Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2014 Feb;4(2):a015594. doi: 10.1101/cshperspect.a015594

Tolerance—Is It Worth It?

Erik B Finger 1, Terry B Strom 2, Arthur J Matas 1
PMCID: PMC3904098  PMID: 24370928

Abstract

We are entering an exciting time in the study of immunologic tolerance. Several cellular and molecular strategies have been developed that show promise in nonhuman transplant models and these approaches are just now appearing in clinical trials. Tolerance strategies that prevent immune rejection and obviate the need for immunosuppressive medications (with inherent risk of cancer, infection, and organ toxicity) would improve both graft and patient survival. Each tolerance protocol brings its own set of associated risks. As the results of these trials become available, we must continue to evaluate their successes and failures. The balance of these outcomes will help us answer the question: “Tolerance—Is it worth it?”


Organ transplantation usually requires lifelong immunosuppression. Tolerance strategies that prevent immune rejection have been pursued. Are the costs of tolerance induction worth the benefits of immunosuppression avoidance?

WHAT IS TOLERANCE?

In the words of Nobel Prize winner Peter Medawar, “‘Immunological tolerance’ may be described as a state of indifference or non-reactivity towards a substance that would normally be expected to excite an immunological response” (Medawar 1964). The concept of tolerance is further clarified by the editors of the American Journal of Transplantation as “a durable state of antigen-specific unresponsiveness, induced by exposure to antigen, in a patient who is otherwise fully immunologically competent” (Halloran et al. 2008).

The experimental evidence for tolerance began with the 1945 studies of Owen (1945) who showed persistence of postnatal red blood cell chimerism in freemartin cattle that share a common placenta and develop cross-placental common circulation. The significance of these observations to transplantation was made apparent by Billingham, Medawar, and colleagues, who showed tolerance to skin grafts from nonidentical dizygotic cattle twins (Anderson et al. 1951; Billingham et al. 1952). Interestingly, these observations occurred in a failed attempt to develop a technique to distinguish monozygotic versus dizygotic twins. The original hypothesis was that skin grafts between dizygotic twins would reject, whereas grafts between single-cell twins would not. To the contrary, skin grafts from both identical and dizygotic twins were accepted without rejection. Similar findings were shown by parabiosis in birds, in which embryonic cross circulation made siblings incapable of making antibodies against each other, or of rejecting skin grafts (Hasek and Hraba 1955).

Billingham et al. (1953) reported the first formal documentation of transplant tolerance by demonstrating that inoculation of fetal mice or chickens with donor cells of a different strain of that species made these animals tolerant to skin grafts of the donor strain. This study is the first report of tolerance induction by mixed chimerism. However, there has been some debate as to whether these experiments show true tolerance (Halloran et al. 2008). Subsequent studies suggest that mixed chimerism may, in itself, be a form of immunosuppression, as this antigen’s nonresponsiveness was not donor-specific (Hofmann et al. 2004). There is disagreement over the interpretation of these findings (Sachs et al. 2008). Additional evidence shows persistence of intact immune function remains with mixed chimerism (Ildstad et al. 1985; Rüedi et al. 1989), and that although nonresponsiveness in these studies was not strain specific, they were at least organ specific (Hofmann et al. 2004).

Despite conflicts in the formal definition of tolerance, a more practical definition may be that of operational tolerance. This may be defined as “a well-functioning graft lacking histological signs of rejection, in the absence of any immunosuppressive drugs (for at least 1 year), in an immunocompetent host” (Ashton-Chess et al. 2007; Orlando et al. 2010). Operational tolerance may further be clarified to include the absence of antibody-mediated rejection.

WHY IS TOLERANCE IMPORTANT?

Transplantation is the best, and in some cases only, treatment for many types of end-stage organ disease (Grinyó 2013). It has been shown to increase both quality and length of life for those affected by organ failure. Unfortunately, for the vast majority of transplant recipients there is an obligate requirement for lifelong immunosuppression. These immunosuppressive medications increase the risk of cancer, infection, and direct drug-associated organ toxicity, all of which contribute to reduced patient survival.

The most common cause of death in patients with functioning renal transplants is cardiovascular disease (24%), followed by infection (16%), and malignancy (12%) (Kahwaji et al. 2011). Each organ has a different distribution of causes of death after transplant, but infection and cancer are frequent etiologies in all (Chapman et al. 2013; Fishman 2013a). There is an ∼50% overall risk of developing some form of cancer (including nonmelanoma skin cancer) attributed to immunosuppressive medications (Scherer et al. 2007). The association of individual cancers with immunosuppression varies, but relative risks of two- to 20-fold have been reported (Kasiske et al. 2004) with the greatest risk being non-Hodgkin’s lymphomas and nonmelanoma skin cancers. The risks of Epstein–Barr virus (EBV)-associated posttransplant lymphoproliferative disease (PTLD) vary based on organ type, age at transplant, and EBV serostatus. A recent report from a large kidney transplant series measured annual incidence of 2.6 cases of PTLD per 1000 patient years and an increase in Hodgkins and non-Hodgkins lymphoma of 5.9 and 7.6 times, respectively, the rate in the general population (Morton et al. 2013).

Transplant patients are also at increased risk for both routine and opportunistic infections (Fishman 2007, 2013a). Of particular concern is the increased susceptibility to viral and fungal infections (Fishman 2002, 2013b; Winston et al. 1995; Winston and Busuttil 2002). Opportunistic infections are even more prevalent in patients undergoing antilymphocyte induction therapy (Issa and Fishman 2009). Infection is second only to cardiovascular disease, which is one of the most common causes of death in kidney transplant recipients (Djamali et al. 2006).

Immunosuppressive agents have a range of associated direct toxicities. Of particular concern is chronic nephropathy of the transplanted allograft in renal transplant recipients or the native kidneys in extrarenal transplant recipients (Ojo et al. 2003). New onset diabetes associated with calcineurin inhibitors is a well-described complication (Kasiske et al. 2003). With strict definition for new onset diabetes, the tacrolimus-associated incidence is in excess of 30% in kidney recipients and even higher in liver recipients (First et al. 2013). The degree of posttransplant hyperglycemia is predictive of cardiovascular and all-cause mortality (Valderhaug et al. 2011).

In addition to the adverse health effects of these medications, there is a continued problem of graft loss due to noncompliance. A review of multiple reports of compliance in kidney transplantation reported that approximately 22% of patients are noncompliant with their medications (Butler et al. 2004). Furthermore, 36% of graft losses are associated with prior noncompliance, and the odds ratio of graft failure is sevenfold higher for nonadherent patients (Butler et al. 2004). A report from Sweden documented that 16% of renal transplant recipients report having missed doses of immunosuppression in the preceding month (Lennerling and Forsberg 2012). The problem is even more pronounced in pediatric recipients, in which there is as much as a 75% nonadherence rate (Fredericks and Dore-Stites 2010). In kidney transplantation, long-term mortality rate was 48%–82% lower among transplant recipients than patients on the waiting list (Wolfe et al. 1999). Graft losses due to noncompliance or direct toxicity of these medications likely contribute to overall shortened patient lifespan.

Successful induction of transplant tolerance would eliminate immune rejection. Even with continued use of therapeutic immunosuppression, graft loss due to immune-mediated acute and chronic rejection shortens both graft and patient survival. Since the first successful kidney transplant in 1954 (Harrison et al. 1956), there has been a continuous improvement in transplant outcomes for all organ types (Organ Procurement 2012). Despite improvements in predominantly short-term outcomes over the last 50 years, improvements in chronic graft loss have not kept pace (Meier-Kriesche et al. 2004; Sykes 2007). Even in the absence of organ dysfunction, there can be subclinical rejection present in nearly half of biopsy specimens. Over time, severe chronic allograft nephropathy develops in 58.4% of patients (Nankivell et al. 2003). This damage, once established, is irreversible, resulting in declining renal function and graft failure. This resulting decline in glomerular filtration rate (GFR) is predictive of mortality following renal transplant (Moranne et al. 2013). In liver, lung, and heart transplants, immune-mediated graft loss leads to patient death unless retransplant is possible.

Tolerogenic strategies that lessen infectious complications, reduce cancer risk, avoid direct pharmacologic toxicity, and prevent graft loss due to acute and chronic immune destruction would improve graft and subsequent patient survival.

HOW CAN TOLERANCE BE ACHIEVED?

Many approaches have been pursued with the objective of tolerance in solid organ transplantation. These approaches can be divided into three main groups: (1) study of noncompliant patients that show spontaneous tolerance following discontinuation of immunosuppression, (2) patients with stable long-term graft function that undergo planned immunosuppression minimization and cessation, and (3) pretransplant-planned protocols for tolerance induction via pharmacologic or cellular therapy (Orlando et al. 2010). There are examples of successful withdrawal of immunosuppression with each of these approaches. Although they may not meet the strict definition of “tolerance,” they fit within a more clinically relevant description of “clinical operational tolerance.” Some of the most promising approaches are summarized below.

Study of Spontaneous Tolerance

There have been many reports of sporadic tolerance identified in noncompliant patients (summarized by Orlando et al. 2010). It is clear that organs differ in their response to cessation of immunosuppression out of noncompliance. There have been many reports of tolerance in kidney transplantation (Orlando et al. 2010), a single report in lung transplant (Di Cocco et al. 2009), and no reports in pancreas transplant. Liver transplant recipients are most likely to have continued graft function after cessation of immunosuppression. Multiple reports document durable liver allograft function after discontinuation by nonadherence (Orlando et al. 2009). There are other case reports or small series of tolerance following discontinuation under circumstances other than noncompliance. One report documented operational tolerance in heart transplant after total lymphoid irradiation for resistant rejection and subsequent withdrawal of all medications, including immunosuppression, because of drug intolerance (Comerci et al. 2009). Similar findings have been found in renal transplants with operational tolerance after total lymphoid irradiation (Strober et al. 1989, 2000). A number of case reports have documented spontaneous tolerance in a subset of patients undergoing immunosuppression withdrawal for treatment of PTLD (Orlando et al. 2010).

Planned Immunosuppression Weaning

Based on the findings of spontaneous tolerance in nonadherent patients, several studies have been performed that sought to wean patients off of immunosuppression as a planned treatment protocol. Orlando reviewed reports of operational tolerance in kidney (Orlando et al. 2010) and liver transplantation (Orlando et al. 2009) following planned immunosuppression withdrawal. In liver transplant, there is an approximate 22% success rate for protocols of withdrawal. In a particularly informative study in liver transplantation, it was found that pediatric patients may be uniquely amenable to immunosuppression weaning (Feng et al. 2012). In this study of planned immunosuppression weaning in patients with stable graft function on single agents after liver transplant for nonviral and nonautoimmune etiologies of ESLD, there was a 60% success rate in immunosuppression withdrawal.

The prevalence of potentially tolerant renal transplant recipients is clearly less than in liver transplant recipients, but the actual percentage of patients is harder to define. Most studies rely on a series of case reports (Roussey-Kesler et al. 2006) or assembled groups of patients already with stable graft function after nonadherence. Approximately 100 cases of operational tolerance in renal transplant have been reported (Orlando et al. 2010) with a denominator of many tens of thousands of total kidney transplants. There are a steadily increasing number of reports of different immune monitoring phenotypes or “signatures” that characterize tolerant recipients (Brouard et al. 2005, 2007; Newell and Larsen 2006; Einecke et al. 2010; Newell et al. 2010; Sagoo et al. 2010). It may be possible to use these combined signatures to identify candidates for immunosuppression weaning.

Pretransplant Tolerance Protocols

Mixed Chimerism

A promising approach developed by several groups makes use of strategies to induce mixed chimerism (Sachs et al. 2013). This is accomplished by a stem-cell infusion in combination with a transplant from the same donor. There are several variations to this approach. The earliest reports were in patients that first had bone marrow/stem cell transplantation and then later had a kidney transplant from the same donor (Sayegh et al. 1991; Helg et al. 1994; Jacobsen et al. 1994; Sorof et al. 1995; Butcher et al. 1999; Sellers et al. 2001; Light et al. 2002). Operational tolerance was reported in these recipients. Subsequent studies have documented some degree of success with a combined or simultaneous stem cell and kidney transplant from the same donor rather than the earlier sequential approach.

The group from Massachusetts General Hospital (MGH) showed tolerance in six patients undergoing simultaneous bone marrow and kidney transplants from human leukocyte antigen (HLA)-identical donors for the treatment of multiple myeloma in patients with renal failure (Fudaba et al. 2006). Three patients showed transient donor chimerism, but were successfully weaned off immunosuppression. Two patients had full chimerism, but immunosuppression was resumed for treatment of graft-versus-host disease (GVHD). One patient developed acute rejection and was treated with resumption of cyclosporine, but was later able to be weaned off immunosuppression once again.

This report was followed by a more rigorous non-HLA-identical series from the same group wherein they transplanted five patients with single haplotype mismatched sibling donors (Kawai et al. 2008). They performed a pretransplant nonmyeloablative conditioning protocol with cyclophosphamide, thymic irradiation, and humanized anti-CD2 monoclonal antibody. Induction therapy also consisted of short-course cyclosporine or tacrolimus (plus rituximab and steroids in a subset of patients). Four out of five patients showed operational tolerance and were weaned off of immunosuppression. The fifth developed early refractory antibody-mediated rejection and had graft failure. The tolerant patients showed transient microchimersim and persisting donor-specific hyporesponsiveness in in vitro assays (Kawai et al. 2008; LoCascio et al. 2010). In subsequent follow up, there were three cases of acute or chronic GVHD, for which two recipients were returned to immunosuppression (Spitzer et al. 2011). Three patients had recurrence of multiple myeloma. This trial has since been extended to 10 patients; seven have shown long-term graft survival without maintenance immunosuppression (Markmann and Kawai 2012). Three failures occurred after withdrawal of immunosuppression from early humoral rejection, thrombotic microangiopathy, and acute rejection.

A group from Stanford reported on their series of 16 patients undergoing simultaneous HLA-identical kidney transplant and same-donor granulocyte-colony stimulating factor (GCSF)-mobilized CD34+ stem-cell infusion (Scandling et al. 2012). They used a nonmyeloablative conditioning regimen of total lymphoid irradiation and antithymocyte globulin. Fifteen out of 16 developed mixed donor/recipient chimerism. Eight of these showed chimerism for more than 6 months and were weaned off immunosuppression. Four patients were undergoing immunosuppression wean. Four patients developed recurrent disease or rejection and were not weaned. All patients had functioning grafts at the end of the reporting period and there was no GVHD.

The groups from Northwestern and University of Louisville reported a novel approach to augment donor stem cell engraftment in conjunction with renal transplant (Leventhal et al. 2012). They make use of a bone marrow-derived CD8+, TCR “facilitator cell” population that promotes the engraftment of allogeneic hematopoietic stem cells in animal models without observed GVHD (Kaufman et al. 1994). To date, data on 15 patients undergoing their protocol have been reported (Leventhal et al. 2012, 2013). Recipients undergo a “low-intensity” conditioning regimen consisting of fludarabine, cyclophosphamide, and 200 cGy total-body irradiation. Subsequently, a combined HLA-mismatched renal transplant with same-donor hematopoietic stem-cell and facilitator-cell infusion is performed. Fourteen out of 15 showed initial macrochimerism that was eventually lost in three patients. There was one engraftment failure in a patient with panel reactive antibody (PRA) of 52%. Initial immunosuppression was tacrolimus and mycophenolate mofetil, which was successfully weaned off at 1 year in all patients with durable macrochimerism (12 patients). One patient lost their graft because of renal artery thrombosis after viral infection and sepsis. There were no cases of GVHD or engraftment syndrome. Of note, all patients that were successfully weaned off all immunosuppression had extremely high levels of donor-cell engraftment (94%–100% donor type), suggesting donor-type replacement rather than mixed chimerism. Two concerns with these reports were the lack of details on the engineered facilitator population and a lack of a control group without facilitator cells (Markmann and Kawai 2012). However, in a similar induction therapy for stem cell transplant, there was an engraftment failure rate of 13% and severe GVHD in 6% (Luznik et al. 2008; Markmann and Kawai 2012).

Regulatory T Cells

A different cellular therapy approach uses regulatory T cells (Treg) (Cobbold and Waldmann 2013). These lymphocytes develop in the thymus (natural Treg) or are induced in the periphery in response to antigen exposure in the presence of TGF-β (adaptive Treg) (reviewed in Josefowicz et al. 2012). Treg are phenotypically described by their expression of the transcription factor FoxP3. When these cells encounter their cognate antigens properly presented by antigen-presenting cells, they suppress the activation and function of nearby effector T cells (Schmidt et al. 2012). Treg are important in a wide variety of counterinflammatory conditions and are a promising target for tolerance-induction strategies.

The first clinical use of Tregs has been in the prevention or treatment of GVHD. Trzonkowski et al. (2009) performed a trial in two patients who were failing standard treatment of GVHD following allogeneic stem cell transplant. Treg were isolated from the stem cell donor and infused back into the recipient. In one patient with chronic GVHD, they were able to stabilize symptoms and wean immunosuppression medications. The long-term durability was not reported. In a second patient with acute GVHD, they were able to temporize symptoms with repeated dosing of Treg. However, they ran out of donor material, were unable to infuse further cells, and the patient relapsed and succumbed to the disease.

A second trial reported by Di Ianni et al. (2011), attempted to use Treg paired with conventional T-cell infusion to prevent GVHD in patients at high risk for GVHD, and to more quickly restore cellular immunity following stem cell transplant. This study consisted of 28 patients with high-risk hematologic malignancies that underwent haploidentical hematopoietic stem cell transplant. The recipients underwent induction conditioning with total-body irradiation and treatment with thiopeta, fludarabine, and cyclophosphamide. GCSF-mobilized, CD34-enriched hematopoietic stem cells from the donors were infused along with ex vivo expanded Treg and conventional T cells. Twenty-six out of 28 had sustained full donor-type engraftment. Two out of 26 developed grade 2, or better, acute GVHD. No chronic GVHD was observed and there was improved T-cell reconstitution and decreased cytomegalovirus compared with standard hematopoietic stem cell transplantation (HST).

A third trial was performed at the University of Minnesota (Brunstein et al. 2011). This study was a phase I dose escalation trial of ex vivo expanded umbilical cord Treg. Patients underwent a conditioning regimen consisting of total-body irradiation and treatment with fludarabine and cyclophosphamide. Patients received two partially HLA-matched umbilical cord blood infusions followed by infusion of Treg isolated and expanded from a third umbilical cord blood sample that was 4-6/6 HLA matched to the patient. Induction immunosuppression was cyclosporine and mycophenolate mofetil (MMF) in the first group of patients and then sirolimus and MMF in the second group. There was no increased risk of infection, relapse of disease, or mortality. There was a decrease in severe GVHD from 61% to 43%.

In an interesting but theoretically contradictory approach, trials are beginning that combine rapamycin and IL-2 administration in diabetes, GVHD, and composite tissue transplants. This combination has a number of Treg-friendly benefits. Treg are IL-2 dependent and IL-2 rather surprisingly serves as a death factor for CD8+ memory T cells (Ku et al. 2000) or newly activated effector T cells (Li et al. 2001) by opposing cytokines. Low-dose IL-2 administration in diabetes-prone nonobese diabetic (NOD) mice promotes Treg survival and protects against the development of diabetes (Tang et al. 2008). The combination of IL-2 and rapamycin favor Treg survival and T effector apoptosis by activating T cells toward an apoptotic pathway rather than clonal expansion (Rabinovitch et al. 2002).

Triple therapy with doses of IL-2Ig that saturate the high affinity, but not intermediate affinity IL-2 receptor, mutant antagonist type IL-15Ig, and rapamycin tilts the balance of immunity from rejection to or toward tolerance in a variety of daunting murine transplant (Zheng et al. 2003) and new onset type 1 diabetes models (Koulmanda et al. 2007). Triple therapy also results in long-term islet allograft survival in nonhuman primates (Koulmanda et al. 2012). The mechanism is hypothesized to be selective depletion of alloreactive T cells and stabilization/expansion of Tregs. Recent clinical trials of low-dose IL-2 therapy in corticosteroid-resistant GVHD (Koreth et al. 2011) and virus-induced vasculitis (Saadoun et al. 2011) have produced exciting results, whereas higher doses produce toxicity by activating innate immune cells bearing the intermediate affinity IL-2 receptor despite expansion of Tregs.

Currently, there are several trials of Treg in solid organ transplantation under way, including a large multinational trial (www.onestudy.org) and other center-based trials. Although there is considerable hope that Treg may be important in the induction of tolerance, many questions persist. How many Treg are needed to prevent rejection? What is the source of Treg? Donor cells? Recipient cells? Do Treg need to be antigen-specific, or would polyclonal expanded cells be sufficient? How long do these cells last once infused? Based on experimental studies in animal models, there may be some benefit to combining Treg with stem-cell infusion. As has been seen with facilitator cells and hematopoetic stem cells, Treg may help with establishment of mixed chimerism and prevent induction of GVHD. Thus, the Treg themselves may not be tolerogenic, but they may support persistence of mixed chimerism that induces tolerant phenotype.

Costimulatory Blockade

Costimulatory molecules provide another attractive target for tolerance-induction protocols (Maltzman and Turka 2013). There are abundant examples in mice in which a blockade of costimulatory molecules prolongs graft survival (reviewed in Kinnear et al. 2013). The three main pathways that have approached as clinical targets have been the CD28:CD80/CD86, CD40:CD154, and LFA-1:ICAM receptor:ligand pairings.

The CD28 costimulatory pathway was the first of these pathways to have a biologic agent approved for clinical use in transplant. In murine experimental models, CTLA-4Ig administration leads to prolonged or indefinite graft survival (Lenschow et al. 1992; Baliga et al. 1994; Pearson et al. 1994, 1997; Judge et al. 1996). CTLA-4Ig works via two pathways to inhibit alloreactive T-cell function. First, CTLA-4Ig binds to CD80 and CD86 on antigen-presenting cells and inhibits binding of these ligands to CD28 molecules on T cells. This inhibition reduces CD28-mediated costimulation during antigen recognition. Second, CTLA-4Ig binding to CD80/86 molecules also induces indoleamine 2,3-dioxygenase (IDO) expression in dendritic cells via an IFN-γ-dependent pathway. Up-regulation of IDO leads to changes in tryptophan metabolism that suppresses T-cell function (Grohmann et al. 2002).

In 2011, belatacept (human CTLA-4Ig) was approved by the Food and Drug Administration for use in kidney transplantation for the prevention of acute rejection in combination with basiliximab, mycophenolate mofetil, and corticosteroids. A number of trials have reported the outcomes of belatacept-based immunosuppression protocols in comparison to cyclosporine-based regimens. One- and 3-year outcomes have shown similar graft and patient survival between cyclosporine and belatacept protocols, but slightly more rejection with belatacept (Durrbach et al. 2010; Vincenti et al. 2010; Rostaing et al. 2011; Pestana et al. 2012). However, belatacept-treated patients had better preservation of kidney function with a higher GFR than the cyclosporine-treated patients. In diabetic patients, there was an increase in patient and graft survival with belatacept, but this did not achieve statistical significance (Rostaing et al. 2011). In each of the studies, there was an increased risk of PTLD with belatacept. However, this was seen predominantly in EBV-naïve patients. Currently, belatacept is not recommended for use in transplant recipients who are EBV negative. In the 3-year follow up, there was an increase in the incidence of tuberculosis infection (Pestana et al. 2012).

A second potential target for costimulatory blockade is the CD40:CD154 pathway. Blockade of this pathway results in prolongation of transplant survival in mouse (Parker et al. 1995; Hancock et al 1996; Larsen et al. 1996; Larsen and Pearson 1997; Shimizu et al. 2000) and nonhuman primate models (Kenyon et al. 1999; Kirk et al. 1999; Koulmanda et al. 2006). However, clinical trials in humans were halted because of safety concerns over thromboembolic events (Sidiropoulos and Boumpas 2004; Couzin 2005). These thromboembolic events were validated in nonhuman primate models and the mechanism was postulated to be caused by unanticipated binding to CD154 on vascular endothelium and platelets (Kawai et al. 2000). Although most preclinical studies reveal anti-CD154 to be more potent than most anti-CD40 mAbs as immune therapy, Reimann has developed an anti-CD40 mAb with immune-inhibitory functions as evidenced by the ability to block IgG production by lipopolysaccharide-activated B cells. This unique antibody has enabled drug-free nonhuman primate islet allograft survival, suggesting that optimization of this approach may have great promise (Lowe et al. 2012).

The third target, efalizumab, is a humanized anti-LFA-1 (CD11a/CD18) monoclonal antibody that was approved for the treatment of psoriasis (Dedrick et al. 2002; Stern 2003), and has been effective at reducing the severity of the disease in moderate to severe psoriasis (Gordon et al. 2003; Lebwohl et al. 2003). The antibody blocks the cell-adhesion molecule LFA-1 from binding to its ligand ICAM-1. LFA-1-mediated cell adhesion is important in a number of steps in the immune process including leukocyte adhesion to endothelium and the strengthening of T-cell/antigen-presenting cell interactions (Dedrick et al. 2002). Anti-LFA-1 was tested in a clinical trial in kidney transplant recipients (Vincenti et al. 2007). It was used in combination with cyclosporine, MMF, and prednisone. Although graft and patient survival was excellent (95% and 97% at 6 months), there was an 11% rejection risk and an 8% PTLD incidence. Other groups have used efalizumab as adjunct therapy in islet transplant trials (Posselt et al. 2010; Turgeon et al. 2010) as part of a maintenance immunosuppression regimen. The use of this agent was halted because of an association with progressive multifocal leukoencephalopathy (PML), a serious and usually fatal central nervous system infection caused by JC polyoma virus (Carson et al. 2009). However, PML has also been associated with other immunosuppressive mediations that remain in common use (Neff et al. 2008; Carson et al. 2009; Weber et al. 2011).

Other Possibilities

A number of additional cellular therapy approaches with promising results in preclinical studies are entering into clinical trial (reviewed in Page et al. 2012; Tang and Bluestone 2013). Tolerogenic macrophage, dendritic cells, and mesenchymal stromal cells have all shown promise in transplant models. Transplant acceptance-inducing cells are a form of regulatory macrophage. Two trials using infusion of donor-derived splenic monocytes in kidney transplant were performed (Hutchinson et al. 2008a,b). Standard immunosuppression induction was initiated and patients were weaned to low-dose monotherapy (or completely off of immunosuppression in one patient). There was an elevated incidence of early acute rejection, but several patients were able to wean down to low-dose single agent. Additional cell types with tolerogenic properties include mesenchymal stromal cells (English and Wood 2013) and tolerogenic dendritic cells (Page et al. 2012). These cell types have been shown effective in animal models and are also entering into clinical testing.

AT WHAT COST?

Highlighted above are a number of approaches that have been, or are soon to be, put into clinical trials as potentially tolerogenic therapies in transplantation. Despite some promising outcomes with several of these approaches, they each pose risk of adverse events and outcomes. For each approach there are specific complications, but these may represent more generalized concerns.

Risks of Stem Cell Infusion

A particular concern of the mixed chimerism approaches is the toxicity of the induction protocols. These protocols make use of chemotherapeutic medications and systemic irradiation as part as their engraftment protocol. These treatments come with both short- and long-term sequella. Although the immediate health outcomes reported seem to decrease with expected outcomes after transplant, we do not know the long-term health impact. An additional risk is the development of GVHD, a potentially life-threatening outcome, which was seen in several of the combined hematopoietic stem cell transplantation (HSCT)/kidney recipients.

Although we do not have long-term outcomes on all patients in the mixed chimerism trials, we can look at similar protocols for HSCT treatment of hematologic malignancies. Apart from recurrent disease, toxicity of the induction protocols and GVHD are the biggest long-term risk factors for these patients (Tichelli et al. 2009, 2012). HSCT recipients have increased risk of endocrine, musculoskeletal, and cardiovascular disorders in addition to increased risk of malignancy. These factors have an impact on morbidity and mortality following treatment. Patients alive at 5 years posttransplant still have a 30% reduction in life expectancy compared with the general population (Bhatia 2011).

Risks of PTLD

The risk of PTLD is increased in several of the trials described, particularly with the use of belatacept and efalizumab. This risk may be inherent to the tolerance approach, as the recipient's immune system is less able to fight off the subset of PTLDs that are donor cell in origin (Olagne et al. 2011). The concern of PTLD is greatest in patients who are EBV-naïve. Avoiding this population of patients may mitigate some of these concerns.

Off-Target Effects

A particular concern in trials of Treg is whether the suppression of allogeneic immune responses will lead to suppression of other off-target immune functions. That is, will Tregs that suppress rejection of liver transplant also suppress antiviral responses to the hepatitis virus that caused the disease (Dolganiuc and Szabo 2008)? Or will they suppress antitumor responses to hepatocellular carcinoma present in the liver explant (Lin et al. 2013)? We know that Treg are capable of bystander suppression (Tang and Bluestone 2008). We do not know if this suppression would be of harm to the transplant recipient (or maybe of benefit if the disease was an autoimmune process).

Other unanticipated off-target effects were seen with costimulation blockade trials. Antibodies against CD154 resulted in increased thromboembolic events. The unexpected complication was likely due to expression of this molecule on unanticipated cell types (platelets and endothelial cells). Other agents that block CD40 may have similar efficacy, but avoid these complications. Use of efalizumab was halted when there was a concern for increased risk of PML. This risk may be common to many immunosuppressant protocols, but serves as an example of another unexpected off-target effect.

Economic Considerations

A final consideration applicable to all of these approaches is an economic one. These protocols are expensive. Each protocol costs several million dollars to enroll a limited number of patients. In our own center, the cost of Treg isolation and expansion alone is ∼$45 K per transplant. Counterbalancing this cost are the expenses of traditional transplants. Financial analysis is most complete for end-stage renal disease (ESRD) and renal transplantation. Data from the USRDS (2012) report shows that the average Medicare cost of ESRD in 2010 was $87,561 per person per year. Despite the cost of the transplant procedure, it is estimated that the transplant-associated savings are approximately $200,000 over 5 years (reviewed in Axelrod 2013). The cost of the transplant procedures themselves would likely be similar for standard therapy and tolerance protocols. The economic benefits of tolerance protocols would be generated by reduction in immunosuppression costs. Annual Medicare costs for injectable medications and immunosuppression following transplant was $10,000 in 2010 (USRDS 2012). Additionally, there would be significant cost savings by potentially avoiding costs associated with long-term viral and bacterial prophylaxis and treatment of opportunistic infections. Tolerance approaches could be financially advantageous if the cost savings associated with avoiding immunosuppression and chemoprophylaxis increased graft survival, and reduced need for retransplants or dialysis exceeds the additional costs of these procedures.

CONCLUSIONS

It is clear that transplantation improves quality and length of life for patients with end-stage organ diseases. Unfortunately, the vast majority of transplant recipients require life-long immunosuppression. This immunosuppression increases risk for cancer, infection, and other direct organ toxicity. Despite continued use of these medications, functional organ lifespans are finite. There is a persisting risk of immune and nonimmune-mediated graft loss that results in decreased patient survival. Strategies that enable long-lasting graft function obviate need for immunosuppression medication, and maintained immune function could increase patient longevity. The question remains, are the costs of these tolerance-induction protocols worth the benefits of immunosuppression avoidance?

There are many considerations when attempting to answer “Is it worth it?” These considerations may not be the same for all organ systems or even for different recipients. For example, it may be worth the risks of total body irradiation and GVHD present in mixed chimerism protocols for a 50-year-old who gets a heart transplant and may not be a candidate for retransplant should their heart fail at the age of 60 from chronic rejection. On the other hand, a 25-year-old living donor kidney recipient might not want to take those risks if they know that if their graft fails they have the option of dialysis, and perhaps have another potential live donor for that eventuality. The consequences of transplant failure for life-saving organs (heart, lung, and liver) may make these recipients more suitable for higher-risk tolerance protocols than for organs with alternative therapies (insulin for pancreas transplants and dialysis for kidney transplants). Additional factors may influence the risk/benefit balance. Highly sensitized patients with donor-specific anti-HLA antibodies may take the risk of a tolerance protocol given their likely reduced graft survival compared with an unsensitized recipient receiving a 0-HLA mismatch graft. It may be one thing to ask a patient with hematologic malignancy to undergo a risky combined stem cell and kidney transplant with irradiation and chemotherapy, but a completely different consideration for a relatively healthy person needing a preemptive kidney transplant. Clearly, each tolerance protocol has a set of risks that must be balanced with the long-term benefits. This risk/benefit analysis would need to be individualized to the organ type, disease process, and even individual patient circumstances, and would also depend on the efficacy and costs of the protocol developed.

The last consideration is: What are the consequences of failure? Under current practice, transplant outcomes are pretty good (at least for short- and medium-term outcomes). Each of the tolerance protocols will have some failure rate. Is the long-term benefit in the successes worth the risks of graft loss, patient death, or other complications in the failures?

The evaluation of each tolerance protocol will be influenced by the success rates and the implications of failure. If protocol failure means loss of graft and need for retransplant in a kidney recipient, the risk may be warranted. If failure means loss of graft and death in a heart transplant recipient, the considerations will be different. If failure means resumption of immunosuppression, the down side is low. If protocol failure means graft loss or death, the implications are more considerable. For example, in the MGH series of patients undergoing combined bone marrow and kidney transplant for multiple myeloma, seven out of 10 patients were successfully weaned off of immunosuppression with stable long-term graft function (Markmann and Kawai 2012). However, three out of 10 failed their protocol and lost their grafts. Is a 70% long-term survival rate free of immunosuppression worth the 30% risk of graft loss? Also, what do we mean by “long-term graft survival?” The MGH experience has shown us that success measured at 1 year may lead to failures after five. Each organ, disease, and patient will have differing answers to these questions.

The answer to “Tolerance—Is it worth it?” is clear—“It depends.” The up side of a nontoxic protocol that enables long-term graft and patient survival without immunosuppression would be tremendous. But currently the risks are significant. As more results of ongoing and future trials become available, we must continually evaluate what are our successes and what are our failures? The decision of “Is it worth it” depends on the balance of the two.

Footnotes

Editors: Laurence A. Turka and Kathryn J. Wood

Additional Perspectives on Transplantation available at www.perspectivesinmedicine.org

REFERENCES

*Reference is also in this collection.

  1. Anderson D, Billingham RE, Lampkin GH, Medawar PB 1951. The use of skin grafting to distinguish between monozygotic and dizygotic twins in cattle. Heredity 5: 379–397 [Google Scholar]
  2. Ashton-Chess J, Giral M, Brouard S, Soulillou JP 2007. Spontaneous operational tolerance after immunosuppressive drug withdrawal in clinical renal allotransplantation. Transplantation 84: 1215–1219 [DOI] [PubMed] [Google Scholar]
  3. Axelrod DA 2013. Economic and financial outcomes in transplantation: Whose dime is it anyway? Curr Opin Organ Transplant 18: 222–228 [DOI] [PubMed] [Google Scholar]
  4. Baliga P, Chavin KD, Qin L, Woodward J, Lin J, Linsley PS, Bromberg JS 1994. CTLA4Ig prolongs allograft survival while suppressing cell-mediated immunity. Transplantation 58: 1082–1090 [PubMed] [Google Scholar]
  5. Bhatia S 2011. Long-term health impacts of hematopoietic stem cell transplantation inform recommendations for follow-up. Exp Rev Hematol 4: 437–452; quiz 453–454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Billingham RE, Lampkin GH, Medawar PB, Williams HLL 1952. Tolerance to homografts, twin diagnosis, and the freemartin condition in cattle. Heredity 6: 201–212 [Google Scholar]
  7. Billingham RE, Brent L, Medawar PB 1953. Actively acquired tolerance of foreign cells. Nature 172: 603–606 [DOI] [PubMed] [Google Scholar]
  8. Brouard S, Dupont A, Giral M, Louis S, Lair D, Braudeau C, Degauque N, Moizant F, Pallier A, Ruiz C, et al. 2005. Operationally tolerant and minimally immunosuppressed kidney recipients display strongly altered blood T-cell clonal regulation. Am J Transplant 5: 330–340 [DOI] [PubMed] [Google Scholar]
  9. Brouard S, Mansfield E, Braud C, Li L, Giral M, Hsieh SC, Baeten D, Zhang M, Ashton-Chess J, Braudeau C, et al. 2007. Identification of a peripheral blood transcriptional biomarker panel associated with operational renal allograft tolerance. Proc Natl Acad Sci 104: 15448–15453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J, Defor T, Levine BL, June CH, Rubinstein P, et al. 2011. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: Safety profile and detection kinetics. Blood 117: 1061–1070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Butcher JA, Hariharan S, Adams MB, Johnson CP, Roza AM, Cohen EP 1999. Renal transplantation for end-stage renal disease following bone marrow transplantation: A report of six cases, with and without immunosuppression. Clin Transplant 13: 330–335 [DOI] [PubMed] [Google Scholar]
  12. Butler JA, Roderick P, Mullee M, Mason JC, Peveler RC 2004. Frequency and impact of nonadherence to immunosuppressants after renal transplantation: A systematic review. Transplantation 77: 769–776 [DOI] [PubMed] [Google Scholar]
  13. Carson KR, Focosi D, Major EO, Petrini M, Richey EA, West DP, Bennett CL 2009. Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: A review from the Research on Adverse Drug Events and Reports (RADAR) Project. Lancet Oncol 10: 816–824 [DOI] [PubMed] [Google Scholar]
  14. *.Chapman JR, Webster AC, Wong G 2013. Cancer in the transplant recipient. Cold Spring Harb Perspect Med 3: a015677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. *.Cobbold SP, Waldmann H 2013. Regulatory cells and transplantation tolerance. Cold Spring Harb Perspect Med 3: a015545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Comerci GD Jr, Williams TM, Kellie S 2009. Immune tolerance after total lymphoid irradiation for heart transplantation: Immunosuppressant-free survival for 8 years. J Heart Lung Transplant 28: 743–745 [DOI] [PubMed] [Google Scholar]
  17. Couzin J 2005. Drug discovery. Magnificent obsession. Science 307: 1712–1715 [DOI] [PubMed] [Google Scholar]
  18. Dedrick RL, Walicke P, Garovoy M 2002. Anti-adhesion antibodies efalizumab, a humanized anti-CD11a monoclonal antibody. Transpl Immunol 9: 181–186 [DOI] [PubMed] [Google Scholar]
  19. Di Cocco P, Bonanni L, D’Angelo M, Clemente K, Greco S, Rizza V, Mazzotta C, Scelzo C, Famulari A, Pisani F, et al. 2009. Clinical operational tolerance after solid organ transplantation. Transplant Proc 41: 1278–1282 [DOI] [PubMed] [Google Scholar]
  20. Di Ianni M, Falzetti F, Carotti A, Terenzi A, Castellino F, Bonifacio E, Del Papa B, Zei T, Ostini RI, Cecchini D, et al. 2011. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood 117: 3921–3928 [DOI] [PubMed] [Google Scholar]
  21. Djamali A, Samaniego M, Muth B, Muehrer R, Hofmann RM, Pirsch J, Howard A, Mourad G, Becker BN 2006. Medical care of kidney transplant recipients after the first posttransplant year. Clin J Am Soc Nephrol 1: 623–640 [DOI] [PubMed] [Google Scholar]
  22. Dolganiuc A, Szabo G 2008. T cells with regulatory activity in hepatitis C virus infection: What we know and what we don’t. J Leukocyte Biol 84: 614–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Durrbach A, Pestana JM, Pearson T, Vincenti F, Garcia VD, Campistol J, Rial Mdel C, Florman S, Block A, Di Russo G, et al. 2010. A phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors (BENEFIT-EXT study). Am J Transplant 10: 547–557 [DOI] [PubMed] [Google Scholar]
  24. Einecke G, Reeve J, Sis B, Mengel M, Hidalgo L, Famulski KS, Matas A, Kasiske B, Kaplan B, Halloran PF 2010. A molecular classifier for predicting future graft loss in late kidney transplant biopsies. J Clin Invest 120: 1862–1872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. *.English K, Wood KJ 2013. Mesenchymal stromal cells in transplantation rejection and tolerance. Cold Spring Harb Perspect Med 3: a015560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Feng S, Ekong UD, Lobritto SJ, Demetris AJ, Roberts JP, Rosenthal P, Alonso EM, Philogene MC, Ikle D, Poole KM, et al. 2012. Complete immunosuppression withdrawal and subsequent allograft function among pediatric recipients of parental living donor liver transplants. JAMA 307: 283–293 [DOI] [PubMed] [Google Scholar]
  27. First MR, Dhadda S, Croy R, Holman J, Fitzsimmons WE 2013. New-onset diabetes after transplantation (NODAT): An evaluation of definitions in clinical trials. Transplantation 96: 58–64 [DOI] [PubMed] [Google Scholar]
  28. Fishman JA 2002. Overview: Fungal infections in the transplant patient. Transpl Infect Dis 4: 3–11 [DOI] [PubMed] [Google Scholar]
  29. Fishman JA 2007. Infection in solid-organ transplant recipients. N Engl J Med 357: 2601–2614 [DOI] [PubMed] [Google Scholar]
  30. *.Fishman JA. Opportunistic infections—Coming to the limits of immunosuppression? Cold Spring Harb Perspect Med. 2013a doi: 10.1101/cshperspect.a015669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Fishman JA 2013b. Overview: Cytomegalovirus and the herpesviruses in transplantation. Am J Transplant 13: 1–8; quiz 8 [DOI] [PubMed] [Google Scholar]
  32. Fredericks EM, Dore-Stites D 2010. Adherence to immunosuppressants: How can it be improved in adolescent organ transplant recipients? Curr Opin Organ Transplant 15: 614–620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Fudaba Y, Spitzer TR, Shaffer J, Kawai T, Fehr T, Delmonico F, Preffer F, Tolkoff-Rubin N, Dey BR, Saidman SL, et al. 2006. Myeloma responses and tolerance following combined kidney and nonmyeloablative marrow transplantation: In vivo and in vitro analyses. Am J Transplant 6: 2121–2133 [DOI] [PubMed] [Google Scholar]
  34. Gordon KB, Papp KA, Hamilton TK, Walicke PA, Dummer W, Li N, Bresnahan BW, Menter A, Efalizumab Study Group 2003. Efalizumab for patients with moderate to severe plaque psoriasis: A randomized controlled trial. JAMA 290: 3073–3080 [DOI] [PubMed] [Google Scholar]
  35. *.Grinyó JM 2013. Why is organ transplantation clinically important? Cold Spring Harb Perspect Med 3: a014985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Grohmann U, Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, Candeloro P, Belladonna ML, Bianchi R, Fioretti MC, et al. 2002. CTLA-4Ig regulates tryptophan catabolism in vivo. Nat Immunol 3: 1097–1101 [DOI] [PubMed] [Google Scholar]
  37. Halloran PF, Bromberg J, Kaplan B, Vincenti F 2008. Tolerance versus immunosuppression: A perspective. Am J Transplant 8: 1365–1366 [DOI] [PubMed] [Google Scholar]
  38. Hancock WW, Sayegh MH, Zheng XG, Peach R, Linsley PS, Turka LA 1996. Costimulatory function and expression of CD40 ligand, CD80, and CD86 in vascularized murine cardiac allograft rejection. Proc Natl Acad Sci 93: 13967–13972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Harrison JH, Merrill JP, Murray JE 1956. Renal homotransplantation in identical twins. Surgical Forum 6: 432–436 [PubMed] [Google Scholar]
  40. Hasek M, Hraba T 1955. Artificial production of immunological tolerance: Immunological effects of experimental embryonal parabiosis. Nature 175: 764–765 [DOI] [PubMed] [Google Scholar]
  41. Helg C, Chapuis B, Bolle JF, Morel P, Salomon D, Roux E, Antonioli V, Jeannet M, Leski M 1994. Renal transplantation without immunosuppression in a host with tolerance induced by allogeneic bone marrow transplantation. Transplantation 58: 1420–1422 [PubMed] [Google Scholar]
  42. Hofmann B, Tao K, Mai L, West LJ 2004. Acceptance of related and unrelated cardiac allografts in neonatally tolerized mice is cardio-specific and transferable by regulatory CD4+ T cells. J Heart Lung Transplant 23: 1069–1076 [DOI] [PubMed] [Google Scholar]
  43. Hutchinson JA, Brem-Exner BG, Riquelme P, Roelen D, Schulze M, Ivens K, Grabensee B, Witzke O, Philipp T, Renders L, et al. 2008a. A cell-based approach to the minimization of immunosuppression in renal transplantation. Transpl Int 21: 742–754 [DOI] [PubMed] [Google Scholar]
  44. Hutchinson JA, Riquelme P, Brem-Exner BG, Schulze M, Matthäi M, Renders L, Kunzendorf U, Geissler EK, Fändrich F 2008b. Transplant acceptance-inducing cells as an immune-conditioning therapy in renal transplantation. Transpl Int 21: 728–741 [DOI] [PubMed] [Google Scholar]
  45. Ildstad ST, Wren SM, Bluestone JA, Barbieri SA, Sachs DH 1985. Characterization of mixed allogeneic chimeras. Immunocompetence, in vitro reactivity, and genetic specificity of tolerance. J Exp Med 162: 231–244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Issa NC, Fishman JA 2009. Infectious complications of antilymphocyte therapies in solid organ transplantation. Clin Infect Dis 48: 772–786 [DOI] [PubMed] [Google Scholar]
  47. Jacobsen N, Taaning E, Ladefoged J, Kristensen JK, Pedersen FK 1994. Tolerance to an HLA-B,DR disparate kidney allograft after bone-marrow transplantation from same donor. Lancet 343: 800. [DOI] [PubMed] [Google Scholar]
  48. Josefowicz SZ, Lu LF, Rudensky AY 2012. Regulatory T cells: Mechanisms of differentiation and function. Ann Rev Immunol 30: 531–564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Judge TA, Tang A, Turka LA 1996. Immunosuppression through blockade of CD28:B7-mediated costimulatory signals. Immunol Res 15: 38–49 [DOI] [PubMed] [Google Scholar]
  50. Kahwaji J, Bunnapradist S, Hsu JW, Idroos ML, Dudek R 2011. Cause of death with graft function among renal transplant recipients in an integrated healthcare system. Transplantation 91: 225–230 [DOI] [PubMed] [Google Scholar]
  51. Kasiske BL, Snyder JJ, Gilbertson D, Matas AJ 2003. Diabetes mellitus after kidney transplantation in the United States. Am J Transplant 3: 178–185 [DOI] [PubMed] [Google Scholar]
  52. Kasiske BL, Snyder JJ, Gilbertson DT, Wang C 2004. Cancer after kidney transplantation in the United States. Am J Transplant 4: 905–913 [DOI] [PubMed] [Google Scholar]
  53. Kaufman CL, Colson YL, Wren SM, Watkins S, Simmons RL, Ildstad ST 1994. Phenotypic characterization of a novel bone marrow-derived cell that facilitates engraftment of allogeneic bone marrow stem cells. Blood 84: 2436–2446 [PubMed] [Google Scholar]
  54. Kawai T, Andrews D, Colvin RB, Sachs DH, Cosimi AB 2000. Thromboembolic complications after treatment with monoclonal antibody against CD40 ligand. Nat Med 6: 114. [DOI] [PubMed] [Google Scholar]
  55. Kawai T, Cosimi AB, Spitzer TR, Tolkoff-Rubin N, Suthanthiran M, Saidman SL, Shaffer J, Preffer FI, Ding R, Sharma V, et al. 2008. HLA-mismatched renal transplantation without maintenance immunosuppression. N Engl J Med 358: 353–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kenyon NS, Chatzipetrou M, Masetti M, Ranuncoli A, Oliveira M, Wagner JL, Kirk AD, Harlan DM, Burkly LC, Ricordi C 1999. Long-term survival and function of intrahepatic islet allografts in rhesus monkeys treated with humanized anti-CD154. Proc Natl Acad Sci 96: 8132–8137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kinnear G, Jones ND, Wood KJ 2013. Costimulation blockade: Current perspectives and implications for therapy. Transplantation 95: 527–535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kirk AD, Burkly LC, Batty DS, Baumgartner RE, Berning JD, Buchanan K, Fechner JH Jr, Germond RL, Kampen RL, Patterson NB, et al. 1999. Treatment with humanized monoclonal antibody against CD154 prevents acute renal allograft rejection in nonhuman primates. Nature Med 5: 686–693 [DOI] [PubMed] [Google Scholar]
  59. Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP III, Armand P, Cutler C, Ho VT, Treister NS, et al. 2011. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med 365: 2055–2066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Koulmanda M, Smith RN, Qipo A, Weir G, Auchincloss H, Strom TB 2006. Prolonged survival of allogeneic islets in cynomolgus monkeys after short-term anti-CD154-based therapy: Nonimmunologic graft failure? Am J Transplant 6: 687–696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Koulmanda M, Budo E, Bonner-Weir S, Qipo A, Putheti P, Degauque N, Shi H, Fan Z, Flier JS, Auchincloss H Jr, et al. 2007. Modification of adverse inflammation is required to cure new-onset type 1 diabetic hosts. Proc Natl Acad Sci 104: 13074–13079 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Koulmanda M, Qipo A, Fan Z, Smith N, Auchincloss H, Zheng XX, Strom TB . 2012. Prolonged survival of allogeneic islets in cynomolgus monkeys after short-term triple therapy. Am J Transplant 12: 1296–1302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Ku CC, Murakami M, Sakamoto A, Kappler J, Marrack P 2000. Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science 288: 675–678 [DOI] [PubMed] [Google Scholar]
  64. Larsen CP, Pearson TC 1997. The CD40 pathway in allograft rejection, acceptance, and tolerance. Curr Opin Immunol 9: 641–647 [DOI] [PubMed] [Google Scholar]
  65. Larsen CP, Elwood ET, Alexander DZ, Ritchie SC, Hendrix R, Tucker-Burden C, Cho HR, Aruffo A, Hollenbaugh D, Linsley PS, et al. 1996. Long-term acceptance of skin and cardiac allografts after blocking CD40 and CD28 pathways. Nature 381: 434–438 [DOI] [PubMed] [Google Scholar]
  66. Lebwohl M, Tyring SK, Hamilton TK, Toth D, Glazer S, Tawfik NH, Walicke P, Dummer W, Wang X, Garovoy MR, et al. 2003. A novel targeted T-cell modulator, efalizumab, for plaque psoriasis. N Engl J Med 349: 2004–2013 [DOI] [PubMed] [Google Scholar]
  67. Lennerling A, Forsberg A 2012. Self-reported non-adherence and beliefs about medication in a Swedish kidney transplant population. Open Nurs J 6: 41–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Lenschow DJ, Zeng Y, Thistlethwaite JR, Montag A, Brady W, Gibson MG, Linsley PS, Bluestone JA 1992. Long-term survival of xenogeneic pancreatic islet grafts induced by CTLA-4Ig. Science 257: 789–792 [DOI] [PubMed] [Google Scholar]
  69. Leventhal J, Abecassis M, Miller J, Gallon L, Ravindra K, Tollerud DJ, King B, Elliott MJ, Herzig G, Herzig R, et al. 2012. Chimerism and tolerance without GVHD or engraftment syndrome in HLA-mismatched combined kidney and hematopoietic stem cell transplantation. Sci Transl Med 4: 124ra28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Leventhal J, Abecassis M, Miller J, Gallon L, Tollerud D, Elliott MJ, Bozulic LD, Houston C, Sustento-Reodica N, Ildstad ST 2013. Tolerance induction in HLA disparate living donor kidney transplantation by donor stem cell infusion: Durable chimerism predicts outcome. Transplantation 95: 169–176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Li XC, Demirci G, Ferrari-Lacraz S, Groves C, Coyle A, Malek TR, Strom TB 2001. IL-15 and IL-2: A matter of life and death for T cells in vivo. Nat Med 7: 114–118 [DOI] [PubMed] [Google Scholar]
  72. Light J, Salomon DR, Diethelm AG, Alexander JW, Hunsicker L, Thistlethwaite R, Reinsmoen N, Stablein DM 2002. Bone marrow transfusions in cadaver renal allografts: Pilot trials with concurrent controls. Clin Transplant 16: 317–324 [DOI] [PubMed] [Google Scholar]
  73. Lin SZ, Chen KJ, Xu ZY, Chen H, Zhou L, Xie HY, Zheng SS 2013. Prediction of recurrence and survival in hepatocellular carcinoma based on two Cox models mainly determined by FoxP3+ regulatory T cells. Clin Transplant 16: 317–324 [DOI] [PubMed] [Google Scholar]
  74. LoCascio SA, Morokata T, Chittenden M, Preffer FI, Dombkowski DM, Andreola G, Crisalli K, Kawai T, Saidman SL, Spitzer TR, et al. 2010. Mixed chimerism, lymphocyte recovery, and evidence for early donor-specific unresponsiveness in patients receiving combined kidney and bone marrow transplantation to induce tolerance. Transplantation 90: 1607–1615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Lowe M, Badell IR, Thompson P, Martin B, Leopardi F, Strobert E, Price AA, Abdulkerim HS, Wang R, Iwakoshi NN, et al. 2012. A novel monoclonal antibody to CD40 prolongs islet allograft survival. Am J Transplant 12: 2079–2087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Luznik L, O’Donnell PV, Symons HJ, Chen AR, Leffell MS, Zahurak M, Gooley TA, Piantadosi S, Kaup M, Ambinder RF, et al. 2008. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant 14: 641–650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. *.Maltzman JS, Turka LA 2013. T cell costimulatory blockade in organ transplantation. Cold Spring Harb Perspect Med 10.1101/cshperspect.a015529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Markmann JF, Kawai T 2012. The quest for transplantation tolerance: Have we finally sipped from the cup? Sci Transl Med 4: 124fs5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Medawar PB 1964. The Nobel Prize in physiology or medicine 1960. Elsevier, Amsterdam [Google Scholar]
  80. Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B 2004. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant 4: 378–383 [DOI] [PubMed] [Google Scholar]
  81. Moranne O, Maillard N, Fafin C, Thibaudin L, Alamartine E, Mariat C 2013. Rate of renal graft function decline after one year is a strong predictor of all-cause mortality. Am J Transplant 13: 695–706 [DOI] [PubMed] [Google Scholar]
  82. Morton M, Coupes B, Roberts SA, Klapper PE, Byers RJ, Vallely PJ, Ryan K, Picton ML 2013. Epidemiology of posttransplantation lymphoproliferative disorder in adult renal transplant recipients. Transplantation 95: 470–478 [DOI] [PubMed] [Google Scholar]
  83. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chapman JR 2003. The natural history of chronic allograft nephropathy. N Engl J Med 349: 2326–2333 [DOI] [PubMed] [Google Scholar]
  84. Neff RT, Hurst FP, Falta EM, Bohen EM, Lentine KL, Dharnidharka VR, Agodoa LY, Jindal RM, Yuan CM, Abbott KC 2008. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation 86: 1474–1478 [DOI] [PubMed] [Google Scholar]
  85. Newell KA, Larsen CP 2006. Tolerance assays: Measuring the unknown. Transplantation 81: 1503–1509 [DOI] [PubMed] [Google Scholar]
  86. Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, Burlingham WJ, Marks WH, Sanz I, Lechler RI, et al. 2010. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest 120: 1836–1847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman AB, Young EW, Arndorfer J, Christensen L, Merion RM 2003. Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med 349: 931–940 [DOI] [PubMed] [Google Scholar]
  88. Olagne J, Caillard S, Gaub MP, Chenard MP, Moulin B 2011. Post-transplant lymphoproliferative disorders: Determination of donor/recipient origin in a large cohort of kidney recipients. Am J Transplant 11: 1260–1269 [DOI] [PubMed] [Google Scholar]
  89. Organ Procurement Transplantation Network (SRTR) SRoTR 2012. OPTN/SRTR 2011 Annual Data Report. Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau, Division of Transplantation, Washington, DC [Google Scholar]
  90. Orlando G, Soker S, Wood K 2009. Operational tolerance after liver transplantation. J Hepatol 50: 1247–1257 [DOI] [PubMed] [Google Scholar]
  91. Orlando G, Hematti P, Stratta RJ, Burke GW III, Di Cocco P, Pisani F, Soker S, Wood K 2010. Clinical operational tolerance after renal transplantation: Current status and future challenges. Ann Surg 252: 915–928 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Owen RD 1945. Immunogenetic consequences of vascular anastomoses between bovine twins. Science 102: 400–401 [DOI] [PubMed] [Google Scholar]
  93. Page EK, Dar WA, Knechtle SJ 2012. Tolerogenic therapies in transplantation. Front Immunol 3: 198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Parker DC, Greiner DL, Phillips NE, Appel MC, Steele AW, Durie FH, Noelle RJ, Mordes JP, Rossini AA 1995. Survival of mouse pancreatic islet allografts in recipients treated with allogeneic small lymphocytes and antibody to CD40 ligand. Proc Natl Acad Sci 92: 9560–9564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Pearson TC, Alexander DZ, Winn KJ, Linsley PS, Lowry RP, Larsen CP 1994. Transplantation tolerance induced by CTLA4-Ig. Transplantation 57: 1701–1706 [PubMed] [Google Scholar]
  96. Pearson TC, Alexander DZ, Corbascio M, Hendrix R, Ritchie SC, Linsley PS, Faherty D, Larsen CP 1997. Analysis of the B7 costimulatory pathway in allograft rejection. Transplantation 63: 1463–1469 [DOI] [PubMed] [Google Scholar]
  97. Pestana JO, Grinyo JM, Vanrenterghem Y, Becker T, Campistol JM, Florman S, Garcia VD, Kamar N, Lang P, Manfro RC, et al. 2012. Three-year outcomes from BENEFIT-EXT: A phase III study of belatacept versus cyclosporine in recipients of extended criteria donor kidneys. Am J Transplant 12: 630–639 [DOI] [PubMed] [Google Scholar]
  98. Posselt AM, Bellin MD, Tavakol M, Szot GL, Frassetto LA, Masharani U, Kerlan RK, Fong L, Vincenti FG, Hering BJ, et al. 2010. Islet transplantation in type 1 diabetics using an immunosuppressive protocol based on the anti-LFA-1 antibody efalizumab. Am J Transplant 10: 1870–1880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Rabinovitch A, Suarez-Pinzon WL, Shapiro AM, Rajotte RV, Power R 2002. Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. Diabetes 51: 638–645 [DOI] [PubMed] [Google Scholar]
  100. Rostaing L, Neumayer HH, Reyes-Acevedo R, Bresnahan B, Florman S, Vitko S, Heifets M, Xing J, Thomas D, Vincenti F 2011. Belatacept-versus cyclosporine-based immunosuppression in renal transplant recipients with pre-existing diabetes. Clin J Am Soc Nephrol 6: 2696–2704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Roussey-Kesler G, Giral M, Moreau A, Subra JF, Legendre C, Noel C, Pillebout E, Brouard S, Soulillou JP 2006. Clinical operational tolerance after kidney transplantation. Am J Transplant 6: 736–746 [DOI] [PubMed] [Google Scholar]
  102. Rüedi E, Sykes M, Ildstad ST, Chester CH, Althage A, Hengartner H, Sachs DH, Zinkernagel RM 1989. Antiviral T cell competence and restriction specificity of mixed allogeneic (P1 + P2–P1) irradiation chimeras. Cell Immunol 121: 185–195 [DOI] [PubMed] [Google Scholar]
  103. Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, Sene D, Cacoub P, Klatzmann D 2011. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365: 2067–2077 [DOI] [PubMed] [Google Scholar]
  104. Sachs DH, Kawai T, Colvin RB, Fishman JA, Sykes M, Cosimi AB 2008. Response to “Tolerance versus immunosuppression: A perspective.” Am J Transplant 8: 1573–1574 [DOI] [PubMed] [Google Scholar]
  105. *.Sachs DH, Kawai T, Sykes M 2013. Induction of tolerance through mixed chimerism. Cold Spring Harb Perspect Med 10.1101/cshperspect.a014985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Sagoo P, Perucha E, Sawitzki B, Tomiuk S, Stephens DA, Miqueu P, Chapman S, Craciun L, Sergeant R, Brouard S, et al. 2010. Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest 120: 1848–1861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Sayegh MH, Fine NA, Smith JL, Rennke HG, Milford EL, Tilney NL 1991. Immunologic tolerance to renal allografts after bone marrow transplants from the same donors. Ann Intern Med 114: 954–955 [DOI] [PubMed] [Google Scholar]
  108. Scandling JD, Busque S, Dejbakhsh-Jones S, Benike C, Sarwal M, Millan MT, Shizuru JA, Lowsky R, Engleman EG, Strober S 2012. Tolerance and withdrawal of immunosuppressive drugs in patients given kidney and hematopoietic cell transplants. Am J Transplant 12: 1133–1145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Scherer MN, Banas B, Mantouvalou K, Schnitzbauer A, Obed A, Krämer BK, Schlitt HJ 2007. Current concepts and perspectives of immunosuppression in organ transplantation. Langenbecks Arch Surg 392: 511–523 [DOI] [PubMed] [Google Scholar]
  110. Schmidt A, Oberle N, Krammer PH 2012. Molecular mechanisms of treg-mediated T cell suppression. Front Immunol 3: 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Sellers MT, Deierhoi MH, Curtis JJ, Gaston RS, Julian BA, Lanier DC Jr, Diethelm AG 2001. Tolerance in renal transplantation after allogeneic bone marrow transplantation-6-year follow-up. Transplantation 71: 1681–1683 [DOI] [PubMed] [Google Scholar]
  112. Shimizu K, Schonbeck U, Mach F, Libby P, Mitchell RN 2000. Host CD40 ligand deficiency long-term allograft survival and donor-specific tolerance in mouse cardiac transplantation but does not prevent graft arteriosclerosis. J Immunol 165: 3506–3518 [DOI] [PubMed] [Google Scholar]
  113. Sidiropoulos PI, Boumpas DT 2004. Lessons learned from anti-CD40L treatment in systemic lupus erythematosus patients. Lupus 13: 391–397 [DOI] [PubMed] [Google Scholar]
  114. Sorof JM, Koerper MA, Portale AA, Potter D, DeSantes K, Cowan M 1995. Renal transplantation without chronic immunosuppression after T cell-depleted, HLA-mismatched bone marrow transplantation. Transplantation 59: 1633–1635 [PubMed] [Google Scholar]
  115. Spitzer TR, Sykes M, Tolkoff-Rubin N, Kawai T, McAfee SL, Dey BR, Ballen K, Delmonico F, Saidman S, Sachs DH, et al. 2011. Long-term follow-up of recipients of combined human leukocyte antigen-matched bone marrow and kidney transplantation for multiple myeloma with end-stage renal disease. Transplantation 91: 672–676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Stern RS 2003. A promising step forward in psoriasis therapy. JAMA 290: 3133–3135 [DOI] [PubMed] [Google Scholar]
  117. Strober S, Dhillon M, Schubert M, Holm B, Engleman E, Benike C, Hoppe R, Sibley R, Myburgh JA, Collins G, et al. 1989. Acquired immune tolerance to cadaveric renal allografts. A study of three patients treated with total lymphoid irradiation. N Engl J Med 321: 28–33 [DOI] [PubMed] [Google Scholar]
  118. Strober S, Benike C, Krishnaswamy S, Engleman EG, Grumet FC 2000. Clinical transplantation tolerance twelve years after prospective withdrawal of immunosuppressive drugs: Studies of chimerism and anti-donor reactivity. Transplantation 69: 1549–1554 [DOI] [PubMed] [Google Scholar]
  119. Sykes M 2007. Immune tolerance: Mechanisms and application in clinical transplantation. J Intern Med 262: 288–310 [DOI] [PubMed] [Google Scholar]
  120. Tang Q, Bluestone JA 2008. The Foxp3+ regulatory T cell: A jack of all trades, master of regulation. Nat Immunol 9: 239–244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. *.Tang Q, Bluestone JA 2013. Regulatory T cell therapy in transplantation: Moving to the clinic. Cold Spring Harb Perspect Med 10.1101/cshperspect.a015552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Tang Q, Adams JY, Penaranda C, Melli K, Piaggio E, Sgouroudis E, Piccirillo CA, Salomon BL, Bluestone JA 2008. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 28: 687–697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Tichelli A, Rovó A, Socié G 2012. Late effects after hematopoietic stem cell transplantation—Critical issues. Curr Probl Dermatol 43: 132–149 [DOI] [PubMed] [Google Scholar]
  124. Tichelli A, Rovó A, Passweg J, Schwarze CP, Van Lint MT, Arat M, Socié G, Late Effects Working Party of the European Group for Blood Marrow Transplantation 2009. Late complications after hematopoietic stem cell transplantation. Expert Rev Hematol 2: 583–601 [DOI] [PubMed] [Google Scholar]
  125. Trzonkowski P, Bieniaszewska M, Juscinska J, Dobyszuk A, Krzystyniak A, Marek N, Myśliwska J, Hellmann A 2009. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+ CD25+ CD127 T regulatory cells. Clin Immunol 133: 22–26 [DOI] [PubMed] [Google Scholar]
  126. Turgeon NA, Avila JG, Cano JA, Hutchinson JJ, Badell IR, Page AJ, Adams AB, Sears MH, Bowen PH, Kirk AD, et al. 2010. Experience with a novel efalizumab-based immunosuppressive regimen to facilitate single donor islet cell transplantation. Am J Transplant 10: 2082–2091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. USRDS 2012. USRDS 2012 Annual Data Report: Atlas of chronic kidney disease and end-stage renal disease in the United States. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD [Google Scholar]
  128. Valderhaug TG, Hjelmesaeth J, Hartmann A, Roislien J, Bergrem HA, Leivestad T, Line PD, Jenssen T 2011. The association of early post-transplant glucose levels with long-term mortality. Diabetologia 54: 1341–1349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Vincenti F, Mendez R, Pescovitz M, Rajagopalan PR, Wilkinson AH, Butt K, Laskow D, Slakey DP, Lorber MI, Garg JP, et al. 2007. A phase I/II randomized open-label multicenter trial of efalizumab, a humanized anti-CD11a, anti-LFA-1 in renal transplantation. Am J Transplant 7: 1770–1777 [DOI] [PubMed] [Google Scholar]
  130. Vincenti F, Charpentier B, Vanrenterghem Y, Rostaing L, Bresnahan B, Darji P, Massari P, Mondragon-Ramirez GA, Agarwal M, Di Russo G, et al. 2010. A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study). Am J Transplant 10: 535–546 [DOI] [PubMed] [Google Scholar]
  131. Weber SC, Uhlenberg B, Raile K, Querfeld U, Muller D 2011. Polyoma virus-associated progressive multifocal leukoencephalopathy after renal transplantation: Regression following withdrawal of mycophenolate mofetil. Ped Transplant 15: E19–E24 [DOI] [PubMed] [Google Scholar]
  132. Winston DJ, Busuttil RW 2002. Randomized controlled trial of oral itraconazole solution versus intravenous/oral fluconazole for prevention of fungal infections in liver transplant recipients. Transplantation 74: 688–695 [DOI] [PubMed] [Google Scholar]
  133. Winston DJ, Emmanouilides C, Busuttil RW 1995. Infections in liver transplant recipients. Clin Infect Dis 21: 1077–1089; quiz 90–91 [DOI] [PubMed] [Google Scholar]
  134. Wolfe RA, Ashby VB, Milford EL, Ojo AO, Ettenger RE, Agodoa LY, Held PJ, Port FK 1999. Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant. N Engl J Med 341: 1725–1730 [DOI] [PubMed] [Google Scholar]
  135. Zheng XX, Sanchez-Fueyo A, Sho M, Domenig C, Sayegh MH, Strom TB 2003. Favorably tipping the balance between cytopathic and regulatory T cells to create transplantation tolerance. Immunity 19: 503–514 [DOI] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES