al. PCR and peptide based PCMV detection in pig – development and application of a combined testing procedure differentiating newly from latent infected pigs. Xenotransplantation 2023;E12803. https://doi.org/10.1111/xen.12803
  97. Denner J, Tönjes RR. Infection barriers to successful xenotransplantation focusing on porcine endogenous retroviruses. Clin Microbiol Rev 2012;25:318-343. https://doi.org/10.1128/cmr.05011-11
  98. Harrison I, Takeuchi Y, Bartosch B, et al. Determinants of high titer in recombinant porcine endogenous retroviruses. J Virol 2004;78:13871-13879. https://doi.org/10.1128/jvi.78.24.13871-13879.2004
  99. Denner J, Specke V, Thiesen U, et al. Genetic alterations of the long terminal repeat of an ecotropic porcine endogenous retrovirus during passage in human cells. Virology 2003;314:125-133. https://doi.org/10.1016/s0042-6822(03)00428-8
  100. Denner J. Why was PERV not transmitted during preclinical and clinical xenotransplantation trials and after inoculation of animals? Retrovirology 2018;15:28. https://doi.org/10.1186/s12977-018-0411-8
  101. Wynyard S, Nathu D, Garkavenko O, et al. Microbiological safety of the first clinical pig islet xenotransplantation trial in New Zealand. Xenotransplantation 2014;21:309-323. https://doi.org/10.1111/xen.12102
  102. 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. https://doi.org/10.1016/j.virusres.2016.08.012
  103. Patience C, Takeuchi Y, Weiss RA. Infection of human cells by an endogenous retrovirus of pigs. Nat Med 1997;3:282-286. https://doi.org/10.1038/nm0397-282
  104. Niu D, Wei HJ, Lin L, et al. Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science 2017;357:1303-1307. https://doi.org/10.1126/science.aan4187
  105. Fu Y, Foden JA, Khayter C, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol 2013;31:822-826. https://doi.org/10.1038/nbt.2623
  106. Thomas M, Burgio G, Adams DJ, et al. Collateral damage and CRISPR genome editing. PLoS Genet 2019;15:E1007994 https://doi.org/10.1371/journal.pgen.1007994
  107. Marks P, Solomon S. Clarifying US regulations on xenotransplantation. Nat Biotechnol 2021;39:1500-1501. https://doi.org/10.1038/s41587-021-01144-7
  108. Chaban R, Cooper DKC, Pierson RN, 3rd. Pig heart and lung xenotransplantation: present status. J Heart Lung Transplant 2022;41:1014-1022. https://doi.org/10.1016/j.healun.2022.04.010
  109. John M, Bailey LL. Neonatal heart transplantation. Ann Cardiothorac Surg 2018;7:118-125. https://doi.org/10.21037/acs.2018.01.05
  110. Morales DLS, Rossano JW, VanderPluym C, et al. Third Annual Pediatric Interagency Registry for Mechanical Circulatory Support (Pedimacs) Report: preimplant characteristics and outcomes. Ann Thorac Surg 2019;107:993-1004. https://doi.org/10.1016/j.athoracsur.2019.01.038
  111. Rohde S, Sandica E, Veen K, et al. Outcomes in small children on Berlin Heart EXCOR support: age and body surface area as clinical predictive factors. European Journal of Cardio-Thoracic Surgery 2022;63:Ezac516. https://doi.org/10.1093/ejcts/ezac516

Downloads

Authors

Eckhard Wolf - Gene Centre and Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians- Universität München, Munich, Germany https://orcid.org/0000-0002-0430-9510

Michael Schmoeckel - Department of Cardiac Surgery, Klinikum Großhadern, Ludwig-Maximilians-Universität München, Munich, Germany

Bruno Reichart - Walter Brendel Centre for Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany https://orcid.org/0000-0003-2859-3664

How to Cite
[1]
Wolf, E., Schmoeckel, M. and Reichart, B. 2023. Cardiac xenotransplantation – from bench to bedside. European Journal of Transplantation. 1, 3 (Oct. 2023), 192–206. DOI:https://doi.org/10.57603/EJT-305.
  • Abstract viewed - 167 times
  • PDF downloaded - 33 times