You are on page 1of 9

Clinica Chimica Acta 514 (2021) 15–23

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/cca

Review

Kidney xenotransplantation: Recent progress in preclinical research


Xiao-Hua Yu a, c, Wen-Yi Deng a, c, Hong-Tao Jiang b, c, Tao Li b, c, Yi Wang a, b, c, *
a
Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 460106, China
b
Department of Organ Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 460106, China
c
The Transplantation Institute of Hainan Medical University, Haikou, Hainan 460106, China

A R T I C L E I N F O A B S T R A C T

Keywords: Kidney transplantation is the most effective treatment for end-stage renal disease, but is limited by the increasing
Kidney xenotransplantation shortage of deceased and living human donor kidneys. Xenotransplantation using pig organs provides the pos­
Pigs sibility to resolve the issue of organ supply shortage and is regarded as the next great medical revolution. In the
Non-human primates
past five years, there have been sequential advances toward the prolongation of life-supporting pig kidney
Genetic modifications
xenograft survival in non-human primates, with the longest survival being 499 days. This progress is due to the
Costimulation blockade
growing availability of pigs with multi-layered genetic modifications to overcome the pathobiological barriers
and the application of a costimulation blockade-based immunosuppressive regimen. These encouraging results
bring the hope to initiate the clinical trials of pig kidney transplantation in the near future. In this review, we
summarized the latest advances regarding pig kidney xenotransplantation in preclinical models to provide a basis
for future investigation and potential clinical translation.

1. Introduction difficulties in breeding. Pig kidneys are similar to human kidneys in size,
structure and major physiological indicators, such as glomerular filtra­
End-stage renal disease (ESRD) is a major cause of mortality across tion rate and endogenous creatinine clearance rate. Pig kidneys are thus
the world. Despite chronic hemodialysis contributes to the prolongation thought to an ideal source of donor organs for clinical transplantation
of survival time, but it is limited to alleviate the symptoms. Kidney [3]. Due to the evolutionary discrepancy between pigs and humans,
transplantation is currently regarded as an optimal option for ESRD wild-type pig kidneys cannot be directly transplanted to humans. Sur­
treatment. However, there is a continuing worldwide shortage of vival of genetically-unmodified pig kidneys in NHPs is generally no
deceased and living human donor kidneys for transplantation. It is longer than a few minutes due to hyperacute rejection (HAR) [4]. Thus,
estimated that more than 100,000 patients are waiting for kidney pigs must be genetically-modified before xenotransplantation. There are
transplants in the US [1]. In 2015, 16,291 kidney transplantation from now an estimated 40 or more genetic alterations that have been pro­
deceased donors was performed in the US, but 4761 patients died when duced in pigs [5]. Moreover, pigs with up to nine genetic manipulations
they were waiting for a kidney [2]. This highlights an urgent need for the are also available [6]. With significant advances in gene editing tech­
unlimited and prompt supply of transplantable kidneys. nology and immunosuppressive therapy, the outcomes of pig-to-NHP
Kidney xenotransplantation provides a feasible approach to resolve kidney xenotransplantation have largely improved in recent years
this problem. Although non-human primates (NHPs) are closer to (Fig. 1), sufficiently encouraging consideration for initial clinical trials
humans than other animals, they are not suitable for organ-source ani­ of pig kidney transplantation [7]. In this review, we summarized the
mals because of ethical concerns, a higher risk of xenozoonosis, and current knowledge about kidney xenotransplantation in preclinical

Abbreviations: ESRD, end-stage renal disease; NHPs, non-human primates; HAR, hyperacute rejection; AHXR, acute humoral xenograft rejection; Gal, galactose-
α1,3-galactose; GGTA1, α1,3-galactosyltransferase; Neu5Gc, N-glycolylneuraminc acid; CMAH, cytidine monophosphate-N-acetylneuraminic acid hydroxylase;
β4GalNT2, β-1,4N-acetylgalactosaminyl transferase 2; PBMCs, peripheral blood mononuclear cells; CRPs, complement regulation proteins; CD55, decay accelerating
factor; hCD46, membrane cofactor protein; CD59, membrane inhibitor of reactive lysis; SP-D, surfactant protein D; TF, tissue factor; CD39, ectonucleoside
triphosphate diphosphohydrolase-1; TBM, thrombomodulin; TFPI, tissue factor pathway inhibitor; EPCR, endothelial protein C receptor; HO-1, hemeoxygenase-1;
PERVs, pig endogenous retroviruses; CTLA4, cytotoxic T lymphocyte-associated protein 4; IL-7R, IL-7 receptor; ATG, anti-thymoglobulin; SLA, swine leukocyte
antigen.
* Corresponding author at: Department of Organ Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 460106, China.
E-mail address: wayne0108@126.com (Y. Wang).

https://doi.org/10.1016/j.cca.2020.11.028
Received 17 April 2020; Received in revised form 26 November 2020; Accepted 30 November 2020
Available online 7 December 2020
0009-8981/© 2020 Elsevier B.V. All rights reserved.
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

research to provide an important framework for future investigation and Table 1


clinical trials. Major genetic modifications for xenotransplantation and their purpose.
Modification Purpose References
2. Genetically-modified pig for xenotransplantation GGTA1KO Deletion of Gal antigen to prevent HAR [18–21]
CMAHKO Deletion of Neu5Gc antigen to counteract AHXR [27]
The high immune incompatibility between the donor and the β4GalNT2KO Deletion of Sda antigen to counteract AHXR [28]
recipient is regarded as a major hurdle for xenotransplantation. Immu­ hCD55 Prevention of complement activation [36]
hCD46 Prevention of complement activation [37]
nological barriers comprise HAR, acute humoral xenograft rejection
hCD59 Prevention of complement activation [38]
(AHXR), cellular xenograft rejection, coagulation dysregulation and hCD47 Protection against cellular xenograft rejection [45]
inflammatory response [8]. With the improvement of gene editing hSP-D Protection against cellular xenograft rejection [46]
techniques, especially CRIPSR/Cas9, a variety of genetically-engineered hCD200 Protection against cellular xenograft rejection [47]
pigs available for xenotransplantation of cells, tissues or organs have hCD274 Protection against cellular xenograft rejection [48]
hCD39 Anticoagulation [54]
been generated to overcome these obstacles (Table 1). hTBM Anticoagulation [55–58]
hTFPI Anticoagulation [60,61]
hEPCR Anticoagulation [62–65]
2.1. Deletion of xenoreactive antigens
hHO-1 Anti-inflammation [73]
hA20 Anti-inflammation [74]
HAR belongs to humoral immune response and is defined as PERV-inactivated Inactivation of PERVs [94]
antibody-mediated complement-dependent cytotoxicity. In this process,
the binding of natural preformed anti-pig antibodies in NHPs or humans
AHXR, which develops within a few days or weeks and has similar
to xenoantigens present in pig vascular endothelial cells activates
histopathological features with HAR [24]. N-glycolylneuraminc acid
complement cascade, leading to rapid graft destruction characterized by
(Neu5Gc) and Sda have been identified as two major non-Gal antigens,
thrombosis, interstitial hemorrhage and edema [9,10]. The most
both of which are produced by cytidine monophosphate-N-
important xenoantigen causing HAR is galactose-α1,3-galactose (Gal),
acetylneuraminic acid hydroxylase (CMAH) and β-1,4N-acetylgalacto­
which is synthesized by the glycosylation enzyme α1,3-galactosyl­
saminyl transferase 2 (β4GalNT2), respectively [25,26]. New World
transferase (GGTA1) [11,12]. Gal is highly expressed in most mammals,
monkeys and Humans do not express Neu5Gc and Sda, while Neu5Gc is
including pigs and New World monkeys, but not Old World monkeys
present in Old World monkeys. Our group recently reported that there
(eg, great apes, baboons) and humans [13]. Lack of Gal results in these
was less human IgG and IgM binding to red blood cells and aortic
primates making antibodies against this foreign antigen. It is believed
endothelial cells (AECs) from GGTA1KO/CMAHKO pigs than those from
that anti-Gal antibodies are induced by Gal-expressing bacteria in the
GGTA1KO pigs [27]. Peripheral blood mononuclear cells (PBMCs) from
primates’ gastrointestinal tract during infancy [14], and account for
GGTA1/CMAH/β4GalNT2 deficient pigs displayed a significant
about 1% of circulating immunoglobulins [15].
decrease in human antibody binding compared with those from pigs
Plasmapheresis and immunoadsorption were first used to remove
lacking GGTA1 and CMAH [28]. After deletion of GGTA1, CMAH and
natural preformed anti-Gal antibodies from the potential recipient’s
β4GalNT2, the immunoreactivity of pig bioprosthetic heart valves was
plasma. However, these methods have been proven to be only partially
significantly reduced [29]. Additionally, simultaneous inactivation of
successful because the antibodies can return rapidly, thereby leading to
these three genes dramatically attenuated the binding of human anti­
AHXR [16,17]. A more effective strategy is knockout of pig GGTA1 gene
bodies to pig renal tissues [30]. These data suggest that GGTA1/CMAH/
(GGTA1KO), representing a milestone event in the xenotransplantation
β4GalNT2 triple knockout provides much greater protection against
field [18]. It has been reported that using GGTA1KO pigs as a donor,
immune rejection. However, whether this genetic manipulation can
cardiac or renal graft survival in baboons is markedly prolonged
prolong kidney graft survival is still unclear and needs to be defined in
[19–21]. GGTA1KO almost completely eliminates Gal-mediated HAR
the future studies.
and thus acts as the basis for further genetic manipulations.
Other non-Gal antigens besides Neu5Gc and Sda are involved in
In addition to Gal antigens, there are a variety of non-Gal antigens on
AHXR [31]. The microvascular system, which is composed of arterioles,
the surface of pig cells [22,23]. It has been demonstrated that non-Gal
capillaries and venules, functions as a major target during AHXR [32].
antigens can also combine with Gal-specific antibodies to trigger

Fig. 1. Maximum survival of life-supporting pig kidney grafts in NHPs (1989–2019). After transplantation of pig kidneys into NHPs, maximum survival time of life-
supporting xenografts has significantly increased from 23 days in 1989 to 499 days in 2019. There were no reports in some years.

16
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

Recently, Zhang et al. used renal microvascular endothelial cells 2.4. Transgenic expression of human coagulation-regulatory proteins
(RMECs) and AECs from a GGTA1KO/CMAHKO pig to immunize cyn­
omolgus monkeys, and found that monkey serum antibody binding and Even after successful control of HAR, DXR and T cell response,
cytotoxicity to RMECs was significantly higher than to AECs [33]. These xenotransplantation is still confronted with thrombotic micro­
authors further revealed that 32 pig gene products were associated with angiopathy and/or consumptive coagulopathy, which can lead to
increased binding of monkey antibodies to RMECs, suggesting that these ischemic injury, graft failure and recipient death [49]. It is well known
proteins might be potential xenoantigens when pig kidneys are trans­ that coagulation dysfunction is predominately caused by (i) pig vascular
planted into NHPs or humans [33]. Future studies will be required to endothelial destruction due to antibody binding, complement deposition
determine which protein plays a critical role in mediating AHXR. This and innate immune cells (eg, monocytes), and (ii) molecular in­
may lead to further donor modification to reinforce resistance to AHXR compatibilities between the pig and primate coagulation-
and further decrease xenograft antigenicity. anticoagulation systems [50–52]. When pig vascular endothelial cells
are impaired, tissue factor (TF) is liberated into circulation and then
combines with clotting factor for thrombus formation [53].
2.2. Transgenic expression of human complement regulation proteins
To overcome this pathobiological barrier, a useful strategy is genetic
(CRPs)
modification of donor animals to express human coagulation-regulatory
proteins, such as ectonucleoside triphosphate diphosphohydrolase-1
Both HAR and AHXR are largely prevented through removal of Gal
(CD39), thrombomodulin (TBM), TF pathway inhibitor (TFPI), and
and non-Gal antigens, but activation of complement cascade still con­
endothelial protein C receptor (EPCR). CD39 is known to block throm­
tributes to immune rejection and xenograft failure [34]. Although pigs
bosis by degrading the platelet agonist ATP. A study by Dwyer et al.
have CRPs similar to humans, their CRPs are insufficient to protect xe­
showed that overexpression of hCD39 dramatically suppressed the
nografts from human complement-mediated injury [35]. Thus, intro­
clotting of human blood [54]. TBM has a stong anticoagulant effect.
duction of hCRPs into organ-source pigs is required to resolve this
Transgenic expression of hTBM was found to decrease thrombin gen­
problem. Indeed, transgenic expression of human decay accelerating
eration and increase coagulation time [55]. Several studies including
factor (hCD55) was shown to prolong pig kidney graft survival from
our own showed that additional hTBM expression prevented coagula­
days to weeks [36]. Prevention of complement activation by inserting
tion dysregulation and consequently contributed to long-term survival
human membrane cofactor protein (hCD46) into pig islets successfully
of cardiac grafts in a pig-to-baboon heterotopic heart transplantation
blocked antibody-mediated rejection and markedly improved the
model [56–58]. TFPI is the most important inhibitor of TF, but there is a
outcome of islet xenotransplantation in diabetic Cynomolgus monkeys,
molecular incompatibility between pig TFPI and human TF [59]. It was
maintaining normoglycemia for over 12 months [37]. Pig articular
reported that pig AECs expressing hTFPI were highly resistant to coag­
chondrocytes expressing human membrane inhibitor of reactive lysis
ulation triggered by antibody binding [60]. In addition, pig bone
(hCD59) were highly resistant to complement-mediated lysis [38].
marrow mesenchymal cells transfected with hTFPI exhibited a pro­
Moreover, insertion of two or three hCRPs provided greater protection
longed recalcification time, suggesting a potential approach to resolve
against complement activation than transgenic expression of a single
the incompatibility of pig TFPI [61].
hCRP [39,40].
EPCR not only has an anticoagulant effect, but also mediates anti-
Recently, Liu et al. generated GGTA1KO Diannan miniature pigs
inflammatory and cytoprotective signaling. Insertion of hEPCR into
expressing hCD55 and hCD59 using T2A-mediated polycistronic vector
pig AECs on a GGTA1KO/hCD46 background was reported to protect
systems and somatic cell nuclear transfer. They found that this triple-
against platelet aggregation [62]. Transgenic expression of hEPCR
gene modification was more successful in inhibiting human serum-
inhibited collagen-induced thrombosis and significantly increased sur­
mediated cytolysis than GGTA1KO alone [41]. Additionally,
vival time of heterotopic cardiac xenografts in the recipients [63]. When
GGTA1KO combined with additional expression of hCD46 or hCD55
a baboon with a kidney graft from a GGTA1KO pig expressing hCD46,
significantly reduced early kidney and heart graft failure in baboons
hCD55 and hEPCR functioned for 136 days until termination due to
[42,43]. Collectively, these observations strongly suggest that
septic shock, there was no evidence of a consumptive coagulopathy
GGTA1KO pig with one or two hCRPs are a more suitable donor for
[64]. Another study from our group showed that additional expression
kidney and other solid organ xenotransplantation.
of hEPCR protected pig kidney xenografts from consumptive coagulop­
athy [65]. These findings suggest that hEPCR overexpression is suffi­
2.3. Transgenic expression of human regulatory factors involving innate cient to avoid coagulation disregulation in kidney xenotransplantation.
or adaptive immune responses
2.5. Transgenic expression of human anti-inflammatory proteins
Cellular xenograft rejection usually occurs days to weeks after
transplantation and is caused by the recipient’s immune cells, including Systemic inflammatory response is another major obstacle to xeno­
natural killer cells, monocytes/macrophages, neutrophils, dendritic cells transplantation [66]. Studies from our team and others have demon­
and T lymphocytes [44]. At present, transgenic pigs that express human strated that a variety of pro-inflammatory cytokines and chemokines are
regulatory factors of innate or adaptive immune responses are available. generated after xenotransplantation [67,68]. Moreover, pro-
For instance, transgenic expression of hCD47, a species-specific immune inflammatory cytokines is able to facilitate coagulation activation,
inhibitory receptor on macrophages, significantly decreased the aggravate immune response, and enhance resistance to immunosup­
phagocytosis of pig endothelial cells and podocytes by baboon and pressive therapy [69–71]. Thus, blockade of systemic inflammation has
human macrophages [45]. This finding suggests that inserting hCD47 multiple beneficial effects on xenotransplantation.
into pig renal glomerular cells may be beneficial to inhibit proteinuria Although anti-inflammatory drugs, such as tocilizumab (IL-6 recep­
after kidney xenotransplantation. Surfactant protein D (SP-D), an oli­ tor blocker) and etanercept (anti-TNF-α agent), have been used to con­
gometric C type lectin, plays a critical role in promoting phagocytosis by trol systemic inflammatory response, their efficacy is not satisfactory in
binding to carbohydrates on microbial surfaces. Introduction of hSP-D pig-NHP-xenotransplantation models [72]. A preferable means is to
into pig endothelial cells was protective against THP-1 macrophage- insert one or more human anti-inflammatory proteins into donor pigs.
mediated phagocytosis [46]. Overexpression of hCD200 inhibited Both hemeoxygenase-1 (HO-1) and A20 play a central role in counter­
macrophage phagocytic and cytotoxic activities against pig endothelial acting inflammation. It was reported that the expression of intercellular
cells [47]. Also, PBMCs from hCD274 transgenic pigs had a significantly cell adhesion molecule-1, vascular cell adhesion molecule-1 and E-
reduced capacity to stimulate proliferation of human CD4+ T cells [48]. selectin was significantly decreased in pig AECs expressing hHO-1 [73].

17
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

Similarly, hA20-transgenic pig AECs showed a significant decrease in approved, the overall efficacy is limited because of their collateral effect
pro-inflammatory cytokine secretion [74]. Importantly, GGTA1KO of simultaneously blocking CTLA-4 coinhibitory signals [96]. As a
combined with transgenic expression of hHO-1 and hA20 protected pig promising alternative to abatacept and belatacept, selective CD28
kidneys from xenograft rejection during Ex vivo perfusion with human blockers, which directly bind to CD28 and do not impede normal coin­
blood [75]. This finding suggests that GGTA1KO/hHO-1/hA20 trans­ hibitory signals mediated by CTLA-4, have been applied to trans­
genic pigs could function as a background for further genetic manipu­ plantation. For instance, anti-CD28 monoclonal antibody (mAb) not
lations toward the production of a donor pig which is clinically relevant only preserved CTLA-4-dependent immune regulation but also inhibited
for kidney xenotransplantation. cardiac and renal graft rejection [97]. FR104 is a novel antagonist
pegylated anti-CD28 Fab’ antibody fragment. Administration of FR104
3. Potential strategies to reduce cross-species transmission of protected against steroid-resistant acute rejection, inhibited donor-
pig endogenous retroviruses (PERVs) specific antibody production, and improved graft survival in NHP kid­
ney allotransplantation model [98,99]. In addition, a humanized pegy­
The transmission of pig microorganism to human recipients repre­ lated anti-CD28 domain antibody termed lulizumab was shown to be
sents another major concern in the xenotransplantation field. Since the effective in preclinical models of allotransplantation [100,101]. More­
organ-source pigs are bred and housed under biosecure isolation con­ over, both FR104 and lulizumab were safe and well tolerated in healthy
ditions, the vast majority of pathogenic microorganisms can be elimi­ subjects [102,103].
nated from their body. However, PERVs are one exception because they CD40/CD40 ligand, also known as CD154, is another important
are integrated into the genome of every pig cell, and consequently will costimulation pathway. Interaction of CD154 with CD40 leads to den­
inevitably be transferred to the recipients with the transplanted organs, dritic cell licensing, B cell clonal expansion, and affinity maturation
tissues or cells [76]. Three replication-competent classes, namely PERV- toward B cell-specific antigen [104]. Blocking the CD40/CD154 axis has
A, PERV-B and PERV-C, have been identified. Both PERV-A and PERV-B been shown to promote long-term survival of pig hearts, kidneys and
are polytropic viruses and can infect pig and human cells, while PERV-C islets in NHPs [58,105,106]. However, anti-CD154 mAb has a risk of
is an ecotropic virus and only infects pig cells [77]. PERV-A/C, a re­ platelet activation and resultant thromboembolism [107,108]. Recently,
combinant form of PERV-A and PERV-C, has more potent infectivity a novel anti-human CD154 domain antibody (BMS-986004) lacking
toward human cells due to increased receptor binding [78]. PERVs have crystallizable fragment binding activity has been developed to avoid
been shown to infect several types of human cells in vitro, such as these adverse effects. In a NHP kidney transplant model, administration
PBMCs, human embryonic kidney cell line HEK-293, and normal human of BMS-986004 effectively blocked CD40/CD154 interaction and
dermal fibroblasts [79–81]. Despite accumulating evidence has markedly extended allograft survival, with no evidence of thromboem­
demonstrated that no transmission of PERVs occurs during preclinical bolism [109]. Moreove, BMS-986004 combined with conventional
and clinical xenotransplantation trials [82–84], a potential infection risk immunosuppressive therapy showed a synergistic role in controlling
should not be neglected when pig organs are transplanted to humans allograft rejection [109]. Thus, BMS-986004 may be a more promising
[85]. blocker of the CD40/CD154 costimulation pathway. There is also an
Several techniques are now available to inactivate PERVs. One is urgent need to determine whether application of BMS-986004 in xen­
RNA interference [86,87]. Using short hairpin RNA targeting pol gene, otransplantation has an efficacy and safety profile similar to
PERV infectivity was inhibited by 70%-80% [88]. Another is virus- allotransplantation.
neutralizing antibodies [89,90]. The third one is microRNAs (miRNAs) Histones are basic structural components of chromatin. However,
targeting various PERV genes [91]. A recent study showed that multi- they can be released into the circulation (known as serum histones) to
targeting miRNA against long terminal region was more successful in induce inflammatory response, coagulation disorder and cytotoxicity
inhibiting PERV replication than single-targeting miRNA [92]. Thus, [110,111]. Our research group reported that serum histone levels were
long terminal region might be a good candidate target for gene silencing significantly increased when baboons undergoing pig heart, liver or
of PERVs. The fourth one is CRISPR/Cas9. In 2015, Yang et al. reported kidney transplantation developed consumptive coagulopathy or infec­
that inactivation of pol gene using CRISPR/Cas9 led to a > 1000-fold tion, and showed a positive correlation with the levels of C-reactive
reduction in the transmission of PERVs to human cells [93]. Subse­ protein, a marker of inflammation [112]. Given that a systemic in­
quently, PERV-inactivated pigs were successfully produced by the same flammatory response in xenograft recipients precedes the the onset of
team [94]. This genetically-engineered pig can effectively avoid the coagulation disturbance [70,113], serum histones may be a potential
cross-species transmission of PERVs and thus serve as the basis for biomarker of systemic inflammation. We also found that administration
further genetic manipulations to provide a safe resource of tissues and of tocilizumab dramatically attenuated serum histone levels, and par­
organs for xenotransplantation. thenolide (an inhibitor of nuclear factor-κB) inhibited histone-induced
platelet aggregation and endothelial cell apoptosis in vitro [112].
4. Progress in immunosuppressive therapy These results suggest that pharmacologically targeting serum histones
could be valuable for controlling inflammatory response to a pig
In addition to gene editing in donor pigs, immunosuppressive ther­ xenograft.
apy in the recipients is necessary for successful xenotransplantation. T cell depletion by antibodies is commonly used as a potent immu­
Conventional immunosuppressive agents include tacrolimus, cyclo­ nosuppressive therapy in organ transplantation. Nevertheless, a resto­
sporine, mycophenolate mofetil, rapamycin, and corticosteroids, all of ration of T cell homeostasis after depletion therapy causes a significant
which are approved by the US Food and Drug Administration (FDA) for increase in memory T cells, thereby leading to graft failure [114,115].
clinical use [95]. To obtain a better efficacy, a number of novel immu­ IL-7 signals through IL-7 receptor (IL-7R), which consists of two chains:
nosuppressive drugs are currently available. α chain (IL-17Rα or CD127) and γ chain. This signaling pathway plays a
T cell activation, a complex process involving multiple cell signaling central role in the proliferation of both naive and memory CD4+ and
pathways, requires primary recognition signal and an additional cos­ CD8+ T cells [115,116]. Recently, Belarif et al. reported that IL-7R
timulatory signal. CD28 and cytotoxic T lymphocyte-associated protein blockade by an anti-IL-7Rα mAb blunted antigen-specific memory T
4 (CTLA4) are prototypal costimulatory and coinhibitory cell surface cell responses and alleviated skin inflammation in baboons [117].
signaling molecules, respectively. They belong to the members of the Importantly, administration of anti-IL-7Rα mAb after T cell depletion led
immunoglobulin superfamily and share the common ligand CD80/86. to marked prolongation of skin allograft survival [118]. Thus, T cell
Although blockade of CD28 costimulation by two CTLA-4 immuno­ depletion in combination with IL-7R blockade may provide greater
globulin fusion proteins (abatacept and belatacept) has been clinically protection to the transplanted tissues and organs than T cell depletion

18
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

alone. suggest that transgenic expression of hEPCR and/or hCD55 in the graft is
critical importance to avoid the occurrence of coagulation
5. Progress in pig-to-NHP kidney transplantation dysregulation.
In 2018, Adams et al. transplanted pig kidneys lacking GGTA1 and
The pig-to-NHP models have been used as the standard experimental β4GalNT2 into six rhesus monkeys with the least reactive cross matches
models in the xenotransplantation field. In 1989, using genetically- [105]. An anti-CD154 mAb-based immunosuppressive regimen was
unmodified (wild-type) pig kidneys, the longest survival time of life- administered to all animals. They found that three kidney xenografts
supporting kidney grafts was 23 days [4]. By 2004, the maximal sur­ were rejected within the first week, but other three ones obtained longer
vival of pig kidney grafts from hCD55 transgenic pigs had been extended survival (35, 100 and 435 days). Pathological analysis showed that
to 90 days in cynomolgus monkeys [119]. After GGTA1KO combined antibody-meditated rejection remained the major cause of graft failure.
with cotransplantation vascularized thymic tissue, pig kidney graft Although the reason for this difference in survival time remains to be
survival in baboons reached 83 days in 2005 [21]. Thereafter, pig-to- determined, a negative crossmatch between pigs and NHPs/human may
NHP kidney xenotransplantation showed only slow improvement until be an important contributor to further prolongation of kidney xenograft
2015 (Table 2). survival.
In 2015, Higginbotham et al. transplanted kidneys from GGTA1KO/ There is increasing evidence that CD4+ T cells play a more important
hCD55 pigs into rhesus macaques with low or high titers of anti-pig role in mediating xenograft rejection than CD8+ T cells [121,122]. More
antibodies [120]. All recipients were subject to T cell depletion using recently, Kim et al. used GGTA1KO/hCD55 pigs as donors and rhesus
anti-CD4 mAb, anti-CD8 mAb combined with costimulation blockade, macaques with low titers of anti-pig antibodies and selective depletion
either anti-CD154 mAb or belatacept. One recipient who received anti- of CD4+ or CD8+ T cells as recipients [123]. They found that kidney
CD154 mAb treatment and had high titers of anti-pig antibodies only xenograft survival was less than 15 days in the presence of CD8+ T cell
survived 6 days, while kidney xenograft survival in two lower-titer re­ depletion, while CD4+ T cell depletion markedly prolonged survival
cipients treated with belatacept was extended to 14 and 21 days. Sur­ time with the longest survival reported to date in a monkey (499 days).
prisingly, costimulation blockade by anti-CD154 mAb led to marked This indicates that CD4+ T cell depletion therapy prior to trans­
prolongation of kidney xenograft survival (>133 and >126 days) in two plantation may be necessary for consistent, long-term kidney xenograft
low-titer animals, without evidence of rejection on biopsies sampled at survival. In addition, their in vitro data demonstrated that the prolifer­
day 100. Thus, pre-transtplant antibody screening and anti-CD154 cos­ ation and cytotoxicity of rhesus CD4+ T cells were dependent on the
timulation blockade may be essential to long-term survival of pig kidney presence of swine leukocyte antigen (SLA) class II [123]. Thus, SLA class
grafts. At the same year, Iwase et al. transplanted a kidney from a II deletion in donor pigs may be an alternative strategy for selective
GGTA1KO/hCD46/hCD55/hTBM/hEPCR/hCD39 pig (both TBM and depletion of CD4+ T cells. Further in vivo studies are needed to confirm
CD39 were very poorly expressed in pig kidney) into a baboon, which this possibility.
received an anti-CD40 mAb-based immunosuppressive regimen and
anti-inflammatory therapy. They found that the baboon survived 136 6. Perspectives on the optimal genetically-modified pigs for
days with a generally stable serum creatinine until termination due to initial clinical kidney xenotransplantation trials
septic shock, with no consumptive coagulopathy, protein-losing ne­
phropathy and an elicited antibody response [64]. This finding suggests With the achievement of increasingly encouraging results in pig-to-
that a combination of multiple gene modifications, effective immuno­ NHP kidney xenotransplantation models during the past few years,
suppressive drugs and anti-inflammatory agents is of critical importance increasing attention is being paid towards initiating clinical trials in the
to prolong kidney xenograft survival. near future. There are now a number of genetically-modified pigs
In 2017, we achieved 237- and 260-day survival in two baboons who available for use as the organ-source, with some pigs expressing up to
underwent kidney transplantation from a GGTA1KO pig transgenic for nine genetic manipulations [6]. It is not clear, however, which combi­
hCD46/hCD55/hEPCR/hTFPI/hCD47 [65]. Immunosuppressive ther­ nation of genetic modifications will be necessary and/or adequate for an
apy was with anti-thymoglobulin (ATG) + anti-CD20 mAb (induction) initial clinical pig kidney transplantation. Further, there are controver­
and anti-CD40 mAb + rapamycin + corticosteroids (maintenance), sial reports on the utility and efficacy of multiple transgenes in pig-to-
which was accompanied by administration with anti-TNF-α and anti-IL- baboon kidney transplantation [124]. Thus, identification of optimal
6R mAbs to block systemic inflammation. Two animals were still healthy combination of genetic modifications in donor pigs is critical importance
with good graft function for >7–8 months, and then died of infectious to successfully initiate a clinical kidney xenotransplantation trial.
complications. There were no features of consumptive coagulopathy and Given that three known glycan antigens (Gal, Neu5Gc and Sda) on
protein-losing nephropathy. In contrast, two baboons with kidneys from pig vascular endothelial cells are regarded as major reasons leading to
a GGTA1KO/hCD46/hTBM pig lost their grafts within 12 days because humoral immune response and graft failure, GGTA1/CMAH/β4GalNT2
of a consumptive coagulopathy despite they received same immuno­ triple knockout should constitute the basis for any clinical trial. Ac­
suppressive and anti-inflammatory therapies. These observations cording to our own work and other studies, additional expression of

Table 2
Preclinical studies of pig-to-NHP kidney xenotransplantation in the past five years.
Year Donor pig Recipient Immunosuppressive regimen Survival time (days) Reference

2015 GGTA1KO/hCD46/hCD55/hTBM/hEPCR/ Baboon (n = 1) ATG, anti-CD20, anti-CD20, CVF, anti-CD40, 136 [64]
hCD39 rapamycin, MP
2015 GGTA1KO/hCD55 Rhesus macaques (n = 5) Anti-CD4, anti-CD8, anti-CD154/belatacept, MMF/ 6, 21, 14,>126, [120]
steroids >133
2017 GGTA1KO/hCD46/hCD55/hEPCR/hTFPI/ Baboon (n = 2) ATG, anti-CD20, CVF, anti-CD40, rapamycin, MP 237, 260 [65]
hCD47
GGTA1KO/hCD46/hTBM Baboon (n = 2) As above 12, 12
2018 GGTA1KO/β4GalNT2KO Rhesus macaques (n = 6) anti-CD4, anti-CD8, anti-CD154, MMF, steroids 5, 6, 6, 35, 100, 435 [105]
2019 GGTA1KO/hCD55 Rhesus macaques (n = anti-CD4/anti-CD8, anti-CD154, MMF, solumedrol 6 to 499 [123]
13)

CVF, cobra venom factor; MP, methylprednisolone; MMF, mycophenolate mofetil.

19
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

several protective human transgenic proteins will be important and References


beneficial to further prevent rejection from developing. First, inserting
hCD46 and hCD55 helps to protect kidney grafts from complement- [1] B. Ekser, D.K.C. Cooper, A.J. Tector, The need for xenotransplantation as a source
of organs and cells for clinical transplantation, Int. J. Surgery 23 (Pt B) (2015)
mediated injury. Second, transgenic expression of hEPCR is to sup­ 199–204.
press thrombotic microangiopathy in the graft and/or consumptive [2] M. Wijkstrom, H. Iwase, W. Paris, H. Hara, M. Ezzelarab, D.K. Cooper, Renal
coagulopathy in the recipient. Third, introduction of hCD47 is to inhibit xenotransplantation: experimental progress and clinical prospects, Kidney Int. 91
(4) (2017) 790–796.
cellular immune response. Fourth, both hHO-1 and hA20 are expressed [3] D.K. Cooper, B. Gollackner, D.H. Sachs, Will the pig solve the transplantation
to avoid systemic inflammatory response. Although there are a variety of backlog? Annu. Rev. Med. 53 (2002) 133–147.
alternative genetic modifications in donor pigs, we believe that a pig [4] D. Lambrigts, D.H. Sachs, D.K. Cooper, Discordant organ xenotransplantation in
primates: world experience and current status, Transplantation 66 (5) (1998)
with above nine genetic manipulations may be optimal as the source of 547–561.
kidneys for a initial clinical trial of xenotransplantation. [5] D.K. Cooper, B. Ekser, J. Ramsoondar, C. Phelps, D. Ayares, The role of
genetically engineered pigs in xenotransplantation research, J. Pathol. 238 (2)
(2016) 288–299.
7. Conclusion and future directions
[6] D.K.C. Cooper, H. Hara, H. Iwase, T. Yamamoto, Q. Li, M. Ezzelarab,
E. Federzoni, A. Dandro, D. Ayares, Justification of specific genetic modifications
Clinical xenotransplantation may provide an ultimate solution to in pigs for clinical organ xenotransplantation, Xenotransplantation 26 (4) (2019),
worldwide shortage of donor organs and is thus regarded as the next e12516.
[7] D.K.C. Cooper, H. Hara, H. Iwase, T. Yamamoto, A. Jagdale, V. Kumar, R.
great medical revolution [69,125]. With the application of gene editing B. Mannon, M.J. Hanaway, D.J. Anderson, D.E. Eckhoff, Clinical pig kidney
and novel immunosuppressive agents, A great advance has been made in xenotransplantation: how close are we? J. Am. Soc. Nephrology: JASN 31 (1)
pig-to NHP kidney transplantation in the past five years, with life- (2020) 12–21.
[8] D.K. Cooper, M.B. Ezzelarab, H. Hara, H. Iwase, W. Lee, M. Wijkstrom, R. Bottino,
supporting pig kidney graft survival extending beyond 1 year. This The pathobiology of pig-to-primate xenotransplantation: a historical review,
progress predominantly results from the increasing availability of donor Xenotransplantation 23 (2) (2016) 83–105.
pigs with multi-layered genetic modifications and the use of novel [9] T. Lu, B. Yang, R. Wang, C. Qin, Xenotransplantation: current status in preclinical
research, Front. Immunol. 10 (2019) 3060.
immunosuppressive agents, particularly costimulation blockers. It is [10] J.L. Platt, R.J. Fischel, A.J. Matas, S.A. Reif, R.M. Bolman, F.H. Bach,
hoped that clinical kidney xenotransplantation trials will become a re­ Immunopathology of hyperacute xenograft rejection in a swine-to-primate model,
ality in the near future. Transplantation 52 (2) (1991) 214–220.
[11] D.K. Cooper, A.H. Good, E. Koren, R. Oriol, A.J. Malcolm, R.M. Ippolito, F.
Undoubtedly, there are still many questions to be resolved before A. Neethling, Y. Ye, E. Romano, N. Zuhdi, Identification of alpha-galactosyl and
initiating such a trial. As mentioned above, a combination of nine ge­ other carbohydrate epitopes that are bound by human anti-pig antibodies:
netic manipulations should be optimal, possibly necessary, for a initial relevance to discordant xenografting in man, Transpl. Immunol. 1 (3) (1993)
198–205.
clinical pig kidney xenotransplantation. Although the beneficial effects
[12] A.H. Good, D.K. Cooper, A.J. Malcolm, R.M. Ippolito, E. Koren, F.A. Neethling,
of each individual manipulation is confirmed by considerable in vitro Y. Ye, N. Zuhdi, L.R. Lamontagne, Identification of carbohydrate structures that
and in vivo studies, whether such a combined manipulation affects the bind human antiporcine antibodies: implications for discordant xenografting in
health of organ-source pigs remains poorly understood and requires humans, Transpl. Proc. 24 (2) (1992) 559–562.
[13] U. Galili, S.B. Shohet, E. Kobrin, C.L. Stults, B.A. Macher, Man, apes, and Old
further characterization. Although PERV-inactivated pigs have been World monkeys differ from other mammals in the expression of alpha-galactosyl
produced, the efficacy and safety of PERV inactivation need to be epitopes on nucleated cells, J. Biol. Chem. 263 (33) (1988) 17755–17762.
evaluated in pig-to-NHP xenotransplantation models. Given that cos­ [14] U. Galili, R.E. Mandrell, R.M. Hamadeh, S.B. Shohet, J.M. Griffiss, Interaction
between human natural anti-alpha-galactosyl immunoglobulin G and bacteria of
timulation blockade agents are not approved by FDA for clinical use, it is the human flora, Infect. Immun. 56 (7) (1988) 1730–1737.
important to explore whether kidney xenograft rejection can be effec­ [15] U. Galili, The alpha-gal epitope and the anti-Gal antibody in xenotransplantation
tively inhibited by FDA-approved pharmacologic or biologic agents. and in cancer immunotherapy, Immunol. Cell Biol. 83 (6) (2005) 674–686.
[16] G. Gannedahl, G. Tufveson, B. Sundberg, C.G. Groth, The effect of plasmapheresis
Current clinical laboratory tests involving transplantation include as and deoxyspergualin or cyclophosphamide treatment on anti-porcine Gal-alpha
much as human leukocyte antigen (HLA) typing, donor-specific HLA (1,3)-Gal antibody titres in humans, Transpl. Proc. 28 (2) (1996) 551.
antibody and MICA testing, renal allograft pathology, blood test for [17] S. Taniguchi, F.A. Neethling, E.Y. Korchagina, N. Bovin, Y. Ye, T. Kobayashi,
M. Niekrasz, S. Li, E. Koren, R. Oriol, et al., In vivo immunoadsorption of antipig
cytomegalovirus, and urinalysis for BK virus infections. Improving the antibodies in baboons using a specific Gal(alpha)1–3Gal column, Transplantation
quality of these laboratory tests and developing novel assays is essential 62 (10) (1996) 1379–1384.
for clinical application of kidney xenotransplantation. During the past [18] C.J. Phelps, C. Koike, T.D. Vaught, J. Boone, K.D. Wells, S.H. Chen, S. Ball, S.
M. Specht, I.A. Polejaeva, J.A. Monahan, et al., Production of alpha 1,3-galacto­
decades, dual kidney transplantation has enabled greater use of mar­
syltransferase-deficient pigs, Science 299 (5605) (2003) 411–414.
ginal kidneys and reduced waiting time. Although there are no reports [19] K. Kuwaki, Y.L. Tseng, F.J. Dor, A. Shimizu, S.L. Houser, T.M. Sanderson, C.
on dual kidney transplantation in pig-to-NHP kidney xeno­ J. Lancos, D.D. Prabharasuth, J. Cheng, K. Moran, et al., Heart transplantation in
transplantation to date, future preclinical research should also explore baboons using alpha1,3-galactosyltransferase gene-knockout pigs as donors:
initial experience, Nat. Med. 11 (1) (2005) 29–31.
the feasibility of this transplantation. Answers to these questions will [20] Y.L. Tseng, K. Kuwaki, F.J. Dor, A. Shimizu, S. Houser, Y. Hisashi, K. Yamada, S.
provide insightful knowledge about xenotransplantation and contribute C. Robson, M. Awwad, H.J. Schuurman, et al., alpha1,3-Galactosyltransferase
to the success of initial clinical pig kidney transplantation trials. gene-knockout pig heart transplantation in baboons with survival approaching 6
months, Transplantation 80 (10) (2005) 1493–1500.
[21] K. Yamada, K. Yazawa, A. Shimizu, T. Iwanaga, Y. Hisashi, M. Nuhn, P. O’Malley,
Declaration of Competing Interest S. Nobori, P.A. Vagefi, C. Patience, et al., Marked prolongation of porcine renal
xenograft survival in baboons through the use of alpha1,3-galactosyltransferase
gene-knockout donors and the cotransplantation of vascularized thymic tissue,
The authors declare that they have no known competing financial Nat. Med. 11 (1) (2005) 32–34.
interests or personal relationships that could have appeared to influence [22] G.W. Byrne, P.G. Stalboerger, E. Davila, C.J. Heppelmann, M.H. Gazi, H.
the work reported in this paper. C. McGregor, P.T. LaBreche, W.R. Davies, V.P. Rao, K. Oi, et al., Proteomic
identification of non-Gal antibody targets after pig-to-primate cardiac
xenotransplantation, Xenotransplantation 15 (4) (2008) 268–276.
Acknowledgments [23] H.D. Tazelaar, G.W. Byrne, C.G. McGregor, Comparison of Gal and non-Gal-
mediated cardiac xenograft rejection, Transplantation 91 (9) (2011) 968–975.
[24] G. Chen, H. Qian, T. Starzl, H. Sun, B. Garcia, X. Wang, Y. Wise, Y. Liu, Y. Xiang,
This work was supported by grant from the major science and
L. Copeman, et al., Acute rejection is associated with antibodies to non-Gal
technology project of Hainan Province, China (ZDKJ2019009). antigens in baboons using Gal-knockout pig kidneys, Nat. Med. 11 (12) (2005)
1295–1298.
[25] K.H. Song, Y.J. Kang, U.H. Jin, Y.I. Park, S.M. Kim, H.H. Seong, S. Hwang, B.
S. Yang, G.S. Im, K.S. Min, et al., Cloning and functional characterization of pig
CMP-N-acetylneuraminic acid hydroxylase for the synthesis of N-

20
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

glycolylneuraminic acid as the xenoantigenic determinant in pig-human [48] A. Buermann, S. Petkov, B. Petersen, R. Hein, A. Lucas-Hahn, W. Baars,
xenotransplantation, Biochem. J. 427 (1) (2010) 179–188. A. Brinkmann, H. Niemann, R. Schwinzer, Pigs expressing the human inhibitory
[26] G.W. Byrne, Z. Du, P. Stalboerger, H. Kogelberg, C.G. McGregor, Cloning and ligand PD-L1 (CD 274) provide a new source of xenogeneic cells and tissues with
expression of porcine beta1,4 N-acetylgalactosaminyl transferase encoding a new low immunogenic properties, Xenotransplantation 25 (5) (2018), e12387.
xenoreactive antigen, Xenotransplantation 21 (6) (2014) 543–554. [49] L. Wang, D.K.C. Cooper, L. Burdorf, Y. Wang, H. Iwase, Overcoming coagulation
[27] B. Gao, C. Long, W. Lee, Z. Zhang, X. Gao, D. Landsittel, M. Ezzelarab, D. Ayares, dysregulation in pig solid organ transplantation in nonhuman primates: recent
Y. Huang, D.K.C. Cooper, et al., Anti-Neu5Gc and anti-non-Neu5Gc antibodies in progress, Transplantation 102 (7) (2018) 1050–1058.
healthy humans, PLoS ONE 12 (7) (2017), e0180768. [50] P.J. Cowan, S.C. Robson, A.J. d’Apice, Controlling coagulation dysregulation in
[28] J.L. Estrada, G. Martens, P. Li, A. Adams, K.A. Newell, M.L. Ford, J.R. Butler, xenotransplantation, Curr. Opin. Organ Transplant. 16 (2) (2011) 214–221.
R. Sidner, M. Tector, J. Tector, Evaluation of human and non-human primate [51] M. Schmelzle, J. Schulte Esch 2nd, S.C. Robson, Coagulation, platelet activation
antibody binding to pig cells lacking GGTA1/CMAH/beta4GalNT2 genes, and thrombosis in xenotransplantation, Curr. Opin. Organ Transplant. 15 (2)
Xenotransplantation 22 (3) (2015) 194–202. (2010) 212–218.
[29] R. Zhang, Y. Wang, L. Chen, R. Wang, C. Li, X. Li, B. Fang, X. Ren, M. Ruan, J. Liu, [52] C.C. Lin, D. Chen, J.H. McVey, D.K. Cooper, A. Dorling, Expression of tissue factor
et al., Reducing immunoreactivity of porcine bioprosthetic heart valves by and initiation of clotting by human platelets and monocytes after incubation with
genetically-deleting three major glycan antigens, GGTA1/beta4GalNT2/CMAH, porcine endothelial cells, Transplantation 86 (5) (2008) 702–709.
Acta Biomater. 72 (2018) 196–205. [53] C.C. Lin, D.K. Cooper, A. Dorling, Coagulation dysregulation as a barrier to
[30] R.G. Wang, M. Ruan, R.J. Zhang, L. Chen, X.X. Li, B. Fang, C. Li, X.Y. Ren, J. xenotransplantation in the primate, Transpl. Immunol. 21 (2) (2009) 75–80.
Y. Liu, Q. Xiong, et al., Antigenicity of tissues and organs from GGTA1/CMAH/ [54] K.M. Dwyer, T.B. Mysore, S. Crikis, S.C. Robson, H. Nandurkar, P.J. Cowan, A.
beta4GalNT2 triple gene knockout pigs, J. Biomed. Res. (2018). J. D’Apice, The transgenic expression of human CD39 on murine islets inhibits
[31] G.W. Byrne, P.G. Stalboerger, Z. Du, T.R. Davis, C.G. McGregor, Identification of clotting of human blood, Transplantation 82 (3) (2006) 428–432.
new carbohydrate and membrane protein antigens in cardiac [55] H. Kim, W.J. Hawthorne, H.J. Kang, Y.J. Lee, J.I. Hwang, S. Hurh, H. Ro, J.
xenotransplantation, Transplantation 91 (3) (2011) 287–292. C. Jeong, B. Cho, J. Yang, et al., Human thrombomodulin regulates complement
[32] M.P. Revelo, D.V. Miller, J. Stehlik, K. Brunisholz, S. Drakos, E.M. Gilbert, activation as well as the coagulation cascade in xeno-immune response,
M. Everitt, D. Budge, R. Alharethi, G. Snow, et al., Longitudinal evaluation of Xenotransplantation 22 (4) (2015) 260–272.
microvessel density in survivors vs. nonsurvivors of cardiac pathologic antibody- [56] A.K. Singh, J.L. Chan, L. DiChiacchio, N.L. Hardy, P.C. Corcoran, B.G.T. Lewis, M.
mediated rejection, Cardiovascular Pathol.: Off. J. Soc. Cardiovasc. Pathol. 21 (6) L. Thomas, A.P. Burke, D. Ayares, K.A. Horvath, et al., Cardiac xenografts show
(2012) 445–454. reduced survival in the absence of transgenic human thrombomodulin expression
[33] J. Zhang, C. Xie, Y. Lu, M. Zhou, Z. Qu, D. Yao, C. Qiu, J. Xu, D. Pan, Y. Dai, et al., in donor pigs, Xenotransplantation 26 (2) (2019), e12465.
Potential antigens involved in delayed xenograft rejection in a Ggta1/Cmah Dko [57] H. Iwase, B. Ekser, V. Satyananda, J. Bhama, H. Hara, M. Ezzelarab, E. Klein,
pig-to-monkey model, Sci. Rep. 7 (1) (2017) 10024. R. Wagner, C. Long, J. Thacker, et al., Pig-to-baboon heterotopic heart
[34] D. Pan, T. Liu, T. Lei, H. Zhu, Y. Wang, S. Deng, Progress in multiple genetically transplantation–exploratory preliminary experience with pigs transgenic for
modified minipigs for xenotransplantation in China, Xenotransplantation 26 (1) human thrombomodulin and comparison of three costimulation blockade-based
(2019), e12492. regimens, Xenotransplantation 22 (3) (2015) 211–220.
[35] J.P. Atkinson, T.J. Oglesby, D. White, E.A. Adams, M.K. Liszewski, Separation of [58] M.M. Mohiuddin, A.K. Singh, P.C. Corcoran, M.L. Thomas Iii, T. Clark, B.G. Lewis,
self from non-self in the complement system: a role for membrane cofactor R.F. Hoyt, M. Eckhaus, R.N. Pierson, A.J. Belli, et al., Chimeric 2C10R4 anti-CD40
protein and decay accelerating factor, Clin. Exp. Immunol. 86 (Suppl 1) (1991) antibody therapy is critical for long-term survival of GTKO.hCD46.hTBM pig-to-
27–30. primate cardiac xenograft, Nat. Commun. 7 (11138) (2016).
[36] E. Cozzi, C. Vial, D. Ostlie, B. Farah, G. Chavez, K.G. Smith, J.R. Bradley, S. Thiru, [59] J. Schulte am Esch, 2nd, X. Rogiers, S.C. Robson, Molecular incompatibilities in
H.F. Davies, J. Wallwork, et al., Maintenance triple immunosuppression with hemostasis between swine and men–impact on xenografting. Ann. Transplant. 6
cyclosporin A, mycophenolate sodium and steroids allows prolonged survival of (3) (2001) 12–16.
primate recipients of hDAF porcine renal xenografts, Xenotransplantation 10 (4) [60] C.C. Lin, M. Ezzelarab, H. Hara, C. Long, C.W. Lin, A. Dorling, D.K. Cooper,
(2003) 300–310. Atorvastatin or transgenic expression of TFPI inhibits coagulation initiated by
[37] D.J. van der Windt, R. Bottino, A. Casu, N. Campanile, C. Smetanka, J. He, anti-nonGal IgG binding to porcine aortic endothelial cells, J. Thrombosis
N. Murase, H. Hara, S. Ball, B.E. Loveland, et al., Long-term controlled Haemostasis: JTH 8 (9) (2010) 2001–2010.
normoglycemia in diabetic non-human primates after transplantation with [61] H. Ji, X. Li, S. Yue, J. Li, H. Chen, Z. Zhang, B. Ma, J. Wang, M. Pu, L. Zhou, et al.,
hCD46 transgenic porcine islets, Am. J. Transplant.: Off. J. Am. Soc. Transplant. Pig BMSCs transfected with human TFPI combat species incompatibility and
Am. Soc. Transplant Surgeons 9 (12) (2009) 2716–2726. regulate the human TF pathway in vitro and in a rodent model, Cell. Physiol.
[38] R. Sommaggio, M. Perez-Cruz, J.L. Brokaw, R. Manez, C. Costa, Inhibition of Biochem.: Int. J. Exp. Cell. Physiol., Biochem., Pharmacol. 36 (1) (2015)
complement component C5 protects porcine chondrocytes from xenogeneic 233–249.
rejection, Osteoarthritis Cartilage 21 (12) (2013) 1958–1967. [62] H. Iwase, B. Ekser, H. Hara, C. Phelps, D. Ayares, D.K. Cooper, M.B. Ezzelarab,
[39] C.Y. Zhou, E. McInnes, L. Copeman, G. Langford, N. Parsons, R. Lancaster, Regulation of human platelet aggregation by genetically modified pig endothelial
A. Richards, C. Carrington, S. Thompson, Transgenic pigs expressing human cells and thrombin inhibition, Xenotransplantation 21 (1) (2014) 72–83.
CD59, in combination with human membrane cofactor protein and human decay- [63] K.F. Lee, B. Lu, J.C. Roussel, L.J. Murray-Segal, E.J. Salvaris, S.J. Hodgkinson, B.
accelerating factor, Xenotransplantation 12 (2) (2005) 142–148. M. Hall, A.J. d’Apice, P.J. Cowan, H. Gock, Protective effects of transgenic human
[40] S. Miyagawa, R. Shirakura, K. Iwata, S. Nakata, G. Matsumiya, H. Izutani, endothelial protein C receptor expression in murine models of transplantation,
H. Matsuda, A. Terado, M. Matsumoto, S. Nagasawa, et al., Effects of transfected Am. J. Transplant.: Off. J. Am. Soc. Transplant. Am. Soc. Transplant Surgeons 12
complement regulatory proteins, MCP, DAF, and MCP/DAE hybrid, on (9) (2012) 2363–2372.
complement-mediated swine endothelial cell lysis, Transplantation 58 (7) (1994) [64] H. Iwase, H. Liu, M. Wijkstrom, H. Zhou, J. Singh, H. Hara, M. Ezzelarab, C. Long,
834–840. E. Klein, R. Wagner, et al., Pig kidney graft survival in a baboon for 136 days:
[41] F. Liu, J. Liu, Z. Yuan, Y. Qing, H. Li, K. Xu, W. Zhu, H. Zhao, B. Jia, W. Pan, et al., longest life-supporting organ graft survival to date, Xenotransplantation 22 (4)
Generation of GTKO diannan miniature pig expressing human complementary (2015) 302–309.
regulator proteins hCD55 and hCD59 via T2A peptide-based bicistronic vectors [65] H. Iwase, H. Hara, M. Ezzelarab, T. Li, Z. Zhang, B. Gao, H. Liu, C. Long, Y. Wang,
and SCNT, Mol. Biotechnol. 60 (8) (2018) 550–562. A. Cassano, et al., Immunological and physiological observations in baboons with
[42] A.M. Azimzadeh, S.S. Kelishadi, M.B. Ezzelarab, A.K. Singh, T. Stoddard, life-supporting genetically engineered pig kidney grafts, Xenotransplantation 24
H. Iwase, T. Zhang, L. Burdorf, E. Sievert, C. Avon, et al., Early graft failure of (2) (2017).
GalTKO pig organs in baboons is reduced by expression of a human complement [66] J. Li, H. Hara, Y. Wang, C. Esmon, D.K.C. Cooper, H. Iwase, Evidence for the
pathway-regulatory protein, Xenotransplantation 22 (4) (2015) 310–316. important role of inflammation in xenotransplantation, J. Inflamm. 16 (2019) 10.
[43] C.G. McGregor, D. Ricci, N. Miyagi, P.G. Stalboerger, Z. Du, E.A. Oehler, H. [67] H. Iwase, B. Ekser, H. Zhou, H. Liu, V. Satyananda, R. Humar, P. Humar, H. Hara,
D. Tazelaar, G.W. Byrne, Human CD55 expression blocks hyperacute rejection C. Long, J.K. Bhama, et al., Further evidence for sustained systemic inflammation
and restricts complement activation in Gal knockout cardiac xenografts, in xenograft recipients (SIXR), Xenotransplantation 22 (5) (2015) 399–405.
Transplantation 93 (7) (2012) 686–692. [68] C.A. Labarrere, J.R. Woods, J.W. Hardin, B.R. Jaeger, M. Zembala, M.C. Deng, G.
[44] S. Le Bas-Bernardet, G. Blancho, Current cellular immunological hurdles in pig-to- S. Kassab, Early inflammatory markers are independent predictors of cardiac
primate xenotransplantation, Transpl. Immunol. 21 (2) (2009) 60–64. allograft vasculopathy in heart-transplant recipients, PLoS ONE 9 (12) (2014),
[45] S. Nomura, Y. Ariyoshi, H. Watanabe, T. Pomposelli, K. Takeuchi, G. Garcia, e113260.
M. Tasaki, D. Ayares, M. Sykes, D. Sachs, et al., Transgenic expression of human [69] B. Ekser, M. Ezzelarab, H. Hara, D.J. van der Windt, M. Wijkstrom, R. Bottino,
CD47 reduces phagocytosis of porcine endothelial cells and podocytes by baboon M. Trucco, D.K. Cooper, Clinical xenotransplantation: the next medical
and human macrophages, Xenotransplantation 27 (1) (2020), e12549. revolution? Lancet 379 (9816) (2012) 672–683.
[46] P. Jiaravuthisan, A. Maeda, C. Takakura, H.T. Wang, R. Sakai, A.M. Shabri, P. [70] M.B. Ezzelarab, B. Ekser, A. Azimzadeh, C.C. Lin, Y. Zhao, R. Rodriguez, G.
C. Lo, R. Matsuura, T. Kodama, H. Eguchi, et al., A membrane-type surfactant J. Echeverri, H. Iwase, C. Long, H. Hara, et al., Systemic inflammation in
protein D (SP-D) suppresses macrophage-mediated cytotoxicity in swine xenograft recipients precedes activation of coagulation, Xenotransplantation 22
endothelial cells, Transpl. Immunol. 47 (2018) 44–48. (1) (2015) 32–47.
[47] J.J. Yan, T.Y. Koo, H.S. Lee, W.B. Lee, B. Kang, J.G. Lee, J.Y. Jang, T. Fang, J. [71] H. Gao, L. Liu, Y. Zhao, H. Hara, P. Chen, J. Xu, J. Tang, L. Wei, Z. Li, D.K.
H. Ryu, C. Ahn, et al., Role of human CD200 overexpression in pig-to-human C. Cooper, et al., Human IL-6, IL-17, IL-1beta, and TNF-alpha differently regulate
xenogeneic immune response compared with human CD47 overexpression, the expression of pro-inflammatory related genes, tissue factor, and swine
Transplantation 102 (3) (2018) 406–416. leukocyte antigen class I in porcine aortic endothelial cells, Xenotransplantation
24 (2) (2017).

21
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

[72] H. Iwase, H. Liu, T. Li, Z. Zhang, B. Gao, H. Hara, M. Wijkstrom, C. Long, R. Saari, [97] N. Poirier, A.M. Azimzadeh, T. Zhang, N. Dilek, C. Mary, B. Nguyen, X. Tillou,
D. Ayares, et al., Therapeutic regulation of systemic inflammation in xenograft G. Wu, K. Reneaudin, J. Hervouet, et al., Inducing CTLA-4-dependent immune
recipients, Xenotransplantation 24 (2) (2017). regulation by selective CD28 blockade promotes regulatory T cells in organ
[73] B. Petersen, W. Ramackers, A. Lucas-Hahn, E. Lemme, P. Hassel, A.L. Queisser, transplantation, Sci. Transl. Med. 2 (17) (2010), 17ra10.
D. Herrmann, B. Barg-Kues, J.W. Carnwath, J. Klose, et al., Transgenic expression [98] N. Poirier, N. Dilek, C. Mary, S. Ville, F. Coulon, J. Branchereau, X. Tillou,
of human heme oxygenase-1 in pigs confers resistance against xenograft rejection V. Charpy, S. Pengam, V. Nerriere-Daguin, et al., FR104, an antagonist anti-CD28
during ex vivo perfusion of porcine kidneys, Xenotransplantation 18 (6) (2011) monovalent fab’ antibody, prevents alloimmunization and allows calcineurin
355–368. inhibitor minimization in nonhuman primate renal allograft, Am. J. Transplant.:
[74] M. Oropeza, B. Petersen, J.W. Carnwath, A. Lucas-Hahn, E. Lemme, P. Hassel, Off. J. Am. Soc. Transplant. Am. Soc. Transplant Surgeons 15 (1) (2015) 88–100.
D. Herrmann, B. Barg-Kues, S. Holler, A.L. Queisser, et al., Transgenic expression [99] S. Ville, N. Poirier, J. Branchereau, V. Charpy, S. Pengam, V. Nerriere-Daguin,
of the human A20 gene in cloned pigs provides protection against apoptotic and S. Le Bas-Bernardet, F. Coulon, C. Mary, A. Chenouard, et al., Anti-CD28 antibody
inflammatory stimuli, Xenotransplantation 16 (6) (2009) 522–534. and belatacept exert differential effects on mechanisms of renal allograft
[75] H.E. Ahrens, B. Petersen, W. Ramackers, S. Petkov, D. Herrmann, J. Hauschild- rejection, J. Am. Soc. Nephrol.: JASN 27 (12) (2016) 3577–3588.
Quintern, A. Lucas-Hahn, P. Hassel, M. Ziegler, W. Baars, et al., Kidneys from [100] D. Liu, S.M. Krummey, I.R. Badell, M. Wagener, L.A. Schneeweis, D.K. Stetsko, S.
alpha1,3-galactosyltransferase knockout/human heme oxygenase-1/human A20 J. Suchard, S.G. Nadler, M.L. Ford, 2B4 (CD244) induced by selective CD28
transgenic pigs are protected from rejection during ex vivo perfusion with human blockade functionally regulates allograft-specific CD8+ T cell responses, J. Exp.
blood, Transplantation direct 1 (6) (2015), e23. Med. 211 (2) (2014) 297–311.
[76] C. Patience, W.M. Switzer, Y. Takeuchi, D.J. Griffiths, M.E. Goward, W. Heneine, [101] S.J. Suchard, P.M. Davis, S. Kansal, D.K. Stetsko, R. Brosius, J. Tamura,
J.P. Stoye, R.A. Weiss, Multiple groups of novel retroviral genomes in pigs and L. Schneeweis, J. Bryson, T. Salcedo, H. Wang, et al., A monovalent anti-human
related species, J. Virol. 75 (6) (2001) 2771–2775. CD28 domain antibody antagonist: preclinical efficacy and safety, J. Immunol.
[77] Y. Takeuchi, C. Patience, S. Magre, R.A. Weiss, P.T. Banerjee, P. Le Tissier, J. 191 (9) (2013) 4599–4610.
P. Stoye, Host range and interference studies of three classes of pig endogenous [102] N. Poirier, G. Blancho, M. Hiance, C. Mary, T. Van Assche, J. Lempoels, S. Ramael,
retrovirus, J. Virol. 72 (12) (1998) 9986–9991. W. Wang, V. Thepenier, C. Braudeau, et al., First-in-human study in healthy
[78] I. Harrison, Y. Takeuchi, B. Bartosch, J.P. Stoye, Determinants of high titer in subjects with FR104, a pegylated monoclonal antibody fragment antagonist of
recombinant porcine endogenous retroviruses, J. Virol. 78 (24) (2004) CD28, J. Immunol. 197 (12) (2016) 4593–4602.
13871–13879. [103] R. Shi, M. Honczarenko, S. Zhang, C. Fleener, J. Mora, S.K. Lee, R. Wang, X. Liu,
[79] V. Specke, S. Rubant, J. Denner, Productive infection of human primary cells and D.E. Shevell, Z. Yang, et al., Pharmacokinetic, pharmacodynamic, and safety
cell lines with porcine endogenous retroviruses, Virology 285 (2) (2001) profile of a novel anti-CD28 domain antibody antagonist in healthy subjects,
177–180. J. Clin. Pharmacol. 57 (2) (2017) 161–172.
[80] S. Yue, Y. Zhang, Y. Gao, A study on the susceptibility of allogeneic human [104] A. Mardomi, N. Mohammadi, H.T. Khosroshahi, S. Abediankenari, An update on
hepatocytes to porcine endogenous retrovirus, Eur. Rev. Med. Pharmacol. Sci. 19 potentials and promises of T cell co-signaling molecules in transplantation, J. Cell.
(18) (2015) 3486–3491. Physiol. 235 (5) (2020) 4183–4197.
[81] M.W. Kimsa, B. Strzalka-Mrozik, M.C. Kimsa, U. Mazurek, C. Kruszniewska-Rajs, [105] A.B. Adams, S.C. Kim, G.R. Martens, J.M. Ladowski, J.L. Estrada, L.M. Reyes,
J. Gola, J. Adamska, M. Twardoch, Differential expression of tripartite motif- C. Breeden, A. Stephenson, D.E. Eckhoff, M. Tector, et al., Xenoantigen deletion
containing family in normal human dermal fibroblasts in response to porcine and chemical immunosuppression can prolong renal xenograft survival, Ann.
endogenous retrovirus infection, Folia Biol. 60 (3) (2014) 144–151. Surg. 268 (4) (2018) 564–573.
[82] J. Denner, Why was PERV not transmitted during preclinical and clinical [106] J.S. Shin, J.M. Kim, B.H. Min, I.H. Yoon, H.J. Kim, J.S. Kim, Y.H. Kim, S.J. Kang,
xenotransplantation trials and after inoculation of animals? Retrovirology 15 (1) J. Kim, H.J. Kang, et al., Pre-clinical results in pig-to-non-human primate islet
(2018) 28. xenotransplantation using anti-CD40 antibody (2C10R4)-based
[83] V.A. Morozov, S. Wynyard, S. Matsumoto, A. Abalovich, J. Denner, R. Elliott, No immunosuppression, Xenotransplantation 25 (1) (2018).
PERV transmission during a clinical trial of pig islet cell transplantation, Virus [107] I. Koyama, T. Kawai, D. Andrews, S. Boskovic, O. Nadazdin, S.L. Wee, H. Sogawa,
Res. 227 (2017) 34–40. D.L. Wu, R.N. Smith, R.B. Colvin, et al., Thrombophilia associated with anti-
[84] L. Scobie, V. Padler-Karavani, S. Le Bas-Bernardet, C. Crossan, J. Blaha, CD154 monoclonal antibody treatment and its prophylaxis in nonhuman
M. Matouskova, R.D. Hector, E. Cozzi, B. Vanhove, B. Charreau, et al., Long-term primates, Transplantation 77 (3) (2004) 460–462.
IgG response to porcine Neu5Gc antigens without transmission of PERV in burn [108] T. Kawai, D. Andrews, R.B. Colvin, D.H. Sachs, A.B. Cosimi, Thromboembolic
patients treated with porcine skin xenografts, J. Immunol. 191 (6) (2013) complications after treatment with monoclonal antibody against CD40 ligand,
2907–2915. Nat. Med. 6 (2) (2000) 114.
[85] M. Guell, D. Niu, Y. Kan, H. George, T. Wang, I.H. Lee, G. Wang, G. Church, [109] S.C. Kim, W. Wakwe, L.B. Higginbotham, D.V. Mathews, C.P. Breeden, A.
L. Yang, PERV inactivation is necessary to guarantee absence of pig-to-patient C. Stephenson, J. Jenkins, E. Strobert, K. Price, L. Price, et al., Fc-silent anti-
PERVs transmission in xenotransplantation, Xenotransplantation 24 (6) (2017). CD154 domain antibody effectively prevents nonhuman primate renal allograft
[86] J. Ramsoondar, T. Vaught, S. Ball, M. Mendicino, J. Monahan, P. Jobst, A. Vance, rejection, Am. J. Transplant.: Off. J. Am. Soc. Transplant. Am. Soc. Transplant
J. Duncan, K. Wells, D. Ayares, Production of transgenic pigs that express porcine Surgeons 17 (5) (2017) 1182–1192.
endogenous retrovirus small interfering RNAs, Xenotransplantation 16 (3) (2009) [110] Y. Alhamdi, C.H. Toh, Recent advances in pathophysiology of disseminated
164–180. intravascular coagulation: the role of circulating histones and neutrophil
[87] A. Karlas, R. Kurth, J. Denner, Inhibition of porcine endogenous retroviruses by extracellular traps, F1000Research 6 (2143) (2017).
RNA interference: increasing the safety of xenotransplantation, Virology 325 (1) [111] J. Xu, X. Zhang, R. Pelayo, M. Monestier, C.T. Ammollo, F. Semeraro, F.B. Taylor,
(2004) 18–23. N.L. Esmon, F. Lupu, C.T. Esmon, Extracellular histones are major mediators of
[88] B. Dieckhoff, B. Petersen, W.A. Kues, R. Kurth, H. Niemann, J. Denner, death in sepsis, Nat. Med. 15 (11) (2009) 1318–1321.
Knockdown of porcine endogenous retrovirus (PERV) expression by PERV- [112] T. Li, W. Lee, H. Hara, C. Long, M. Ezzelarab, D. Ayares, H. Huang, Y. Wang, C.
specific shRNA in transgenic pigs, Xenotransplantation 15 (1) (2008) 36–45. T. Esmon, D.K.C. Cooper, et al., An investigation of extracellular histones in pig-
[89] A. Waechter, M. Eschricht, J. Denner, Neutralization of porcine endogenous to-baboon organ xenotransplantation, Transplantation 101 (10) (2017)
retrovirus by antibodies against the membrane-proximal external region of the 2330–2339.
transmembrane envelope protein, J. General Virol. 94 (Pt 3) (2013) 643–651. [113] M.B. Ezzelarab, D.K.C. Cooper, Systemic inflammation in xenograft recipients
[90] A. Waechter, J. Denner, Novel neutralising antibodies targeting the N-terminal (SIXR): a new paradigm in pig-to-primate xenotransplantation? Int. J. Surgery 23
helical region of the transmembrane envelope protein p15E of the porcine (Pt B) (2015) 301–305.
endogenous retrovirus (PERV), Immunol. Res. 58 (1) (2014) 9–19. [114] Z. Wu, S.J. Bensinger, J. Zhang, C. Chen, X. Yuan, X. Huang, J.F. Markmann,
[91] H.C. Chung, V.G. Nguyen, H.J. Moon, H.K. Kim, S.J. Park, J.H. Lee, M.G. Choi, A. A. Kassaee, B.R. Rosengard, W.W. Hancock, et al., Homeostatic proliferation is a
R. Kim, B.K. Park, Inhibition of porcine endogenous retrovirus in PK15 cell line by barrier to transplantation tolerance, Nat. Med. 10 (1) (2004) 87–92.
efficient multitargeting RNA interference, Transplant Int.: Off. J. Eur. Soc. Organ [115] D.C. Neujahr, C. Chen, X. Huang, J.F. Markmann, S. Cobbold, H. Waldmann, M.
Transplant. 27 (1) (2014) 96–105. H. Sayegh, W.W. Hancock, L.A. Turka, Accelerated memory cell homeostasis
[92] H.C. Chung, V.G. Nguyen, W.T. Oh, T.M. Huynh, H.J. Moon, J.H. Lee, H.K. Kim, during T cell depletion and approaches to overcome it, J. Immunol. 176 (8)
S.J. Park, B.K. Park, Inhibition of porcine endogenous retrovirus by multi- (2006) 4632–4639.
targeting micro RNA against long terminal region, Transpl. Proc. 49 (9) (2017) [116] J. Sprent, C.D. Surh, Normal T cell homeostasis: the conversion of naive cells into
2225–2232. memory-phenotype cells, Nat. Immunol. 12 (6) (2011) 478–484.
[93] L. Yang, M. Guell, D. Niu, H. George, E. Lesha, D. Grishin, J. Aach, E. Shrock, [117] L. Belarif, C. Mary, L. Jacquemont, H.L. Mai, R. Danger, J. Hervouet, D. Minault,
W. Xu, J. Poci, et al., Genome-wide inactivation of porcine endogenous V. Thepenier, V. Nerriere-Daguin, E. Nguyen, et al., IL-7 receptor blockade blunts
retroviruses (PERVs), Science 350 (6264) (2015) 1101–1104. antigen-specific memory T cell responses and chronic inflammation in primates,
[94] D. Niu, H.J. Wei, L. Lin, H. George, T. Wang, I.H. Lee, H.Y. Zhao, Y. Wang, Y. Kan, Nat. Commun. 9 (1) (2018) 4483.
E. Shrock, et al., Inactivation of porcine endogenous retrovirus in pigs using [118] H.L. Mai, F. Boeffard, J. Longis, R. Danger, B. Martinet, F. Haspot, B. Vanhove,
CRISPR-Cas9, Science 357 (6357) (2017) 1303–1307. S. Brouard, J.P. Soulillou, IL-7 receptor blockade following T cell depletion
[95] K.L. Hardinger, D.C. Brennan, Novel immunosuppressive agents in kidney promotes long-term allograft survival, J. Clin. Investig. 124 (4) (2014)
transplantation, World J. Transplant. 3 (4) (2013) 68–77. 1723–1733.
[96] R.L. Crepeau, M.L. Ford, Challenges and opportunities in targeting the CD28/ [119] N. Baldan, P. Rigotti, F. Calabrese, R. Cadrobbi, A. Dedja, I. Iacopetti, M. Boldrin,
CTLA-4 pathway in transplantation and autoimmunity, Expert Opin. Biol. Ther. M. Seveso, L. Dall’Olmo, L. Frison, et al., Ureteral stenosis in HDAF pig-to-primate
17 (8) (2017) 1001–1012. renal xenotransplantation: a phenomenon related to immunological events? Am.

22
X.-H. Yu et al. Clinica Chimica Acta 514 (2021) 15–23

J. Transplant.: Off. J. Am. Soc. Transplant. Am. Soc. Transplant Surgeons 4 (4) [123] S.C. Kim, D.V. Mathews, C.P. Breeden, L.B. Higginbotham, J. Ladowski,
(2004) 475–481. G. Martens, A. Stephenson, A.B. Farris, E.A. Strobert, J. Jenkins, et al., Long-term
[120] L. Higginbotham, D. Mathews, C.A. Breeden, M. Song, A.B. Farris 3rd, C.P. Larsen, survival of pig-to-rhesus macaque renal xenografts is dependent on CD4 T cell
M.L. Ford, A.J. Lutz, M. Tector, K.A. Newell, et al., Pre-transplant antibody depletion, Am. J. Transplant.: Off. J. Am. Soc. Transplant. Am. Soc. Transplant
screening and anti-CD154 costimulation blockade promote long-term xenograft Surgeons 19 (8) (2019) 2174–2185.
survival in a pig-to-primate kidney transplant model, Xenotransplantation 22 (3) [124] S. Le Bas-Bernardet, X. Tillou, N. Poirier, N. Dilek, M. Chatelais, J. Devalliere,
(2015) 221–230. B. Charreau, D. Minault, J. Hervouet, K. Renaudin, et al., Xenotransplantation of
[121] R.N. Pierson 3rd, H.J. Winn, P.S. Russell, H. Auchincloss Jr., Xenogeneic skin galactosyl-transferase knockout, CD55, CD59, CD39, and fucosyl-transferase
graft rejection is especially dependent on CD4+ T cells, J. Exp. Med. 170 (3) transgenic pig kidneys into baboons, Transpl. Proc. 43 (9) (2011) 3426–3430.
(1989) 991–996. [125] D.K.C. Cooper, Introduction: the present status of xenotransplantation research,
[122] R.N. Pierson 3rd, H.J. Winn, P.S. Russell Jr., H. Auchincloss Jr., CD4+ Methods Mol. Biol. 2110 (2020) 1–25.
lymphocytes play a dominant role in murine xenograft rejection, Transpl. Proc. 21
(1 Pt 1) (1989) 519.

23

You might also like