CC BY-NC-ND 4.0 · Thorac Cardiovasc Surg
DOI: 10.1055/a-2235-8854
Review Article

Current Status of Cardiac Xenotransplantation: Report of a Workshop of the German Heart Transplant Centers, Martinsried, March 3, 2023

Michael Schmoeckel
1   Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
,
Matthias Längin
2   Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Bruno Reichart
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Jan-Michael Abicht
2   Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Martin Bender
2   Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Sebastian Michel
1   Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Christine-Elena Kamla
1   Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
,
Joachim Denner
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
4   Institut für Virologie, Fachbereich für Veterinärmedizin, Freie Universität Berlin, Berlin, Germany
,
Ralf Reinhard Tönjes
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
5   Paul-Ehrlich-Institut, Langen, Germany
,
Reinhard Schwinzer
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
6   Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
,
Georg Marckmann
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
7   Institut für Ethik, Geschichte und Theorie der Medizin, LMU München, Germany
,
Eckhard Wolf
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
8   Lehrstuhl für Molekulare Tierzucht und Biotechnologie, Genzentrum der LMU München, Germany
,
Paolo Brenner
1   Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
3   DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
,
Christian Hagl
1   Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
9   DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), Partner Site Munich, Germany
› Author Affiliations
 

Abstract

This report comprises the contents of the presentations and following discussions of a workshop of the German Heart Transplant Centers in Martinsried, Germany on cardiac xenotransplantation. The production and current availability of genetically modified donor pigs, preservation techniques during organ harvesting, and immunosuppressive regimens in the recipient are described. Selection criteria for suitable patients and possible solutions to the problem of overgrowth of the xenotransplant are discussed. Obviously microbiological safety for the recipient and close contacts is essential, and ethical considerations to gain public acceptance for clinical applications are addressed. The first clinical trial will be regulated and supervised by the Paul-Ehrlich-Institute as the National Competent Authority for Germany, and the German Heart Transplant Centers agreed to cooperatively select the first patients for cardiac xenotransplantation.


#

Introduction

Novel medical treatments for advanced heart failure have proven to be highly effective.[1] However, in cases where all other treatment options have been exhausted, heart transplantation (HTx) remains the preferred approach for patients with end-stage heart disease, offering a strong likelihood of extended life in good health. Unfortunately, the shortage of available human organs for transplantation has led to extensive waiting lists, with annual demand far exceeding the actual number of transplants performed.

Exploring alternative solutions, researchers have considered taking increased risks in donor selection, such as the acceptance of hepatitis C-positive brain-dead persons.[2] Another avenue under investigation is donation after circulatory death (DCD);[3] [4] [5] however, DCD is not permitted in Germany.

At present, mechanical assist devices serve as the primary alternative to HTx, but these devices come with a high complication rate and offer only moderate improvements in patients' quality of life. The 1- and 5-year survival rates for patients on these devices are 83 and 52%, respectively, which are significantly worse when compared to allogeneic heart transplants. After implantation of assist devices, hospital readmission rates are high, primarily due to infections and bleeding events, with 36 and 68% occurring at 3 and 12 postoperative months, respectively. The main cause of death in these cases is withdrawal of care.[6]

Encouragingly, significant progress has been made in the field of pig-to-primate cardiac xenotransplantation. This progress is attributed to genetically modified (GM) donor pigs, improved preservation techniques, optimized transplantation models, and effective immunosuppressive regimens.[7] [8] [9] [10] A milestone was reached in January 2022 when the first compassionate use xenotransplantation (XT) of a GM pig heart into a patient with terminal heart failure took place at the University of Maryland, Baltimore.[11] [12] Although the patient passed away after 2 months due to various complications, this achievement marked a crucial step in demonstrating the feasibility of clinical cardiac XT by sustaining normal heart function for over 45 days.

Subsequent to this, in June and July 2022, two orthotopic HTx were performed at New York University using the same 10 × GM pigs (United Therapeutics/Revivicor, Blacksburg, Virginia, United States) as donors, allowing the hearts to beat for 72 hours without signs of rejection.[13] It is worth noting that while these short-term experiments provide valuable insights, the unstable condition of brain-dead recipients limits longer observation times.[14] [15] For more reliable data, XT must be conducted in living patients.

On September 20, 2023, the Baltimore group performed a second pig-to-human heart transplant in a 58-year-old patient ineligible for an allogeneic heart transplant due to severe peripheral vascular disease and complications with internal bleeding. The patient died 40 days after transplant presumably due to initial signs of rejection.


#

Genetic Modification of Source Pigs to Alleviate the Pathobiology of Pig Heart Xenotransplantation

The complexity of the pathobiology in organ XT surpasses that of allotransplantation, with innate immune responses playing a more prominent role ([Table 1]).[16] In essence, during infancy, both humans and nonhuman primates (NHPs) produce antibodies that react to carbohydrate antigens present on the surface of unaltered pig cells. Consequently, when a normal pig organ is transplanted into a human or baboon, these antibodies quickly attach to the vascular endothelial cells of the graft. This triggers the activation of the complement cascade and attracts leukocytes that infiltrate the porcine heart through various mechanisms, ultimately leading to the rejection of the graft within minutes to hours. This rapid rejection, dependent on antibodies, is known as “hyperacute rejection” and is characterized by histopathological features such as venous thrombosis, loss of vascular integrity, interstitial hemorrhage, edema, and the infiltration of innate immune cells.

Table 1

Genetic modifications of clinically available genetically modified pigs

Genetic modifications

Rationale

Reference

Knockout of α-1,3-galactosyltransferase (GGTA1-KO)

Knockout to prevent hyperacute rejection, as galactose-α-(1,3)-galactose (αGal) is the major xenoantigen causing hyperacute rejection in pig-to-human/primate xenotransplantation

[19]

Knockout of cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH-KO)

CMAH is the enzyme responsible for the synthesis of Neu5Gc. Knockout removes the major non-αGal xenoreactive antigen, against which humans have an innate immune response

[20] [21]

Knockout of β-1,4-N-acetyl-galactosaminyl transferase 2 (B4GALNT2-KO)

Removes the glycan resembling the human Sd(a), against which humans/primates develop preformed antibodies

[22]

Expression of human CD46[a]

CD46 is a complement regulatory protein (CRP), downregulating complement activation. Express to suppress complement activation

[23]

Expression of human CD55[a]

CD55 is a CRP, similar role as CD46

[24]

Expression of human CD59[a]

CD59 is a CRP, similar role as CD46

[25]

Expression of human thrombomodulin (hTBM)

Human TBM is an anticoagulant protein, necessary to overcome coagulation incompatibilities after pig-to-primate/human xenotransplantations

[29]

Expression of human endothelial protein C receptor (hEPCR)b

Human EPCR is an anticoagulant protein, supports the formation of the TBM-thrombin complex

[30]

a Probably one CPRP (complement pathway regulatory protein) is sufficient.


b additional hEPCR to hTBM is not necessary.


Hyperacute (and subsequently acute) rejections of pig organs in humans or NHPs primarily occur due to preexisting antibodies targeting galactose-α-(1,3)-galactose (αGal). Humans also have 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.[17] [18]

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),[19] cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH),[20] [21] and β-1,4-N-acetyl-galactosaminyl transferase 2 (B4GALNT2)/B4GALNT2-like (B4GALNT2L)[22] genes were generated, resulting in what is commonly referred to as “triple-knock-out (TKO) pigs.” ([Fig. 1])

Zoom Image
Fig. 1 Mechanisms of hyperacute xenograft rejection and strategies to overcome them. Created with BioRender.com.

However, complement activation can also occur through pathways unrelated to antibody binding, such as ischemia–reperfusion injury. To address this issue, additional human complement pathway regulatory (inhibitory) proteins (CPRPs), namely CD46,[23] CD55,[24] and CD59,[25] have been expressed in pigs by genetic engineering. Organs derived from animals with transgenic expression of one or more human CPRPs show a substantial level of protection against further complement-mediated injury in humans or NHPs. When combined with TKO pigs, these “humanized” porcine organs exhibit significantly reduced cell injury.[26]

Dysregulation of the coagulation pathway represents another facet of the pathobiology associated with XT of pig organs.[27] [28] This dysregulation is influenced by several factors, including the previously mentioned immune responses, which promote inflammation and vascular damage, ultimately leading to a procoagulant state in the pig's endothelium. A significant contributing factor to this issue is the molecular incompatibility between coagulation regulators in pigs and those in humans or NHPs, leading to thrombotic microangiopathy even when using clinically approved anticoagulation therapy. Physiologically, thrombomodulin (TBM) on endothelial cells binds thrombin from the circulation, and the TBM–thrombin complex—with the help of an endothelial protein C receptor (EPCR)—activates protein C that has an anticoagulation effect ([Fig. 2]). After organ xenotransplantation, porcine TBM on the transplant's endothelial cells can bind human or NHP thrombin, but the complex appears not to effectively activate human or NHP protein C. As a consequence, harmful fibrin clots form within the capillary system of the donor organ, finally leading to thrombotic microangiopathy. This can be effectively prevented by using source pigs expressing human TBM on their vascular endothelial cells.[29]

Zoom Image
Fig. 2 Activation of protein C by the thrombin–thrombomodulin complex after allogeneic (top) and xenogeneic transplantation (bottom). EPCR = endothelial protein C receptor; PC = protein C; PCa = activated protein C; TBM = thrombomodulin; Va = activated factor V; VIIIa = activated factor VIII. Created with BioRender.com.

Despite the compatibility of the porcine EPCR in facilitating protein C activation in the human or NHP protein C pathway, transgenic pigs have been developed to express human EPCR.[30] This modification aims to elevate EPCR levels and, consequently, may enhance protective thromboregulation.


#

Further Prerequisites for Successful Xenotransplantation

Nonischemic Perfusion Technique of the Porcine Donor Heart

For over two decades, preclinical outcomes following orthotopic xenogeneic HTx were inconsistent, with a perioperative mortality rate ranging from 40 to 60%.[31] This unpredictability was attributed to “perioperative cardiac xenograft dysfunction” (PCXD), believed to be linked to ischemia/reperfusion injury.[17] [32] Porcine hearts are notably less resistant to ischemia compared to human hearts. Since 2015, PCXD has been consistently prevented through continuous, nonischemic perfusion of grafts with an 8°C hyperoncotic, oxygenated cardioplegic (Steen) solution containing erythrocytes, nutrients, and hormones.[33] [34] This perfusion preservation technique was also utilized in the already mentioned first clinical case at the University of Maryland, Baltimore.[11] [12]


#

Development of a Nonnephrotoxic Immunosuppressive Regimen with CD40 or CD154 Costimulation Blockade

Initial pig-to-baboon cardiac XT studies employed conventional immunosuppressive regimens without long-term success. Since 2000, costimulation blockade, initially with anti-CD154 monoclonal antibodies (mAb), has been applied.[35] [36] However, due to thrombotic complications in humans, a chimeric anti-CD40 mAb (2C10)-based regimen was introduced instead, contributing to longer cardiac xenograft survivals in baboons.[7] [10] [37] In the recent Maryland case, a humanized version of the anti-CD40 antibody (KPL-404, Kiniksa Pharmaceuticals, Lexington, MA, United States) was used, along with cortisone, ATG, and rituximab (anti-CD20). Maintenance included tapering down cortisone, mycophenolate mofetil, and/or rapamycin for graft overgrowth control.[11]


#

Postimplantation Growth Control of the Xenoheart

Pig breeds used for XT experiments, such as German Landrace or Large White, weigh outgrown 200 to 300 kg, resulting in proportionately large hearts of approximately 1 kg, much too big for a human recipient, not to mention a baboon weighing between 15 and 20 kg. While it was previously believed that grafts would adapt to recipient growth regulation, recent findings[7] [38] indicate that donor organ growth is genetically regulated: the porcine donor heart behaves as if it is still in a fast-growing pig's body; additionally, elevated afterload in baboon recipients causes concentric myocardial hypertrophy of juvenile porcine grafts. In combination, these intrinsic (donor-specific) and extrinsic (recipient-specific) factors led to extensive cardiac overgrowth and the development of dynamic outflow tract obstruction in preclinical experiments.[38] This “overgrowth” phenomenon was also observed after xenogeneic kidney transplantation experiments.[39] [40] Strategies to prevent cardiac overgrowth in a preclinical setting include lowering blood pressure, early discontinuation of cortisone, and treatment with sirolimus, a ubiquitous growth inhibitor.

In the future, smaller donor animal breeds, such as Auckland Island pigs from New Zealand, with a weight range of 70 to 90 kg, may be preferred for clinical applications, and consequently, a small porcine endogenous retrovirus-C (PERV-C) free herd near Munich, within the experimental LMU-farm, has been established ([Fig.3]).

Zoom Image
Fig. 3 Auckland Island pigs in the Center for Innovative Medical Models (CiMM; www.lmu.de/cimm/) at LMU Munich.

#
#

Identifying “Low-Risk” Donor–Recipient Combinations for Clinical Xenotransplantation

The level of histocompatibility between donor and recipient is an important parameter determining the risk for rejection in the course after allo- and xenotransplantation. High titers of antibodies to donor antigens in a prospective recipient are associated with an enhanced risk for antibody-mediated rejection. The existence of antidonor antibodies is usually demonstrated in vitro by incubating the serum of a prospective recipient with cells from a prospective donor (cross-matching). Antibody binding to donor cells can be visualized by flow cytometry or by antibody-induced complement activation resulting in cytotoxicity.[41] [42] An assessment of the level of anti-pig antibodies by previous cross-match studies has been performed in recent pig-to-human heart and kidney xenotransplantations in deceased human recipients.[13] [43] Incompatibility between donor and recipient is not only the reason for the deleterious effects of antibodies, but in addition, it also influences the intensity of T cell responses against a transplant. Thus, high numbers of human leukocyte antigen (HLA) class-I and/or class-II mismatches between donor and recipient have been associated with a poorer outcome in the long-term course after kidney and heart allotransplantation.[44] [45] [46]

Preformed IgM and IgG antibodies directed against the three carbohydrate antigens on porcine cells mentioned above are present in all individuals.[47] [48] [49] Binding of these antibodies to their targets is the key event to induce hyperacute rejection of xenografts. With the generation of TKO pigs,[22] it could be revealed that 30% of patients have very low or no IgM and IgG binding to TKO peripheral blood mononuclear cells.[50] Based on these findings, it was recommended to use pigs as donors for initial clinical studies where the TKO platform is combined with additional genetic modifications.[51] [52]

Nevertheless, the question arises whether a low-risk organ can also be provided for those 70% of recipients having a positive cross-match with TKO cells.[50] A possible solution for this problem was provided by the characterization of the specificity of anti-TKO antibodies. These studies revealed that some of the residual antibody binding to TKO cells is mediated by anti-HLA antibodies which cross-react on porcine MHC molecules (SLA, swine leucocyte antigen[50] [53] [54]). The existence of antibodies in human serum with reactivity to porcine SLA is also supported by recent data characterizing the antibody repertoire against TKO cells.[55] To define the level of anti-SLA antibodies in prospective recipients of xenografts, flow cytometry cross-match could be performed using genetically engineered cells expressing individual SLA-I orSLA-II antigens.[56] [57] Based on the observed reactivity patterns (e.g., dominance of anti-SLA antibodies) organ-source pigs with genetic modifications (e.g. SLA-I knockout) could be selected to avoid damaging effects of anti-SLA antibodies.[58] Organs from pigs expressing neither SLA-I nor SLA-II[59] may be of further advantage for recipients with antibodies against a broad spectrum of different SLA alleles.

Detailed characterization of some anti-SLA-I and -SLA-II antibodies revealed that single-amino acid epitopes are responsible for antibody cross-reactivity with HLA and SLA. This observation could be of great relevance for clinical XT because we also found that mutation of the amino acid eliminated antibody binding.[53] [57] It has been discussed that SLA-I/II mutated xenografts may be sufficient to prevent anti-SLA antibody binding[60] instead of using grafts with complete absence of SLA. For individuals who have antibodies directed to other carbohydrates than αGal, Neu5Gc, and Sd(a), there is currently no genetically engineered pig available to avoid the binding of such antibodies. Thus, it would be safer to exclude these patients from initial studies ([Fig. 4]).

Zoom Image
Fig. 4 Flowchart to achieve optimal donor–recipient-combinations for clinical xenotransplantation. Cross-matching of sera from potential recipients should be performed by using cells from TKO pigs lacking αGal, Neu5Gc, and Sd(a). Negative cross-match: Organs from TKO donor pigs combined with additional genetic modifications will be used as previously explained. In case of a positive cross-match, further characterization of antipig antibodies will be required. Recipients expressing anti-SLA antibodies may be transplanted with organs from SLA-I/II knockout pigs or pigs expressing mutated SLA to avoid antibody binding (both on “TKO plus” platform). Cross-matching may be complemented by HLA–SLA matching to identify donor–recipient combinations with low level T cell reactivity.[94]

#

Microbiological Safety

XT may be associated with the transmission of porcine microorganisms, for example, viruses, bacteria, fungi, and parasites.[61] Whereas bacteria, fungi, and parasites can be easily eliminated from the donor pigs, the situation with viruses is more complicated but can be solved.

It is important to remember in this context, that during allotransplantation, human viruses such as human immunodeficiency virus 1, human cytomegalovirus, rabies virus, and others have been transmitted to the recipient due to lack of time and methods of detection. In contradistinction, pigs as donor animals can be screened for viruses carefully long before surgery, and consequently, XT will be safer compared with allotransplantation.

Whereas the total number of viruses in pigs—their virome—is high,[62] the actual number of viruses able to infect humans and, ultimately cause diseases in humans, is still unknown. Diseases induced by animal viruses in humans after XT are called xenozoonoses.[63] The risks of infections should be negligible if state-of-the-art knowledge is applied.

First preclinical trials in nonhuman primates, and first clinical trials transplanting pig tissues into more than 200 human recipients, demonstrated that the number of xenozoonotic viruses was low.[64] At present, the hepatitis E virus genotype 3 is most important. It is transmitted to humans by eating undercooked pork or by contact with pigs. In immunosuppressed individuals, chronic infections are induced, preexisting liver diseases are aggravated.[65] [66] A herpes virus type, the porcine cytomegalovirus (PCMV), is another possibly dangerous microorganism. PCMV is actually a porcine roseolovirus (PRV) related to the human herpesviruses 6 and 7.[67] Of note PCMV/PRV is not closely related to the human cytomegalovirus, which causes major pulmonary complications when transmitted during allotransplantation.[68] Until recently PCMV/PRV was shown to be harmful only for NHPs: transmission of the virus to baboons and rhesus monkeys significantly reduced the survival time of the transplant.[69] However, when a GM pig heart was transplanted into the first patient in Baltimore, PCMV/PRV was transmitted and obviously contributed to his death.[11] Although there is no evidence that PCMV/PRV infects NHP and human cells, consumptive coagulopathy and multiorgan failure were observed in the infected transplanted baboons and the patient. The levels of interleukin-6, tumor necrosis factor α, tissue plasminogen activator, and plasminogen activator inhibitor 1 were significantly increased when compared to noninfected baboons.[69] The virus obviously interacts directly with the recipient's immune system and endothelial cells. Therefore, a major lesson learned from the study in Baltimore is that viral safety is pivotal for the success of XT and that testing should be done with assays of the highest quality and following an optimal strategy.[70]

Since there are no antivirals or vaccines available, a preventive strategy was developed by the Munich group: both viruses, PCMV/PRV[71] [72] and HEV have been eliminated from the pig facility in Munich by applying “early weaning,” which means, the piglets did not drink milk from their mother which may transmit the viruses during that time via its snout.

This strategy cannot be used to eliminate the risk of PERVs, which are integrated in the genome of all pigs[73]: PERV-A and -B are present in all pigs, but they are able to infect human cells only in vitro (under experimental conditions), PERV-C infects only porcine cells and is indeed not present in all pigs: in Munich imported Auckland-Island pigs were selected and were PERV-C free. Why is the absence of PERV-C so important? PERV-A and -C can recombine and the resulting recombinants can infect human cells.[74] [75] [76] Until now PERV transmission has never been observed neither in preclinical nor clinical XT studies.[77]


#

Ethical Considerations

As a novel treatment strategy, XT raises several ethical issues[78] [79] [80] [81] which require thorough scrutiny before entering a first clinical trial. The ethical assessment should proceed in a transparent and structured manner.[82] [Table 2] shows relevant criteria for the ethical evaluation and its justifications. While it is beyond the scope of this report to give a full assessment of all criteria, we highlight how the most important ethical concerns can be addressed appropriately.

Table 2

Criteria for the ethical evaluation of clinical xenotransplantation with their justification

Evaluation criteria

Ethical justification

Expected patient benefit of XT

Principle of beneficence

Potential harm of XT for patient

Principle of nonmaleficence

Promotion and respect of patient autonomy

Principle respect for autonomy

Potential harm for third-parties

Principle of nonmaleficence

Fair access to XT

Principle of justice

Efficiency of XT

Principle of utility maximization

Burden for animals as organ source

Animal welfare

Abbreviation: XT, xenotransplantation.


First of all, the heart XT recipients must have a benefit with sufficient certainty. Due to the persistent shortage of human donor organs, patients with terminal heart failure are in high need of an allograft. Some even die on the waiting list or experience detrimental side effects. In contradistinction, the risk of hyper-acute/humoral rejection of a cardiac xenograft could be reduced significantly due to multiple genetic modifications of the donor pigs.[7] [18] In 2000, the Xenotransplantation Advisory Committee of the International Society of Heart and Lung Transplantation set up criteria, when the first clinical trial should be considered.[83] The required preclinical results have been met: consistent survival of two-third of the life-supporting porcine heart replacements in NHPs, in good health for up to a minimum of 3 months (has recently been extended for 6 months, or in single case longer).[7] [8] [10] [84]

Taken together so far, a heart XT can be expected to have a rather large benefit with sufficient certainty for patients with terminal heart failure, given the highly unmet need for human donor hearts. And, the higher quality of xenografts compared to an average allograft from a brain-dead donor is an additional benefit, also the elective planning of the XT.

On the contrary, the risk of potential harm of the XT, especially the risk of xenogeneic infections, could substantially be reduced over the last years[61]: the donor pigs are screened with highly sensitive methods to prevent transmissions of xenogeneic viruses. A transmission of PERVs has never been observed, neither in preclinical nor in clinical studies.[61] [77] If sufficiently sensitive tests are used, the risk of transmission of other viruses, like PCMV, can also be controlled sufficiently.[85] With appropriate sensitive screening for xenogeneic infections, the potential harm for third parties, hospital staff, and close relatives does not appear to represent an obstacle from an ethical perspective.

Due to the multiple genetic modifications, XT patients may need less aggressive immunosuppressive (even nonnephrotoxic) treatment and may therefore suffer less side-effects. Negative psychological effects of XT cannot be excluded completely, but appear rather unlikely: potential xenograft recipients are more concerned with the benefit–risk ratio than the source of the graft.[86] Nevertheless, XT patients should receive appropriate psychological support.[81]

Given the novel aspects of the treatment strategy, promoting and respecting patient autonomy must play an important role in the first XT clinical trials. Patients should especially be informed about the expected benefits and risks of a heart XT compared to allotransplantation. While some experts suggest that patients who do not have access to an allotransplant should primarily be selected for a first XT trial,[81] [83] participation should also be considered for patients who are on the transplant waiting list and therefore have the (later) option to receive a human allograft in case a xenograft fails (bridge-to-allotransplantation[84] [86] [87]). These patients would ultimately have a real choice between waiting for an allograft and receiving a xenograft—which could foster their autonomous decision about participating in a first-in-human XT trial.

Overall, heart XT seems to have a considerable expected benefit for terminal heart failure patients, while the potential risks appear comparatively low. While not all uncertainties can be eliminated in preclinical studies, first-in-human XT pivotal (pilot) trials seem to be justified according to the expected benefit–harm ratio. However, the benefits and risks of such a regulated study (in contradistinction to the unregulated compassionate use case of the two Baltimore cases[11] [12]) must be documented thoroughly.[88] The risk of the transmission of xenogeneic infections seems to be manageable.


#

Regulatory Aspects

In the European Union (EU), guidelines and ordinances on advanced therapy medicinal products (ATMP), pharmacovigilance, and clinical trials form a regulatory framework for XT. The framework adequately protects the fundamental rights of both animals as donors and humans as recipients of organs, tissues, and cells. Furthermore, in the 27 EU member states, national laws may be implemented, such as the German AMG (Arzneimittelgesetz, Medicinal Products Act).

The ATMP regulation on XT displays some limitations in regard to animal organs, which are not explicitly mentioned, even though they are (in this case) derived from GM animals. The definition of somatic cell therapeutics, as well that of tissue-engineered products of animal origin, is based on tissues or cells; however, it excludes organs. Naturally, organs derived from GM animals contain tissues and cells. To this end, the European Medicines Agency (EMA, Amsterdam, Netherlands) has published the guideline on xenogeneic cell-based medicinal products.

Central elements of the ATMP regulation include

  1. designation of the EMA to grant marketing authorization for XT products within the EU

  2. requirement for xenograft traceability from creation through clinical use and ultimate disposition, and

  3. hospital exemption for medicinal products that are not routinely prepared.

In the EU, regulatory pathways to yield marketing authorizations for medicinal products, including those under ATMP regulation, are based on data that cover product quality, nonclinical assessment (i.e., preclinical trials), as well as clinical trials. Data must be summarized by the applicant, often the pharmaceutical entrepreneur working in partnership with clinical investigators and their medical institution(s), in dossiers including an internationally standardized set of Common Technical Documents. The application is checked by the European National Competent Authorities (NCA, in Germany the Paul-Ehrlich-Institut, Langen) that are nominated as rapporteur and co-rapporteur by EMA.

The documents are expected to show consistent data on the quality, safety, and efficacy of the particular product. Beforehand, EMA and NCA offer scientific recommendations on the classification of ATMP. Concerning the state-of-the-art of research, appropriate regulations will be adopted.

In the United States of America, the Food and Drug Administration sets the hallmarks for the regulation of medical and other products. There, the Center for Biologics Evaluation and Research (CBER) regulates biological products for human use under applicable federal laws, including the Public Health Service Act and the Federal Food, Drug and Cosmetic Act. CBER is responsible for ensuring the safety and effectiveness of biologics, including XT products. The Center for Veterinary Medicine (CVM) is responsible for assessing GMs in the source pigs.

CVM and CBER collaborate on their assessments of animals used for xenotransplantation. Submission of an Investigational New Drug application is required for the approval of clinical trials; preclinical experimental data must be submitted which demonstrate the safety and effectiveness of the GM porcine hearts for its intended human use.[89]


#

What Experimental Results Would Justify a Formal Clinical Trial?

In 2000, the Xenotransplantation Advisory Committee of the International Society for Heart and Lung Transplantation recommended that consistent survival of NHPs supported by orthotopic porcine heart transplants for 3 months would be sufficient to warrant a clinical trial.[83] However, advancements in the field have raised the bar for evidence, with some suggesting that consistent survival of up to 6 months without irreversible rejection or infection would be more appropriate for initiating clinical trials in carefully selected patients.[7] [10] [84] Extending survival durations to nine or even 12 months with one or two recipients would provide further assurance. It is imperative that clinical trials involve teams with expertise in both clinical orthotopic HTx and the preclinical pig-to-NHP model.


#

Selection of the First Patients

Selection of the initial patients for clinical trials of cardiac XT requires meticulous consideration to justify the inherent risks and ensure highly favorable outcomes. Potential candidates may include individuals in intensive care units who are unsuitable for mechanical circulatory support. This category encompasses patients with conditions like hypertrophic cardiomyopathy, prior mechanical or biological valve replacements, and postinfarction ventricular septal defects. These high-risk patients often experience increasing instability due to their reliance on inotropic medications and the presence of arrhythmias. It is imperative to assess the potential reversibility of secondary liver and kidney damage and the treatability of pulmonary hypertension in these cases[37] (see [Table 3] for further details).

Table 3

Potential indications for the initial clinical trials of pig heart transplantation

1.

Relative or absolute contraindications to mechanical circulatory support, e.g.

(a) restrictive or hypertrophic cardiomyopathy

(b) presence of a dysfunctional mechanical valve prosthesis or degenerated bioprosthesis

(c) atrial or ventricular septal defects

2.

High titres of broad panel-reactive anti-HLA antibodies (high PRA) that do not cross-react with swine leukocyte antigens (SLA) of the donor animal (see also chapter on “low-risk” donor-recipient combinations)

3.

Chronic rejection after cardiac allotransplantation

4.

Heart transplantation after successful carcinoma treatment

5.

• Hypoplastic left heart syndrome (particularly with reduced ejection fraction of the systemic right ventricle and/or severe tricuspid regurgitation)

• Other single ventricle patients with AV-valve regurgitation

• Pulmonary atresia with intact ventricular septum and right ventricular-dependent coronary circulation

• Unstable neonatal Ebstein

• Failed initial palliation (after Norwood or Glenn procedure)

• Cardiomyopathies with biventricular heart failure

Abbreviations: AV, atrioventricular; HLA, human leukocyte antigen; PRA, panel reactive antibody.


Source: Based on[93]


Neonates and infants with complex congenital heart diseases may benefit most from cardiac XT due to the lack of donors and the difficulties and poor outcomes of mechanical circulatory support in this age group.

Although there has been some progress in the field of mechanical circulatory support in patients with complex congenital heart disease like hypoplastic left heart syndrome or other forms of single ventricle physiology (e.g., pulmonary atresia with intact ventricular septum and right ventricular-dependent coronary circulation due to sinusoids),[90] [91] mortality after ventricular assist device (VAD) implantation as a bridge to transplant is still high (30% at 6 months).[92]

Therefore, we think that children with congenital heart disease not amenable to biventricular repair and with a high risk for palliative procedures or poor outcomes after VAD therapy would be candidates for cardiac XT as a bridge to allotransplantation. The readily available xenograft would overcome the high waiting list mortality in this age group ([Table 3]).

An advantage in the pediatric population will be the immature immune system of the neonate in combination with the thymectomy at the time of heart transplant. This environment would be ideal to induce immunological tolerance.


#

Anticipating the Future of Cardiac Xenotransplantation in the Next 5 to 10 Years

It is crucial to acknowledge that allografts will always be the preferred choice for individuals with advanced/terminal myocardial disease. However, due to the long waiting lists for donor hearts, we estimate that pig heart xenografts will be in clinical practice within the next 2 to 3 years. Initially, this might occur as a bridge to allotransplantation on an individual compassionate basis but ideally as part of a formal clinical trial. We foresee the approval of trials for both infant and adult patients. With successful long-term outcomes, cardiac XT may eventually become an accepted form of destination therapy.

We firmly believe that the field of XT will witness significant advancements in the next decade, surpassing those in mechanical assist devices, stem cell technology, and regenerative medicine.


#

Key Messages

  1. Significant progress in the field of xenotransplantation has been made and allowed for the first xenotransplantation of pig hearts into two patients in the United States (compassionate use), who died after 60 and 40 days, respectively.

  2. Nevertheless, in preclinical studies extended survival with clinically acceptable immunosuppression has been achieved, organ overgrowth could be controlled, appropriate donor-recipient matching is now established.

  3. Microbiological safety is no longer a prohibitive concern.

  4. Ethical considerations allow for a cautious start of clinical trials.

  5. Regulation and surveillance on a national and European level have been established.

  6. The German Heart Transplant Centers agreed to cooperatively select the first patients for a first clinical trial as soon as suitable donor pigs become available.


#
#

Conflict of Interest

None declared.

List of Participants

Abteilung für Herzchirurgie, Kerckhoff-Klinik GmbH Bad Nauheim -


Prof. Dr. Yeong-Hoon Choi


Dr. Nemanja Zukic


Klinik für Thorax- und kardiovaskuläre Chirurgie, Herz und Diabeteszentrum Bad Oeynhausen


Prof. Dr. Jan Gummert


Prof. Dr. René Schramm


Deutsches Herzzentrum der Charité Berlin


Prof. Dr. Christoph Knosalla


Freie Universität Berlin


Dr. Joachim Denner


Herzzentrum Dresden


Dr. Stefan Brose


Prof. Dr. Sems Malte Tugtekin


Pascal Tugtekin


Klinik für Herzchirurgie, Universitätsklinikum Düsseldorf


Prof. Dr. Artur Lichtenberg


Prof. Dr. Udo Boeken


Herzchirurgische Klinik, Uniklinikum Erlangen


Prof. Dr. Christian Heim


Kinderherzchirurgische Abteilung, Uniklinikum Erlangen


Prof. Dr. Oliver Dewald


Dr. Ariawan Purbojo


Klinik für Thorax- und kardiovaskuläre Chirurgie, Universitätsmedizin Essen


Prof. Dr. Arjang Ruhparwar (seit 01.04.2023 Klinik für Herz-, Thorax-, Transplantationschirurgie, Medizinische Hochschule Hannover)


PD Dr. Bastian Schmack (seit 01.04.2023 Klinik für Herz-, Thorax-, Transplantationschirurgie, Medizinische Hochschule Hannover)


Universitäts-Herzzentrum Freiburg/Bad Krozingen


Prof. Dr. Martin Czerny


PD Dr. David Schibilsky


Klinik für Herz-, Thorax- und Gefäßchirurgie, Universitätsmedizin Göttingen


Prof. Dr. Ingo Kutschka


Klinik für Herz- und Gefäßchirurgie, Universitätsklinikum Hamburg-Eppendorf


Prof. Dr. Hermann Reichenspurner


PD Dr. Alexander Bernhard


Klinik und Poliklinik für Kinderherzmedizin und EMAH, Universitätsklinikum Hamburg-Eppendorf


Prof. Dr. Michael Hübler


Dr. Samira Hübler


Klinik für Herz-, Thorax-, Transplantations- und Gefäßchirurgie, Medizinische Hochschule Hannover


Prof. Dr. Axel Haverich


Dr. Tobias Goecke


Klinik für Herzchirurgie, Universitätsklinikum Heidelberg


PD Dr. Rawa Arif


Klinik für Herz- und Thoraxchirurgie, Universitätsklinikum Jena


Prof. Dr. Torsten Doenst


PD Dr. Gloria Färber


Klinik für Herz- und Gefäßchirurgie, Universitätsklinikum Schleswig-Holstein, Campus Kiel


Prof. Dr. Jochen Cremer


Prof. Dr. Assad Haneya


Paul-Ehrlich Institut, Bundesinstitut für Impfstoffe und biomedizinische Arzneimittel, Langen


Prof. Dr. Ralf Reinhard Tönjes


Herzzentrum Leipzig


Dr. Uwe Schulz


Klinikum der Universität München


Prof. Dr. Christian Hagl


Prof. Dr. Michael Schmoeckel


Prof. Dr. Bruno Reichart


PD Dr. Matthias Längin


PD Dr. Jan-Michael Abicht


Dr. Michael Bender


Prof. Dr. Paolo Brenner


Dr. Carola Grinninger


Dr. Christine-Elena Kamla


Prof. Dr. Georg Marckmann


Prof. Dr. Sebastian Michel


Dr. Daniel Reichart


Reinhard Ellgass


Genzentrum und Centre for Innovative Medical Models (CiMM) der LMU München


Prof. Dr. Eckhard Wolf


Prof. Dr. Elisabeth Kempter


Dr. Barbara Keßler


Granzer Regulatory Consulting & Services, München


Dr. Stefan Blesse


Klinik und Poliklinik für Herz-, Thorax- und herznahe Gefäßchirurgie, Universitätsklinikum Regensburg


Prof. Dr. Christof Schmid


PD Dr. Simon Schopka


  • References

  • 1 Bauersachs J. Heart failure drug treatment: the fantastic four. Eur Heart J 2021; 42 (06) 681-683
  • 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
  • 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 (10) 1385-1390
  • 4 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 (02) 361-371
  • 5 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; 42 (08) 1120-1130
  • 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 (02) 311-327
  • 7 Längin M, Mayr T, Reichart B. et al. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 2018; 564 (7736) 430-433
  • 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 (08) 751-757
  • 9 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
  • 10 Mohiuddin MM, Goerlich CE, Singh AK. et al. Progressive genetic modifications of porcine cardiac xenografts extend survival to 9 months. Xenotransplantation 2022; 29 (03) e12744
  • 11 Griffith BP, Goerlich CE, Singh AK. et al. Genetically modified porcine-to-human cardiac xenotransplantation. N Engl J Med 2022; 387 (01) 35-44
  • 12 Mohiuddin MM, Singh AK, Scobie L. et al. Graft dysfunction in compassionate use of genetically engineered pig-to-human cardiac xenotransplantation: a case report. Lancet 2023; 402 (10399): 397-410
  • 13 Moazami N, Stern JM, Khalil K. et al. Pig-to-human heart xenotransplantation in two recently deceased human recipients. Nat Med 2023; 29 (08) 1989-1997
  • 14 Novitzky D. Electrocardiographic, hemodynamic and endocrine changes occurring during experimental brain death in the Chacma baboon. J Heart Transplant 1984; 4: 63-69
  • 15 Bery A, Marklin G, Itoh A. et al. Specialized donor care facility model and advances in management of thoracic organ donors. Ann Thorac Surg 2022; 113 (06) 1778-1786
  • 16 Cooper DKC, Hara H. Xenotransplantation—a basic science perspective. Kidney360 2023; 4 (08) 1147-1149
  • 17 Byrne GW, McGregor CG. Cardiac xenotransplantation: progress and challenges. Curr Opin Organ Transplant 2012; 17 (02) 148-154
  • 18 Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2019; 4 (41) eaau6298
  • 19 Phelps CJ, Koike C, Vaught TD. et al. Production of alpha 1,3-galactosyltransferase-deficient pigs. Science 2003; 299 (5605) 411-414
  • 20 Kwon DN, Lee K, Kang MJ. et al. Production of biallelic CMP-Neu5Ac hydroxylase knock-out pigs. Sci Rep 2013; 3: 1981
  • 21 Lutz AJ, Li P, Estrada JL. et al. Double knockout pigs deficient in N-glycolylneuraminic acid and galactose α-1,3-galactose reduce the humoral barrier to xenotransplantation. Xenotransplantation 2013; 20 (01) 27-35
  • 22 Estrada JL, Martens G, Li P. et al. Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/β4GalNT2 genes. Xenotransplantation 2015; 22 (03) 194-202
  • 23 Diamond LE, Quinn CM, Martin MJ, Lawson J, Platt JL, Logan JS. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation 2001; 71 (01) 132-142
  • 24 Cozzi E, White DJG. The generation of transgenic pigs as potential organ donors for humans. Nat Med 1995; 1 (09) 964-966
  • 25 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 (23) 11153-11157
  • 26 Galli C. Animal engineering for xenotransplantation: genome editing and cloning in pigs for xenotransplantation. EJT 2023; 1 (03) 182-191
  • 27 Cowan PJ, Robson SC. Progress towards overcoming coagulopathy and hemostatic dysfunction associated with xenotransplantation. Int J Surg 2015; 23 (Pt B): 296-300
  • 28 Pierson III RN, Fishman JA, Lewis GD. et al. Progress toward cardiac xenotransplantation. Circulation 2020; 142 (14) 1389-1398
  • 29 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 (02) 138-147
  • 30 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 (03) 211-220
  • 31 Shu S, Ren J, Song J. Cardiac xenotransplantation: a promising way to treat advanced heart failure. Heart Fail Rev 2022; 27 (01) 71-91
  • 32 Mohiuddin MM, Reichart B, Byrne GW, McGregor CGA. Current status of pig heart xenotransplantation. Int J Surg 2015; 23 (Pt B): 234-239
  • 33 Steen S, Paskevicius A, Liao Q, Sjöberg T. Safe orthotopic transplantation of hearts harvested 24 hours after brain death and preserved for 24 hours. Scand Cardiovasc J 2016; 50 (03) 193-200
  • 34 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 (01) e12636
  • 35 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 (09) 1323-1331
  • 36 Samy KP, Butler JR, Li P, Cooper DKC, Ekser B. The role of costimulation blockade in solid organ and islet xenotransplantation. J Immunol Res 2017; 2017: 8415205
  • 37 Reichart B, Längin M, Denner J, Schwinzer R, Cowan PJ, Wolf E. Pathways to clinical cardiac xenotransplantation. Transplantation 2021; 105 (09) 1930-1943
  • 38 Längin M, Buttgereit I, Reichart B. et al. Xenografts show signs of concentric hypertrophy and dynamic left ventricular outflow tract obstruction after orthotopic pig-to-baboon heart transplantation. Transplantation 2023; 107 (12) e328-e338
  • 39 Soin B, Ostlie D, Cozzi E. et al. Growth of porcine kidneys in their native and xenograft environment. Xenotransplantation 2000; 7 (02) 96-100
  • 40 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 (07) 1778-1790
  • 41 Arrunátegui AM, Ramón DS, Viola LM, Olsen LG, Jaramillo A. Technical and clinical aspects of the histocompatibility crossmatch assay in solid organ transplantation. Biomédica 2022; 42 (02) 391-413
  • 42 Guillaume N. Improved flow cytometry crossmatching in kidney transplantation. HLA 2018; 92 (06) 375-383
  • 43 Porrett PM, Orandi BJ, Kumar V. et al. First clinical-grade porcine kidney xenotransplant using a human decedent model. Am J Transplant 2022; 22 (04) 1037-1053
  • 44 Wekerle T, Segev D, Lechler R, Oberbauer R. Strategies for long-term preservation of kidney graft function. Lancet 2017; 389 (10084): 2152-2162
  • 45 Ansari D, Bućin D, Nilsson J. Human leukocyte antigen matching in heart transplantation: systematic review and meta-analysis. Transpl Int 2014; 27 (08) 793-804
  • 46 Ladowski JM, Houp J, Hauptfeld-Dolejsek V, Javed M, Hara H, Cooper DKC. Aspects of histocompatibility testing in xenotransplantation. Transpl Immunol 2021; 67: 101409
  • 47 Galili U. Discovery of the natural anti-Gal antibody and its past and future relevance to medicine. Xenotransplantation 2013; 20 (03) 138-147
  • 48 Padler-Karavani V, Varki A. Potential impact of the non-human sialic acid N-glycolylneuraminic acid on transplant rejection risk. Xenotransplantation 2011; 18 (01) 1-5
  • 49 Byrne GW, Stalboerger PG, Du Z, Davis TR, McGregor CG. Identification of new carbohydrate and membrane protein antigens in cardiac xenotransplantation. Transplantation 2011; 91 (03) 287-292
  • 50 Martens GR, Reyes LM, Li P. et al. Humoral reactivity of renal transplant-waitlisted patients to cells from GGTA1/CMAH/B4GalNT2, and SLA Class I knockout pigs. Transplantation 2017; 101 (04) e86-e92
  • 51 Cooper DKC, Hara H, Iwase H. et al. Justification of specific genetic modifications in pigs for clinical organ xenotransplantation. Xenotransplantation 2019; 26 (04) e12516
  • 52 Ladowski J, Martens G, Estrada J, Tector M, Tector J. The desirable donor pig to eliminate all xenoreactive antigens. Xenotransplantation 2019; 26 (04) e12504
  • 53 Ladowski JM, Martens GR, Reyes LM. et al. Examining the biosynthesis and xenoantigenicity of Class II swine leukocyte antigen proteins. J Immunol 2018; 200 (08) 2957-2964
  • 54 Ladowski JM, Hara H, Cooper DKC. The role of SLAs in xenotransplantation. Transplantation 2021; 105 (02) 300-307
  • 55 He S, Li T, Feng H. et al. Incidence of serum antibodies to xenoantigens on triple-knockout pig cells in different human groups. Xenotransplantation 2023; (e-pub ahead of print). DOI: 10.1111/xen.12818.
  • 56 Ladowski JM, Reyes LM, Martens GR. et al. Swine leukocyte antigen Class II is a xenoantigen. Transplantation 2018; 102 (02) 249-254
  • 57 Martens GR, Ladowski JM, Estrada J. et al. HLA Class I-sensitized renal transplant patients have antibody binding to SLA Class I epitopes. Transplantation 2019; 103 (08) 1620-1629
  • 58 Fischer K, Rieblinger B, Hein R. et al. Viable pigs after simultaneous inactivation of porcine MHC class I and three xenoreactive antigen genes GGTA1, CMAH and B4GALNT2. Xenotransplantation 2020; 27 (01) e12560
  • 59 Fu R, Fang M, Xu K. et al. Generation of GGTA1-/-β2M-/-CIITA-/- pigs using CRISPR/Cas9 technology to alleviate xenogeneic immune reactions. Transplantation 2020; 104 (08) 1566-1573
  • 60 Ladowski JM, Martens GR, Reyes LM, Hauptfeld-Dolejsek V, Tector M, Tector J. Examining epitope mutagenesis as a strategy to reduce and eliminate human antibody binding to class II swine leukocyte antigens. Immunogenetics 2019; 71 (07) 479-487
  • 61 Denner J. Virus safety of xenotransplantation. Viruses 2022; 14: 1926
  • 62 Denner J. The porcine virome and xenotransplantation. Virol J 2017; 14 (01) 171
  • 63 Denner J. Zoonosis and xenozoonosis in xenotransplantation: A proposal for a new classification. Zoonoses Public Health 2023; 70 (06) 578-579
  • 64 Denner J, Schuurman HJ, Patience C. 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 2009; 16 (04) 239-248
  • 65 Meng XJ. Hepatitis E virus: animal reservoirs and zoonotic risk. Vet Microbiol 2010; 140 (3-4(: 256-265
  • 66 Denner J. Xenotransplantation and hepatitis E virus. Xenotransplantation 2015; 22 (03) 167-173
  • 67 Denner J, Bigley TM, Phan TL, Zimmermann C, Zhou X, Kaufer BB. Comparative analysis of Roseoloviruses in humans, pigs, mice, and other species. Viruses 2019; 11 (12) 1108
  • 68 Kotton CN, Torre-Cisneros J, Aguado JM. et al; International CMV Symposium Faculty. Cytomegalovirus in the transplant setting: where are we now and what happens next? A report from the International CMV Symposium 2021. Transpl Infect Dis 2022; 24 (06) e13977
  • 69 Denner J. Reduction of the survival time of pig xenotransplants by porcine cytomegalovirus. Virol J 2018; 15 (01) 171
  • 70 Denner J, Längin M, Reichart B. et al. Impact of porcine cytomegalovirus on long-term orthotopic cardiac xenotransplant survival. Sci Rep 2020; 10 (01) 17531
  • 71 Denner J, Schuurman HJ. Early testing of porcine organ xenotransplantation products in humans: microbial safety as illustrated for porcine cytomegalovirus. Xenotransplantation 2022; 29 (06) e12783
  • 72 Egerer S, Fiebig U, Kessler B. et al. Early weaning completely eliminates porcine cytomegalovirus from a newly established pig donor facility for xenotransplantation. Xenotransplantation 2018; 25 (04) e12449
  • 73 Denner J, Tönjes RR. Infection barriers to successful xenotransplantation focusing on porcine endogenous retroviruses. Clin Microbiol Rev 2012; 25 (02) 318-343
  • 74 Halecker S, Krabben L, Kristiansen Y. et al. Rare isolation of human-tropic recombinant porcine endogenous retroviruses PERV-A/C from Göttingen minipigs. Virol J 2022; 19 (01) 30
  • 75 Denner J, Schuurman HJ. High prevalence of recombinant porcine endogenous retroviruses (PERV-A/Cs) in minipigs: a review on origin and presence. Viruses 2021; 13 (09) 1869
  • 76 Karlas A, Irgang M, Votteler J. et al. Characterisation of a human cell-adapted porcine endogenous retrovirus PERV-A/C. Ann Transplant 2010; 15 (02) 45-54
  • 77 Denner J. Why was PERV not transmitted during preclinical and clinical xenotransplantation trials and after inoculation of animals?. Retrovirology 2018; 15 (01) 28
  • 78 Entwistle JW, Sade RM, Drake DH. Clinical xenotransplantation seems close: Ethical issues persist. Artif Organs 2022; 46 (06) 987-994
  • 79 Caplan A, Parent B. Ethics and the emerging use of pig organs for xenotransplantation. J Heart Lung Transplant 2022; 41 (09) 1204-1206
  • 80 Johnson LSM. Existing ethical tensions in xenotransplantation. Camb Q Healthc Ethics 2022; 31 (03) 355-367
  • 81 Silverman H, Odonkor PN. Reevaluating the ethical issues in porcine-to-human heart xenotransplantation. Hastings Cent Rep 2022; 52 (05) 32-42
  • 82 Marckmann G, Schmidt H, Sofaer N, Strech D. Putting public health ethics into practice: a systematic framework. Front Public Health 2015; 3: 23
  • 83 Cooper DKC, Keogh AM, Brink J. et al; Xenotransplantation Advisory Committee of the International Society for Heart and Lung Transplantation. 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 (12) 1125-1165
  • 84 Reichart B, Cooper DKC, Längin M, Tönjes RR, Pierson III RN, Wolf E. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023; 118 (18) 3499-3516
  • 85 Halecker S, Hansen S, Krabben L, Ebner F, Kaufer B, Denner J. How, where and when to screen for porcine cytomegalovirus (PCMV) in donor pigs for xenotransplantation. Sci Rep 2022; 12 (01) 21545
  • 86 Kögel J, Thiersch S, Ludwig B, Seissler J, Marckmann G. What does it take to consent to islet cell xenotransplantation?: Insights from an interview study with type 1 diabetes patients and review of the literature. BMC Med Ethics 2021; 22 (01) 37
  • 87 Pierson III RN, Allan JS, Cooper DKC. et al. Expert opinion special feature: patient selection for initial clinical trials of pig organ transplantation. Transplantation 2022; 106 (09) 1720-1723
  • 88 Kögel J, Marckmann G. [First-of-its-kind xenotransplantation: need for ethical reflection in science and society]. Ethik Med 2023; 35 (01) 137-143
  • 89 Marks P, Solomon S. Clarifying US regulations on xenotransplantation. Nat Biotechnol 2021; 39 (12) 1500-1501
  • 90 Bleiweis MS, Philip J, Peek GJ. et al. Palliation plus ventricular assist device insertion in 15 neonates and infants with functionally univentricular circulation. Ann Thorac Surg 2022; 114 (04) 1412-1418
  • 91 Philip J, Powers E, Machado D. et al. Pulsatile ventricular assist device as a bridge to transplant for the early high-risk single-ventricle physiology. J Thorac Cardiovasc Surg 2021; 162 (02) 405-413.e4
  • 92 Rossano JW, VanderPluym CJ, Peng DM. et al; Pedimacs Investigators. Fifth Annual Pediatric Interagency Registry for Mechanical Circulatory Support (Pedimacs) Report. Ann Thorac Surg 2021; 112 (06) 1763-1774
  • 93 Chaban R, Cooper DKC, Pierson III RN. Pig heart and lung xenotransplantation: present status. J Heart Lung Transplant 2022; 41 (08) 1014-1022
  • 94 Hundrieser J, Hein R, Pokoyski C. et al. Role of human and porcine MHC DRB1 alleles in determining the intensity of individual human anti-pig T-cell responses. Xenotransplantation 2019; 26 (04) e12523

Address for correspondence

Michael Schmoeckel, MD, PhD
Department of Cardiac Surgery, LMU Klinikum Grosshadern
Marchioninistr. 15, 81377 Munich
Germany   

Publication History

Received: 25 September 2023

Accepted: 22 November 2023

Accepted Manuscript online:
28 December 2023

Article published online:
12 February 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Bauersachs J. Heart failure drug treatment: the fantastic four. Eur Heart J 2021; 42 (06) 681-683
  • 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
  • 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 (10) 1385-1390
  • 4 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 (02) 361-371
  • 5 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; 42 (08) 1120-1130
  • 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 (02) 311-327
  • 7 Längin M, Mayr T, Reichart B. et al. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 2018; 564 (7736) 430-433
  • 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 (08) 751-757
  • 9 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
  • 10 Mohiuddin MM, Goerlich CE, Singh AK. et al. Progressive genetic modifications of porcine cardiac xenografts extend survival to 9 months. Xenotransplantation 2022; 29 (03) e12744
  • 11 Griffith BP, Goerlich CE, Singh AK. et al. Genetically modified porcine-to-human cardiac xenotransplantation. N Engl J Med 2022; 387 (01) 35-44
  • 12 Mohiuddin MM, Singh AK, Scobie L. et al. Graft dysfunction in compassionate use of genetically engineered pig-to-human cardiac xenotransplantation: a case report. Lancet 2023; 402 (10399): 397-410
  • 13 Moazami N, Stern JM, Khalil K. et al. Pig-to-human heart xenotransplantation in two recently deceased human recipients. Nat Med 2023; 29 (08) 1989-1997
  • 14 Novitzky D. Electrocardiographic, hemodynamic and endocrine changes occurring during experimental brain death in the Chacma baboon. J Heart Transplant 1984; 4: 63-69
  • 15 Bery A, Marklin G, Itoh A. et al. Specialized donor care facility model and advances in management of thoracic organ donors. Ann Thorac Surg 2022; 113 (06) 1778-1786
  • 16 Cooper DKC, Hara H. Xenotransplantation—a basic science perspective. Kidney360 2023; 4 (08) 1147-1149
  • 17 Byrne GW, McGregor CG. Cardiac xenotransplantation: progress and challenges. Curr Opin Organ Transplant 2012; 17 (02) 148-154
  • 18 Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2019; 4 (41) eaau6298
  • 19 Phelps CJ, Koike C, Vaught TD. et al. Production of alpha 1,3-galactosyltransferase-deficient pigs. Science 2003; 299 (5605) 411-414
  • 20 Kwon DN, Lee K, Kang MJ. et al. Production of biallelic CMP-Neu5Ac hydroxylase knock-out pigs. Sci Rep 2013; 3: 1981
  • 21 Lutz AJ, Li P, Estrada JL. et al. Double knockout pigs deficient in N-glycolylneuraminic acid and galactose α-1,3-galactose reduce the humoral barrier to xenotransplantation. Xenotransplantation 2013; 20 (01) 27-35
  • 22 Estrada JL, Martens G, Li P. et al. Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/β4GalNT2 genes. Xenotransplantation 2015; 22 (03) 194-202
  • 23 Diamond LE, Quinn CM, Martin MJ, Lawson J, Platt JL, Logan JS. A human CD46 transgenic pig model system for the study of discordant xenotransplantation. Transplantation 2001; 71 (01) 132-142
  • 24 Cozzi E, White DJG. The generation of transgenic pigs as potential organ donors for humans. Nat Med 1995; 1 (09) 964-966
  • 25 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 (23) 11153-11157
  • 26 Galli C. Animal engineering for xenotransplantation: genome editing and cloning in pigs for xenotransplantation. EJT 2023; 1 (03) 182-191
  • 27 Cowan PJ, Robson SC. Progress towards overcoming coagulopathy and hemostatic dysfunction associated with xenotransplantation. Int J Surg 2015; 23 (Pt B): 296-300
  • 28 Pierson III RN, Fishman JA, Lewis GD. et al. Progress toward cardiac xenotransplantation. Circulation 2020; 142 (14) 1389-1398
  • 29 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 (02) 138-147
  • 30 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 (03) 211-220
  • 31 Shu S, Ren J, Song J. Cardiac xenotransplantation: a promising way to treat advanced heart failure. Heart Fail Rev 2022; 27 (01) 71-91
  • 32 Mohiuddin MM, Reichart B, Byrne GW, McGregor CGA. Current status of pig heart xenotransplantation. Int J Surg 2015; 23 (Pt B): 234-239
  • 33 Steen S, Paskevicius A, Liao Q, Sjöberg T. Safe orthotopic transplantation of hearts harvested 24 hours after brain death and preserved for 24 hours. Scand Cardiovasc J 2016; 50 (03) 193-200
  • 34 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 (01) e12636
  • 35 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 (09) 1323-1331
  • 36 Samy KP, Butler JR, Li P, Cooper DKC, Ekser B. The role of costimulation blockade in solid organ and islet xenotransplantation. J Immunol Res 2017; 2017: 8415205
  • 37 Reichart B, Längin M, Denner J, Schwinzer R, Cowan PJ, Wolf E. Pathways to clinical cardiac xenotransplantation. Transplantation 2021; 105 (09) 1930-1943
  • 38 Längin M, Buttgereit I, Reichart B. et al. Xenografts show signs of concentric hypertrophy and dynamic left ventricular outflow tract obstruction after orthotopic pig-to-baboon heart transplantation. Transplantation 2023; 107 (12) e328-e338
  • 39 Soin B, Ostlie D, Cozzi E. et al. Growth of porcine kidneys in their native and xenograft environment. Xenotransplantation 2000; 7 (02) 96-100
  • 40 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 (07) 1778-1790
  • 41 Arrunátegui AM, Ramón DS, Viola LM, Olsen LG, Jaramillo A. Technical and clinical aspects of the histocompatibility crossmatch assay in solid organ transplantation. Biomédica 2022; 42 (02) 391-413
  • 42 Guillaume N. Improved flow cytometry crossmatching in kidney transplantation. HLA 2018; 92 (06) 375-383
  • 43 Porrett PM, Orandi BJ, Kumar V. et al. First clinical-grade porcine kidney xenotransplant using a human decedent model. Am J Transplant 2022; 22 (04) 1037-1053
  • 44 Wekerle T, Segev D, Lechler R, Oberbauer R. Strategies for long-term preservation of kidney graft function. Lancet 2017; 389 (10084): 2152-2162
  • 45 Ansari D, Bućin D, Nilsson J. Human leukocyte antigen matching in heart transplantation: systematic review and meta-analysis. Transpl Int 2014; 27 (08) 793-804
  • 46 Ladowski JM, Houp J, Hauptfeld-Dolejsek V, Javed M, Hara H, Cooper DKC. Aspects of histocompatibility testing in xenotransplantation. Transpl Immunol 2021; 67: 101409
  • 47 Galili U. Discovery of the natural anti-Gal antibody and its past and future relevance to medicine. Xenotransplantation 2013; 20 (03) 138-147
  • 48 Padler-Karavani V, Varki A. Potential impact of the non-human sialic acid N-glycolylneuraminic acid on transplant rejection risk. Xenotransplantation 2011; 18 (01) 1-5
  • 49 Byrne GW, Stalboerger PG, Du Z, Davis TR, McGregor CG. Identification of new carbohydrate and membrane protein antigens in cardiac xenotransplantation. Transplantation 2011; 91 (03) 287-292
  • 50 Martens GR, Reyes LM, Li P. et al. Humoral reactivity of renal transplant-waitlisted patients to cells from GGTA1/CMAH/B4GalNT2, and SLA Class I knockout pigs. Transplantation 2017; 101 (04) e86-e92
  • 51 Cooper DKC, Hara H, Iwase H. et al. Justification of specific genetic modifications in pigs for clinical organ xenotransplantation. Xenotransplantation 2019; 26 (04) e12516
  • 52 Ladowski J, Martens G, Estrada J, Tector M, Tector J. The desirable donor pig to eliminate all xenoreactive antigens. Xenotransplantation 2019; 26 (04) e12504
  • 53 Ladowski JM, Martens GR, Reyes LM. et al. Examining the biosynthesis and xenoantigenicity of Class II swine leukocyte antigen proteins. J Immunol 2018; 200 (08) 2957-2964
  • 54 Ladowski JM, Hara H, Cooper DKC. The role of SLAs in xenotransplantation. Transplantation 2021; 105 (02) 300-307
  • 55 He S, Li T, Feng H. et al. Incidence of serum antibodies to xenoantigens on triple-knockout pig cells in different human groups. Xenotransplantation 2023; (e-pub ahead of print). DOI: 10.1111/xen.12818.
  • 56 Ladowski JM, Reyes LM, Martens GR. et al. Swine leukocyte antigen Class II is a xenoantigen. Transplantation 2018; 102 (02) 249-254
  • 57 Martens GR, Ladowski JM, Estrada J. et al. HLA Class I-sensitized renal transplant patients have antibody binding to SLA Class I epitopes. Transplantation 2019; 103 (08) 1620-1629
  • 58 Fischer K, Rieblinger B, Hein R. et al. Viable pigs after simultaneous inactivation of porcine MHC class I and three xenoreactive antigen genes GGTA1, CMAH and B4GALNT2. Xenotransplantation 2020; 27 (01) e12560
  • 59 Fu R, Fang M, Xu K. et al. Generation of GGTA1-/-β2M-/-CIITA-/- pigs using CRISPR/Cas9 technology to alleviate xenogeneic immune reactions. Transplantation 2020; 104 (08) 1566-1573
  • 60 Ladowski JM, Martens GR, Reyes LM, Hauptfeld-Dolejsek V, Tector M, Tector J. Examining epitope mutagenesis as a strategy to reduce and eliminate human antibody binding to class II swine leukocyte antigens. Immunogenetics 2019; 71 (07) 479-487
  • 61 Denner J. Virus safety of xenotransplantation. Viruses 2022; 14: 1926
  • 62 Denner J. The porcine virome and xenotransplantation. Virol J 2017; 14 (01) 171
  • 63 Denner J. Zoonosis and xenozoonosis in xenotransplantation: A proposal for a new classification. Zoonoses Public Health 2023; 70 (06) 578-579
  • 64 Denner J, Schuurman HJ, Patience C. 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 2009; 16 (04) 239-248
  • 65 Meng XJ. Hepatitis E virus: animal reservoirs and zoonotic risk. Vet Microbiol 2010; 140 (3-4(: 256-265
  • 66 Denner J. Xenotransplantation and hepatitis E virus. Xenotransplantation 2015; 22 (03) 167-173
  • 67 Denner J, Bigley TM, Phan TL, Zimmermann C, Zhou X, Kaufer BB. Comparative analysis of Roseoloviruses in humans, pigs, mice, and other species. Viruses 2019; 11 (12) 1108
  • 68 Kotton CN, Torre-Cisneros J, Aguado JM. et al; International CMV Symposium Faculty. Cytomegalovirus in the transplant setting: where are we now and what happens next? A report from the International CMV Symposium 2021. Transpl Infect Dis 2022; 24 (06) e13977
  • 69 Denner J. Reduction of the survival time of pig xenotransplants by porcine cytomegalovirus. Virol J 2018; 15 (01) 171
  • 70 Denner J, Längin M, Reichart B. et al. Impact of porcine cytomegalovirus on long-term orthotopic cardiac xenotransplant survival. Sci Rep 2020; 10 (01) 17531
  • 71 Denner J, Schuurman HJ. Early testing of porcine organ xenotransplantation products in humans: microbial safety as illustrated for porcine cytomegalovirus. Xenotransplantation 2022; 29 (06) e12783
  • 72 Egerer S, Fiebig U, Kessler B. et al. Early weaning completely eliminates porcine cytomegalovirus from a newly established pig donor facility for xenotransplantation. Xenotransplantation 2018; 25 (04) e12449
  • 73 Denner J, Tönjes RR. Infection barriers to successful xenotransplantation focusing on porcine endogenous retroviruses. Clin Microbiol Rev 2012; 25 (02) 318-343
  • 74 Halecker S, Krabben L, Kristiansen Y. et al. Rare isolation of human-tropic recombinant porcine endogenous retroviruses PERV-A/C from Göttingen minipigs. Virol J 2022; 19 (01) 30
  • 75 Denner J, Schuurman HJ. High prevalence of recombinant porcine endogenous retroviruses (PERV-A/Cs) in minipigs: a review on origin and presence. Viruses 2021; 13 (09) 1869
  • 76 Karlas A, Irgang M, Votteler J. et al. Characterisation of a human cell-adapted porcine endogenous retrovirus PERV-A/C. Ann Transplant 2010; 15 (02) 45-54
  • 77 Denner J. Why was PERV not transmitted during preclinical and clinical xenotransplantation trials and after inoculation of animals?. Retrovirology 2018; 15 (01) 28
  • 78 Entwistle JW, Sade RM, Drake DH. Clinical xenotransplantation seems close: Ethical issues persist. Artif Organs 2022; 46 (06) 987-994
  • 79 Caplan A, Parent B. Ethics and the emerging use of pig organs for xenotransplantation. J Heart Lung Transplant 2022; 41 (09) 1204-1206
  • 80 Johnson LSM. Existing ethical tensions in xenotransplantation. Camb Q Healthc Ethics 2022; 31 (03) 355-367
  • 81 Silverman H, Odonkor PN. Reevaluating the ethical issues in porcine-to-human heart xenotransplantation. Hastings Cent Rep 2022; 52 (05) 32-42
  • 82 Marckmann G, Schmidt H, Sofaer N, Strech D. Putting public health ethics into practice: a systematic framework. Front Public Health 2015; 3: 23
  • 83 Cooper DKC, Keogh AM, Brink J. et al; Xenotransplantation Advisory Committee of the International Society for Heart and Lung Transplantation. 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 (12) 1125-1165
  • 84 Reichart B, Cooper DKC, Längin M, Tönjes RR, Pierson III RN, Wolf E. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023; 118 (18) 3499-3516
  • 85 Halecker S, Hansen S, Krabben L, Ebner F, Kaufer B, Denner J. How, where and when to screen for porcine cytomegalovirus (PCMV) in donor pigs for xenotransplantation. Sci Rep 2022; 12 (01) 21545
  • 86 Kögel J, Thiersch S, Ludwig B, Seissler J, Marckmann G. What does it take to consent to islet cell xenotransplantation?: Insights from an interview study with type 1 diabetes patients and review of the literature. BMC Med Ethics 2021; 22 (01) 37
  • 87 Pierson III RN, Allan JS, Cooper DKC. et al. Expert opinion special feature: patient selection for initial clinical trials of pig organ transplantation. Transplantation 2022; 106 (09) 1720-1723
  • 88 Kögel J, Marckmann G. [First-of-its-kind xenotransplantation: need for ethical reflection in science and society]. Ethik Med 2023; 35 (01) 137-143
  • 89 Marks P, Solomon S. Clarifying US regulations on xenotransplantation. Nat Biotechnol 2021; 39 (12) 1500-1501
  • 90 Bleiweis MS, Philip J, Peek GJ. et al. Palliation plus ventricular assist device insertion in 15 neonates and infants with functionally univentricular circulation. Ann Thorac Surg 2022; 114 (04) 1412-1418
  • 91 Philip J, Powers E, Machado D. et al. Pulsatile ventricular assist device as a bridge to transplant for the early high-risk single-ventricle physiology. J Thorac Cardiovasc Surg 2021; 162 (02) 405-413.e4
  • 92 Rossano JW, VanderPluym CJ, Peng DM. et al; Pedimacs Investigators. Fifth Annual Pediatric Interagency Registry for Mechanical Circulatory Support (Pedimacs) Report. Ann Thorac Surg 2021; 112 (06) 1763-1774
  • 93 Chaban R, Cooper DKC, Pierson III RN. Pig heart and lung xenotransplantation: present status. J Heart Lung Transplant 2022; 41 (08) 1014-1022
  • 94 Hundrieser J, Hein R, Pokoyski C. et al. Role of human and porcine MHC DRB1 alleles in determining the intensity of individual human anti-pig T-cell responses. Xenotransplantation 2019; 26 (04) e12523

Zoom Image
Fig. 1 Mechanisms of hyperacute xenograft rejection and strategies to overcome them. Created with BioRender.com.
Zoom Image
Fig. 2 Activation of protein C by the thrombin–thrombomodulin complex after allogeneic (top) and xenogeneic transplantation (bottom). EPCR = endothelial protein C receptor; PC = protein C; PCa = activated protein C; TBM = thrombomodulin; Va = activated factor V; VIIIa = activated factor VIII. Created with BioRender.com.
Zoom Image
Fig. 3 Auckland Island pigs in the Center for Innovative Medical Models (CiMM; www.lmu.de/cimm/) at LMU Munich.
Zoom Image
Fig. 4 Flowchart to achieve optimal donor–recipient-combinations for clinical xenotransplantation. Cross-matching of sera from potential recipients should be performed by using cells from TKO pigs lacking αGal, Neu5Gc, and Sd(a). Negative cross-match: Organs from TKO donor pigs combined with additional genetic modifications will be used as previously explained. In case of a positive cross-match, further characterization of antipig antibodies will be required. Recipients expressing anti-SLA antibodies may be transplanted with organs from SLA-I/II knockout pigs or pigs expressing mutated SLA to avoid antibody binding (both on “TKO plus” platform). Cross-matching may be complemented by HLA–SLA matching to identify donor–recipient combinations with low level T cell reactivity.[94]