Withdrawal of immunosuppression in liver transplantation and the mechanism of tolerance
 
Chi-Xian Zhang, Pei-Hao Wen and Yu-Ling Sun
Zhengzhou, China
 
 
Author Affiliations: Institute of Hepatobiliary and Pancreatic Diseases, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhengzhou University, School of Medicine, Zhengzhou 450052, China (Zhang CX, Wen PH and Sun YL)
Corresponding Author: Yu-Ling Sun, MD, PhD, Institute of Hepatobiliary and Pancreatic Diseases, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhengzhou University, School of Medicine, Zhengzhou 450052, China (Tel:+86-371-66862122; Fax: +86-371-66964927; Email: ylsun@zzu.edu.cn)
 
© 2015, Hepatobiliary Pancreat Dis Int. All rights reserved.
doi: 10.1016/S1499-3872(15)60411-8
Published online September 9, 2015.
 
 
Contributors: SYL proposed the study. ZCX and WPH performed the study and wrote the first draft. All authors have read and approved the final manuscript. SYL is the guarantor.
Funding: This study was supported by grants from the National Natural Science Foundation of China (81100304) and Science and Technology Bureau of Zhengzhou (131PLJRC658).
Ethical approval: Not needed.
Competing interest: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
 
 
BACKGROUND: Immunosuppression reagents have side effects and cause considerable long-term morbidity and mortality in patients after liver transplantation. Sufficient evidences showed that minimization or withdrawal of immunosuppression reagents does not deteriorate the recipient's immune response and physiological function and therefore, is feasible in some recipients of liver transplantation. However, the mechanisms are not clear. The present review was to update the current status of immunosuppression in liver transplantation and the mechanism of minimization or withdrawal of immunosuppression in liver recipients.
 
DATA SOURCES: We searched articles in English on minimization or withdrawal of immunosuppression in liver transplantation in PubMed. We focused on the basic mechanisms of immune tolerance in liver transplantation. Studies on immunosuppression minimization or withdrawal protocols and biomarker in tolerant recipients were also analyzed.
 
RESULTS: Minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance, which may not be permanent and can be affected by various factors. However, accurately evaluating immune status post-transplant is a prerequisite to achieve individualized immunosuppression. Numerous mechanisms for immune tolerance have been found, including immunophenotypic shift of memory CD8+ T cells and CD4+ T cell subsets. Activation of the inflammasome through apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) in dendritic cells is associated with rejection after liver transplantation.
 
CONCLUSIONS: Minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance via different mechanisms. This process could be affected by immunophenotypic shift of memory CD8+ T cells and CD4+ T cell subsets, which may be correlated with activation of the inflammasome through ASC in dendritic cells.
 
(Hepatobiliary Pancreat Dis Int 2015;14:470-476)
 
KEY WORDS: liver transplantation; immunosuppression; minimization; withdrawal; tolerance
 
 
Introduction
Liver transplantation is the only effective treatment for end-stage liver disease. However, rejection after transplantation results in short-term graft loss. Although immunosuppressive drugs prevent early graft rejection, long-term complications including infections, renal toxicity, neurotoxicity and bone marrow suppression are common.[1-4] To address the side effects of long-term immunosuppression, scientists developed strategies on minimization or withdrawal of immunosuppressive drugs in liver recipients without rejecting liver grafts while maintaining the normal immune response. In addition, physicians can provide individualized treatment plans with varying drug concentrations based on the patient's immune status. Despite these research advances in immunosuppression,[5-7] the failure in the process of withdraw of immunosupression happened and there were no biological markers for monitoring the immune status. Furthermore, the mechanism of operational tolerance was still not clear. The present review was to explore the application of minimization or withdrawal of immunosuppression in liver recipients and the possible mechanisms of immune tolerance in this population.
 
 
Clinical application of minimization or withdrawal of immunosuppression
Minimization or withdrawal of immunosuppressant after liver transplantation is much easier in children because their immature immune systems allow reshaping of the immune response. In a study[8] performed at the University of Pittsburgh, 20% of children had successfully withdrawn from immunosuppression after liver transplantation, and 37% were in the process of withdrawal. In a pilot study reported by Feng et al,[9] 60% of pediatric recipients of parental living donor liver transplants remained off immunosuppression therapy for at least 1 year with normal graft function and stable allograft histology. Because of the inherent tolerogenic property of the liver and careful scrutiny of accumulated clinical experience, immunosuppression weaning is feasible in almost 20% of selected liver transplant recipients.[10-12] Although minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance, tolerance may not be permanent and can be affected by various factors.[9] It is therefore necessary to monitor the immune status of these patients. However, no reliable biological markers are currently available.
 
The timing and specific approach to minimization or withdrawal of immunosuppression are important factors influencing outcomes. In clinical practice, initiation of immunosuppressive minimization or withdrawal ranges from 3 months to 2 or even 5 years after transplantation. However, in most patients minimization of immunosuppression was initiated 1 year after the last rejection occurred.[13, 14] The successful rate of earlier withdrawal (in 6 months after transplantation) is very low (1/18). However, this rate is much higher and the procedure is safer when minimization or withdrawal was initiated 1 year after the transplant.[15] The process of immunosuppressive minimization or withdrawal generally begins with glucocorticoid and extends to calcineurin inhibitor until complete withdrawal of immunosuppressive drugs.[16-18] One study on long-term outcomes of 600 living donor liver transplants for pediatric patients at a single center concluded that it is easier to induce immune tolerance and achieve a high success rate with minimization or withdrawal by prolonging maintenance immunosuppression rather than frequently decreasing the doses during minimization. Furthermore, graft rejection is typically mild and easy to control.[19] These data are summarized in Table.
 
After complete withdrawal of immunosuppression, patients who underwent living donor liver transplantation often experienced some degree of liver fibrosis accompanied by small bile duct hyperplasia and luminal narrowing; these complications often ameliorated after the use of immunosuppression via an unknown mechanism.[20] Egawa and colleagues[21] demonstrated that if minimization or withdrawal of immunosuppression results in rejection, original immunosuppressive therapy should be resumed.
 
 
Mechanisms of operative tolerance in liver transplantation
With or without immunosuppression, immune tolerance to self-major histocompatibility complex (MHC) antigens has been a limitation in organ transplantation, with no satisfactory method to induce tolerance in humans.[22] Although it is common to completely withdraw immunosuppressive drugs after liver transplantation because of the immunologically privileged status of the liver, immunologic tolerance induction in organ transplantation is not yet fully understood. Numerous mechanisms for immune tolerance have been found, including immune incompetence of antigen-specific T cells induced by antigen-presenting cells and suppression of memory cell reactions by the generation of regulatory T cells (Tregs).[23,24] Studies[25, 26] have also found that soluble MHC-related molecules, Fas protein, leukocytes from the donor liver and hematopoietic chimerism are all involved in the formation of immune tolerance.
 
To achieve minimization or withdrawal of immunosuppression after liver transplantation, transplant centers have used a variety of methods to induce immune tolerance. In 2003, Starzl et al[27] successfully achieved minimization of immunosuppression by clearing lymphocytes before transplantation to induce immune tolerance. In this study, 82 patients awaiting for transplantation of the kidney, liver, pancreas or intestine were pretreated with a broadly reacting rabbit antithymocyte globulin in several hours. Post-transplant immunosuppression was restricted to tacrolimus unless additional drugs were needed to treat breakthrough rejection. After 4 months, patients on tacrolimus monotherapy were considered for dose-spacing to every other day or longer intervals. 95% (78/82) patients survived for 1 year or for 13-18 months. Graft survival rate was 89% (73/82) at 1 year and 88% (72/82) at 13-18 months. Of the 72 recipients with surviving grafts, 43 were on spaced doses of tacrolimus monotherapy: every other day (n=6), three times per week (n=11), twice per week (n=15), or once per week (n=11). Inspired by Starzl's method, other researchers used rabbit anti-thymocyte globulin to induce immune tolerance in transplant patients. These patients not only successfully achieved steroid withdrawal, but the dose of tacrolimus was also significantly reduced.[28] During early liver transplantation, ursodeoxycholic acid can reduce the expression of MHC-associated molecules on biliary and central vein epithelial cells. However, attempts to use ursodeoxycholic acid to induce immune tolerance were not encouraging.[14, 29] Other researchers injected hematopoietic stem cells into three transplanted liver to induce hematopoietic chimerism.[30] In 2 cases, in vitro studies showed indices of immunological tolerance as assessed by specific hyporesponsiveness to donor alloantigens in mixed lymphocytes culture. In the third patient, acute rejection rapidly occurred after discontinuation of immunosuppression, and minimal immunosuppression has to be maintained in a long-term follow-up. In a child with familial hemophagocytic lymphohistiocytosis, infusion of hematopoietic stem cells from the donor mother before transplantation enabled the complete withdrawal of immunosuppression.[31] Similar results have been reported by a number of research institutions.[32-34]
 
 
Estimation of immune status and biological markers
Dose and concentration of immunosuppressants given after liver transplantation are closely related to the patient's immune status. Therefore, accurately evaluating immune status post-transplant is a prerequisite for the achievement of individualized immunosuppression. Currently, γδT cell phenotypes, natural killer cell receptors, precursor dendritic cell (DC) numbers, ratio of plasma cells to myeloid DCs, peripheral blood CD4+CD25+ T cells, CD8+ Tregs, chimerism, and low titers of donor-specific antibodies contribute to the induction of immune tolerance to achieve minimization or withdrawal of immunosuppression.[20, 35-38] However, the relationship between these biological markers and dynamic changes in immune status is not clear, because these markers are found only in patients who have already established tolerance and cannot accurately predict the patient's immune status over time.
 
Normal liver function alone after successful reduction or withdrawal of immunosuppression cannot serve as a valid biological marker, because these patients often experience chronic liver fibrosis, requiring occasional liver biopsies. Additionally, the risk of chronic rejection in these patients is also significantly increased.[39]
 
 
Memory CD8+ T lymphocytes and immune tolerance
Although minimization or withdrawal of immunosuppression in children and adults has been achieved, the presence of memory T cells can interfere with this stable immune status through different mechanisms.[40,41] Memory CD8+ T cells are the main effector cells in rejection and are important target cells for immune tolerance in transplantation. The authors[40] found that the phenotypes of memory CD8+ T cell subsets in recipients with stable immune status after liver transplantation change regularly with time. After reducing the dose and concentration of immunosuppressive drugs, the patients experienced immune rejection, and the phenotypes of memory CD8+ T cell subsets also changed. However, with enhanced immunosuppressant therapy, the main proportion of CD8+ cells were dominated by naive CD8+ cells and then rapidly restored to the immunophenotypes of memory T cells after effective treatment. Namely, the memory CD8+ T cell subsets were rapidly restored to their original phenotypes. These results demonstrated that immunophenotypic shift of memory CD8+ T cells was closely related to the change of the immune status in the patients after liver transplantation. Monitoring the immunophenotypic shift of memory CD8+ T cells is of great importance in the prediction for allograft rejection and treatment effectiveness after liver transplantation.[42] The mechanisms underlying rejection after minimization or withdrawal of immunosuppression are not clear. In view of the important role of CD8+ memory T cells in immune rejection of the transplanted liver, understanding the differentiation and function of these cells is essential to the immunosuppressive treatment. Therefore, developing new methods to identify the role of memory CD8+ T cells and their differential roles in rejection may be key issues for new immunosuppressive treatments in liver transplantation.
 
Additionally, one study[41] showed that immunosuppression has an important effect on the formation and function of memory T cells. Immunosuppression not only effectively suppresses T cell function and reduces the secretion of cytokines associated with rejection but also has an important role in the generation and maintenance of memory T cells. An in vitro experiment[43] showed that most immunosuppressants can inhibit the production and function of effector cells, with tacrolimus showing the strongest effect, but this inhibitory effect can be reversed by the CD28 co-stimulatory pathway. However, the role of rapamycin is different. Although it can promote the generation of memory CD8+ T cells, rapamycin fails to prevent the activation of memory CD8+ T cells, and its ability to suppress the function of memory CD8+ T cells function is also limited.[44]
 
 
CD4+ T lymphocytes and immune tolerance
CD4+ T cells represent a heterogeneous cell population.[45] A better understanding of CD4+ T cell subsets in the immunology of rejection will be instrumental for the development of effective strategies to induce clinical transplantation tolerance. CD4+CD25+Foxp3+ Tregs, a minor population of CD4+ T cells, play an important role in the control of immunologic tolerance to foreign and alloantigens.[46] Tregs include two major groups: naturally occurring Tregs and adaptive Tregs. The former are produced in a central lymphoid organ and the latter are generated in the periphery after antigenic stimulation in a specific tolerogenic microenvironment.[47]
 
Over the past decade, compelling evidence has shown the importance of Tregs in the promotion of transplant tolerance in animal models and patients.[48-50] In 2002, Graca et al[51] reported that T cell suppression of graft rejection is an active process that operates beyond secondary lymphoid tissue, and involves the persistent presence of regulatory T cells at the site of the tolerated transplant in skin grafts. In 2008, Li et al[52] found that Foxp3+CD25+CD4+ T cells were increased in liver grafts and recipient spleens from day 5 to day 100 post-transplantation, associated with enhanced CTLA4 and TGF-beta expression and IL-4 production. Depletion of recipient CD25+CD4+ T cells using anti-CD25 mAb (250 µg/day) induced acute liver allograft rejection. This was associated with a decreased ratio of Foxp3+ Treg: T effector cells, decreased IL-4 and elevated IL-10 and IL-2 production by graft-infiltrating T cells, and reduced apoptotic activity of graft-infiltrating CD4+ and CD8+ T cells in anti-CD25-mAb-treated recipients. Almost at the same time, the study from Pons et al[15] also demonstrated a significant increase in the number of peripheral Tregs in liver transplant recipients who developed tolerance versus those who did not. The mechanism of Tregs in establishing transplantation tolerance was associated with the suppression of all subsets of CD4+ T-helper cells in inflammation and transplantation.[45]
 
In addition to the therapeutic role of Tregs in the induction of allograft tolerance, gene expression profiling examining Treg molecules (Foxp3 and CTLA4) may be useful for identifying transplant patients experiencing tolerance and transplant patients experiencing rejection,[53, 54] but it should be pointed out that Foxp3 expression in humans, not like in mice, is not specific for cells with a regulatory phenotype and may be a consequence of activation status.[55]
 
Future research is needed to identify the mechanisms of different subsets of CD4+ T cells in the complex of transplantation rejection and tolerance. The mechanistic studies will help the clinician to devise the strategies to achieve transplantation tolerance in clinical practice.
 
 
Other factors
Genetic polymorphism of cytokines and chemokines is closely related to rejection in liver transplantation and may also affect post-transplant immunosuppression.[56] In addition, donor-related and unrelated antibody levels, tissue-specific immune responses, and endothelium-related immune stimulation are closely related to immune tolerance.
 
Inflammation is a main process of rejection, and activation of the inflammasome in DCs is associated with rejection after transplantation.[57] The up-regulation of the inflammation-associated molecule apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) is directly related to rejection in rat liver transplantation models.[58] Inflammasome activation involves nucleotide-binding oligomerization domain-like receptors in the cytoplasm to activate the linker protein containing a C-terminal caspase recruitment domain (ASC) and caspase-1, which cleaves IL-1β from its precursor.[59] The study[58] has confirmed that activation of the inflammasome in DCs is closely related to T cell immunity and plays an important role in transplantation tolerance. The inflammasome in DCs accelerates the proliferation of memory CD8+ T cells and regulates their function through different signaling pathways. Memory CD8+ T cells also inhibit DC activation and expression of the inflammasome.[60, 61]
 
 
Summary
Minimization or withdrawal of immunosuppression can be achieved by the induction of immune tolerance, which may not be permanent and can be affected by various factors. The process of immunosuppressive minimization or withdrawal generally begins with glucocorticoid, extends to calcineurin inhibitor and finally achieves complete withdrawal of immunosuppression. Numerous mechanisms for immune tolerance have been found, including immune incompetence of antigen-specific T cells induced by antigen-presenting cells and suppression of memory cell reactions by the generation of Tregs, soluble MHC-related molecules, Fas protein, leukocytes from the donor liver and hematopoietic chimerism. However, accurately evaluating immune status post-transplant is a prerequisite for the achievement of individualized immunosuppression. Immunophenotypic shift of memory CD8+ T cells and CD4+ T cell subsets is closely related to the change of the immune status. Activation of the inflammasome through ASC in DCs is associated with rejection after transplantation. However, the relationship between immunophenotypic shift of memory CD8+ T cells and activation of the inflammasome through ASC is not clear. Future research is needed to identify the mechanisms of transplantation rejection and tolerance to facilitate the development of strategies to achieve transplantation tolerance in liver recipients.
 
 
References
 
1 Wimmer CD, Angele MK, Schwarz B, Pratschke S, Rentsch M, Khandoga A, et al. Impact of cyclosporine versus tacrolimus on the incidence of de novo malignancy following liver transplantation: a single center experience with 609 patients. Transpl Int 2013;26:999-1006. PMID: 23952102
2 Teperman L, Moonka D, Sebastian A, Sher L, Marotta P, Marsh C, et al. Calcineurin inhibitor-free mycophenolate mofetil/sirolimus maintenance in liver transplantation: the randomized spare-the-nephron trial. Liver Transpl 2013;19:675-689. PMID: 23775875
3 Zivkovi? SA, Abdel-Hamid H. Neurologic manifestations of transplant complications. Neurol Clin 2010;28:235-251. PMID: 19932384
4 Mucha K, Foroncewicz B, Or?owski T, Religioni J, Bobek-Billewicz B, Jarz?b B, et al. Atypical presentation of invasive pulmonary aspergillosis in a liver transplant recipient. Ann Transplant 2013;18:238-242. PMID: 23792526
5 Benítez C, Londoño MC, Miquel R, Manzia TM, Abraldes JG, Lozano JJ, et al. Prospective multicenter clinical trial of immunosuppressive drug withdrawal in stable adult liver transplant recipients. Hepatology 2013;58:1824-1835. PMID: 23532679
6 Barbier L, Garcia S, Cros J, Borentain P, Botta-Fridlund D, Paradis V, et al. Assessment of chronic rejection in liver graft recipients receiving immunosuppression with low-dose calcineurin inhibitors. J Hepatol 2013;59:1223-1230. PMID: 23933266
7 de la Garza RG, Sarobe P, Merino J, Lasarte JJ, D'Avola D, Belsue V, et al. Trial of complete weaning from immunosuppression for liver transplant recipients: factors predictive of tolerance. Liver Transpl 2013;19:937-944. PMID: 23784747
8 Mazariegos GV, Sindhi R, Thomson AW, Marcos A. Clinical tolerance following liver transplantation: long term results and future prospects. Transpl Immunol 2007;17:114-119. PMID: 17306742
9 Feng S, Ekong UD, Lobritto SJ, Demetris AJ, Roberts JP, Rosenthal P, et al. Complete immunosuppression withdrawal and subsequent allograft function among pediatric recipients of parental living donor liver transplants. JAMA 2012;307:283-293. PMID: 22253395
10 Liu XQ, Hu ZQ, Pei YF, Tao R. Clinical operational tolerance in liver transplantation: state-of-the-art perspective and future prospects. Hepatobiliary Pancreat Dis Int 2013;12:12-33. PMID: 23392795
11 Lerut J, Sanchez-Fueyo A. An appraisal of tolerance in liver transplantation. Am J Transplant 2006;6:1774-1780. PMID: 16889539
12 Martínez-Llordella M, Lozano JJ, Puig-Pey I, Orlando G, Tisone G, Lerut J, et al. Using transcriptional profiling to develop a diagnostic test of operational tolerance in liver transplant recipients. J Clin Invest 2008;118:2845-2857. PMID: 18654667
13 Tisone G, Orlando G, Cardillo A, Palmieri G, Manzia TM, Baiocchi L, et al. Complete weaning off immunosuppression in HCV liver transplant recipients is feasible and favourably impacts on the progression of disease recurrence. J Hepatol 2006;44:702-709. PMID: 16473433
14 Assy N, Adams PC, Myers P, Simon V, Minuk GY, Wall W, et al. Randomized controlled trial of total immunosuppression withdrawal in liver transplant recipients: role of ursodeoxycholic acid. Transplantation 2007;83:1571-1576. PMID: 17589339
15 Pons JA, Revilla-Nuin B, Baroja-Mazo A, Ramírez P, Martínez-Alarcón L, Sánchez-Bueno F, et al. FoxP3 in peripheral blood is associated with operational tolerance in liver transplant patients during immunosuppression withdrawal. Transplantation 2008;86:1370-1378. PMID: 19034005
16 Samonakis DN, Mela M, Quaglia A, Triantos CK, Thalheimer U, Leandro G, et al. Rejection rates in a randomised trial of tacrolimus monotherapy versus triple therapy in liver transplant recipients with hepatitis C virus cirrhosis. Transpl Infect Dis 2006;8:3-12. PMID: 16623815
17 Lerut J, Mathys J, Verbaandert C, Talpe S, Ciccarelli O, Lemaire J, et al. Tacrolimus monotherapy in liver transplantation: one-year results of a prospective, randomized, double-blind, placebo-controlled study. Ann Surg 2008;248:956-967. PMID: 19092340
18 Bourdeaux C, Pire A, Janssen M, Stéphenne X, Smets F, Sokal E, et al. Prope tolerance after pediatric liver transplantation. Pediatr Transplant 2013;17:59-64. PMID: 23171043
19 Ueda M, Oike F, Ogura Y, Uryuhara K, Fujimoto Y, Kasahara M, et al. Long-term outcomes of 600 living donor liver transplants for pediatric patients at a single center. Liver Transpl 2006;12:1326-1336. PMID: 16773638
20 Koshiba T, Li Y, Takemura M, Wu Y, Sakaguchi S, Minato N, et al. Clinical, immunological, and pathological aspects of operational tolerance after pediatric living-donor liver transplantation. Transpl Immunol 2007;17:94-97. PMID: 17306739
21 Egawa H, Miyagawa-Hayashino A, Haga H, Teramukai S, Yoshizawa A, Ogawa K, et al. Non-inflammatory centrilobular sinusoidal fibrosis in pediatric liver transplant recipients under tacrolimus withdrawal. Hepatol Res 2012;42:895-903. PMID: 22524409
22 McCaughan GW, Bowen DG, Bertolino P. Operational tolerance in liver transplantation: shall we predict or promote? Liver Transpl 2013;19:933-936. PMID: 23913809
23 Bilate AM, Lafaille JJ. Induced CD4+Foxp3+ regulatory T cells in immune tolerance. Annu Rev Immunol 2012;30:733-758. PMID: 22224762
24 Xie J, Wang Y, Bao J, Ma Y, Zou Z, Tang Z, et al. Immune tolerance induced by RelB short-hairpin RNA interference dendritic cells in liver transplantation. J Surg Res 2013;180:169-175. PMID: 23196258
25 Verdonk RC, Haagsma EB, Jongsma T, Porte RJ, Roozendaal C, van den Berg AP, et al. A prospective analysis of the natural course of donor chimerism including the natural killer cell fraction after liver transplantation. Transplantation 2011;92:e22-24. PMID: 21814126
26 Chen Y, Liu Z, Liang S, Luan X, Long F, Chen J, et al. Role of Kupffer cells in the induction of tolerance of orthotopic liver transplantation in rats. Liver Transpl 2008;14:823-836. PMID: 18508376
27 Starzl TE, Murase N, Abu-Elmagd K, Gray EA, Shapiro R, Eghtesad B, et al. Tolerogenic immunosuppression for organ transplantation. Lancet 2003;361:1502-1510. PMID: 12737859
28 Eason JD, Cohen AJ, Nair S, Alcantera T, Loss GE. Tolerance: is it worth the risk? Transplantation 2005;79:1157-1159. PMID: 15880061
29 Assy N, Adams PC, Myers P, Simon V, Ghent CN. A randomised controlled trial of total immunosuppression withdrawal in stable liver transplant recipients. Gut 2007;56:304-306. PMID: 17303606
30 Donckier V, Troisi R, Le Moine A, Toungouz M, Ricciardi S, Colle I, et al. Early immunosuppression withdrawal after living donor liver transplantation and donor stem cell infusion. Liver Transpl 2006;12:1523-1528. PMID: 17004249
31 Matthes-Martin S, Peters C, Königsrainer A, Fritsch G, Lion T, Heitger A, et al. Successful stem cell transplantation following orthotopic liver transplantation from the same haploidentical family donor in a girl with hemophagocytic lymphohistiocytosis. Blood 2000;96:3997-3999. PMID: 11090093
32 Tryphonopoulos P, Tzakis AG, Weppler D, Garcia-Morales R, Kato T, Madariaga JR, et al. The role of donor bone marrow infusions in withdrawal of immunosuppression in adult liver allotransplantation. Am J Transplant 2005;5:608-613. PMID: 15707417
33 Mathew JM, Garcia-Morales RO, Carreno M, Jin Y, Fuller L, Blomberg B, et al. Immune responses and their regulation by donor bone marrow cells in clinical organ transplantation. Transpl Immunol 2003;11:307-321. PMID: 12967784
34 Hematti P. Role of mesenchymal stromal cells in solid organ transplantation. Transplant Rev (Orlando) 2008;22:262-273. PMID: 18656340
35 Bishop GA, Ierino FL, Sharland AF, Hall BM, Alexander SI, Sandrin MS, et al. Approaching the promise of operational tolerance in clinical transplantation. Transplantation 2011;91:1065-1074. PMID: 21544029
36 Li L, Wozniak LJ, Rodder S, Heish S, Talisetti A, Wang Q, et al. A common peripheral blood gene set for diagnosis of operational tolerance in pediatric and adult liver transplantation. Am J Transplant 2012;12:1218-1228. PMID: 22300520
37 Mazariegos GV, Zahorchak AF, Reyes J, Chapman H, Zeevi A, Thomson AW. Dendritic cell subset ratio in tolerant, weaning and non-tolerant liver recipients is not affected by extent of immunosuppression. Am J Transplant 2005;5:314-322. PMID: 15643991
38 Castellaneta A, Mazariegos GV, Nayyar N, Zeevi A, Thomson AW. HLA-G level on monocytoid dendritic cells correlates with regulatory T-cell Foxp3 expression in liver transplant tolerance. Transplantation 2011;91:1132-1140. PMID: 21423069
39 Orlando G, Soker S, Wood K. Operational tolerance after liver transplantation. J Hepatol 2009;50:1247-1257. PMID: 19394103
40 Jones ND. Memory T cells: how might they disrupt the induction of tolerance? Transplantation 2009;87:S74-77. PMID: 19424014
41 Donckier V, Craciun L, Miqueu P, Troisi RI, Lucidi V, Rogiers X, et al. Expansion of memory-type CD8+ T cells correlates with the failure of early immunosuppression withdrawal after cadaver liver transplantation using high-dose ATG induction and rapamycin. Transplantation 2013;96:306-315. PMID: 23799424
42 Sun Y, Yin S, Xie H, Zhou L, Wang Y, Wu L, et al. Immunophenotypic shift of memory CD8 T cells identifies the changes of immune status in the patients after liver transplantation. Scand J Clin Lab Invest 2009;69:789-796. PMID: 19929722
43 Jones DL, Sacks SH, Wong W. Controlling the generation and function of human CD8+ memory T cells in vitro with immunosuppressants. Transplantation 2006;82:1352-1361. PMID: 17130785
44 Mineharu Y, Kamran N, Lowenstein PR, Castro MG. Blockade of mTOR signaling via rapamycin combined with immunotherapy augments antiglioma cytotoxic and memory T-cell functions. Mol Cancer Ther 2014;13:3024-3036. PMID: 25256739
45 Liu Z, Fan H, Jiang S. CD4(+) T-cell subsets in transplantation. Immunol Rev 2013;252:183-191. PMID: 23405905
46 Sakaguchi S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 2005;6:345-352. PMID: 15785760
47 Shalev I, Schmelzle M, Robson SC, Levy G. Making sense of regulatory T cell suppressive function. Semin Immunol 2011;23:282-292. PMID: 21592823
48 Yu X, Huang C, Song B, Xiao Y, Fang M, Feng J, et al. CD4+CD25+ regulatory T cells-derived exosomes prolonged kidney allograft survival in a rat model. Cell Immunol 2013; 285:62-68. PMID: 24095986
49 Fan H, Cao P, Game DS, Dazzi F, Liu Z, Jiang S. Regulatory T cell therapy for the induction of clinical organ transplantation tolerance. Semin Immunol 2011;23:453-461. PMID: 21920772
50 Wood KJ, Bushell A, Hester J. Regulatory immune cells in transplantation. Nat Rev Immunol 2012;12:417-430. PMID: 22627860
51 Graca L, Cobbold SP, Waldmann H. Identification of regulatory T cells in tolerated allografts. J Exp Med 2002;195:1641-1646. PMID: 12070291
52 Li W, Kuhr CS, Zheng XX, Carper K, Thomson AW, Reyes JD, et al. New insights into mechanisms of spontaneous liver transplant tolerance: the role of Foxp3-expressing CD25+CD4+ regulatory T cells. Am J Transplant 2008;8:1639-1651. PMID: 18557727
53 Xie L, Ichimaru N, Morita M, Chen J, Zhu P, Wang J, et al. Identification of a novel biomarker gene set with sensitivity and specificity for distinguishing between allograft rejection and tolerance. Liver Transpl 2012;18:444-454. PMID: 22162188
54 Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, et al. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity 2000;12:431-440. PMID: 10795741
55 Morgan ME, van Bilsen JH, Bakker AM, Heemskerk B, Schilham MW, Hartgers FC, et al. Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol 2005;66:13-20. PMID: 15620457
56 Krüger B, Schröppel B, Murphy BT. Genetic polymorphisms and the fate of the transplanted organ. Transplant Rev (Orlando) 2008;22:131-140. PMID: 18631866
57 Critchley WR, Fildes JE. Graft rejection - endogenous or allogeneic? Immunology 2012;136:123-132. PMID: 22260525
58 Zhang M, Zhang S, Wu J, Sun Y, Li L, Du W, et al. The immunosuppressant protosappanin A promotes dendritic cell-mediated expansion of alloantigen-specific tregs and prolongs allograft survival in rats. PLoS One 2013;8:e66336. PMID: 22732093
59 Guo X, Dhodapkar KM. Central and overlapping role of Cathepsin B and inflammasome adaptor ASC in antigen presenting function of human dendritic cells. Hum Immunol 2012;73:871-878. PMID: 23840445
60 Kupz A, Guarda G, Gebhardt T, Sander LE, Short KR, Diavatopoulos DA, et al. NLRC4 inflammasomes in dendritic cells regulate noncognate effector function by memory CD8+ T cells. Nat Immunol 2012;13:162-169. PMID: 22231517
61 Beynon V, Quintana FJ, Weiner HL. Activated human CD4+CD45RO+ memory T-cells indirectly inhibit NLRP3 inflammasome activation through downregulation of P2X7R signalling. PLoS One 2012;7:e39576. PMID: 22768094
 
Received August 15, 2014
Accepted after revision June 25, 2015