Hanyang Med Rev.  2014 Nov;34(4):197-201. 10.7599/hmr.2014.34.4.197.

Clinical Immune Tolerance in Liver Transplantation: Present and Future

Affiliations
  • 1Division of transplantation, Department of Surgery, Samsung Medical Center, School of Meidicine Sunkyunkwan University, Seoul, Korea. medi9370@gmail.com

Abstract

Liver transplantation is the most effective treatment for end-stage liver diseases (ESLD) with satisfactory clinical results and so is considered as the treatment of choice for ESLD and early hepatocellular carcinoma with cirrhotic liver. Unfortunately, adverse effects of life-long immunosuppression prevent the development of alternative strategies to achieve better long-term outcome. Achieving clinical operational tolerance is one of the ultimate goals in the clinical transplantation field. Around 15% of liver transplantation recipients develop spontaneous operational tolerance after immunosuppression withdrawal, and the percentage may be even higher in pediatric living donor liver transplantation recipients. One of the possible explainable mechanisms is a T cell fatigue from large amount of antigen loaded. Despite continuing progress, clinical operational tolerance is still rare in liver transplantation. Reprogramming the recipient immune system by creating chimerism and utilizing regulatory cell therapies are among the newer promising means to achieve clinical liver transplantation tolerance in the future. In animal studies, administration of donor specific regulatory T cells allows a prolonged survival without immunosuppressive agents. In this review, proposed mechanisms for clinical tolerance will be offered and current experimental trial will be introduced.

Keyword

Liver Transplantation; Immune Tolerance; End Stage Liver Disease

MeSH Terms

Animals
Carcinoma, Hepatocellular
Chimerism
End Stage Liver Disease
Fatigue
Humans
Immune System
Immune Tolerance*
Immunosuppression
Immunosuppressive Agents
Liver
Liver Diseases
Liver Transplantation*
Living Donors
T-Lymphocytes, Regulatory
Tissue Donors
Immunosuppressive Agents

Cited by  1 articles

Cutting Edge Technologies in Organ Transplantation
Dongho Choi
Hanyang Med Rev. 2014;34(4):143-144.    doi: 10.7599/hmr.2014.34.4.143.


Reference

1. Calne RY, Sells RA, Pena JR, Davis DR, Millard PR, Herbertson BM, et al. Induction of immunological tolerance by porcine liver allografts. Nature. 1969; 223:472–476.
Article
2. Kamada N, Brons G, Davies HS. Fully allogeneic liver grafting in rats induces a state of systemic nonreactivity to donor transplantation antigens. Transplantation. 1980; 29:429–431.
Article
3. Kamada N, Davies HS, Roser B. Reversal of transplantation immunity by liver grafting. Nature. 1981; 292:840–842.
Article
4. Subbotin V, Sun H, Aitouche A, Valdivia LA, Fung JJ, Starzl TE, et al. Abrogation of chronic rejection in a murine model of aortic allotransplantation by prior induction of donor-specific tolerance. Transplantation. 1997; 64:690–695.
Article
5. Ramos HC, Reyes J, Abu-Elmagd K, Zeevi A, Reinsmoen N, Tzakis A, et al. Weaning of immunosuppression in long-term liver transplant recipients. Transplantation. 1995; 59:212–217.
Article
6. Reyes J, Zeevi A, Ramos H, Tzakis A, Todo S, Demetris AJ, et al. Frequent achievement of a drug-free state after orthotopic liver transplantation. Transplant Proc. 1993; 25:3315–3319.
7. Graeb C, Justl M, Scherer MN, Andrassy J, Frank E, Zuelke C, et al. Use of an adenoviral vector to express soluble donor-major histocompatibility complex molecules capable of suppressing the immune response in rat transplant recipients. Hum Immunol. 2002; 63:844–852.
Article
8. Priestley CA, Dalchau R, Sawyer GJ, Fabre JW. A detailed analysis of the potential of water-soluble classical class I MHC molecules for the suppression of kidney allograft rejection and in vitro cytotoxic T cell responses. Transplantation. 1989; 48:1031–1038.
Article
9. Tsui TY, Deiwick A, Ko S, Schlitt HJ. Specific immunosuppression by postoperative infusion of allogeneic spleen cells: requirement of donor major histocompatibility complex expression and graft-versus-host reactivity. Transplantation. 2000; 69:25–30.
Article
10. Yan Y, Shastry S, Richards C, Wang C, Bowen DG, Sharland AF, et al. Posttransplant administration of donor leukocytes induces long-term acceptance of kidney or liver transplants by an activation-associated immune mechanism. J Immunol. 2001; 166:5258–5264.
Article
11. Solgi G, Gadi V, Paul B, Mytilineos J, Pourmand G, Mehrsai A, et al. Five-year clinical effects of donor bone marrow cells infusions in kidney allograft recipients: improved graft function and higher graft survival. Chimerism. 2013; 4:87–94.
Article
12. Bertolino P, McCaughan GW, Bowen DG. Role of primary intrahepatic T-cell activation in the 'liver tolerance effect'. Immunol Cell Biol. 2002; 80:84–92.
Article
13. Benseler V, Warren A, Vo M, Holz LE, Tay SS, Le Couteur DG, et al. Hepatocyte entry leads to degradation of autoreactive CD8 T cells. Proc Natl Acad Sci U S A. 2011; 108:16735–16740.
Article
14. Bowen DG, Zen M, Holz L, Davis T, McCaughan GW, Bertolino P. The site of primary T cell activation is a determinant of the balance between intrahepatic tolerance and immunity. J Clin Invest. 2004; 114:701–712.
Article
15. Kamada N, Shinomiya T. Clonal deletion as the mechanism of abrogation of immunological memory following liver grafting in rats. Immunology. 1985; 55:85–90.
16. Lappe MA, Graff RG, Snell GD. The importance of target size in the destruction of skin grafts with non-H-2 incompatibility. Transplantation. 1969; 7:372–377.
Article
17. Sun J, Sheil AG, Wang C, Wang L, Rokahr K, Sharland A, et al. Tolerance to rat liver allografts: IV. Acceptance depends on the quantity of donor tissue and on donor leukocytes. Transplantation. 1996; 62:1725–1730.
18. Sun J, Bishop G, Wang C, Wang L, McCaughan G, Sheil R. Quantity of donor tissue and rat allograft survival. Transplant Proc. 1997; 29:1143–1144.
Article
19. Wang C, Sheil AG, Sun J. Outcome of different models of multiorgan transplantation in rats. Microsurgery. 1999; 19:318–323.
Article
20. Madsen JC, Sachs DH, Fallon JT, Weissman NJ. Cardiac allograft vasculopathy in partially inbred miniature swine. I. Time course, pathology, and dependence on immune mechanisms. J Thorac Cardiovasc Surg. 1996; 111:1230–1239.
Article
21. Madsen JC, Yamada K, Allan JS, Choo JK, Erhorn AE, Pins MR, et al. Transplantation tolerance prevents cardiac allograft vasculopathy in major histocompatibility complex class I-disparate miniature swine. Transplantation. 1998; 65:304–313.
Article
22. Yamada K, Mawulawde K, Menard MT, Shimizu A, Aretz HT, Choo JK, et al. Mechanisms of tolerance induction and prevention of cardiac allograft vasculopathy in miniature swine: the effect of augmentation of donor antigen load. J Thorac Cardiovasc Surg. 2000; 119:709–719.
Article
23. Jones ND, Turvey SE, Van Maurik A, Hara M, Kingsley CI, Smith CH, et al. Differential susceptibility of heart, skin, and islet allografts to T cell-mediated rejection. J Immunol. 2001; 166:2824–2830.
Article
24. Steger U, Denecke C, Sawitzki B, Karim M, Jones ND, Wood KJ. Exhaustive differentiation of alloreactive CD8+ T cells: critical for determination of graft acceptance or rejection. Transplantation. 2008; 85:1339–1347.
Article
25. Ganbold A, Andersen S, Tay SS, Cunningham E, Ilie V, Krishnan S, et al. Expression of common gamma chain signalling cytokines and their receptors distinguishes rejection from tolerance in a rat organ transplant model. Transpl Immunol. 2012; 27:89–94.
Article
26. Cunningham EC, Tay SS, Wang C, Rtshiladze M, Wang ZZ, McGuffog C, et al. Gene therapy for tolerance: high-level expression of donor major histocompatibility complex in the liver overcomes naive and memory alloresponses to skin grafts. Transplantation. 2013; 95:70–77.
27. Sharland A, Logan GJ, Bishop A, Alexander IE. Liver-directed gene expression using recombinant AAV 2/8 vectors--a tolerogenic strategy for gene delivery? Discov Med. 2010; 9:519–527.
28. Laurence JM, Wang C, Zheng M, Cunningham S, Earl J, Tay SS, et al. Overexpression of indoleamine dioxygenase in rat liver allografts using a high-efficiency adeno-associated virus vector does not prevent acute rejection. Liver Transpl. 2009; 15:233–241.
Article
29. Simpson N, Cho YW, Cicciarelli JC, Selby RR, Fong TL. Comparison of renal allograft outcomes in combined liver-kidney transplantation versus subsequent kidney transplantation in liver transplant recipients: Analysis of UNOS Database. Transplantation. 2006; 82:1298–1303.
Article
30. Neumann UP, Lang M, Moldenhauer A, Langrehr JM, Glanemann M, Kahl A, et al. Significance of a T-lymphocytotoxic crossmatch in liver and combined liver-kidney transplantation. Transplantation. 2001; 71:1163–1168.
Article
31. Gutierrez A, Crespo M, Mila J, Torregrosa JV, Martorell J, Oppenheimer F. Outcome of simultaneous liver-kidney transplantation in highly sensitized, crossmatch-positive patients. Transplant Proc. 2003; 35:1861–1862.
Article
32. Olausson M, Mjornstedt L, Norden G, Rydberg L, Molne J, Backman L, et al. Successful combined partial auxiliary liver and kidney transplantation in highly sensitized cross-match positive recipients. Am J Transplant. 2007; 7:130–136.
Article
33. Rana A, Robles S, Russo MJ, Halazun KJ, Woodland DC, Witkowski P, et al. The combined organ effect: protection against rejection? Ann Surg. 2008; 248:871–879.
34. Lee WP, Yaremchuk MJ, Pan YC, Randolph MA, Tan CM, Weiland AJ. Relative antigenicity of components of a vascularized limb allograft. Plast Reconstr Surg. 1991; 87:401–411.
Article
35. Cunningham EC, Tay SS, Wang C, Rtshiladze M, Wang ZZ, McGuffog C, et al. Gene therapy for tolerance: high-level expression of donor major histocompatibility complex in the liver overcomes naive and memory alloresponses to skin grafts. Transplantation. 2013; 95:70–77.
Full Text Links
  • HMR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr