Open access

Modern Immunosuppression Regimens in Kidney Transplantation

Written By

Cheguevara Afaneh, Meredith J. Aull and Sandip Kapur

Submitted: 20 June 2012 Published: 19 December 2012

DOI: 10.5772/54092

From the Edited Volume

Current Concepts in Kidney Transplantation

Edited by Sandip Kapur, Cheguevara Afaneh and Meredith J. Aull

Chapter metrics overview

2,815 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Historical Background of Induction Therapy

The initial results of kidney transplantation were significantly affected by a high rate of acute rejection as well as significant perioperative morbidity.Historically, the armamentarium of the transplant physician consisted of glucocorticoids and azathioprine. Significant improvements in the science and understanding of kidney transplantation immunology have lead to the development of induction therapy agents. Early induction therapy agents possessed little specificity and delivered a broad spectrum of effects;however, their potent ability to prevent early acute rejection episodes led to their widespread use [1].

The extensive use of these formulations exposed their flaws. The cross-reactivity with hematopoietic cells revealed dose-limiting side effects including thrombocytopenia, anemia, and neutropenia [2, 3]. Moreover, the lack of standardized preparation led to variations in dosing. In addition, these formulations had significant antigenic properties as a result of using horse or rabbit based formulations, which lead to significant side effects, such as serum sickness, cytokine release syndrome, or even anaphylaxis [4-6].

The development of specific, monoclonal antibodies by Kohler and Milstein circumvented many of the drawbacks of polyclonal formulations, including lack of specificity and variability in preparation [7]. Muromonab, or OKT3, was the first monoclonal antibody prepared from mouse specific for cluster of differentiation 3 (CD3) [8]. OKT3 was effective at specifically depleting T cells from the circulation, and became widely used as a valuable tool to combat acute rejection episodes [9, 10]. Nevertheless, these monoclonal formulations still maintained some of the similar side effect profile of the polyclonal formulations, including an antigenic response to the protein or cytokine release syndrome, which lead to limited dosing in some patients [11].

The 1980’s marked an important era in transplantation with new advances in genetic engineering. Monoclonal antibodies became more sophisticated, targeting specific T cell populations and allowing blockade of T cell activation, such as the interleukin 2 receptor (IL-2R) or CD25 [12]. Moreover, the ability to avoid antigenic proteins by encoding genetic sequences of DNA binding sites of animal proteins onto human antibodies led to the development of chimeric monoclonal antibodies [13-15]. Using these techniques, soluble fusion proteins can be formed by merging nonantibody receptors with the Fc portion of antibodies.

1.2. Antibodies

Understanding the structure and function of antibodies is critical to understanding the efficacy of antibody induction therapy. Antibodies are composed of two identical heavy chains (either µ, γ, α, ε, or δ) and two identical light chains (either κ or λ). The heavy and light chain portions create two identical antigen binding sites (Fab fragment) which are held together by the common region, termed the Fc portion [16]. The type of heavy chain differentiates the immunoglobulin type as IgM, IgG, IgA, IgE, and IgD. In clinical transplantation, the IgG molecule is typically utilized, as it’s readily produced and structurally feasible to manipulate (Fig. 1).

Figure 1.

Basic antibody structure. Depicted is a standard IgG molecule. The heavy chains are colored in blue, while the light chains are colored in green. The yellow lines signify the disulfide bonds.

Antibodies are present on the surface of B cells. Upon secretion into the serum, antibodies are able to neutralize circulating antigens. Antibodies maintain their effector functions irrespective of species. Antibodies are capable of various functions, including mimicking activating ligands of receptors and serving as receptor inhibitors by blocking the ligand binding site [17, 18]. In some instances, antibody binding can lead to both activation and inhibition by inducing surface molecule internalization, whereby the molecule is removed from the surface of the cell [19]. This results in a negligible net effect. A major limitation of antibody use is the inability to directly bind intracellular molecules.

Antibodies have the ability to deplete target cells through two basic mechanisms. First, antibodies can activate the complement system resulting in complement-mediated lysis of target cells. Second, certain cells with Fc region receptors have the ability to phagocytosecells covered with antibodies through a mechanism termed antibody-dependent cellular cytotoxicity (ADCC) (Fig.2). The efficacy with which this occurs depends upon the Fab fragment and the Fc region [20]. It is important to note that cells which have significantly matured, or memory cells, are somewhat resistant to antibody-dependent depletion mechanisms, possibly due to increased expression of anti-apoptotic or complement regulatory genes [21].

Figure 2.

Antibody-dependent cellular cytotoxicity (ADCC). The Fc receptor on the macrophage is used to bind the constant Fc portion of antibodies to facilitate engulfment of cells coated with antibodies.

1.3. Classifying induction therapy agents

Induction therapy agents can be classified into two groups: depleting agents and non-depleting agents (Table 1). This distinction is based on the ability to target specific antigens or cells, leading to a decrease in the total expression or cell count. Most depleting agents are relatively potent with potential for toxicity with prolonged administration, while non-depleting agents are generally well-tolerated. In addition, the use of induction therapy agents has decreased the rates of acute rejection in the first 6 months compared to no induction therapy [22]. Although these short-term benefits appear promising, long-term outcomes, including patient and graft survival rates, have not been shown to be altered by the use of induction therapy, possibly the effect of long-term maintenance immunosuppressive therapy or even patient co-morbidities.

The overall success of a kidney transplant is contingent on both surgical technique and potent immunosuppressive medications. Although induction therapy has not affected surgical morbidity, the rate of allograft thrombosis has been shown to be reduced in children with the use of induction agents [23, 24]. However, not all medications used are FDA-approved for induction therapy. Additionally, these medications are not without risks, including infectious complications and the development of post-transplantlymphoproliferative disorder (PTLD), which has been well-described with the use of OKT3 and maintenance immunosuppression [25, 26]. Because of the effects of depleting agents on T cells, appropriate infectious prophylaxis should be instituted for all transplant recipients.

In 1995 induction therapy was used in less than half of all kidney transplants in the United States, while 10 years later, approximately 70% of all kidney transplant recipients received induction therapy [27]. Given the availability of various potent, specific induction agents in modern transplantation, the clinical dilemma lies in selecting the most appropriate agent for a given patient, taking into account co-morbidities, donor quality, immunological status, and planned immunosuppression maintenance therapy.

AgentClonalityTargetsDosingHalflifeDuration of effectsCytokine Release Syndrome?
rATG1PolyclonalVarious immune targets, especially T cellsMultiple doses (POD2#0-4)29.8-37.7 daysMonths to yearsYes
BasiliximabMonoclonalCD25 (predominantly activated T cells)2 doses (POD2#0 & 4)7.2 daysWeeksNo
DaclizumabMonoclonalCD25 (predominantly activated T cells)Multiple doses (POD2#0, then every 2 weeks)20 daysWeeksNo
AlemtuzumabMonoclonalCD52 (naïve T cells, some B cells, and monocytes)Typically 1 dose (POD2#0)12 daysMonths to yearsYes (less than rATG1)

Table 1.

Pharmacological Comparison of Induction Therapy

Advertisement

2. Induction therapy agents

2.1. Depleting agents

2.1.1. Rabbit Antithymocyte globulin (rATG)

2.1.1.1. Mechanism of Action

Rabbit antithymocyte globulin (rATG) is a polyclonal heterologous antibody produced from immunizing rabbits with human thymocytes, which serve as the immunogens (Fig. 3) [28]. The rabbit serum is then gathered and purified to remove antibodies with potentially detrimental effects and only the IgGisotypes are collected. Despite these purification techniques, it is possible that the majority of antibodies in these formulations serve no therapeutic purpose [29, 30]. When administered to humans, the rATG antibody formulations bind all antigens that the rabbits were exposed to during the immunization process.

rATG binds multiple T cell surface antigens and receptors involved in antigen recognition, adhesion and costimulation, including CD2, CD3, CD4, CD5, CD8, CD28, CD45, and CD40L. In addition, rATG may also bind non-T cell molecules such as CD16, CD20, CD56, and the major histocompatibility molecules (class I and II) [28-30]. The depleting effect of rATG occurs within 24 hours of administration and can persist with a prolonged serum half-life of several weeks [31, 32]. The effects of lymphocyte depletion are persists for years following administration, as evidenced by selectively low CD4+ T cell counts [33, 34].

Figure 3.

Polyclonal antibodies. Polyclonal antibodies are non-specific and bind multiple antigens as shown in the figure.

2.1.1.2. Clinical applications

rATG has been approved for use as an induction agent and for the treatment of acute rejection in Europe since 1984 [35]. However, in the United States, it is only indicated for the treatment of acute rejection. Nevertheless, it is routinely administered as induction therapy in many centers in the United States. Although early studies demonstrated an increased infectious risk and post-transplant malignancy when administered in conjunction with cyclosporine [36], improvements in infectious prophylaxis and lower doses have significantly alleviated these risks.

rATG administration improves early outcomes in kidney transplantation. Although the exact mechanism leading to this is unclear, rATG may minimize ischemia-reperfusion injury and potentially prevent the development of delayed graft function, which has been associated with poorer outcomes [37]. rATG has been used in patients at higher risk of developing delayed graft function, including recipients of donation after cardiac death donors, and recipients of extended criteria donors [38-40]. It is also administered in patients at higher immunologic risk, such as retransplants. Finally, it may help minimize the need for maintenance immunosuppression therapy facilitating early corticosteroid withdrawal [40, 41].

2.1.1.3. Adverse effects

Patients treated with rATG may experience a variety of side effects. It has been associated with a syndrome called cytokine release syndrome (Fig. 4), which is common to many polyclonal antibody formulations. Patients may experience mild flu-like symptoms, such as fever, chills, nausea, urticaria, rash, and headache [32]. This occurs as a result of increased production of tumor necrosis factor-α, IL-1, and IL-6 [28, 32, 42]. Premedication with corticosteroids, antipyretics, and antihistamines can prevent or treat these flu-like symptoms. In some cases, patients may develop more severe shock-like reactions, such as dyspnea, severe hypotension, pulmonary edema, or even anaphylaxis. Although patients frequently experience the mild flu-like symptoms and not the more severe reactions, recipient co-morbid conditions, such as cardiac or pulmonary disease, should be considered when selecting rATG as an induction agent. Serum sickness has also been associated with rATG administration in up to 7-10% of patients [43, 44].

Figure 4.

Antibody activation and cytokine release. Antibodies can bind antigens resulting in activation of the cell and cytokine release as illustrated in the figure.

Hematological adverse events may occur, including leucopenia and thrombocytopenia. It is important to monitor white blood cell, lymphocyte, and platelet counts daily. Effectively, these adverse events may lead to an increase in infectious complications, including cytomegalovirus (CMV), herpes simplex virus, Epstein-Barr virus (EBV), and varicella [45, 46].

2.1.2. Alemtuzumab

2.1.2.1. Mechanism of action

Alemtuzumab, or Campath-1H, is a monoclonal antibody to rat antihuman CD52 (Fig. 5). It is an IgG1 humanized molecule [47]. CD52 is present in high abundance on most lymphocytes, including T cell, B cells, and monocytes, but not hematopoietic precursors [48]. It effectively depletes T cells, and some B cells and monocytes in the circulation as well as the allograft [49].

Figure 5.

Monoclonal antibodies. Monoclonal antibodies are specific and bind a single antigen as shown in the figure.

2.1.2.2. Clinical applications

Alemtuzumab has not been approved for use as an induction agent; however, this is a common off-label use. At this time, it is only approved to treat lymphogenous malignancies. As an off-label induction agent, it’s been used with various immunosuppression regimens, including steroid-sparing regimens. Effectively, it depletes lymphocytes at the time of transplantation and last for several months to a year before the immune system is reconstituted [50]. Alemtuzumab is given at a dose of 30 mg or 0.3 mg/kg through a peripheral line over 3 hours. Sometimes 2 doses are given, although T cells are expectedly removed within 1 hour of initial administration [21, 49].

Alemtuzumab depletes all T cell subsets, but has a predilection for more naïve T cells [21]. Memory T cell subsets may not be depleted with this therapy, but these cell types are especially susceptible to calcineurin inhibitors. Because of the prompt and intense depletion, alemtuzumab is especially appealing to use in patients with delayed graft function, as calcineurin inhibitor therapy can be withheld to avoid concomitant calcineurin-induced renal insults.

Early studies of alemtuzumab demonstrated its efficacy as a treatment therapy for acute rejection; however, it was associated with significant infectious morbidity and mortality [47]. Patients were significantly over-immunosuppressed, especially on a triple maintenance therapy. More recent literature has been small studies or anecdotal data [51-53]. Because its efficacy is greatest against naïve T cells, its use in sensitized patients may-be limited.

In a recent study, alemtuzumab was prospectively compared to basiliximab and rATG as an induction agent in patients on a steroid-sparing immunosuppressive regimen [54]. Alemtuzumab demonstrated lower short-term rates of acute rejection compared to basiliximab in patients at low-risk of developing acute rejection. At 3-years, however, the rates of acute rejection were no different between alemtuzumab and rATG. Patients receiving alemtuzumab did not experience an increased incidence of adverse events.

2.1.2.3. Adverse effects

Similar to rATG, alemtuzumab has been associated with cytokine release syndrome, but to a lesser extent. With adequate premedication with methylprednisolone, acetaminophen, and diphenhydramine, the cytokine release is blunted. Rash is one of the most common manifestations, while anaphylaxis and hypotension have been reported. It has been linked to the development of autoimmune thyroiditis in patients treated with alemtuzumab for multiple sclerosis [55]. This has also been reported in a renal transplant recipient treated with alemtuzumab [56].

2.2. Non-depleting agents

2.2.1.Basiliximab

2.2.1.1. Mechanism of action

Basiliximab is a chimeric mouse-human monoclonal IgG1 antibody to CD25, the α-subunit of the IL-2 receptor. Basiliximab inhibition of IL-2 binding occurs through steric hindrance (Fig. 6). Effectively, basiliximab causes prevention of early T cell activation, as opposed to T cell depletion [50].

2.2.1.2. Clinical applications

Basiliximab targets naïve T cells, limiting its role to induction therapy. The first dose is administered on the day of transplant with the final dose administered on postoperative day 4 (20 mg per dose) via a peripheral line. Its use has been associated with decreased rates of acute cellular rejection compared to no formal induction agent on either triple or double drug immunosuppression regimens [57, 58]. Additional studies comparing basiliximab induction to polyclonal antibody depleting induction agents in the setting of triple maintenance immunosuppression regimens have shown similar outcomes with respect to acute rejection rates and delayed graft function [59, 60]. Basiliximab induction therapy has been successfully used in steroid avoidance immunosuppression regimens [61]. In the setting of monotherapy or calcineurin inhibitor free regimens; however, basiliximab has not been shown to be effective in preventing early immunologic events [62, 63]. In cases of excellent human leukocyte antigen (HLA)-matching (i.e. 2-haplotype matches), it’s been used as an effective induction agent with steroid avoidance immunosuppressive regimens [61]. Given the relatively mild side effect profile, basiliximab is well-tolerated in all patients, even those with significant cardiac or pulmonary co-morbidities.

2.2.1.3. Adverse effects

The side effect profile of basiliximab is relatively mild [57, 58]. Because of the lack of T cell activation or stimulation, cytokine release syndrome does not occur. The most serious adverse event is hypersensitivity, which is rare (<1%) [50]. There is no increased risk of infectious complications or PTLD compared to no induction therapy [64].

Figure 6.

Antibody blockade. In this figure the antibody functions by blocking the antigen from binding to the receptor.

2.2.2. Daclizumab

2.2.2.1. Mechanism of Action

Daclizumab, like basiliximab, is a CD25 antagonist; however, it is a humanized IgG1 antibody. The CD25 molecule was the first humanized monoclonal antibody to be successfully targeted in the field of transplantation [65]. The mechanism of action of daclizumab essentially duplicates that of basiliximab.

2.2.2.2. Clinical applications

Daclizumab has been shown to decrease the incidence of acute cellular rejection when administered as an induction agent [66, 67]. Given the favorable side effect profile, it is well tolerated, irrespective of co-morbid conditions. The main disadvantage of daclizumab, as compared to basiliximab, is that it is more costly and requires repeated administrations [50]. Given the low demand for the medication, it has been discontinued by the manufacturer.

2.2.2.3. Adverse effects

The generally favorable side effect profile resembles that of basiliximab. Cytokine release is not typically associated with this agent [66, 67]. Like basiliximab, the risk of infectious complications or PTLD is not significantly increased with use [64].

Advertisement

3. Desensitizing agents

3.1. Rituximab

3.1.1. Mechanism

Rituximab is a monoclonal chimeric antibody to the CD20 molecule. CD20 is a glycoproteinon the cell surface of circulating, mature B cells. Rituximab effectively depletes CD20+ cellsfrom the circulation by inducing apoptosis [68]. These cells are precursors toantibody-producing plasma cells, and their role in transplantation is only partiallycharacterized. They may play a role in acute rejection, as B cells can act as antigen presenting cells.

3.1.2. Applications

Rituximab is approved for use in various lymphomas, leukemias, PTLD, and rheumatoidarthritis [50, 69]. Peripheral veins can be used foradministration and dosing is dependent on the indication. A recent study examining therole of rituximab as an induction agent found no benefit compared to placebo [70]. However, it does play a role as a desensitizing agent in patients with preformeddonor specific antibodies (DSA), in conjunction with total plasmapheresis and/orintravenous immunoglobulin (IVIG) [71, 72].

Additionally, it has been used to aid in transplanting across blood group barriers in donorrecipient pairs and in patients with positive crossmatches following antibody elimination.

Rituximab is increasingly being used to treat episodes of vascular rejection and antibodymediatedrejections [73, 74]. Finally, rituximab is a provenand effective agent in the treatment of PTLD [75]. Administration does notreplace immunosuppression reduction or chemotherapy, but rather supplements the othermodalities.

3.1.3. Adverse effects

Rituximab is generally well-tolerated with minimal side effects. Anaphylaxis remains atheoretical concern, as is the case with most agents. Reports on infectious complicationsrelated to rituximab have been variable [76-78]. In some instances there was no difference in bacterial, viral, or fungal infections inkidney transplant recipients treated with rituximab, however, this remains controversial.

3.2. Bortezomib

3.2.1. Mechanism

Bortezomib is a proteasomal inhibitor that causes apoptosis of plasma cells. It binds the 26Ssubunit of the proteasome [79]. Proteasome inhibition ultimately leads toapoptosis during mitosis. Bortezomib selectively causes apoptosis in CD138+ plasma cells [80]. Additionally, Bortezomib may block T cell cycling and decrease the numberof circulating B cells by reducing bone marrow levels of IL-6 [81].

3.2.2. Applications

Bortezomib has not been approved for use in kidney transplantation; however, it has beenused in sensitized patients [80]. Bortezomib has been successfully used todecrease DSA levels, which may play a role in acute antibody-mediated rejection (AMR)Induction Therapy in Renal Transplant Recipients [82]. Furthermore, in vivo data has demonstrated a decrease in thepercentage of bone marrow plasma cells, antibody production, and allospecificities ofplasma cells in bone marrow aspirates of patients treated with bortezomibi in the setting ofAMR [80].

3.2.3. Adverse events

Bortezomib has been associated with various side effects. Although gastrointestinal sideeffects are the most common, peripheral neuropathy has also been reported, especially inpatients with a pre-existing history of neuropathy [79]. Moreover,myelosuppression and shingles has been reported.

3.3. Intravenous Immunoglobulin (IVIG)

3.3.1. Mechanism

Intravenous immunoglobulin, or IVIG, is pooled polyclonal antibodies from differenthuman donors. These are high-dose human IgG fractions with a wide range of specificities.These are non-T cell specific formulations and have no specific cell targets [83]. It is able to bind activated complement components or even inhibit complementactivation [84]. IVIG may also modulate the alloimmune response by bindingto the Fc receptor of antigen-presenting cells, effectively quelling the alloimmune response [85].

3.3.2. Applications

Despite the inability to deplete T cells, IVIG is an effective treatment of acute cellularrejection. Early studies showed that IVIG was as effective as OKT3 in reversing steroidresistantacute rejection episodes [86]. In the setting of antibody-mediatedrejection, IVIG has been shown to be beneficial when used in conjunction with plasma pheresis and/or rituximab [87-88]. As adesensitization agent alone, no study has demonstrated a clear benefit [88, 89]. Definitive reduction of antibody was not shown and a survivaladvantage was not evident.

3.3.3. Adverse effects

The side-effect profile of IVIG increases with dosing. High-dose IVIG is associated withmore infusion-related complications, such as headache, thrombotic incidents, hemolysis, bronchospasms, osmotic nephropathy, or even aseptic meningitis [83, 90]. Sucrose-based and high osmolality products have a higher risk ofdeveloping osmotic nephropathy as opposed to other preparation. Nevertheless, it istypically well-tolerated, especially at lower doses and most patients report only headache.

Advertisement

4. Maintenance immunosuppression regimens

4.1. Historical background

The initial transplant armamentarium consisted only of azathioprine and steroids for maintenance immunosuppression in renal transplantation until the 1980’s, when the first calcineurin inhibitor, cyclosporine became available. Over the next 20 years, azathioprine had been largely replaced by mycophenolate (MMF), an antiproliferative agent. Standard therapy in most modern immunosuppression regimens now consists of a calcineurin inhibitor, mycophenolate, with or without steroid maintenance.

Minimizing global immunosuppression in the modern era of transplantation has become an important goal. The use of induction therapy has allowed for steroid avoidance immunosuppression regimens. The goal of steroid avoidance immunosuppression is to decrease the negative cardiovascular profile associated with long-term administration of steroids. Specifically, steroid-free regimens should decrease the negative effects on blood pressure control as well serum glucose and lipid metabolism [91]. Moreover, the leading cause of death in kidney transplant patients is cardiovascular events [92].

4.2. Steroid maintenance versus withdrawal

Advocates of steroid-maintenance regimens suggest that steroids may allow for lower doses of calcineurin inhibitors, such as cyclosporine or tacrolimus. Moreover, steroids may decrease the incidence of nephrotoxicity perioperatively. However, there has been insufficient data to support either conclusion [93].

The effectiveness of steroid-withdrawal and cyclosporine-based therapy has been clearly associated with timing. Early studies of cyclosporine-based regimens demonstrated that cessation of steroids prior to the 6 month period post-transplantation increased the risk of acute rejection [94]. Furthermore, a meta-analysis of seven randomized-controlled trials of steroid avoidance and/or withdrawal demonstrated an increased risk of acute rejection with steroid avoidance or early withdrawal (most steroids were withdrawn in the first 3 months post-transplant) [95]. However, patient and graft survival were not adversely affected in the meta-analysis.

The ability to withdrawal steroids appears to be better with tacrolimus-based immunosuppression regimens. An early report by Shapiro et al. demonstrated that patients receiving tacrolimus and steroid-sparing immunosuppression had excellent early and intermediate-term patient and graft survival compared to kidney transplant recipients receiving standard steroid-maintenance immunosuppression [96]. Later, various randomized-controlled trials were undertaken to assess the initial outcomes. A meta-analysis of six randomized, controlled-trials comparing a calcineurin inhibitor-based immunosuppression regimen with MMF demonstrated a slightly increased risk of acute rejection once steroids were discontinued; however, this did not affect the incidence of graft failure [97]. Shortly thereafter, a randomized trial from Europe assigned low immunologic risk patients to receive either triple immunosuppression with tacrolimus, MMF, and steroids, a tacrolimus-based steroid withdrawal regimen, or a tacrolimus-based steroid-maintenance regimen without MMF [98]. At 6 months, the incidence of acute rejection was not different between the groups. Furthermore, the steroid withdrawal group benefited from an improved lipid profile. Kumar et al. reported on a series of 300 kidney transplant recipients receiving basiliximab induction therapy followed by steroid maintenance or withdrawal at 2 days post-transplant [99]. Maintenance therapy for all patients consisted of a calcineurin inhibitor and MMF or sirolimus. At 3 years, the incidence of biopsy-proven acute rejection, patients and graft survival, chronic allograft nephropathy, or graft function was not significantly different. Moreover, the steroid withdrawal group benefited from a lower rate of new-onset diabetes after transplantation.

Successful avoidance of steroids is contingent upon the use of calcineurin inhibitors. In 2006 Gelens and colleagues performed a single-center, randomized, trial of three parallel groups, which were: tacrolimus and sirolimus (group one), tacrolimus and MMF (group two), and sirolimus and MMF with daclizumab induction [100]. During an interim analysis when 50% of the patients were included, group one had a significantly increased rejection free survival (82%) compared to group three (34%, P=0.03) and between groups one and two (tacrolimus-based, 76%) and group three (34%, P=0.04). The study was halted prematurely. Despite the current armamentarium of antibody-depleting medications, steroid withdrawal seems feasible only with a calcineurin inhibitor-based regimen.

4.3. Induction therapy and steroid withdrawal

The possible minimization of maintenance immunosuppression has been studied using basiliximab and rATG without compromising allograft outcomes. In the Astellas Steroid Withdrawal Study, patients assigned to the steroid-withdrawal arm and treated with rATG experienced a lower cumulative incidence of biopsy-proven acute rejection at 5 years compared to patients treated with basiliximab [101]. Selection bias; however, may have marred this study, given that the investigators selected which antibody induction agent was used. Our transplant center’s experience utilizing induction therapy to enable steroid withdrawal has been very successful in a diverse population, using rATG in the majority of patients [102] and basiliximab in well-matched living donor recipients [61]. In a study by Cantarovichet al., patients administered rATG and steroid-maintenance immunosuppression had significantly lower acute rejection rates compared to patients on a steroid-free immunosuppression regimen, although the incidence of malignancy, de novo diabetes, and hyperlipidemia were higher in steroid-maintenance group [103]. Patient survival, graft survival, and infection rates were not significantly different between the two groups at 1 year.

Alemetuzumaband steroid-free regimens have been compared to both basiliximab and rATG. In the study by Hanaway et al., acute rejection rates were relatively low in low-risk patients receiving alemtuzumab compared to basiliximab, although the reduced immunologic risk profile of alemtuzumab was not evident in high risk patients treated with rATG[54]. The overall rate of adverse events with alemtuzumabwas similar to that of basiliximab or rATG over the 3 year study period (53% versus 50%, respectively; p=0.46). Moreover, the rate of cardiovascular events of all alemtuzumab treated patients compared to basiliximab or rATG was also similar (7% versus 10%, respectively; p=0.26), although the similarity was less evident in the high-risk immunologic group treated with rATG compared to alemtuzumab (12% versus 3%, respectively; p=0.06). Cai et al. analyzed the United Network for Organ Sharing registry and found that recipients of alemtuzumab in conjunction with steroid-maintenance therapy had the lowest risk of graft failure, while patients administered an interleukin-2 receptor antagonist on a steroid-free immunosuppression regimen had the highest risk of graft failure [104]. In a single-center, open-label randomized trial of 200 kidney transplant recipients, low dose dual induction therapy ofrATG and daclizumabwas compared to lose dose dual therapy ofrATG and alemtuzumab in patients maintained on steroid-free maintenance immunosuppression [105]. Patient and graft survival rates as well as acute rejection and infectious complication rates were not significantly different. In addition, no patient developed post-transplant lymphoproliferative disorder.

Advertisement

5. New and experimental agents

5.1. Siplizumab (MEDI-507)

Originally described as BTI-322, siplizumab is a monoclonal humanized antibody to CD2. It is an IgG1k molecule derived from rat [106]. CD2, or lymphocyte function-associated antigen-2 (LFA-2) is an important T cell adhesion molecule that binds to CD58, or LFA-3. This is a transmembrane signal transduction molecule that facilitates T cell receptor binding. Early studies examined the use of siplizumab as an induction agent and treatment modality for acute rejection in solid organ transplantation as well as graft-versus-host disease [106, 107]. The first human study of siplizumab demonstrated the safety and feasibility in kidney transplantation, as compared to placebo; however, current endeavors are focused on investigating its use in nonmyeloablative conditioning regimens to achieve mixed chimerism [106, 108, 109]. In addition, it is being investigated for the treatment of plaque psoriasis [110].

5.2. Alefacept

Alefacept is a dimeric fusion protein (Fig.7) constituted from LFA-3 and the human Fc portion of IgG1. Studies have demonstrated inhibition of T cell proliferation and depletion of effector memory T cells [111, 112]. Currently, alefacept is approved to treat plaque psoriasis. Preclinical studies in nonhuman primates have demonstrated a survival benefit of alefacept, when used in conjunction with costimulatory blockade, but not alone; however in human trials have never shown a benefit [113].

Figure 7.

Mimicry. In this figure, the antibody is fused with a protein structural similar to the intended antigen, which can serve as activating or inhibitory.

5.3. Costimulatory blockade

5.3.1. Abatacept

Abatacept is a recombinant cytotoxic T-lymphocyte antigen 4 (CTLA4) fused with the Fc portion of IgG1 [114, 115]. Animal models demonstrated its ability to delay or even prevent the onset of allograft rejection, which is comparable to basiliximab and some polyclonal antibody therapies [114-116]. It has been approved for treatment of rheumatoid arthritis [117, 118]. Further investigations of this medication are not currently under development.

5.3.2. Belatacept

Belatacept is the improved version of abatacept, providing selective blockade of T cell activation as a fusion protein. Two amino acids have been changed to improve dissociation rates when binding to CD80 and CD86 [119, 120]. In the phase II trial comparing belatacept to cyclosporine, acute rejection rates were similar, while allograft function was significantly improved in patients receiving belatacept [119]. In the phase III trial of kidney transplantation, patients receiving belatacept experienced improved allograft function at 12 months; however, acute rejection rates and severity of acute rejection episodes were significantly higher in the belatacept arm of the study. Additionally, the incidence of PTLD was greater in patients receiving belatacept [120]. An additional study investigating the efficacy of belatacept in kidney transplantation of extended criteria donors demonstrated similar results, with a predilection towards central nervous system (CNS) forms of PTLD [121]. The novelty of costimulation blockade is the ability to avoid calcineurin inhibitors, especially in allografts at increased risk of delayed graft function. Belatacept has recently been approved for the prophylaxis of organ rejection in adult patients receiving a kidney transplant, in combination with basiliximab induction, mycophenolatemofetil, and corticosteroids [122]. Current recommendations include using it only in patients who are EBV seropositive; however, patients should be monitored for an increased risk of infectious complications and Progressive Multifocal Leukoencephalopathy.

5.4. Eculizumab

Recently, a new medication called eculizumab has emerged as a humanized monoclonal antibody to complement component 5 (C5) to mediate complement-mediated injury [123]. Blocking complement activation, especially the last step of the complement cascade, has important implications in kidney transplantation. However, the role of eculizumab appears to be more applicable to cases of clear complement-mediated destruction, such as antibody-mediated rejection and desensitization protocols [124]. Furthermore, the logistics of administration may further hinder its’ use as a maintenance immunosuppression agent, as it must be administered biweekly or weekly intravenously at least for the first 1-2 months upon initiation of therapy. Currently, it is only approved for the treatment of patients with paroxysmal nocturnal hemoglobinuria [123].

Acknowledgement

The authors gratefully acknowledge the expert assistance of Ms. Johanna Martin in creating all figures depicted in this chapter.

References

  1. 1. CosimiA. BThe clinical value of antilymphocyte antibodies.Transplant Proc, 131March 1981462468
  2. 2. HenricssonAHusbergBBergentzS. EThe mechanism behind the effect of ALG on platelets in vivo.ClinExpImmunol, 293September 1977515522
  3. 3. RosenbergJ. CLekasNLyszKet al1975Effect of antithymocyte globulin and other immune reactants on human platelets.Surgery774April 1975), 520529
  4. 4. NiblackGJohnsonKWilliamsTet alAntibody formation following administration of antilymphocyte serum.Transplant Proc, 191February 198718961897
  5. 5. Prin MathieuC.; Renoult, E.; Kennel De March, A., et al. Serum anti-rabiit and anti-horse IgG, IgA, and IgM in kidney transplant recipients. Nephrol Dial Transplant, 1210October 199721332139
  6. 6. TatumA. HBollingerR. RSanfilippoFRapid serological diagnosis of serum sickness from antilymphocyte globulin therapy using enzyme immunoassay. Transplantation, 386December 1984582586
  7. 7. KohlerGMilsteinCContinuous cultures of fused cells secreting antibody of predefined specificity.Nature2565517August 1975495497
  8. 8. CosimiA. BBurtonR. CColvinR. Bet alTreatment of acute renal allograft rejection with OKT3 monoclonal antibody.Transplantation326December 1981535539
  9. 9. Ortho Multicenter Transplant Study GroupA randomized clinical trial of OKT3 monoclonal antibody for acute rejection of cadaveric renal transplants. N Engl J Med, 3136August 1985337342
  10. 10. PonticelliCRivoltaETarantinoAet alTreatment of severe rejection of kidney transplant with OKT3.PAN. Transplant Proc, 11February 198719081909
  11. 11. JaffersG. JFullerT. CCosimiA. Bet alMonoclonal antibody therapy: anti-idiotype and non-anti-idiotype antibodies to OKT3 arising despite intense immunosuppression. Transplantation, 415May 1986572578
  12. 12. VincentiFLantzMBirnbaumJet alA phase I trial of humanized anti-interleukin 2 receptor antibody in renal transplantation.1997631January 19973338
  13. 13. BoulianneG. LHozumiNShulmanM. Jet alProduction of functional chimaeric mouse/human antibody.Nature3125995December 1984643646
  14. 14. JonesP. TDearP. HFoote, et alReplacing the complementarity-determining regions in human antibody with those from a mouse.Nature3216069May 1986522525
  15. 15. MorrisonS. LJohnsonM. JHerzenbergL. Aet alChimeric human antibody molecules: mouse antigen-binding domains with human constant region domains.ProcNatlAcadSci U S A, 8121November 198468516855
  16. 16. CapraJ. DEdmundsonA. BThe antibody combining site.Sci Am, 2361January 19775059
  17. 17. TiteJ. PSloanAJanewayC. JThe role of L3T4 in T cell activation: L3T4 may be both an Ia-binding protein and a receptor that transduces a negative signal.J Mol Cell Immunol, 241986179190
  18. 18. WongJ. TEylathA. AGhobrialIet alThe mechanism of anti-CD3 monoclonal antibodies: mediation of cytolysis by inter-T cell bridging.Transplantation504October 1990683689
  19. 19. KerrP. GAtkinsR. CThe effects of OKT3 therapy on infiltrating lymphocytes in rejecting renal allografts.Transplantation481July 19893336
  20. 20. FerrantJ. LBenjaminC. DCutlerA. Het alThe contribution of the Fc effector mechanisms in the efficacy of anti-CD154 immunotherapy depends on the nature of the immune challenge. IntImmunol, 1611Novemeber 200415831594
  21. 21. PearlJ. PParrisJHaleD. Aet alImmunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletionAm J Transplant, 53March 2005465474
  22. 22. SzczechL. ABerlinJ. AAradhyeSet alEffect of anti-lymphocyte induction therapy on renal allograft survival: a meta-analysisJ Am SocNephrol, 811November 199717711777
  23. 23. HumarARamcharanTDennyRet alAre wound complications after a kidney transplant more common with modern immunosuppression?Transplantation7212December 200119201923
  24. 24. SinghAStableinDTejaniARisk factors for vascular thrombosis in pediatric renal transplantation: a special report of the North American Pediatric Renal Transplant Cooperative Study.Transplantation639May 199712631267
  25. 25. BustamiR. TOjoA. OWolfeR. Aet alImmunosuppression and the risk of post-transplant malignancy among cadaveric first kidney transplant recipientsAm J Transplant, 41January 20048793
  26. 26. JamilBNichollsKBeckerG. Jet alImpact of acute rejection therapy on infections and malignancies in renal transplant recipients.Transplantation6810November 199915971603
  27. 27. Meier-kriescheH. ULiSGruessnerR. Wet alImmunosuppression: evolution in practice and trends, 1994-2004Am J Transplant, 65200611111131
  28. 28. HardingerK. LRabbit antithymocyte globulin induction therapy in adult renal transplantation.Pharmacotherpay, 2612December 200617711783
  29. 29. Bonnefoy-berardNVincentCRevillardJAntibodies against the functional leukocyte surface molecules in polyclonal antilymphocyte and antithymocyte globulins. Transplantation513March 1991669673
  30. 30. BourdageJ. SHamlinD. MComparative polyclonal antithymocyte globulin and antilymphocyte/antilymphoblast globulin anti-CD antigen analysis by flow cytometryTransplantation598April 199511941200
  31. 31. BunnDLeaC. KBevanD. Jet alThe pharmacokinetics of anti-thymocyte globulin (ATG) following intravenous infusion in man.Clin Nephrol, 451January 19962932
  32. 32. GuttmannR. DCaudrelierPAlbericiGet alPharmacokinetics, foreign protein immune response, cytokine release, and lymphocyte subsets in patients receiving thymoglobuline and immunosuppression.Transplant Proc, 297A24S246
  33. 33. BrennanD. CFlavinKLowellJ. Aet alA randomized,double-blinded comparison of thymoglobulin versus Atgam for induction immunosuppressive therapy in adult renal transplant recipients. Transplantation, 677April 199910111018
  34. 34. HardingerK. LSchnitzlerM. AMillerBet alFive-year follow up of thymoglobulin versus ATGAM induction in adult renal transplantation.Transplantation2004136 EOF41 EOF
  35. 35. IMTIX-SangStatThymoglobuline (rabbit antithymocyte globulin) prescribing information. Lyon, France; 2003
  36. 36. MerionRWhiteD. JThiruSet alCyclosporine: five years experience in cadaveric renal transplantation.N Engl J Med, 3103January 1984148154
  37. 37. ShoskesD. AHalloranP. FDelayed graft function in renal transplantation: etiology, management and long-term significance.J Urol, 1556June 199618311840
  38. 38. Beiras-fernandezAChappellDHammerCet alInfluence of polyclonal anti-thymocyte globulins upon ischemia reperfusion injury in a non-human primate modelTransplImmunol, 154April 2006273279
  39. 39. CeckaJ. MGjertsonDTerasakiPDo prophylactic antilymphocyte globulins (ALG and OKT3) improve renal transplant in recipient and donor high-risk groups? Transplant Proc, 251February 1993548549
  40. 40. ShieldC. FEdwardsE. BDaviesD. Bet alAntilymphocyte induction therapy in cadaver renal transplantationTransplantation639May 199712571263
  41. 41. SchafferDLangoneANylanderW. Aet alA pilot protocol of a calcineurin-inhibitor free regimen of kidney transplant recipients of marginal donor kidneys or with delayed graft function. Clin Transplant, 17920033134
  42. 42. DebetsJ. M. HLeunissenK. M. LVan HooffH. Jet alEvidene of involvement of tumor necrosis factor in adverse reactions during treatment of kidney allograft rejection with antithymocyte globulin. Transplantation, 473March 1989487492
  43. 43. BuchlerMHurault de Ligny, B.; Madec, C., et al. Induction therapy by anti-thymocyte globulin (rabbit) in renal transplantation: a 1-yr follow-up of safety and efficacyClin Transplant, 176December 2003539545
  44. 44. MouradGGarrigueVSquiffletJ. Pet alInduction versus noninduction in renal transplant recipients with tacrolimus-based immunosuppression.Transplantation726September 200110501055
  45. 45. AbottK. CHypoliteI. OViolaRet alHospitalizations for cytomegalovirus disease after renal transplantation in the United States. Ann Epidemiol, 126August 2002402409
  46. 46. GourishankarSMcdermidJ. CJhangriG. Set alHerpes zoster infection following solid organ transplantation: incidence, risk factors and outomes in the current immunosuppressive era.Am J Transplant, 41January 2004108115
  47. 47. HaleGWaldmannHFriendPet alPilot study of CAMPATH-1, a rat monoclonal antibody that fixes human complement, as an immunosuppressant in organ transplantation.Transplantation423September 1986308311
  48. 48. HaleGTheC. Dantigen and development of the CAMPATH antibodies. Cytotherapy332001137143
  49. 49. KirkA. DHaleD. AMannonR. Bet alResults from a human renal allograft tolerance trial evaluating the humanized CD52-specific monoclonal antibody alemtuzumab (CAMPATH-1H).Transplantation761July 2003120129
  50. 50. GabardiSMartinS. TRobertsK. Let alInduction immunosuppressive therapies in renal transplantationAm J Health Syst Pharm, 683February 2011211218
  51. 51. ClatworthyM. RFriendP. JCalneR. Yet alAlemtuzumab (CAMPATH-1H) for the treatment of acute rejection in kidney transplant recipients: long-term follow-upTransplantation877April 200910921095
  52. 52. CsapoZBenavides-viverosCPodderHet alCampath-1H as rescue therapy for the treatment of acute rejection in kidney transplant patients.Transplant Proc, 375June 200520322036
  53. 53. JirasirithamSKhunprakantRTechawathanawannaNet alTreatment of simultaneous acute antibody-mediated rejection and acute cellular rejection with alemtuzumab in kidney transplantation: a case report. Transplant Proc, 423April 2010987989
  54. 54. HanawayM. JWoodleE. SMulgaonkarSet alAlemtuzumab induction in renal transplantationN Engl J Med, 36420May 201119091919
  55. 55. ColesA. JWingMSmithSet alPulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis.Lancet3549191Novemeber 199916911695
  56. 56. KirkA. DHaleD. ASwansonS. Jet alAutoimmune thyroid disease after renal transplantation using depletional induction with alemtuzumab.Am J Transplant, 65May 200610841085
  57. 57. KahanB. DRajagopalanP. RHallMReduction of the occurrence of acute cellular rejection among renal allograft recipients treated with basiliximab, a chimeric anti-interleukin-2-receptor monoclonal antibody. Transplantation672276284
  58. 58. NashanBMooreRAmlotPet alRandomized trial of basiliximab versus placebo for control of acute cellular rejection in renal allograft recipients. CHIB201 International Study Group. Lancet, 3509086October 199711931198
  59. 59. LebranchuYBridouxFBuchlerMet alImmunoprophylaxis with basiliximab compared with antithymocyte globulin in renal transplant patients receiving MMF-containing triple therapyAm J Transplant, 214856
  60. 60. MouradGRostaingLLegendreCet alSequential protocols using basiliximab versus antithymocyte globulins in renal-transplant patients receiving mycophenolatemofetil and steroids. Transplantation, 784August 2004584590
  61. 61. AfanehCHalpernJChengEet alSteroid avoidance in two-haplotype-matched living donor renal transplants with basiliximab induction therapyTransplant Proc, 4210December 201045264529
  62. 62. ParrottN. RHammadA. QWatsonC. Jet alMulticenter, randomized study of the effectiveness of basiliximab in avoiding addition of steroids to cyclosporine a monotherapy in renal transplant recipients.Transplantation793February 2005344348
  63. 63. VincentiFRamosEBrattstromCet alMulticenter trial exploring calcineurin inhibitors avoidance in renal transplantation.Transplantation719May 201112821287
  64. 64. CherikhW. SKauffmanH. MMcbrideM. Aet alAssociation of the type of induction immunosuppression with posttransplant lymphoproliferative disorder, graft survival, and patient survival after primary kidney transplantation.Transplantation769Novemeber 200312891293
  65. 65. KirkmanR. LShapiroM. ECarpenterC. Bet alA randomized prospective trial of anti-Tac monoclonal antibody in human renal transplantation.Transplantation, 511January 1991107113
  66. 66. HershbergerR. EStarlingR. CEisenH. Jet alDaclizumab to prevent rejection after cardiac transplantation.N Engl J Med, 35226June 200527052713
  67. 67. NashanBLightSHardieI. Ret alReduction of acute renal allograft rejection by daclizumabTransplantation, 671January 1999110115
  68. 68. DeansJ. PLiHPolyakM. Jet alCD20-mediated apoptosis: signaling through lipid rafts.Immunology1072October 2002176182
  69. 69. Grillo-lopezA. JWhiteC. AVarnsCet alOverview of the clinical development of rituximab: first monoclonal antibody approved for the treatment of lymphoma.SeminOncol, 265October 19996673
  70. 70. TydenGGenbergHTollemarJet alA randomized, double-blind, placebo controlled, study of single-dose rituximab as induction in renal transplantation. Transplantation879May 200913251329
  71. 71. FuchinoueSIshiiYSawadaTet alThe 5-year outcome of ABO-incompatible kidney transplantation with rituximab inductionTransplantation9118April 2011853857
  72. 72. SonnendayC. JWarrenD. SCooperMet alPlasmapheresis, CMV, hyperimmune globulin, and anti-CD20 allow ABO-incompatible renal transplantation without splenectomy. Am J Transplant, 48August 200413151322
  73. 73. BeckerY. TSamaniego-picotaMSollingerH. WThe emerging role of rituximab in organ transplantationTransplInt, 198August 2006621628
  74. 74. FehrTRüsiBFischerAet alRituximab and intravenous immunoglobulin treatment of chronic antibody-mediated kidney allograft rejectionTransplantation8712June 200918371834
  75. 75. SvobodaJKotloffRTsaiD. EManagement of patients with post-transplant lymphoproliferative disorder: the role of rituximabTransplInt, 194April 2006259269
  76. 76. GrimS. APhamTThielkeJet alInfectious complications associated with the use of rituximab for ABO-incompatible and positive cross-match renal transplant recipientsClin Transplant, 215September 2007628632
  77. 77. KamarNMiliotoOPuissant-lubranoBet alIncidence and predictive factors for infectious disease after rituximab therapy in kidney-transplant patientsAm J Transplant, 101July 20098998
  78. 78. NishidaHIshidaHTanakaTet alCytomegalovirus infection following renal transplantation in patients administered low-dose rituximab induction therapy, TransplInt, 2210July 2009961969
  79. 79. BonviniPZorziEBassoGet alBortezomib-mediated 26S proteasome inhibition causes cell-cycle arrest and induces apoptosis in CD-30+ anaplastic large cell lymphoma.Leukemia214April 2007838842
  80. 80. PerryD. KBurnsJ. MPollingerH. Set alProteasome inhibition causes apoptosis of normal human plasma cells preventing alloantibody productionAm J Transplant, 91January 2009201209
  81. 81. San MiguelJ.F.; Schlag, R.; Khuageva, N.K., et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myelomaN Engl J Med, 3599August 2008906917
  82. 82. TrivediH. LTerasakiP. IFerozAet alAbrogation of anti-HLA antibodies via proteasome inhibitionTransplantation8710May 200915551561
  83. 83. JordanS. CToyodaMKahwajiJet alClinical Aspects of Intravenous Immunoglobulin Use in Solid Organ Transplant Recipients.Am J Transplant, 112January 2011196202
  84. 84. JordanS. CToyodaMVoA. AIntravenous immunoglobulin a natural regulator of immunity and inflammationTransplantation881July 200916
  85. 85. KazatchkineM. DKaveriS. VImmunomodulation of autoimmune and inflammatory disease with intravenous immune globulin.N Engl J Med, 34510September 2001747755
  86. 86. CasadeiD. Hdel C Rial, M.; Opelz, G., et al. A randomized and prospective study comparing treatment with high-dose intravenous immunoglobulin with monoclonal antibodies for rescue of kidney grafts with steroid-resistant rejection.Transplantation711January 20015358
  87. 87. LefaucheurCNochyDAndradeJet alComparison of combination plasmapheresis/IVIG/Anti-CD20 versus high dose IVIG in the treatment of antibody-mediated rejectionAm J Transplant, 95May 200910991107
  88. 88. ShehataNPaldaVMeyerRet alThe use of immunoglobulin therapy for patients undergoing solid organ transplantation: an evidence-based practice guideline. Transf Med Rev, 241January 2010S727
  89. 89. PisaniB. AMullenG. MMalinowskaKet alPlasma- pheresis with intravenous immunoglobulin G is effective in patients with elevated panel reactive antibody prior to cardiac transplantation. J Heart Lung Transplant, 187July 1999701706
  90. 90. KahwajiJBarkerEPepkowitzSet alAcute hemolysis after high dose IVIG therapy in highly HLA sensitized patients. Clin J Am SocNephrol, 412October 200919931997
  91. 91. KnightS. RMorrisP. JSteroid avoidance or withdrawal after renal transplantation increases the risk of acute rejection but decreases cardiovascular risk: a meta analysisTransplantation891January 2010785794
  92. 92. OjoA. OHansonJ. AWolfeR. Aet alLong-term survival in renal transplant recipients with draft function. Kidney Int, 571January 2000307313
  93. 93. GriffinP. JDa Costa, C.A.; Salaman, J.R. A controlled trial of steroids in cyclosporine-treated renal transplant recipients. Transplantation434April 1987505508
  94. 94. HricikD. EWhalenC. CLautmanJet alWithdrawal of steroids after renal transplantation- clinical predictors of outcome.Transplantation531January 19924145
  95. 95. HricikD. EOTooleM. ASchulakJ.A., et alSteroid-free immunosuppression in cyclosporine-treated renal transplant recipients: a meta-analysis.J Am SocNephrol, 4199313001305
  96. 96. ShapiroRJordanM. LScantleburyV. Pet alOutcome after steroid withdrawal in renal transplant patients receiving tacrolimus-based immunosuppression.Transplant Proc, 304June 199813751377
  97. 97. PascualJQueradaCZamoraJet alSteroid withdrawal in renal transplant patients on triple therapy with a calcineurin inhibitor and mycophenolatemofetil: a meta-analysis of randomized, controlled trials. Transplantation, 7810November 200415481556
  98. 98. VanrenterghemYVan HooffJ. PSquiffletJ. Pet alMinimization of immunosuppressive therapy after renal transplantation: results of a randomized controlled trialAm J Transplant, 51January 20058795
  99. 99. KumarM. SHeifetsMMoritzM. Jet alSafety and efficacy of steroid withdrawal two days after kidney transplantation: analysis of results at three years.Transplantation816March 2006832839
  100. 100. GelensM. AChristiaansM. HVan HeurnE. Let alHigh rejection rate during calcineurin inhibitor-free and early steroid withdrawal immunosuppression in renal transplantation.Transplantation829November 200612211223
  101. 101. WoodleE. SFirstM. RPirschJet alA prospective, randomized, double-blind placebo-controlled multicenter trial comparing early (7 day) corticosteroid cessation versus long-term, low-dose corticosteroid therapy.Ann Surg, 2484October 2008564577
  102. 102. AullM. JDadhaniaDAfanehCet alEarly corticosteroid withdrawal in high risk recipients of renal allografts: a single center report of ethnically diverse recipients and recipients of marginal deceased donor kidneys. Transplantation, 2012Sep 20. DOI:TP.0b013e318265c461 [Epub ahead of print]
  103. 103. CantarovichDRostaingLKamarNet alCorticosteroid avoidance in adult kidney transplant recipients under rabbit anti-T-lymphocyte globulin, mycophenolatemofetil and delayed cyclosporine microemulsion introduction.TransplInt, 233March 2010313324
  104. 104. CaiJTerasakiP. IInduction immunosuppression improves long-term graft and patient outcome in organ transplantation: an analysis of united network for organ sharing registry dataTransplantation9012December 201015111515
  105. 105. CiancioGGaynorJ. JSageshimaJet alRandomized trial of dual antibody induction therapy with steroid avoidance in renal transplantationTransplantation9212December 201113481357
  106. 106. PruettT. LMcgoryR. WWrightF. Het alSafety profile, pharmacokinetics, and pharmacodynamics of siplizumab, a humanized anti-CD2 monoclonal antibody, in renal allograft recipientsTransplant Proc, 419November 200936553661
  107. 107. SquiffletJ. PBesseTMalaiseJet alBTI-322 for induction therapy after renal transplantation: a randomized study.Transplant Proc, 291-2February 1997317319
  108. 108. KawaiTCosimiA. BSpitzerT. Ret alHLA-mismatched renal transplantation without maintenance immunosuppressionN Engl J Med, 3584January 2008353361
  109. 109. SpitzerT. RMcafeeS. LDeyB. Ret alNonmyeloablativehaploidentical stem-cell transplantation using anti-CD2 monoclonal antibody (MEDI-507)-based conditioning for refractory hematologic malignancies. Transplantation, 7510May 200317481751
  110. 110. LangleyR. GPappKBissonnetteRet alSafety profile of intravenous and subcutaneous siplizumab, an anti-CD2 monoclonal antibody, for the treatment of plaque psoriasis: results of two randomized, double-blind, placebo-controlled studiesInt J Dermatol, 497July 2010818828
  111. 111. EllisC. NKruegerG. GAlefacept Clinical Study Group. Treatment of chronic plaque psoriasis by selective targeting of memory effector T lymphocytes. N Engl J Med, 3454July 2001248255
  112. 112. GordonK. BVaishnawA. KOGormanJ., et alTreatment of psoriasis with alefacept: correlation of clinical improvement with reductions of memory T-cell counts.Arch Dermatol, 13912December 200315631570
  113. 113. WeaverT. ACharafeddineA. HAgarwalAet alAlefacept promotes co-stimulation blockade based allograft survival in nonhuman primatesNat Med, 157July 2009746749
  114. 114. LenschowD. JZengYThistlethwaiteJ. Ret alLong-term survival of xenogeneic pancreatic islet grafts induced with CTLA4Ig. Science, 2575071August 1992789792
  115. 115. TurkaL. ALinsleyP. SLinHet alT-cell activation by the CD28 ligand B7 is required for cardiac allograft rejection in vivo.ProcNatlAcadSci U S A, 8922November 19921110211105
  116. 116. KirkA. DHarlanD. MArmstrongN. Net alCTLA4-Ig and anti-CD40 ligand prevent renal allograft rejection in primates.ProcNatlAcadSci U S A, 9416August 199787898794
  117. 117. GenoveseM. CBeckerJ. CSchiffMet alAbatacept for rheumatoid arthritis refractory to tumor necrosis alpha inhibition. N EnglJ Med, 3531111141123
  118. 118. NogidAPhamD. QRole of abatacept in the management of rheumatoid arthritis.ClinTher, 2811November 200617641778
  119. 119. VincentiFLarsenCDurrbachAet alCostimulation blockade with belatacept in renal transplantation.N Engl J Med, 3538August 2005770781
  120. 120. VincentiFCharpentierBVanrenterghemYet alA phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study)Am J Transplant, 103March 2010535546
  121. 121. DurrbachAPestanaJ. MPearsonTet alA phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors (BENEFIT-EXT study)Am J Transplant, 103March 2010547557
  122. 122. Bristol-Myers Squibb CompanyNulojix® (belatacept) prescribing information. Princeton, New Jersey, USA; (2011
  123. 123. FehrTGaspertAAntibody-mediated kidney allograft rejection: therapeutic options and their experimental rationale.TransplInt, 256June 2012623632
  124. 124. LarreaC. FCofanFOppenheimerFet alEfficacy of eculizumab in the treatment of recurrent atypical hemolytic-uremic syndrome after renal transplantationTransplantation897April 2010903904

Written By

Cheguevara Afaneh, Meredith J. Aull and Sandip Kapur

Submitted: 20 June 2012 Published: 19 December 2012