Open access

Operational Tolerance after Renal Transplantation in the Regenerative Medicine Era

Written By

Giuseppe Orlando, Pierpaolo Di Cocco, Lauren Corona, Tommaso Maria Manzia, Katia Clemente, Antonio Famulari and Francesco Pisani

Submitted: 10 November 2010 Published: 23 August 2011

DOI: 10.5772/20769

From the Edited Volume

Kidney Transplantation - New Perspectives

Edited by Dr.(Md) Magdalena Trzcinska

Chapter metrics overview

2,478 Chapter Downloads

View Full Metrics

1. Introduction

An immunosuppression [IS]-free state is the ultimate goal to achieve in solid organ transplantation [SOT] because IS-related toxicity tremendously impact on overall clinical outcome [Orlando, Hematti et al., 2010]. The IS-free state substantiates into what is referred to as clinical operational tolerance [COT], defined as the condition in which a SOT recipient retains stable graft function and lacks histological signs of rejection, after having been completely off all IS for at least 1 year. The patient in question is an immunocompetent host capable of responding to other immune challenges, including infections or transplantation of organs from third party donors [Orlando, Hematti et al., 2010; Orlando et al., 2008]. However, experimental and clinical information collected so far show that COT is extremely difficult to achieve and is organ-dependent. In fact, the majority of cases of COT reported so far relate to liver graft recipients, in reason of the well known immune-privileged status of the liver determined by factors that remain obscure.

In this chapter, we will provide evidence that the achievement of COT after RT remains exceptional and that tolerogenic strategies are not yet available for daily clinical practice. To do so, we will illustrate and discuss the most relevant cases of COT described so far in the English literature. However, by briefly touching the recent outstanding progress achieved in regenerative medicine, we will bring the attention of the reader on the fact that regenerative medicine may offer a valuable approach to generate immediate, stable and durable COT in SOT recipients.

Advertisement

2. Classification

Several classifications have been proposed to categorize all strategies proposed so far to induce COT in RT recipients [Fehr & Sykes, 2004; Orlando et al, 2008; Orlando, Hematti et al, 2010]. Nevertheless, given the interest and enthusiasm generated by recent investigations in the field of stem cell biology, we believe that so-called tolerogenic strategies – namely, protocols implemented to induce COT – may be divided into two new groups, cell-based and non-cell-based, according to whether or not stem cell strains showing some immunomodulatory effect in vitro are administered in the preoperative period in addition to standard IS.

2.1. Cell-based approach

2.1.1. Metachronous bone-marrow transplant for hematologic disorders

Bone marrow transplantation [BMT], results in the total replacement of the recipient’s bone marrow by the donor’s bone marrow hematopoietic cells, a condition referred to as full chimerism [Delis et al, 2004]. If the patient receives an organ from the same bone marrow donor late after BMT, then the reconstituted white cell compartment may recognize the new organ as self; theoretically, no response will be mounted by the host immune system and the new graft will be tolerated without need for any IS. However, despite several cases of BMT plus solid organ transplants [RT included] have been reported [Butcher et al, 1999; Chiang & Lazarus, 2003; Helg et al, 1994; Jacobsen et al, 1994; Light et al, 2002; Sayegh et al, 1991; Sellers et al, 2001; Sorof et al, 1995], yet COT has been an exceptional finding. Importantly, according to what reported by all series, COT developed in patients in which the transplant was performed several years after BMT, but never where BMT and RT were performed simultaneously.

2.1.2. Perioperative infusion of HSC

Somehow, the above reported data conflicted with what described in mice by Monaco and Wood, namely that the addition of donor bone marrow to a strong lymphocyte depleting regimen may result in the long-lasting survival of skin allografts from the same donor, without any maintenance IS [Monaco et al, 1970; Monaco et al, 1976; Monaco et al, 1985]. These experimental findings represented the platform for translational investigations aimed to prove that the peroperative infusion of donor-derived bone marrow cells can prolong allograft survival [Sykes, 2009]. The group at Massachusetts General Hospital adopted such strategy in 7 adult patients with renal failure due to multiple myeloma, who received combined human leukocyte antigen [HLA]-matched kidney and bone marrow transplant after a non-myeloablative conditioning regimen consisting of cyclophosphamide, antithymocyte globulin, and pretransplant thymic irradiation [Buhler et al, 2002; Fudaba et al, 2006; Spitzer et al, 1999; Spitzer et al, 2011]. Maintenance IS consisted of cyclosporine which was discontinued as early as day 73 post-transplant. Patients were followed for a mean of 7.4 (range 4-12) years. Two of them died for relapse of the baseline disease and therapy-related acute myeloid leukemia after 7.7 and 4.2 years, respectively; noteworthy, both were IS-free at the time of death. Of the 5 patients who are still alive [4 of whom with no evidence of myeloma recurrence], 3 are IS-free, and show serum creatinine levels of 0.63, 1.14 and 1.7 mg/dL, respectively. The remaining 2 individuals are receiving either prednisone or tacrolimus for graft-versus-host disease [but no rejection!], and present serum creatinine levels of 0.9 and 1.42 mg/dL.

More recently, the same group reported the short-term results of a seminal trial in which non-cancer patients received renal grafts and bone marrow from HLA haploidentical donors following nonmyeloablative conditioning to induce renal allograft tolerance [Kawai et al, 2008; Porcheray et al, 2009]. Even in this case, cyclosporine was the only immunosuppressant used for maintenance therapy. IS could be successfully weaned in 4 out of 5 patients, whereas the remaining patient experienced acute rejection and lost his graft. Cyclosporine could be withdrawn after 294 (range 240-272) days. However, matters of concern are represented by two major findings. First, the reported most recent mean creatinine clearance (67 ml/min, range 60-75) and serum creatinine levels (1.5 mg/dl, range 1.2-1.8) levels in the 4 IS-free patients were slightly abnormal. Second, 2 of these patients have shown evidence of de novo antibodies reactive to donor antigens between 1 and 2 years after the transplant [Porcheray et al, 2009]. Overall, this series demonstrates that B-cell immunity can occur in the context of T-cell tolerance to donor antigens through unknown mechanisms, otherwise said T-cell unresponsiveness is not synonymous of tolerance of both arms of the adaptive immune system. This is consistent with information reported by Nantes. Soulillou et al. extensively studied IS-free RT recipients [Ashton-Chess et al., 2007; Baeten et al., 2005; Ballet et al., 2006; Brouard et al., 2005; Louis et al., 2006; Roussey-Kesler et al., 2006;] and found that COT may occur in the presence of anti-donor class II antibodies, as well as in patients who have previously experienced acute rejection. Moreover, they showed that IS-free RT recipients are more likely to have received grafts from donors younger than 30 years of age, are low-responders to blood transfusion, may develop graft dysfunction at any time after the weaning of IS, and show an incidence of infectious diseases comparable to that in normal individuals.

The Stanford group has implemented a similar strategy in two distinct small series, where patients received a renal graft followed by the infusion of HSC from the same HLA-mismatched (x4) or -matched (x6) donors, respectively [Millan et al., 2002; Scandling et al., 2008]. IS consisted of induction with total lymphoid irradiation and rabbit anti-thymocyte globulin, followed by cyclosporine and prednisone as maintenance therapy. After steroid discontinuation on day 10, mycophenolate mofetil was introduced and administered for 1 month. Intravenous injection of HSC was repeated during the third postoperative week. Results were poor. In the first trial, only one patient could be weaned off IS, while however showing slightly elevated serum creatinine leves (1.4 mg/dL). In the second trial, one individual could be weaned off IS but renal function tests are not shown. Two further patients rejected, while the remaining 3 were still under weaning at the time of publication.

Overall, based on the reported information, the above series show the lack of safety of hematopoietic stem cell-based regimens. Despite this approach may allow minimization of overall IS, however further studies are required to improve safety and effectiveness. Importantly, most of the IS-free patients reported above do not comply with the stringent criteria for COT adopted in the present chapter.

2.1.3. Perioperative infusion of alternative types of immunomodulatory cells

Transplant-acceptance-inducing cells (TAIC) are immunoregulatory macrophages which have shown some capacity of establishing a state of alloantigen-specific partial tolerance of solid organ transplants in renal transplant recipients [Hutchinson, Brem-Exner et al., 2008; Hutchinson, Riquelme et al., 2008; Riquelme P et al., 2009]. TAIC’s presumed tolerogenic properties have been tested in two safety trials differing in the method for TAIC preparation, numbers of cells infused, induction IS, and timing of infusions. None of the 17 patients enrolled in either study could be weaned off IS, most of them rejected, 2 dropped out the study for non-immunological causes, while only 2 individuals did well yet under tacrolimus monotherapy at the end of the study. Overall, although these trials demonstrated that the infusion of TAIC is feasible, major concerns remain regarding the efficacy and safety of such an approach. It is unclear whether or not this approach confers any benefit in the establishment of minimal IS in RT patients when compared to the protocols currently adopted, as clearly stated by authors in the discussion. Lastly, the optimal dose and timing of cell infusions, as well as the most appropriate concomitant IS regimen, is yet to be determined.

For their ability for tissue repair and immunomodulation, mesenchymal stem cells (MSCs) are currently being evaluated for a wide variety of clinical applications including the treatment of disorders characterized by a dysfunction of immune regulation, such as graft versus host disease after bone marrow transplantation and rejection after cell or organ transplantation [Crop et al., 2009; Ding et al., 2009; Hematti, 2008; Le Blanc et al., 2008; Le Blanc & Ringden, 2007]. As MSCs seem to be able to promote engraftment of allogeneic cells, tissues and organs, as well as to prevent or treat rejection in pre-clinical models, they provide an exciting opportunity for further research in the field of IS minimization and withdrawal in SOT [Hematti, 2008; Hoogduijn MJ et al., 2010].

It is worth to mention that, in addition to stem cells, the therapeutic properties of various other immune cells like regulatory T cells or dendritic cells are currently being explored for SOT purposes [Issa, Hester et al., 2010; Issa, Schiopu et al., 2010; Orlando, Hematti et al., 2010].

2.2. Non cell-based protocols

2.2.1. Non-adherence to IS

The majority of cases of COT described so far after RT is represented by patients who spontaneously decided to stop IS for non-compliance to treatment [Ashton-Chess et al., 2007; Baeten et al., 2005; Ballet et al., 2006; Brouard et al., 2005; Hussey, 1975; Louis et al., 2006; Najarian, 1975; Newell et al., 2010; Owens et al., 1975; Roussey-Kesler et al., 2006; Sagoo et al., 2010; Uehling et al., 1976; Zoller et al., 1980]. These patients have been the object of numerous investigations conducted at different times by diverse authors with methods presenting dissimilar accuracy. However, we believe that it is worth to report the investigations by Burlingham et al. In an attempt to identify patients who may be weaned off IS at no risk, he recurred to the human-to-mouse trans-vivo delayed-type hypersensitivity assay [Geissler et al., 2001; Van Buskirk et al., 2000]. By observing that 2 renal allograft recipients failed to exhibit donor-reactive delayed type hypersensitivity responses, although they frequently develop donor-reactive alloantibodies, the authors demonstrated that this pattern of immune responses is not due to an absence of allosensitization, but to the development of an immune mechanism that actively inhibits anti-donor delayed-type (i.e., cell-mediated) immune responses. In other words, authors provided evidence that COT develops following the onset of regulatory, rather than suppressing mechanisms.

Of interest is the case of spontaneous interruption of IS by a 24-year old woman who had received a deceased donor RT at the age of 13 for membranoproliferative glomerulonephritis. When the woman found out to be pregnant, she stopped all IS to protect the fetus from any possible IS-induced teratogenic effect [Fischer et al., 1996]. Interestingly, she did not resume any IS after delivery, and no immunological response was observed for 9 years after the withdrawal of IS, while the patient was showing perfectly normal renal function tests.

2.2.2. Molecule-based tolerogenic protocols

A molecule-based tolerogenic regimen was implemented by Starzl a decade ago [Starzl et al., 2003]. The working hypothesis was that the need for continual high dose IS could be avoided in most cases with the use of a strong lymphocyte-depleting regimen prior to engraftment, followed by the administration of low dose tacrolimus monotherapy. The rationale was to remove the non-specific clones of immune cells responsible for rejection before contact with foreign donor antigens occurs. Once the donor antigens are in place after implantation of the new organ, repletion of immune cells occurs, favored by the homeostatic expansion triggered by leukocyte depletion. In addition, minimization of maintenance IS was implemented to further reduce the anti-donor response with just enough treatment to prevent irreversible immune damage to the graft, but not with such heavy treatment that the donor-specific clonal exhaustion-deletion process is precluded.

Broadly reacting rabbit antithymocyte globulins were administered preoperatively, followed by tacrolimus monotherapy, to 82 adult kidney, liver, pancreas, and intestinal transplant recipients. After a mean follow-up time of 18-months, 1-year patient and graft survival rates were 95% and 82% overall, respectively, IS-related morbidity was virtually eliminated, and 48/72 surviving patients were receiving spaced doses of tacrolimus monotherapy. Noteworthy, 25/39 (64%) renal, 12/17 (70%) liver, 5/12 (42%) pancreas, as well as 6/11 (54%) intestinal transplant recipients, resulted on tacrolimus spaced doses at the time of publication. However, IS could not be weaned off in any patient. Other protocols based on a similar strategy – i.e., leukocyte depletion followed by the administration of low dose single-drug IS - have been implemented after RT [Agarwal et al., 2008; Calne et al., 2000; Calne et al., 1999; Calne et al., 1998; Ciancio & Burke, 2008; Clatworthy et al., 2009; Kirk et al., 2003; Kirk et al., 2005; Orlando, Hematti et al., 2010; Trzonkowski et al., 2006; Trzonkowski et al., 2008; Watson et al., 2005] and liver, the organ most capable of developing COT, transplantation [Eason et al., 2005]. However, none of these protocols has proved effective or safe in producing COT nor has they shown any convincing impact on overall outcomes.

2.2.3. Total lymphoid irradiation

Three cases of COT have been described in RT recipients subjected to total lymphoid irradiation, a perioperative course of antithymocyte globulin, and maintenance prednisone that was gradually weaned and eventually stopped [Strober et al., 1989; Strober et al., 2000; Strober et al., 2004]. Two patients remained IS-free for 12 years and 69 months, while the third developed severe urinary tract obstruction 10 months after the IS withdrawal, for which he was eventually retransplanted. Importantly, the complete original series comprised 28 RT recipients, 25 of whom did not develop COT.

2.2.4. COT developed after intentional IS withdrawal for lymphoproliferative disorders

Two cases of COT developing after the intentional withdrawal of IS due to the development of post transplant lymphoproliferative disorders (PTLD) have been described, mainly within a larger series [Roussey-Kesler et al., 2006]. A third case relates to a 21-year-old man who received 2 haplo-identical RTs, the first at age 11 from his mother and the second at age 15 from his father [Christensen et al., 1998]. Three years after the second RT, he developed PTLD. IS was promptly interrupted and the PTLD subsequently resolved. At the time of publication, IS had been withdrawn for 3 years with no rejection.

ClassificationSubtypeCenterType of studyClaimed cases of COTLength of follow up from the time of the withdrawal of the IS
Cell-basedPrevious BMTBoston, Brigham and Women Hospital [Sayegh et al, 1991]Case report2*7 and 3 years
Geneva [Helg et al, 1994]Case report12 years
Copenhagen [Jacobsen et al, 1994]Case report11 year
San Francisco [Sorof et al, 1995]Case report12 year
Milwaukee [Bucther et al, 1999]Case report33, 11, 18 years
Birmingham, US [Sellers et al, 2001]Case report17
HSCBoston, Massachussetts General Hospital [Spitzer et al, 1999; Buhler et al, 2002; Fudaba et al, 2006; Spitzer et al, 2011]Prospective, non comparative3/612.1, 7.7, 4, 6.8 years
Boston, Massachussetts General Hospital [Kawai et al, 2008; Porcheray et al, 2009]Prospective, non comparative4/54.6, 3.4, 2.2 and 1.2 years
Stanford [Millan et al, 2002]Prospective, non comparative1/414 years
Stanford [Scandling et al, 2008]Prospective, non comparative1/328 months
Miami [Ciancio et al, 2004]Prospective, non randomized, comparative0/63-
TACIKiel [Hutchinson et al, 2008]Prospective, non comparative0/12-
Kiel [Hutchinson et al, 2008]Prospective, non comparative0/5-
Non cell-basedNon-adherence to ISColumbus-Madison [VanBuskirk et al, 2000; Geissler et al, 2001]Case report33 years
Madison [Uehling et al, 1976]Case report5, but 4 rejected after few months5 years
Los Angeles [Owens et al, 1975]Case report4, but eventually 1 rejected after 18 months17, 23, 52 months
Minneapolis [Najarian, 1975]Case report6, but 5 rejected after few monthsNA
Madison [Hussey, 1975]Case report8, but 7 rejected after few months40
TBIStanford [Strober et al, 1989; Strober et al, 2000]Prospective3/28, but 1 rejected after 10 months for non-immunological reasons144 and 69 months
PregnancyErlangen-Munster [Fischer et al, 1996]Case report19 years
PTLDAarhus [Christensen et a, 1998]Case report1"/>3 years
Molecule-basedPittsburgh [Starzl et al, 2003]Prospective, non comparative0/39-
Cambridge [Clatworthy et al, 2009; Watson et al, 2005; Calne et al, 2000; Calne et al, 1999; Calne et al, 1998]Prospective, comparative, non randomized0/33-
Oxford [Trzonkoski et al, 2008]Prospective, non comparative0/13-
Bethesda [Kirk et al, 2003]Prospective, non comparative 0/7-
Bethesda [Kirk et al, 2005]Prospective, non comparative 0/5-
SurveysBoston [Zoller et al, 1980]Descriptive, observational13"/>1 year
Nantes [Roussey-Kesler et al, 2006]Descriptive, investigative 10, but 2 rejected after 7 and 13 years9 years [mean time, range 1-20]
Nantes [Braud et al, 2008; Sivozhelezov et al, 2008]Descriptive, investigative 8"/>1 year
Nantes [Bouard et al, 2007]Descriptive, investigative 17"/>1 year
Los Angeles [Owens et al, 1975]Descriptive, observational24, but 22 rejected after few months9, 36 months
European Consortium for tolerance [Sagoo et al, 2010]Descriptive, investigative 11"/>1 year
American network for immune tolerance [Newell et al, 2010]Descriptive, investigative 25"/>1 year

Table 1.

Synoptic view of all successful and unsuccessful cases of COT described after RT. The mismatch between the number of cases claimed by numerous authors and the real effectice number of COT calculated according to the definition of COT adopted in the present manuscript is evident [116 vs. 108, respectively].

Advertisement

3. Immune monitoring

As illustrated above, most cases of COT have developed in individuals who have spontaneously terminated IS. When all clinical trials in which a presumed tolerogenic protocol has been implemented are taken into consideration, it is frustrating to observe that COT could be obtained in only 6 out of 248 patients, accounting for a poor, unacceptable success rate of 2.5% [Orlando, Hematti et al, 2010]. As a corollary, the tolerogenic regimens attempted so far in RT recipients are not efficient and, more importantly, lack safety. Ideally, before implementing any of such regimens, investigators should rely on parameters able to predict with high accuracy whether patients would tolerate the weaning process without any risk to reject. Unfortunately, no parameter as such is available for routine practice, not even renal biopsy. For example, Burlingham reported on a late graft rejection in a patient who received a RT 9.5 years earlier from his mother and who had been IS-free for 7 years; a gradual rise in serum creatinine level to 2.0 mg/dl prompted a biopsy that ruled out rejection, yet 10 months later severe cellular rejection arose [Burlingham et al., 2000].

Investigators have obtained promising results in the field of liver transplantation where this problem has been circumvented by the identification of so-called markers of tolerance [Martinez-Llordella et al., 2008; Martinez-Llordella et al., 2007], defined as functional and molecular correlates of immune reactivity whose purpose is to provide clinically useful information for therapeutic decision-making in view of IS withdrawal [Ashton-Chess et al., 2009]. Investigations in tolerant liver transplant recipients resulted in the discovery and validation of a tolerance-associated transcriptional patterns, consisting of several gene signatures and multiple peripheral blood lymphocyte subsets capable of identifying tolerant and non-tolerant recipients with high accuracy. As these data suggested that transcriptional profiling of peripheral blood can be employed to identify recipients who can discontinue immunosuppressive therapy at no risk for rejection, RT researchers are currently exploring a signature of COT after RT which may allow the selection of those patients who may be more prone to develop an IS-free state with no or quasi no risk for rejection.

The group in Nantes has pioneered investigations aimed to the identification of specific biological signatures of COT [Braud et al., 2008; Brouard et al., 2007; Sivozhelezov et al., 2008]. Brouard et al. identified a set of 49 genes and differentially expressed gene transcripts using gene-expression profiling of peripheral blood from 17 tolerant RT recipients, with tolerance class prediction scores of >90% [Brouard et al., 2007]. This fingerprint is expected to identify patients who might be eligible for a progressive tapering of their immunosuppressive medications and, more importantly, those who instead need to stay on their current IS regimen. The same group has also exploited the capabilities of high throughput microarray technology to study peripheral blood specific gene expression profiles and corresponding molecular pathways associated with operational tolerance [Braud et al., 2008; Sivozhelezov et al., 2008]. Investigations revealed that tolerant patients display a set of 343 differentially expressed genes, mainly immune and defense genes, in their peripheral blood mononuclear cells (PBMC), of which 223 were also different from healthy volunteers. Using the expression pattern of these 343 genes, they were able to correctly classify >80% of the patients in a cross-validation analysis and correctly identified all of the samples over time. All together, this study identified a unique PBMC gene signature associated with human operational tolerance in kidney transplantation [Orlando, Hematti et al., 2010].

Investigations have been conducted in parallel in Europe and the United States. The European Union Indices of Tolerance Network showed that IS-free RT patients present a distinctive expansion of peripheral blood B lymphocytes and natural killer cells and differential expression of several immune-relevant genes in the absence of donor-specific antibodies [Sagoo et al., 2010]. Similar population expansion of B immune cells and selective expression of B cell-related genes in samples obtained from tolerant individuals were noted by the National Institute of Health’s Immune Tolerance Network [Newell et al., 2010].

Advertisement

4. The potential of regenerative medicine

The term regenerative medicine refers to that field in the health sciences which aims to replace or regenerate human cells, tissues, or organs in order to restore or establish normal function [Mason & Dunnill, 2008; Orlando, Baptista et al., 2010; Orlando et al., 2011]. The process of regenerating body parts can occur in vivo or ex vivo and may require cells, natural or artificial scaffolding materials, growth factors, or different combinations of all three elements. Instead, the term tissue engineering refers to a field which is narrower in scope and strictly limited to the production of body parts ex vivo, by seeding cells either on or into a supporting scaffold [Orlando et al., 2011].

In the last decade, investigators in the field have been able to produce and implant in patients relatively simple hollow organs like skin [Naughton et al., 1999], vessels [Hibino et al., 2010; L’Heureux N et al., 2007; Mc Allister et al., 2009; Shinoka et al., 2001; Shinoka et al., 2005;], bladders [Atala et al., 2006], windpipes [Macchiarini et al., 2008] and urethras [Raya-Rivera et al., 2011]. Importantly, all constructs were manufactured by either combining autologous cells with scaffolding material, or through the engineering of autologous cells per se [Orlando, Baptista et al., 2010; Orlando et al., 2011]. Importantly, none of the patients did require IS at any time after implantation of the bioengineered body part. Therefore, regenerative medicine has shown the potential to offer a valuable approach to COT. Importantly, as the above mentioned implanted body parts were bioengineered from autologous cells, recipients never required IS and therefore we can conclude that regenerative medicine offers the only possible approach to immediate, stable and durable COT. In fact, as illustrated throughout the whole chapter, all cases of COT reported so far in the English literature developed at least several weeks after the transplants and always required some IS, the exception being RT between identical twins [Orlando, Hematti et al., 2010]. Moreover, in several circumstances, patients who could be weaned off IS eventually rejected and required resumption of IS.

Bioengineering technology has been implemented to manufacture heart [Ott et al., 2008], liver [Badylak et al., 2011; Baptista et al., 2011; Uydun et al., 2010;] and lung [Ott et al., 2010; Petersen et al., 2010;] organoids from rodent organs [Orlando, Baptista et al, 2010; Orlando et al, 2011]. In extenso, rat or ferret organs were decellularized with detergents and repopulated with cells from unrelated species, humans included. In the ideal scenario, when and if this technology will be perfected, the cellular compartment will be reconstituted – even in this case – from patient’s autologous cells, whereas the supporting scaffold will be represented by an acellular porcine or non-human primate organ depleted of its native cells.

Advertisement

5. Conclusions

When all the above information is taken together, it is clear that, although a wealth of knowledge exists, little progress has been made in developing a sure-fire strategy towards attaining COT. Despite tolerance has been widely investigated for decades, efforts to understand the mechanisms underlying this phenomenon and how to achieve it have thus far been to no avail. In addition to the failure of all molecule-based strategies, we have learned that stem cells do exert some modulatory effect on the immune system but we do not know why and how this occurs. Therefore, we cannot predict when the opportunities for COT to develop in a patient the greatest.

As it stands, with the technology that is currently available, the withdrawal of IS after RT cannot yet be encouraged because it is neither effective nor safe and must be considered as still in an experimental phase. Efforts to identify a peripheral blood transcriptional biomarker panel associated with COT after RT are certainly laudable but, provided the safety for the withdrawal of IS is not guaranteed, any clinical implementation should be banned outside specialized cutting-edge center. The lack of safety of all tolerogenic strategies implemented so far remains their major weakness. Recent revolutionary progress in regenerative medicine has revealed the immense potential of the field pertinent to COT. In a foreseeable future, regenerative medicine will meet the two major needs of SOT, namely that of a potentially inexhaustible source of organs and COT itself [Orlando, 2011].

References

  1. 1. AgarwalA.ShenL. Y.KirkA. D.2008The role of alemtuzumab in facilitating maintenance immunosuppression minimization following solid organ transplantation. Transplant Immunology, 201-26110966-3274
  2. 2. Ashton-ChessJ.GiralM.SoulillouJ. P.BrouardS.2009Can immune monitoring help to minimize immunosuppression in kidney transplantation? Transplant International: official journal of the European Society for Organ Transplantation, 2211101190934-0874
  3. 3. Ashton-ChessJ.GiralM.BrouardS.SoulillouJ. P.2007Spontaneous operational tolerance after immunosuppressive drug withdrawal in clinical renal allotransplantation. Transplantation, 8410121512190041-1337
  4. 4. AtalaA.BauerS. B.SokerS.YooJ. J.RetikA. B.2006Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet, 3679518124112460140-6736
  5. 5. BadylakS.F. ZhangL. MedberryC.J. FukumitsuK.FaulkD. JiangH.ReingJ.E. GramignoliR. KomoriJ. RossM. NagayaM.LagasseE. Beer StolzD. StromS. FoxI.J. (2011). A Whole Organ Regenerative Medicine Approach for Liver Replacement. Tissue Engineering. Part C, Methods, 1937-338419373384
  6. 6. BaetenD.LouisS.BraudC.BraudeauC.BalletC.MoizantF.PallierA.GiralM.BrouardS.SoulillouJ. P.2005Phenotypically and functionally distinct CD8+ lymphocyte populations in long-term drug-free tolerance and chronic rejection in human kidney graft recipients. Journal of the American Society of Nephrology: JASN, 1712943041046-6673
  7. 7. BalletC.Roussey-KeslerG.AubinJ. T.BrouardS.GiralM.MiqueuP.LouisS.Van der WerfS.SoulillouJ. P.2006Humoral and cellular responses to influenza vaccination in human recipients naturally tolerant to a kidney allograft. American Journal of Transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 611279628011600-6135
  8. 8. BaptistaP. M.SiddiquiM. M.LozierG.RodriguezS. R.AtalaA.SokerS.2011The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology: official journal of the American Association for the study of Liver Diseases, 5326046170270-9139
  9. 9. BraudC.BaetenD.GiralM.PallierA.Ashton-ChessJ.BraudeauC.ChevalierC. LebarsA. LégerJ. MoreauA.PechkovaE. NicoliniC.SoulillouJ.P. BrouardS. (2008). Immunosuppressive drug-free operational immune tolerance in human kidney transplant recipients: Part I. Blood gene expression statistical analysis. Journal of cellular biochemistry, 0730-2312610316811692
  10. 10. BrouardS.DupontA.GiralM.LouisS.LairD.BraudeauC.DegauqueN.MoizantF.PallierA.RuizC.GuilletM.LaplaudD.SoulillouJ. P.2005Operationally tolerant and minimally immunosuppressed kidney recipients display strongly altered blood T-cell clonal regulation. American Journal of Transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 523303401600-6135
  11. 11. BrouardS.MansfieldE.BraudC.LiL.GiralM.HsiehS. C.BaetenD.ZhangM.Ashton-ChessJ.BraudeauC.HsiehF.DupontA.PallierA.MoreauA.LouisS.RuizC.SalvatierraO.SoulillouJ. P.SarwalM.2007Identification of a peripheral blood transcriptional biomarker panel associated with operational renal allograft tolerance. Proceedings of the National Academy of Sciences of the United States of America, 1043915448154530027-8424
  12. 12. BühlerL. H.SpitzerT. R.SykesM.SachsD. H.DelmonicoF. L.Tolkoff-RubinN.SaidmanS. L.SacksteinR.Mc AfeeS.DeyB.ColbyC.CosimiA. B.2002Induction of kidney allograft tolerance after transient lymphohematopoietic chimerism in patients with multiple myeloma and end-stage renal disease. Transplantation, 7410140514090041-1337
  13. 13. BurlinghamW. J.Jankowska-GanE.Van BuskirkA.OroszC. G.LeeJ. H.KusakaS.2000Loss of tolerance to a maternal kidney transplant is selective for HLA class II: evidence from trans-vivo DTH and alloantibody analysis. Human Immunology, 6112139514020198-8859
  14. 14. ButcherJ. A.HariharanS.AdamsM. B.JohnsonC. P.RozaA. M.CohenE. P.1999Renal transplantation for end-stage renal disease following bone marrow transplantation: a report of six cases, with and without immunosuppression. Clinical Transplantation, 1343303350902-0063
  15. 15. CalneR.MoffattS. D.FriendP. J.JamiesonN. V.BradleyJ. A.HaleG.FirthJ.BradleyJ.SmithK. G.WaldmannH.2000Prope tolerance with induction using Campath 1H and low-dose cyclosporin monotherapy in 31 cadaveric renal allograft recipients. Nippon Geka Gakkai Zasshi, 10133013060301-4894
  16. 16. CalneR.MoffattS. D.FriendP. J.JamiesonN. V.BradleyJ. A.HaleG.FirthJ.BradleyJ.SmithK. G.WaldmannH.1999Campath IH allows low-dose cyclosporine monotherapy in 31 cadaveric renal allograft recipients. Transplantation, 6810161316160041-1337
  17. 17. CalneR.FriendP.MoffattS.BradleyA.HaleG.FirthJ.BradleyJ.SmithK.WaldmannH.1998Prope tolerance, perioperative Campath 1H, and low-dose cyclosporin monotherapy in renal allograft recipients. Lancet, 3519117170117020140-6736
  18. 18. ChiangK. Y.LazarusH. M.2003Should we be performing more combined hematopoietic stem cell plus solid organ transplants? Bone marrow transplantation, 3186336420268-3369
  19. 19. ChristensenL. L.GrunnetN.RüdigerN.MøllerB.BirkelandS. A.1998Indications of immunological tolerance in kidney transplantation. Tissue Antigens, 5166376440001-2815
  20. 20. CiancioG.BurkeG. W.2007Alemtuzumab (Campath-1H) in kidney transplantation. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 8115201600-6135
  21. 21. CiancioG.BurkeG. W.MoonJ.Garcia-MoralesR.RosenA.EsquenaziV.MathewJ.JinY.MillerJ.2004Donor bone marrow infusion in deceased and living donor renal transplantation. Yonsei medical journal, 45699810030513-5796
  22. 22. ClatworthyM. R.FriendP. J.CalneR. Y.RebelloP. R.HaleG.WaldmannH.WatsonC. J..2009Alemtuzumab (CAMPATH-1H) for the Treatment of Acute Rejection in Kidney Transplant Recipients: Long-Term Follow-Up. Transplantation, 877109210950041-1337
  23. 23. CropM.BaanC.WeimarW.HoogduijnM.2009Potential of mesenchymal stem cells as immune therapy in solid-organ transplantation. Transplant International: official journal of the European Society for Organ Transplantation, 2243653760934-0874
  24. 24. DelisS.CiancioG.BurkeG. W.Garcia-MoralesG.MillerJ.2004Donor bone marrow transplantation, chimerism and tolerance. Transplant Immunology, 1321051150966-3274
  25. 25. DingY. XuD.FengG.BushellA.MuschelR.J. WoodK.J. (2009). Mesenchymal stem cells prevent the rejection of fully allogenic islet grafts by the immunosuppressive activity of matrix metalloproteinase-2 and-9. Diabetes, 0012-179785817971806
  26. 26. EasonJ. D.CohenA. J.NairS.AlcanteraT.LossG. E.2005Tolerance: is it worth the risk? Transplantation, 799115711590041-1337
  27. 27. FehrT.SykesM.2004Tolerance induction in clinical transplantation. Transplant Immunology, 1321171300966-3274
  28. 28. FischerT.SchobelH.BarenbrockM. (1996). Specific immune tolerance during pregnancy after renal transplantation. European journal of obstetrics, gynecology, and reproductive biology, 0301-2115270217219
  29. 29. FudabaY. SpitzerT.R. ShafferJ.KawaiT.FehrT. DelmonicoF. PrefferF.Tolkoff-RubinN. DeyB.R. SaidmanS.L. KrausA. BonnefoixT.McAfeeS. PowerK. KattlemanK. ColvinR.B. SachsD.H. CosimiA.B. SykesM. (2006). Myeloma responses and tolerance following combined kidney and nonmyeloablative marrow transplantation: in vivo and in vitro analyses. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 1600-61359621212133
  30. 30. GeisslerF.Jankowska-GanE.De Vito-HaynesL. D.RheinT.KalayogluM.SollingerH. W.BurlinghamW. J.2001Human liver allograft acceptance and the "tolerance assay": in vitro anti-donor T cell assays show hyporeactivity to donor cells, but unlike DTH, fail to detect linked suppression. Transplantation, 7245715800041-1337
  31. 31. HelgC.ChapuisB.BolleJ. F.MorelP.SalomonD.RouxE.AntonioliV.JeannetM.LeskiM.1994Renal transplantation without immunosuppression in a host with tolerance induced by allogeneic bone marrow transplantation. Transplantation, 5812142014220041-1337
  32. 32. HutchinsonJ. A.Brem-ExnerB. G.RiquelmeP.RoelenD.SchulzeM.IvensK.GrabenseeB.WitzkeO.PhilippT.RendersL.HumpeA.SotnikovaA.MatthäiM.HeumannA.GövertF.SchulteT.KabelitzD.ClaasF. H.GeisslerE. K.KunzendorfU.FändrichF.2008A cell-based approach to the minimization of immunosuppression in renal transplantation. Transplant International: official journal of the European Society for Organ Transplantation, 2187427540934-0874
  33. 33. HutchinsonJ. A.RiquelmeP.Brem-ExnerB. G.SchulzeM.MatthäiM.RendersL.KunzendorfU.GeisslerE. K.FändrichF.2008Transplant acceptance-inducing cells as an immune-conditioning therapy in renal transplantation. Transplant International: official journal of the European Society for Organ Transplantation, 2187287410934-0874
  34. 34. HemattiP. (2008). Role of mesenchymal stromal cells in solid organ transplantation. Transplantation reviews, 0095-5470422262273
  35. 35. HibinoN.Mc GillicuddyE.MatsumuraG.IchiharaY.NaitoY.BreuerC.ShinokaT.2010Late-term results of tissue-engineered vascular grafts in humans. The Journal of thoracic and cardiovascular surgery, 1392431436e1-2, 0022-5223
  36. 36. HoogduijnM. J.PoppF. C.GrohnertA.CropM. J.van RhijnM.RowshaniA. T.EggenhoferE.RennerP.ReindersM. E.RabelinkT. J.van der LaanL. J.DorF. J.IjzermansJ. N.GeneverP. G.LangeC.DurrbachA.HoutgraafJ. H.ChristB.SeifertM.ShagidulinM.DonckierV.DeansR.RingdenO.PericoN.RemuzziG.BartholomewA.SchlittH. J.WeimarW.BaanC. C.DahlkeM. H.2010Advancement of mesenchymal stem cell therapy in solid organ transplantation (MISOT). Transplantation, 9021241260934-0874
  37. 37. HusseyJ. L.1976Letter: Discontinuance of immunosuppression. Archives of surgery, 11156140272-5533
  38. 38. IssaF.HesterJ.GotoR.NadigS. N.GoodacreT. E.WoodK.2010Ex vivo-expanded human regulatory T cells prevent the rejection of skin allografts in a humanized mouse model. Transplantation, 9012132113270934-0874
  39. 39. IssaF.SchiopuA.WoodK. J.2010Role of T cells in graft rejection and transplantation tolerance. Expert review of clinical immunology, 611551690174-4666X
  40. 40. JacobsenN.TaaningE.LadefogedJ.KristensenJ. K.PedersenF. K.1994Tolerance to an HLA-B,DR disparate kidney allograft after bone-marrow transplantation from same donor. Lancet, 3438900800 EOF0140-6736
  41. 41. KawaiT.CosimiA. B.SpitzerT. R.Tolkoff-RubinN.SuthanthiranM.SaidmanS. L.ShafferJ.PrefferF.DingR.SharmaV.FishmanJ. A.DeyB.KoD. S.HertlM.GoesN. B.WongW.WilliamsW. W.ColvinR. B.SykesM.SachsD. H.2008HLA-mismatched renal transplantation without maintenance immunosuppression. The New England journal of medicine, 35843533610028-4793
  42. 42. KirkA. D.MannonR. B.KleinerD. E.SwansonJ. S.KampenR. L.CendalesL. K.ElsterE. A.WakefieldT.ChamberlainC.HoffmannS. C.HaleD. A.2005Results from a human renal allograft tolerance trial evaluating T-cell depletion with alemtuzumab combined with deoxyspergualin. Transplantation, 808105110590934-0874
  43. 43. KirkA. D.HaleD. A.MannonR. B.KleinerD. E.HoffmannS. C.KampenR. L.CendalesL. K.TadakiD. K.HarlanD. M.SwansonS. J.2003Results from a human renal allograft tolerance trial evaluating the humanized CD52-specific monoclonal antibody alemtuzumab (CAMPATH-1H). Transplantation, 7611201290934-0874
  44. 44. Le BlancK.FrassoniF.BallL.LocatelliF.RoelofsH.LewisI.LaninoE.SundbergB.BernardoM. E.RembergerM.DiniG.EgelerR. M.BacigalupoA.FibbeW.RingdénO.2008Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet, 3719624157915860140-6736
  45. 45. Le BlancK.RingdénO.2007Immunomodulation by mesenchymal stem cells and clinical experience. Journal of internal medicine, 26255095250954-6820
  46. 46. L’HeureuxN.Mc AllisterT. N.de la FuenteL. M.2007Tissue-engineered blood vessel for adult arterial revascularization. The New England journal of medicine, 35714145114530028-4793
  47. 47. LightJ.SalomonD. R.DiethelmA. G.AlexanderJ. W.HunsickerL.ThistlethwaiteR.ReinsmoenN.StableinD. M.2007Bone marrow transfusions in cadaver renal allografts: pilot trials with concurrent controls. Clinical transplantation, 35714145114530902-0063
  48. 48. LouisS.BraudeauC.GiralM.DupontA.MoizantF.RobillardN.MoreauA.SoulillouJ. P.BrouardS.2006Contrasting CD25hiCD4+T cells/FOXP3 patterns in chronic rejection and operational drug-free tolerance. Transplantation, 8133984070934-0874
  49. 49. MacchiariniP. JungebluthP. GoT.AsnaghiM.A. ReesL.E. CoganT.A.DodsonA.MartorellJ. BelliniS. ParnigottoP.P. DickinsonS.C. HollanderA.P. ManteroS. ConconiM.T. BirchallM.A. (2008). Clinical transplantation of a tissue-engineered airway. Lancet, 0140-6736965537220232030
  50. 50. Martínez-LlordellaM.LozanoJ. J.Puig-PeyI.OrlandoG.TisoneG.LerutJ.BenitezC.PonsJ. A.ParrillaP.RamirezP.BrugueraM.RimolaA.Sánchez-FueyoA.2008Towards a diagnostic test of operational tolerance in liver transplantation employing transcriptional profiling. The Journal of clinical investigation, 1188284528570021-9738
  51. 51. Martínez-LlordellaM.Puig-PeyI.OrlandoG.TisoneG.RamoniM.LerutJ.RimolaA.NavasaM.MargaritC.BilbaoI.Hernández-FuentesM.SoulillouJ. P.Sánchez-FueyoA.2007Multiparameter immune profiling of operational tolerance in liver transplantation. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 723093191600-6135
  52. 52. MasonC.DunnillP.2008A brief definition of regenerative medicine. Regenerative medicine, 31151746-0751
  53. 53. Mc AllisterT. N.MaruszewskiM.GarridoS. A.WystrychowskiW.DusserreN.MariniA.ZagalskiK.FiorilloA.AvilaH.ManglanoX.AntonelliJ.KocherA.ZembalaM.CierpkaL.de la FuenteL. M.L’heureuxN.2009Effectiveness of haemodialysis access with an autologous tissue-engineered vascular graft: a multicentre cohort study. Lancet, 3739673144014460140-6736
  54. 54. MillanM. T.ShizuruJ. A.HoffmannP.Dejbakhsh-JonesS.ScandlingJ. D.GrumetF. C.TanJ. C.SalvatierraO.HoppeR. T.StroberS.2002Mixed chimerism and immunosuppressive drug withdrawal after HLA-mismatched kidney and hematopoietic progenitor transplantation. Transplantation, 7391386 EOF91 EOF0934-0874
  55. 55. MonacoA. P.WoodM. L.MakiT.MadrasP. N.SahyounA. I.SimpsonM. A.1985Attempt to induce unresponsiveness to human renal allografts with antilymphocyte globulin and donor-specific bone marrow. Transpl Proc ;2713121314
  56. 56. MonacoA. P.ClarkA. W.WoodM. L.SahyounA. I.CodishS. D.BrownR. W.1976Possible active enhancement of a human cadaver renal allograft with antilymphocyte serum (ALS) and donor bone marrow: case report of an initial attempt. Surgery, 7943843920039-6060
  57. 57. MonacoA. P.WoodM. L.1970Studies on heterologous antilymphocyte serum in mice. VII. Optimal cellular antigen for induction of immunologic tolerance with antilymphocyte serum. Transplantation proceedings, 244894960041-1345
  58. 58. NajarianJ. S.1975Editorial comment. Arch Surg ;110:1451.
  59. 59. NaughtonG.1999The Advanced Tissue Sciences story. Scientific American, 280484850036-8733
  60. 60. NewellK.A. AsareA. KirkA.D.GislerT.D. BourcierK. SuthanthiranM.BurlinghamW.J. MarksW.H. SanzI. LechlerR.I. Hernandez-FuentesM.P. TurkaL.A. Seyfert-MargolisV.L. Immune Tolerance Network ST507 Study Group. (2010). Identification of a B cell signature associated with renal transplant tolerance in humans. The Journal of clinical investigation, 0021-9738612018361847
  61. 61. OrlandoG.WoodK. J.StrattaR. J.YooJ.AtalaA.SokerS.(n.dRegenerativemedicine.organtransplantation.Pastpresent.futureTransplantation, in press, 0934-087409340874
  62. 62. OrlandoG.2011Transplantation as a subfield of regenerative medicine. An interview by Lauren Constable. Expert review of clinical immunology, 7No.???, 1371410174-4666X
  63. 63. OrlandoG.BaptistaP.BirchallM.De CoppiP.FarneyA.Guimaraes-SouzaN. K.OparaE.RogersJ.SeliktarD.Shapira-SchweitzerK.StrattaR. J.AtalaA.WoodK. J.SokerS.2010Regenerative medicine as applied to solid organ transplantation: current status and future challenges. Transplant International: official journal of the European Society for Organ Transplantation, 2432232320934-0874
  64. 64. OrlandoG.HemattiP.StrattaR. J.BurkeG. W.Di CoccoP.PisaniF.SokerS.WoodK. J.2010Clinical operational tolerance after renal transplantation: current status and future challenges. Annals of surgery, 25269159280003-4932
  65. 65. OrlandoG.SokerS.WoodK.2009Operational tolerance after liver transplantation. Journal of hepatology, 506124712570168-8278
  66. 66. OttH. C.ClippingerB.ConradC.SchuetzC.PomerantsevaI.IkonomouL.KottonD.VacantiJ. P.2010Regeneration and orthotopic transplantation of a bioartificial lung. Nature medicine, 1689279331078-8956
  67. 67. OttH. C.MatthiesenT. S.GohS. K.BlackL. D.KrenS. M.NetoffT. I.TaylorD. A.2008Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nature medicine, 1422132211078-8956
  68. 68. OwensM. L.MaxwellG.GoodnightJ.WolcottM. W.1975Discontinuance of immunosuppression in renal transplant patients. Archives of surgery, 11012145014510272-5533
  69. 69. PetersenT. H.CalleE. A.ZhaoL.LeeE. J.GuiL.RaredonM. B.GavrilovK.YiT.ZhuangZ. W.BreuerC.HerzogE.NiklasonL. E.2010Tissue-Engineered Lungs for in Vivo Implantation. Science, 3295995385410193-4511
  70. 70. Raya-RiveraA.EsquilianoD. R.YooJ. J.Lopez-BayghenE.SokerS.AtalaA.2011Tissue-engineered autologoug urethras for patients who need reconstruction: an observational study. Lancet, (n.d.) 0140-673601406736
  71. 71. RiquelmeP.GövertF.GeisslerE. K.FändrichF.HutchinsonJ. A.2009Human transplant acceptance-inducing cells suppress mitogen-stimulated T cell proliferation. Transplant immunology, 2131621650966-3274
  72. 72. Roussey-KeslerG.GiralM.MoreauA.SubraJ. F.LegendreC.NoelC.PilleboutE.BrouardS.SoulillouJ. P.2006Clinical operational tolerance after kidney transplantation. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 647367461600-6135
  73. 73. SagooP.PeruchaE.SawitzkiB. TomiukS. StephensD.A. MiqueuP.ChapmanS.CraciunL. SergeantR. BrouardS.RovisF.JimenezE.BallowA.GiralM.Rebollo-MesaI. Le MoineA.BraudeauC.HiltonR.GerstmayerB. BourcierK. SharifA.KrajewskaM.LordG.M. RobertsI.GoldmanM.WoodK.J. NewellK.Seyfert-MargolisV. WarrensA.N.JanssenU. VolkH.D. SoulillouJ.P. Hernandez-FuentesM.P. LechlerR.I. (2010). Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. The Journal of clinical investigation, 0021-9738612018481861
  74. 74. SayeghM. H.FineN. A.SmithJ. L.RennkeH. G.MilfordE. L.TilneyN. L.1991Immunologic tolerance to renal allografts after bone marrow transplants from the same donors. Annals of internal medicine, 114119549550003-4819
  75. 75. ScandlingJ. D.BusqueS.Dejbakhsh-JonesS.BenikeC.MillanM. T.ShizuruJ. A.HoppeR. T.LowskyR.EnglemanE. G.StroberS.2008Tolerance and chimerism after renal and hematopoietic-cell transplantation. The New England journal of medicine, 35843623680028-4793
  76. 76. SellersM. T.DeierhoiM. H.CurtisJ. J.GastonR. S.JulianB. A.LanierD. C.DiethelmA. G.2001Tolerance in renal transplantation after allogeneic bone marrow transplantation-6-year follow-up. Transplantation, 7111168116830041-1337
  77. 77. ShinokaT.MatsumuraG.HibinoN.NaitoY.WatanabeM.KonumaT.SakamotoT.NagatsuM.KurosawaH.2005Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells. The Journal of thoracic and cardiovascular surgery, 1296133013380022-5223
  78. 78. ShinokaT.ImaiY.IkadaY.2001Transplantation of a Tissue-Engineered Pulmonary Artery. The New England journal of medicine, 34475325330028-4793
  79. 79. SivozhelezovV.BraudC. GiacomelliL.PechkovaE.GiralM.SoulillouJ.P. BrouardS. NicoliniC. (2008). Immunosuppressive drug-free operational immune tolerance in human kidney transplants recipients. Part II. Non-statistical gene microarray analysis. Journal of cellular biochemistry, 0730-2312610316931906
  80. 80. SorofJ. M.KoerperM. A.PortaleA. A.PotterD.De SantesK.CowanM.1995Renal transplantation without chronic immunosuppression after T cell-depleted, HLA-mismatched bone marrow transplantation. Transplantation, 5911163316350041-1337
  81. 81. SpitzerT. R.DelmonicoF.Tolkoff-RubinN.Mc AfeeS.SacksteinR.SaidmanS.ColbyC.SykesM.SachsD. H.CosimiA. B.1999Combined histocompatibility leukocyte antigen-matched donor bone marrow and renal transplantation for multiple myeloma with end stage renal disease: the induction of allograft tolerance through mixed lymphohematopoietic chimerism. Transplantation, 6844804840041-1337
  82. 82. StarzlT. E.MuraseN.Abu-ElmagdK.GrayE. A.ShapiroR.EghtesadB.CorryR. J.JordanM. L.FontesP.GayowskiT.BondG.ScantleburyV. P.PotdarS.RandhawaP.WuT.ZeeviA.NalesnikM. A.WoodwardJ.MarcosA.TruccoM.DemetrisA. J.FungJ. J.2003Tolerogenic immunosuppression for organ transplantation. Lancet, 3619368150215100140-6736
  83. 83. StroberS.LowskyR. J.ShizuruJ. A.ScandlingJ. D.MillanM. T.2004Approaches to transplantation tolerance in humans. Transplantation, 7769329360041-1337
  84. 84. StroberS.BenikeC.KrishnaswamyS.EnglemanE. G.GrumetF. C.2000Clinical transplantation tolerance twelve years after prospective withdrawal of immunosuppressive drugs: studies of chimerism and anti-donor reactivity. Transplantation, 698154915540041-1337
  85. 85. StroberS.DhillonM.SchubertM.HolmB.EnglemanE.BenikeC.HoppeR.SibleyR.MyburghJ. A.CollinsG.LevinB.1989Acquired immune tolerance to cadaveric renal allografts. A study of three patients treated with total lymphoid irradiation. The New England journal of medicine, 321128330028-4793
  86. 86. SykesM.2009Hematopoietic cell transplantation for tolerance induction: animal models to clinical trials. Transplantation, 7769329360041-1337
  87. 87. TrzonkowskiP.ZilvettiM.ChapmanS.WieckiewiczJ.SutherlandA.FriendP.WoodK. J.2008Homeostatic repopulation by CD28-CD8+ T cells in alemtuzumab-depleted kidney transplant recipients treated with reduced immunosuppression. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 823383471600-6135
  88. 88. UehlingD. T.HusseyJ. L.WeinsteinA. B.WankR.BachF. H.1976Cessation of immunosuppression after renal transplantation. Surgery, 7932782820039-6060
  89. 89. UygunB. E.Soto-GutierrezA.YagiH.IzamisM. L.GuzzardiM. A.ShulmanC.MilwidJ.KobayashiN.TillesA.BerthiaumeF.HertlM.NahmiasY.YarmushM. L.UygunK.2010Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nature medicine, 1678148201078-8956
  90. 90. Van BuskirkA. M.BurlinghamW. J.Jankowska-GanE.ChinT.KusakaS.GeisslerF.PelletierR. P.OroszC. G.2000Human allograft acceptance is associated with immune regulation. The Journal of clinical investigation, 10611451550021-9738
  91. 91. WatsonC. J.BradleyJ. A.FriendP. J.FirthJ.TaylorC. J.BradleyJ. R.SmithK. G.ThiruS.JamiesonN. V.HaleG.WaldmannH.GalneR.2005Alemtuzumab (CAMPATH 1H) induction therapy in cadaveric kidney transplantation--efficacy and safety at five years. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, 56134713531600-6135
  92. 92. ZollerK. M.ChoS. I.CohenJ. J.HarringtonJ. T.1980Cessation of immunosuppressive therapy after successful transplantation: a national survey. Kidney international, 1811101140085-2538

Written By

Giuseppe Orlando, Pierpaolo Di Cocco, Lauren Corona, Tommaso Maria Manzia, Katia Clemente, Antonio Famulari and Francesco Pisani

Submitted: 10 November 2010 Published: 23 August 2011