Accepted Manuscript Current status of pig liver xenotransplantation Burcin Ekser, MD, PhD, James F. Markmann, MD, PhD, A.Joseph Tector, MD, PhD PII:

S1743-9191(15)00373-8

DOI:

10.1016/j.ijsu.2015.06.083

Reference:

IJSU 2018

To appear in:

International Journal of Surgery

Received Date: 26 June 2015 Accepted Date: 28 June 2015

Please cite this article as: Ekser B, Markmann JF, Tector AJ, Current status of pig liver xenotransplantation, International Journal of Surgery (2015), doi: 10.1016/j.ijsu.2015.06.083. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

CURRENT STATUS OF PIG LIVER XENOTRANSPLANTATION

PhD1

RI PT

Burcin Ekser, MD, PhD1, James F. Markmann, MD, PhD2, A. Joseph Tector, MD,

(1) Transplant Division, Department of Surgery, Indiana University School of

Medicine, Indianapolis, IN, USA; (2) Division of Transplantation, Department of

M AN U

SC

Surgery, Massachusetts General Hospital, Boston, MA, USA

Short Title: Liver xenotransplantation

Address correspondence to: Burcin Ekser, MD, PhD

TE D

Transplant Division, Department of Surgery, Indiana University School of Medicine, 550 University Blvd, Room 4601 Indianapolis, IN, USA

EP

Telephone: 317-944-4370; Fax 317-948-3268

AC C

Email: [email protected]

KEY WORDS: acute liver failure, liver, pig, nonhuman primate, xenotransplantation

(Word counts: Abstract 161; Text 1,967, Tables 1; Figures 5)

ACCEPTED MANUSCRIPT

ABBREVIATIONS β4GalNT2 = beta-1,4-N-acetyl galactosaminyl transferase 2

GTKO = α1,3-galactosyltransferase gene knock-out hDAF = human decay-accelerating factor LSEC = liver sinusoidal endothelial cell

SC

MELD= model for end-stage liver disease

TE D

CONFLICT OF INTEREST

M AN U

Neu5Gc = N-glycolylneuraminic acid WT = wild-type

RI PT

GE = genetically-engineered

AC C

EP

The authors declare no conflict of interest.

1

ACCEPTED MANUSCRIPT

ABSTRACT

The shortage of organs from deceased human donors is a major problem limiting the

RI PT

number of organs transplanted each year and results in the death of thousands of

patients on the waiting list. Pigs are currently the preferred species for clinical organ xenotransplantation. Progress in genetically-engineered (GE) pig liver

SC

xenotransplantation increased graft and recipient survival from hours with unmodified pig livers to up to 9 days with normal to near-normal liver function. Deletion of genes

M AN U

such as GGTA1 (Gal-knockout pigs) or adding genes such as human complement regulatory proteins (hCD55, hCD46 expressing pigs) enabled hyperacute rejection to be overcome. Although survival up to 9 days was recorded, extended pig graft survival was not achieved due to lethal thrombocytopenia. The current status of GE pig liver

TE D

xenotransplantation with world experience, potential factors causing thrombocytopenia, new targets on pig endothelial cells, and novel GE pigs with more genes deletion to avoid remaining antibody response, such as beta1,4-N-acetyl galactosaminyl

AC C

EP

transferase 2 (β4GalNT2), are discussed.

2

ACCEPTED MANUSCRIPT

INTRODUCTION

Liver transplantation is the only curative therapy for end-stage liver disease. One-year,

RI PT

3-year, and 5-year patient survival rates post liver transplantation are 90%, >80%, and >70%, respectively [1]. The current problem with liver transplantation like any other organ transplantation is organ shortage. In the USA, in February 2015, the United

SC

Network for Organ Sharing waiting list for liver transplantation approached 16,000

patients. This number remains unchanged in the last decade [2]. In 2014, 3,165 patients

M AN U

were removed from the waiting list due to death (n= 1,553) or because they became too sick to transplant for the surgical procedure (n=1,612) [2]. In the last 20 years, the total number of patients removed from the liver waiting list without receiving a transplant is

TE D

more than 51,000 [2].

Xenotransplantation, using pig organs, could resolve the shortage of suitable donor organs [3-5]. We have reviewed the current status of pig liver xenotransplantation with

EP

recent advances in genetically-engineered (GE) pigs and especially in preclinical trials

AC C

with nonhuman primates.

PROGRESS WITH GENETICALLY-ENGINEERED PIGS First studies in experimental preclinical pig-to-nonhuman primate liver xenotransplantation were performed by Calne et al in 1968 [6] using livers from unmodified (wild-type, WT) pigs. The status of WT pig liver xenotransplantation was discussed elsewhere [7-9]. Using GE pigs in xenotransplantation is more limited and not

3

ACCEPTED MANUSCRIPT

as encouraging as other GE pig organ or cell xenotransplantation, though it cleverly raised graft survival from hours with WT pigs to up to 9 days with GE pigs. The world

RI PT

experience of GE pig liver xenotransplantation is shown in Table 1.

The first report of GE pig-to-nonhuman primate liver xenotransplantation was by

Ramirez et al [10], where human CD55 (decay-accelerating factor, DAF) expressing

SC

pigs were used. Two baboon recipients survived 4 and 8 days, respectively. One

baboon recipient died due to the aspiration pneumonia on day 4 and the other baboon

M AN U

died due to sepsis and coagulopathy on post-transplant day 8 [10]. By the end of experiments, there were significant increases in direct and total bilirubin (5.4 and 18.9 mg/dl, respectively), indicating cholestatis. In the 8-day survivor baboon, an acute rejection episode on day 2 was treated with three boluses of methylprednisolone [10].

TE D

Thrombocytopenia was evident starting from day 1 in both recipients. On post-operative day 8, the platelet count was 34,000/mm3, associated with a significant reduction in fibrinogen and an elevation of d-dimer, suggesting the development of consumptive

EP

coagulopathy. Histology of grafts showed no evidence of rejection in the 4-day survivor, but there was evidence of some patchy focal ischemia in the 8-day survivor [10]. The

AC C

same group subsequently reported the use of livers from pigs transgenic for CD55, CD59, and H-transferase [11]. The five recipient baboons survived for only 13 to 24 hours.

The first experience using GTKO (α1,3-galactosyltransferase gene knock-out) pig livers was reported by the Pittsburgh team [12]. Ten orthotopic liver xenotransplants were

4

ACCEPTED MANUSCRIPT

performed in baboons, with two livers being obtained from GTKO pigs and eight from GTKO pigs transgenic for human CD46 (GTKO.hCD46). The Pittsburgh group emphasized the importance of size matching between pig liver and recipient baboon in

RI PT

which the pig should be almost 60% smaller in weight than the recipient baboon [12]. Six of 10 baboons survived for 4 to 7 days with normal to near-normal liver function (except for some cholestasis), as evidenced by tests of detoxification, complement

SC

activity, coagulation parameters, and production of coagulation factors (Figure 1) [13]. Increased bilirubin levels were observed as in the Ramirez et al.’s study [10], but this

M AN U

was not associated with mechanical obstruction since viscous bile was present in bile ducts [14]. Kobayashi et al. [15] showed that although human and pig hepatic bile showed similarities, baboon bile is less viscous, containing lower levels of sodium, potassium, calcium, and magnesium, as well as less cholesterol and bilirubin, with

TE D

significantly higher levels of sodium bicarbonate. These observations indicate that bile viscosity seen in pig-to-baboon liver xenotransplants will not be a problem after pig-tohuman liver xenotransplants. All baboons eventually died or euthanized due to severe

EP

spontaneous bleeding caused by lethal thrombocytopenia (Figure 2) [14, 16].

AC C

More recent GTKO pig-to-baboon liver xenotransplantation studies come from the Boston group. Kim et al were able to achieve 9-day survival using GTKO pig livers in baboons (Table 1) [17]. Three transplanted baboons survived 6, 8, and 9 days. The Boston group preferred to use the same immunosuppressive regimen as in their previous studies, including thymoglobulin and cobra venom factor induction with antiCD154mAb and tacrolimus maintenance (Table 1). The administration of Amicar

5

ACCEPTED MANUSCRIPT

(aminocaproic acid) in 2 cases led to the partial resolution of thrombocytopenia, maintaining platelet counts over 40,000/mm3 throughout the study (Figure 2). Good liver function (normal to near-normal transaminases, coagulation parameters with

RI PT

coagulation factors) was documented. However, both recipients died due to bleeding and enterococcal infection. In all three cases, there was no evidence of rejection [17].

SC

In a more recent study by Yeh et al, the Boston group studied the contribution of

anticoagulant production and clotting pathway deficiencies to fatal bleeding using

M AN U

auxiliary GTKO pig liver xenotransplantation in baboons (Table 1) [18]. The donor liver was transplanted in the left upper quadrant by anastomosing the donor portal vein to the confluence of the recipient inferior mesenteric vein and splenic vein (ligating the splenic vein distally post-splenectomy) and the donor infrahepatic inferior vena cava to the

TE D

recipient left renal vein and inferior vena cava confluence. The arterial supply of the auxiliary pig liver was achieved by anastomosing the donor infrahepatic aorta to the recipient infrarenal aorta [18]. There were 3 auxiliary liver xenotransplants with 6, 9, and

EP

15 days of graft survival. One recipient didn’t receive thymoglobulin induction in order to reduce the infectious complications, however lost the xenograft on day 6 post-transplant

AC C

due to severe acute cellular rejection. This particular baboon recipient survived with his native liver post pig auxiliary liver graftectomy. Interestingly, severe thrombocytopenia recovered immediately after xenograft removal [18]. Pig coagulation factors and proand anti-coagulant levels were measured and compared with orthotopic liver xenotransplantation from the same group [17]. The conclusion was that massive hemorrhage in the setting of orthotopic pig liver xenotransplantation might be avoided

6

ACCEPTED MANUSCRIPT

by supplementation with primate clotting components. However, the remaining native liver continues to be the source of antibodies and coagulation factors which makes the

RI PT

model very challenging to make a solid conclusion [18].

The same group recently published a study where they sought to determine the effects of exogenous administration of human coagulation factors infusing human Factor VIIa

SC

or human prothrombin concentrate complex in GTKO pig-to-baboon liver

xenotransplantation model [19]. In the control baboon which received no human

M AN U

coagulation factor, thrombocytopenia was evident throughout the study. Interestingly, the control baboon survived 6 days, however 2 baboons with human prothrombin concentrate complex infusion survived 2 and 4 days. Baboons (n=3) receiving human Factor VIIa infusion survived 6, 6, and 7 days, respectively. Two of them recovered

TE D

platelet counts at the end of the study but eventually died on post-transplant day 6. The longest survived baboon (7 days) with human Factor VIIa infusion had prominent thrombocytopenia at the end of the study [19]. In 3 baboons with human Factor VIIa

EP

infusion, the histopathology showed minimal to mild inflammation with preserved hepatic architecture on post-operative day 4. Patchy and centrilobular necrosis (10%)

AC C

was also observed with concerns for rejection in one baboon [19]. Unfortunately, there was no data on immunosuppressive regimen and any major bleeding due to thrombocytopenia [19].

FACTORS INFLUENCING THE DEVELOPMENT OF THROMBOCYTOPENIA

7

ACCEPTED MANUSCRIPT

In vivo, ex vivo perfusion, and in vitro studies by the Pittsburgh, Boston, and Indianapolis groups identified several factors influencing the development of survival

RI PT

limiting thrombocytopenia and consumptive coagulopathy in GE pig liver xenografts.

The Pittsburgh group found; (i) tissue factor on recipient platelets and PBMCs

(peripheral blood mononuclear cells) (possibly not with activation of donor endothelial

SC

cells) initiates thrombocytopenia and consumptive coagulopathy independently than immune response starting from 2h post-reperfusion (Figure 3) [16], (ii) no form of

M AN U

rejection in 2h biopsies excluding hyperacute rejection, and biopsies at necropsy showed hemorrhagic necrosis, platelet aggregation, platelet-fibrin thrombi, monocyte/macrophage margination mainly in liver sinusoids, and vascular endothelial cell hypertrophy, minimal deposition of IgM, and a near absence of IgG, C3, C4d, C5b-

TE D

9, or cellular infiltrate, suggesting that neither antibody- nor cell-mediated rejection played a major role [12, 14, 16], and (iii) pig von Willebrand factor (vWF) expression on the hepatic vascular endothelium in 2h biopsies and at euthanasia in GTKO pig livers

EP

and also in WT pig liver when hyperacute rejection occurred [16].

AC C

The Indianapolis group identified; (i) human platelet phagocytosis by pig liver sinusoidal endothelial cells (LSEC) and hepatocytes (Figure 4) [20] via asialoglycoprotein receptor 1 (ASGR1) [21, 22], (ii) differences in human and porcine platelet oligosaccharides influencing platelet phagocytosis by LSECs in vitro [23], and (iii) involvement of CD18 receptor in platelet phagocytosis by porcine Kupffer cells [24]. They also confirmed previous finding of interspecies incompatibilities in CD47/Signal Regulatory Protein

8

ACCEPTED MANUSCRIPT

alpha (SIRPα) self-signaling impacting human platelet uptake by pig LSECs (Figure 5) [25, 26].

RI PT

The Boston group, besides confirming the normal to near-normal liver function with good coagulation post GE pig liver xenotransplantation in nonhuman primates [17], showed (i) the partial and complete recovery of thrombocytopenia using aminocapric

SC

acid [17] and human Factor VIIa infusion (Figure 2) [19]. They also found that massive hemorrhage but not thrombocytopenia post pig liver xenotransplantation could be

M AN U

partially ameliorated with auxillary GE pig liver xenotransplantation [18]. (iii) In an in vitro study [27], they confirmed baboon platelet phagocytosis by pig aortic endothelial cells, which was previously shown by the Indianapolis group [20]. However, the pathway

TE D

identified in this work [27] was integrin adhesion pathway involving vWF and MAC-1.

Other significant works published by the Toledo group showing that blocking porcine sialoadhesin improves extracorporeal porcine liver xenoperfusion with human blood

EP

[28], and by the Maryland group in extracorporeal pig-to-baboon liver xenoperfusion model revealing that glycoprotein Ib receptor blockade through anti-GPIb antibody and

AC C

D-arginine vasopressin improve thrombocytopenia [29].

CONCLUSION AND FUTURE VIEW ON CLINICAL PERSPECTIVE Several groups demonstrated good liver function and the production of porcine proteins and coagulation factors post GE pig liver xenotransplantation in nonhuman primates

9

ACCEPTED MANUSCRIPT

with minimal evidence of rejection [10, 12, 13, 17]. Xenograft survival has been limited by lethal thrombocytopenia and coagulopathy [12, 17, 30] causing hemorrhage in native

RI PT

organs as well as the xenografted liver [12, 14, 17].

Recent in vitro and in vivo studies have shed a light on understanding of

thrombocytopenia post pig liver xenotransplantation. Major hemorrhage and/or necrosis

SC

of pig livers causes limited graft survival despite partial or complete resolution of thrombocytopenia via aminocaproic acid [17] or human Factor VIIa infusion [19]

M AN U

indicates that (i) recipient platelets, despite fully present in the peripheral circulation, become non-functional with a shear-stress via porcine LSECs either via ASGR1 receptor or interspecies incompatibility of CD47/SIRPα and/or CD18, MAC-1, vWF/GPIb, or both, and (ii) despite anti-Gal antibodies and complement-related

TE D

immunological injury are mainly avoided via GTKO pigs and human complement regulatory protein expressing pigs, other preformed antibodies (e.g. anti-nonGal antibodies) against proteins, such as β4GalNT2 (beta-1,4-N-acetyl galactosaminyl

EP

transferase 2) and Neu5Gc (N-glycolylneuraminic acid) may be sufficient to induce an innate immune response, therefore causing an antibody mediated injury in the

AC C

xenograft.

With growing evidence on ASGR1 receptor involvement in xenogeneic platelet phagocytosis in vitro and in a human-to-pig ex vivo perfusion model using porcine forelimbs and livers by Bongoni et al [31], an ASGR1 knockout pig-to-nonhuman primate liver xenotransplantation experiment is of great interest. Recent advancement in

10

ACCEPTED MANUSCRIPT

generating multi-gene knockout pigs, such as GGTA1/CMAH/β4GalNT2 genes [32] using a CRISPR/Cas9 technology [33], or multi-gene expressing pigs such as human CD46, human CD55, human Thrombomodulin, human CD39 [34] will reduce antibody

RI PT

binding and may correct consumptive coagulopathy. New preclinical studies in GE pigto-nonhuman primate liver xenotransplantation are warranted using newly available GE pigs, such as ASGR1 knockout, β4GalNT2 knockout, and human SIRPα transgenic

M AN U

SC

pigs.

Funding None.

TE D

Ethical approval None.

Research Registration Not required for review article.

EP

Author contribution

AC C

BE, JFM, AJT – all participated in review of the literature, writing of the manuscript, and final approval of the manuscript

Guarantor

Burcin Ekser, MD, PhD

11

ACCEPTED MANUSCRIPT

REFERENCES

[1] OPTN - Organ Procurement and Transplantation Network.

RI PT

http://optn.transplant.hrsa.gov (last accessed on February 10th, 2015).

[2] UNOS - United Network for Organ Sharing (UNOS). http://www.unos.org (last accessed on February 10th, 2015).

M AN U

backlog? Annu. Rev. Med. 53 (2002) 133–147.

SC

[3] D.K. Cooper, B. Gollackner, D.H. Sachs, Will the pig solve the transplantation

[4] B. Ekser, M. Ezzelarab, H. Hara, D.J. van der Windt, M. Wijkstrom, R. Bottino, et al., Clinical xenotransplantation: the next medical revolution? Lancet. 379 (2012) 672-683.

[5] B. Ekser, B. Gridelli, A.J. Tector, D.K. Cooper, Pig liver xenotransplantation as a bridge to allotransplantation: which patients might benefit? Transplantation. 88 (2009)

TE D

1041-1049.

[6] R.Y. Calne, H.J. White, B.M. Herbertson, et al., Pig to baboon liver xenografts,

EP

Lancet. 7553 (1968) 1176-1178.

[7] H. Hara, B. Gridelli, Y.J. Lin, A. Marcos, D.K.C. Cooper, Liver xenografts for the

AC C

treatment of acute liver failure: clinical and experimental experience and remaining immunological barriers, Liver Transplant. 14 (2008) 425-434.

[8] B. Ekser, B. Gridelli, M. Veroux, D.K.C. Cooper, Clinical pig liver xenotransplantation: how far do we have to go? Xenotransplantation. 18 (2011) 158167.

[9] D.K. Cooper, V. Satyananda, B. Ekser, D.J. van der Windt, H. Hara, M.B. Ezzelarab, et al., Progress in pig-to-non-human primate transplantation models (1998-2013): a

12

ACCEPTED MANUSCRIPT

comprehensive review of the literature, Xenotransplantation. 21 (2014) 397-419.

[10] P. Ramirez, R. Chavez, M. Majado, et al., Life-supporting human complement regulator decay accelerating factor transgenic pig liver xenograft maintains the

RI PT

metabolic function and coagulation in the nonhuman primate for up to 8 days, Transplantation. 70 (2000) 989-998.

[11] P. Ramirez, M.J. Montoya, A. Rios, et al., Prevention of hyperacute rejection in a

SC

model of orthotopic liver xenotransplantation from pig to baboon using polytransgenic

M AN U

pig livers (CD55, CD59, and H-transferase), Transplant. Proc. 37 (2005) 4103-4106.

[12] B. Ekser, C. Long, G.J. Echeverri, et al., Impact of thrombocytopenia on survival of baboons with genetically-modified pig liver transplants: clinical relevance, Am. J. Transplant. 10 (2010) 273-285.

[13] B. Ekser, G.J. Echeverri, A.C. Hassett, M.H. Yazer, C. Long, M. Meyer, et al.,

TE D

Hepatic function after genetically engineered pig liver xenotransplantation in baboons, Transplantation. 90 (2010) 483-493.

[14] B. Ekser, E. Klein, J. He, et al., Genetically-engineered pig-to-baboon liver

AC C

(2012) e29720.

EP

xenotransplantation: histopathology of xenografts and native organs, PLoS ONE. 7

[15] T. Kobayashi, S. Taniguchi, Y. Ye, et al., Comparison of bile chemistry between humans, baboons, and pigs: implications for clinical and experimental liver xenotransplantation, Lab. Animal. Sci. 48 (1998) 197-200.

[16] B. Ekser, C.C. Lin, C. Long, et al., Potential factors influencing the development of thrombocytopenia and consumptive coagulopathy after genetically modified pig liver xenotransplantation, Transplant Int. 25 (2012) 882-896.

13

ACCEPTED MANUSCRIPT

[17] K. Kim, C. Schuetz, N. Elias, G.R. Veillette, I. Wamala, M. Varma, et al, Up to 9-day survival and control of thrombocytopenia following alpha1,3-galactosyl transferase knockout swine liver xenotransplantation in baboons, Xenotransplantation. 19 (2012)

RI PT

256-264.

[18] H. Yeh, Z. Machaidze, I. Wamala, J.W. Fraser, N. Navarro-Alvarez, K. Kim, et al., Increased transfusion-free survival following auxiliary pig liver xenotransplantation,

SC

Xenotransplantation. 21 (2014) 454-464.

[19] J. Shah, N. Navarro-Alvarez, H. Yeh, et al., Infusion of human Factor 7a restores

M AN U

circulating platelets and prevents TMA in pig-to-baboon liver xenotransplantation, Am. J. Transplant. 15 Suppl 3 (2015) (Abstract# 70).

[20] C. Burlak, L.L. Paris, R.K. Chihara, et al., The fate of human platelets perfused through the pig liver: implications for xenotransplantation, Xenotransplantation. 17

TE D

(2010) 350-361.

[21] L.L. Paris, R.K. Chihara, R.A. L.M. Reyes, et al., ASGR1 expressed by procine enriched liver sinusoidal endothelial cells mediates human platelet phagocytosis in vitro,

EP

Xenotransplantation. 18 (2011) 245-251.

[22] L.L. Paris, J.L. Estrada, P. Li, et al., Reduced human platelet uptake by pig livers

AC C

deficient in the asialoglycoprotein receptor 1 protein, Xenotransplantation. 22 (2015) 203-210.

[23] L.L. Paris, R.K. Chihara, R.A. Sidner, A.J. Tector, C. Burlak, Differences in human and porcine platelet oligosaccharides may influence phagocytosis by liver sinusoidal cells in vitro, Xenotransplantation. 19 (2012) 31-39.

14

ACCEPTED MANUSCRIPT

[24] R.K. Chihara, L.L. Paris, L.M. Reyes, et al., Primary porcine Kupffer cell phagocytosis of human platelets involves the CD18 receptor, Transplantation. 92 (2011) 739-744.

RI PT

[25] L.L. Paris, L.M. Reyes, A.J. Tector, SIRPalpha interspecies incompatibilities lead to xenogeneic phagocytosis of platelets by liver cells, Xenotransplantation. 18 (2011) Abstract# 411

M AN U

Xenotransplantation. 17 (2010) 267-273.

SC

[26] Y.G. Yang, CD47 in xenograft rejection and tolerance induction,

[27] Q. Peng, H.Yeh, L.Wei, et al., Mechanisms of xenogeneic baboon platelet aggregation and phagocytosis by porcine liver sinusoidal endothelial cells, PLoS ONE. 7 (2012) e47273.

[28] J.P. Waldman, T. Vogel, C. Burlak, et al., Blocking porcine sialoadhesin improves

(2013) 239-251.

TE D

extracorporeal porcine liver xenoperfusion with human blood, Xenotransplantation. 20

[29] J.C. LaMattina, L. Burdorf, T. Zhang, et al., Pig-to-baboon liver xenoperfusion

AC C

(2014) 274-286.

EP

utilizing GalTKO.hCD46 pigs and glycoprotein Ib blockade, Xenotransplantation. 21

[30] M. Ezzelarab, B. Ekser, B. Gridelli, et al., Thrombocytoepnia after pig-t-baboon liver xenotransplantation: where do platelets go? Xenotransplantation. 18 (2011) 320-327.

[31] A.K. Bongoni, D. Kiermeir, J. Denoyelle, et al., Porcine extrahepatic vascular endothelial asialoglycoprotein receptor 1 mediates xenogeneic platelet phagocytosis in vitro and in vivo human-to-pig ex vivo xenoperfusion, Transplantation. 99 (2015) 693701.

15

ACCEPTED MANUSCRIPT

[32] J.L. Estrada, G. Martens, P. Li, et al., Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/βGalNT2 genes, Xenotransplantation. 22 (2015) 194-202.

RI PT

[33] P. LI, J.L Estrada, C. Burlak, et al., Efficient generation of genetically distinc pigs in a single pregnancy using multiplexed single-guide RNA and carbonhydrate selection, Xenotransplantation. 22 (2015) 20-31.

SC

[34] H. Iwase, H. Liu, M. Wijkstrom, H. Zhou, J. Singh, H. Hara, et al., Pig kidney graft survival in a baboon for 136 days: longest life-supporting organ graft survival to date,

M AN U

Xenotransplantation. (2015) in press.

[35] B. Ekser, C. Burlak, J.P. Waldman, et al., Immunobiology of liver

AC C

EP

TE D

xenotransplantation, Expert. Rev. Clin. Immunol. 8 (2012) 621-634.

16

ACCEPTED MANUSCRIPT

Table 1: World experience of preclinical GE pig-to-nonhuman primate liver xenotransplantation Recipient

Type of

N. of

Tx

Tx

Immunosuppression

Survival

Author

Year

Ref

(hours)

RI PT

Pig Type

Baboon

Orthotopic

2

CyP+CsA+Cs

hCD55.CD59.

Baboon

Orthotopic

5

CyP+Daclizumab+Rituximab+

HT

CsA+MMF+Cs

SC

hCD55

#

96, 192

Ramirez

2000

10

13, 18, 20, 21,

Ramirez

2005

11

24

Baboon

Orthotopic

2

ATG+Tacrolimus+MMF+Cs

3, 144

Ekser

2010

12

GTKO.hCD46

Baboon

Orthotopic

8

ATG+Tacrolimus+MMF+Cs

3, 20, 24, 96,

Ekser

2010

12

(n=5),

120, 144, 144,

144, 192, 216

Kim

2012

17

144, 216, 360

Yeh

2014

18

48, 96, 144,

Shah

2015

19

M AN U

GTKO

CyP+Tacrolimus+MMF+Cs (n=3) Baboon

Orthotopic

3

ATG+LoCD2b+CVF+anti-

TE D

GTKO

168

CD154mAb+Azathioprine+Cs

Baboon

Auxiliary

3

GTKO

Baboon

Orthotopic

6

ATG+CVF+Tacrolimus+Cs Not available

144, 144, 168

AC C

EP

GTKO

Legend: ATG = anti-thymocyte globulin, CD46 = membrane cofactor protein, CD55 = decay accelerating factor, CD59 = homologous restriction factor (protectin), Cs = corticosteroids, CsA = cyclosporine, CyP = cyclophosphamide, GTKO = α1,3-galactosyltransferase gene-knockout, h = human, HT = H-transferase (α1,2-fucosyltransferase), MMF = mycophenolate mofetil, Tx = transplantation, WT = wild-type.

17

ACCEPTED MANUSCRIPT

Figure Legend: Figure 1: Evidence of genetically-engineered pig liver function in nonhuman primates. A) alanine transaminase pre- and post-transplant in pig-to-baboon liver

RI PT

xenotransplantation model; B) International normalized ratio pre- and post-liver

xenotransplantation; C) Coagulation factor Factor V in pre- and post-transplant in pig-tobaboon liver xenotransplantation model with pig (P), baboon (B), and human (H)

average values; D) Western blotting assay to show pig proteins in baboon blood.

SC

(Obtained with permission from Ekser et al. Transplantation. 90 (2010) 483-493 [13]).

M AN U

Figure 2: Platelet counts after in vivo orthotopic GE pig liver xenotransplantation. Platelet counts after pig-to-baboon liver xenotransplantation: Ramirez et al [10]; average of two WT.hCD55 pig liver xenotransplantations. Ekser et al [12]; average of WT (n=1), GTKO (n=2), and GTKO.hCD46 (n=8) pig liver xenotransplantation. Kim et al [17]; average of 3 GTKO pig liver xenotransplantation (estimated from Kim et al. [17]). Shah et al [19]; average of 6 GTKO pig liver xenotransplantation (with available data from the

TE D

abstract)

Figure 3: Tissue factor activity on recipient platelets and PBMCs. Tissue factor activity was measured using the recalcified clotting assay pre-

EP

transplantation (TX) and 2h after reperfusion of the pig liver (2h) (# = p

Current status of pig liver xenotransplantation.

The shortage of organs from deceased human donors is a major problem limiting the number of organs transplanted each year and results in the death of ...
998KB Sizes 0 Downloads 5 Views