<<

REvIEWS

Advances in for renal transplantation Antoine Durrbach, Helene Francois, Severine Beaudreuil, Antoine Jacquet and Bernard Charpentier

Abstract | The development of immunosuppressants with minimal adverse and nephrotoxic effects is important to improve outcomes, such as acute and chronic -mediated rejection, after organ transplantation. In addition, the application of expanded criteria for donors and transplantation in immunized patients necessitates the development of new therapies. Drug development over the past 10 years has generally been disappointing, but several new promising compounds have been or are being developed to prevent acute and chronic transplant rejection. In this Review, we report on several compounds that have been developed to remove allogenic T cells and/or to inhibit T-cell activation. We also discuss compounds that interfere with antibody-mediated rejection.

Durrbach, A. et al. Nat. Rev. Nephrol. 6, 160–167 (2010); published online 2 February 2010; doi:10.1038/nrneph.2009.233

Introduction Renal transplantation has specific features that make or as a result of previous transplantation, has increased it different from transplantation procedures for other over the past decade. In addition, ABO­incompatible organs. For example, outcomes can be affected by grafts are becoming more frequently used. Together, these common states, such as donor and/or recipient age, high factors have led to a rise in the number of ‘immuno logically blood pressure, diabetes mellitus, metabolic disturbances at­risk’ kidney transplantations. Few immunosuppressants (such as high LDL cholesterol) and abnormalities in fluid targeted to B cells have, however, been available to control and electrolyte balance. The kidney is also very sensitive the antibody­mediated response. to injury by ischemia and reperfusion and by the use of New immunosuppressants are needed. These should nephrotoxic drugs. ideally be free from any nephrotoxic effects, be strong In the 1970s, immunosuppressants were not nephro­ enough to act on T­cell and B­cell pathways, interfere toxic (for example, , steroids, anti with the immunological components of chronic graft globulins). The 1980s saw the introduction of a new nephropathy, and prevent or treat antibody­mediated generation of cyclic molecules that had anticalcineurin rejection. Many molecules have been newly selected for activity (for example, and anti­ drug development on the basis of their ability to affect biotics with strong inhibitory properties against T­cell activated T­cell pathways, but over the past 10 years, most activation, such as tacro limus). These drugs, which were of these have failed in clinical development. Agents that included in additive, multidrug, immuno suppressive can address the changing challenges of organ transplanta­ regimens, reduced acute allograft rejection rates and tion will, however, almost certainly become available. In markedly increased overall graft survival. Use of these this Review, we discuss the different drugs that have been drugs, however, notably increases the risk of chronic dys­ developed to remove allogenic T cells and other new com­ function, with immunological (chronic allograft rejec­ pounds that inhibit T­cell activation. We also report on tion) and nonimmuno logical (nephrotoxic effects from results obtained for agents that interfere with antibody­ inhibitors) compo nents, which can lead to mediated rejection. The different agents are summarized irreversible graft failure and a return to dialysis. in Table 1 and the pathways are described in Figure 1. Nephrology and Transplantation Other phenomena also impact on the success rates of Department, Hopital renal transplantation. For example, organs from deceased Strategies to target T cells Bicetre, UMR U542 donors with extended criteria (that is, age >65 years, renal The use of agents to deplete T cells is only proposed for INSERM/University Paris-Sud XI, 78 Rue du grafts with reduced glomerular filtration rates, donors with the induction phase of transplantation. Although some Général Leclerc, Paris, risk factors for cardiovascular disease) are increasingly studies have highlighted benefits with such treatments, for France (A. Durrbach, H. Francois, transplanted, which has resulted in an increased risk of example prolonging survival of transplanted organs, their S. Beaudreuil, recipients developing chronic renal dysfunction. Moreover, long­term use is not recommended. Immunodepletion of A. Jacquet, B. Charpentier) the number of recipients who are immunized before trans­ alloreactive T cells during sequential immunosuppression plantation, such as through pregnancy, blood transfusions may be beneficial for graft outcome by avoiding initial Correspondence to: acute rejection. Noncontrolled use of T­cell­depleting B. Charpentier bernard.charpentier@ Competing interests agents is associated with several complica tions, such bct.aphp.fr The authors declare no competing interests. as and tumor development. For instance,

160 | MARCH 2010 | voluMe 6 www.nature.com/nrneph © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 160 8/2/10 15:29:12 revIewS

increasing numbers of tumors are reported in registries.1 Key points The development of tumors is a multifactorial event: ■ Allogenic organ transplantation is limited by drug-associated toxicity and the long­term exposure to immunosuppressants, the dura­ occurrence of antibody-mediated or chronic rejection tion of the trans plantation, patient’s age, the number of ■ Improved understanding of the molecular mechanisms of rejection has led to transplantations and genetic predisposition or epigenetic the development of agents that regulate T-cell function, complement activation factors can all influence the occurrence of tumors. The and/or the survival of plasma and B cells in immunized or naive patients use of depleting agents can, however, lower this risk and ■ These new agents may control acute, antibody-mediated rejection and chronic improve management. rejection to increase long-term renal transplantation outcomes ■ The safety of these new agents remains to be evaluated to ensure that they do Clonal deletion with alefacept not increase the risk of infections or tumor formation in transplanted patients Depletion of allogenic T cells by inducing of activated T cells (clonal deletion) has emerged as a useful immunomodulatory strategy. Alefacept is a Table 1 | Immunological effects of promising new immunosuppressants that binds to the immunological synapse­associated Inhibitory Target Molecule type Method of Main function protein CD2. CD2 is expressed on T cells and normally molecule administration interacts with lymphocyte­function­associated antigen­3 Alefacept CD2 Fusion protein Intravenous Deletion of activated (LFA­3) on antigen­presenting cells to participate in the T cells formation and stabilization of the immune synapse. CD80 Fusion protein Intravenous Inhibition of the second LFA­3–CD2 signaling has important roles in the activa­ and signal of activation tion of memory–effector T cells, increased proliferative CD86 responses, and cytotoxic T­cell effector functions.2,3 By Sotrastaurin PKC Small molecule Oral Inhibition of PKC contrast, the use of paired or monoclonal anti­ CP-690,550 JAK-3 Small molecule Oral JAK-3 inhibitor bodies (mAbs) against CD2 prevents interaction with LFA C5 Antibody Intravenous Inhibition of complement and induces caspase­dependent and caspase­independent activation 4–9 apoptosis of alloreactive T cells. Use of monoclonal Bortezomib NFκB Small molecule Intravenous Depletion of plasmocytes 9 antibodies in mice prevents and treats acute rejection. and inhibition of NFκB

similarly, a human fusion protein, comprising the first Abbreviations: JAK, Janus kinase 3; NFκB, nuclear factor κB; PKC, protein kinase C. extracellular domain of LFA­3 fused to two constant domains of human IgG1, has been designed to prevent interaction between LFA­3 and CD2, and thus to interfere LFA­3–Ig. The modest decrease in lymphocyte count is with the activation of T cells.10–12 In addition, the IgG1 attributable to the selective depletion of memory T­cell domain can activate the complement cascade and inter­ levels without altering the number of naive T cells. acts with CD16 (Fc γ receptor type III) on natural killer Alefacept has also been used to treat chronic exten­ cells to induce granzyme­mediated apoptosis of memory sive graft­versus­host disease (cGvHD) in humans.16,17 T cells and/or, preferentially, alloreactive T cells.11 Among 12 patients with steroid­resistant cGvHD, Alefacept was initially developed to treat , a eight (75%) responded to alefacept therapy. six of the lymphocyte­dependent disease. In animal models this 12 patients were still alive more than 30 months after drug prevents psoriasis­like lesions. A randomized, therapy; the others died from progression of cGvHD. double­blind, placebo­controlled, multicenter, phase III Alefacept has also been shown to control cGvHD that study demonstrated that, compared with placebo, occurs after liver transplantation.17 These findings indi­ intravenous alefacept improved cutaneous lesions.13 cate that alefacept could be useful in the control of allo­ Intramuscular administration of this agent was also genic in transplantation and could be used associ ated with a dose­dependent reduction in memory as an induction therapy, or to treat cellular rejection. T cells (CD4+CD45RO+ and CD8+CD45RO+), whereas levels of naive CD4+CD45RA+ and CD8+CD45RA+CD19+ regulation of T‑cell activation with belatacept B cells, and CD16+CD56+ natural killer cells were unaf­ second­signal activation has an important role in the fected.14 After 12 weeks, total CD4+ lymphocyte counts activa tion of T cells. The second­signal family is com­ returned to within the normal range. Currently available posed of two subfamilies—CD28 and tumor necrosis co­ stimulation, blockade­based immunosuppression regi­ factor receptors.18,19 These subfamilies induce positive and mens do not prevent allograft rejection driven by memory negative signals in activated T cells. The CD28 pathway T cells, therefore, alefacept has been evaluated in associa­ has been extensively studied. Blockade of this pathway has tion with CTLA4–Ig recombinant molecules and siro limus been evaluated in human transplantation.20 to prevent allograft rejection in a nonhuman primate CD28 is a disulfide­bound molecule that belongs to model of heart transplantation.15 This regimen was associ­ the immunoglobulin superfamily and is constitutively ated with prolonged graft survival in 50% of cases when expressed on T cells. CD28 interacts with B7.1 (CD80) compared with other combinations, a greater reduction in and B7.2 (CD86) molecules expressed on the surface of CD8+ cells, and an increase in the CD4+:CD8+ ratio when antigen­presenting cells and induces full T­cell activa­ compared with other regimens . In macaques treated with tion.19 The molecule shares 20% sequence identity with the LFA­3–Ig, these results were reversed after this therapy inhibitory receptor CTLA4 (CD152), which is upregulated was withdrawn, compared with that in those not receiving during T­cell activation. CD28 and CTLA4 both bind to

NATURe RevIeWs | nepHrology vOLUMe 6 | MARCH 2010 | 161 © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 161 8/2/10 15:29:12 revIewS

■ Alefacept: T-cell apoptosis longer periods of kidney graft survival (up to 6 months) Belatacept: T-cell activation ■ with humanized CD80 and CD86 antibodies than did ■ Sotrastaurin: T-cell activation untreated monkeys.27,31 All the monkeys that received LFA-3 ■ Eculizumab: Complement activation ■ Bortezomib: B-cell survival treatment developed donor­specific antibodies and had renal infiltrates, which indicates that CD28 blockade CD2 has to be used in combination. The agent belatacept was Alefa cept improved by the introduction of two point mutations to APC increase its avidity for CD80 and CD86. This drug has been evaluated in a clinical trial of human renal transplant Sotrastaurin 20 IL-2 recipients. This phase II, multicenter study included CD80 IL-2 or CD86 PKC 218 adults who were randomly assigned to either an intensive regimen of belatacept (without ciclosporin), a CTLA4 less­intensive regimen of belatacept without ciclosporin, JAK-3 Belatacept or ciclosporin alone (control). All patients received a Fc (IgG1) ­based induction therapy, mycophenolate mofetil and steroids. The incidence of acute rejection at

CP-690,550 6 months was similar in the three groups: 7% for intensive belatacept, 6% for less­intensive belatacept, and 8% for ciclosporin alone. subclinical rejection at 12 months (as Complement assessed by routine biopsy) was more common with less­ intensive belatacept therapy (20%) than with intensive Eculizumab Bortezomib IL-6 production Anti-HLA antibodies belatacept (9%) or ciclosporin (11%). Glomerular filtra­ Figure 1 | Intracellular signaling and inhibition by novel compounds. The JAK3 tion rate at 12 months was markedly elevated in patients inhibitor (CP-690,550), PKC inhibitor (sotrastaurin) and CD28 inhibitor receiving belatacept compared with those treated with (belatacept) inhibit T-cell activation whereas LFA-3–Ig depletes allogenic T cells. ciclosporin (66.3 and 62.1 ml/min/1.73 m2 in patients Bortezomib impairs plasma cell production of anti-HLA antibodies and eculizumab receiving intensive and nonintensive belatacept, respec­ inhibits complement activation mediated by anti-HLA antibodies. Abbreviations: tively, versus 53.5 ml/min/1.73 m2 in ciclosporin­treated APC, antigen-presenting cell; IL, interleukin; JAK-3, Janus kinase 3; LFA-3, patients). By month 12, the incidence of chronic allograft lymphocyte-function-associated antigen-3; PKC, protein kinase C. nephropathy was lower in both belatacept groups (20% for the intensive group and 29% for the less­intensive CD80+ and CD86+ molecules, although the latter does so group) than in the control group (44%). The incidence of with higher kinetic energy than the former and induces a infections or tumors did not differ between groups. positive signal.21 By contrast, CTLA4 exerts a negative effect Two additional phase III studies have been developed on T cells, which leads to a decrease in the signal trans­ with standard donors (BeNeFIT study) or with donors with mitted by T­cell receptors. Belatacept was created by fusing extended criteria (BeNeFIT extended study).32,33 These the ecto domain of CTLA4 with two constant domains of trials showed that, compared with ciclosporin, belata­ human IgG1 (CTLA4–Ig), enabling dissociation of CD28 cept was associated with improved renal function and a from CD80+CD86+ molecules, thus impairing T­cell decreased risk of cardiovascular risk factors in recipients. activa tion. This effect is also associ ated with modifica­ A trend towards more acute rejections, however, was seen tion of the activa tion status of the target cells. The binding in patients receiving belatacept than in those receiving of CTLA4–Ig induces tryptophan cata bolism through ciclosporin. In addition, more cases of post­transplant the stimulation of indoleamine 2,3­dioxygenase, which were reported in patients treated with belata­ degrades tryptophan and, therefore, impairs T­cell prolifera­ cept than in the controls, although the overall number tion and promotes the extension of CD4+CD25+FOXP3+ was low and most cases occurred in patients who had 22,23 T­regulatory (TReG) cells. In addition, CTLA4–Ig well­known risk factors for lymphoma, such as a negative binding favors the synthesis of the regulatory human status for epstein–Barr virus before transplantation and leuko cyte antigen (HLA) G molecule by antigen­ the use of T­cell­depleting agents. presenting cells. Furthermore, in vivo allogenic regulation In a trial of belatacept in renal transplant recipients, + + + + + by TReG cells (CD4 CD25 FOXP3 ) that express CTLA4 patients did not exhibit expansion of TReG (CD4 CD25 requires B7 expression on antigen­presenting cells.24 bright) cells or upregulation of FOXP3+ T cells.24 Prolonged graft survival and donor­specific tolerance Moreover, two patients in this trial in whom belatacept has been induced by CTLA4–Ig in a human pancreatic treatment was stopped, developed chronic rejection, sug­ islet cell xenograft in rats, in a heterotopic cardiac allograft gesting that in humans short­term administration of this model in rats and mice, in a rat renal allograft model and in drug does not induce long­term tolerance. a mouse skin allograft model.25–29 In addition, CTLA4–Ig reduced the incidence of GvHD induced by allogeneic Inhibition of protein kinase C with sotrastaurin bone marrow transplantation and its related mortality. Protein kinase C (PKC) isoforms have a pivotal role Co­stimulation blockade in nonhuman primate models in signal transmission in T­cell­receptor engagement has bridged the gap between small­animal models and (signal 1) and CD28 co­activation (signal 2); inhibition of clinical protocols.30 Adolescent rhesus monkeys displayed PKC blocks early T­cell activation.34,35 The PKC family is

162 | MARCH 2010 | voluMe 6 www.nature.com/nrneph © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 162 8/2/10 15:29:15 revIewS

very large and is separated into three subfamilies accord­ of sotrastaurin so far seems acceptable for each of the ing to the needs of their cofactors.36–38 Based on findings combinations tested.49,50 from animal studies—mostly in PKC isoenzyme­specific several phase II trials have been started in the past 2 years knockout mice—at least three isoforms (PKC­α, ­β and to evaluate calcineurin inhibitor withdrawal at 3 months ­θ), have assigned importance in T­cell and B­cell signal­ after transplantation in patients receiving tacro limus plus ing.39–43 PKC­θ is mostly found in T cells and is a trans­ sotrastaurin.49,50 At 3 months after trans plantation, tacro­ mission factor between nuclear factor κB (NFκB) and limus was replaced with . However, the transcription factor activator protein 1, and has an both these trials were stopped because of marked increases important role in inducing the production of interleukin in episodes of acute rejection. A third phase II trial was (IL)­2.42–45 For example, notable impairment of T­cell conducted as a multicenter, open­label study to assess activation is seen in PKC­θ knockout mice.46 use of a calcineurin­free regimen of 300 mg sotras taurin sotrastaurin is a novel immunosuppressant that blocks and 720 mg mycophenolic acid twice daily (n = 44) in early T­cell activation via inhibition of PKC. This agent comparison with a control regimen of tacro limus and is a highly potent and selective inhibitor of the classical mycophenolic acid (n = 81).49,50 All patients also received (isoforms α and β) and novel (isoforms δ, ε, η and θ) PKC basiliximab and steroids. The primary end points were subfamilies. At the cellular level, early T­cell activa tion, acute rejection (assessed by biopsy), graft loss, death, or but not T­cell proliferation, is strongly inhibited by sotras­ loss to follow­up at 3 months. At 1 month, the sotras taurin taurin, which is reflected in raised IL­2 secretion.34 By regimen seemed as efficacious as the control regimen, by contrast, T­cell proliferation driven by IL­2 is not affected. 3 months, efficacy had fallen by 26% in the sotrastaurin sotrastaurin potently inhibits allogenic­ stimulated T­cell group for the composite end point compared with a decline proliferation in mixed lymphocyte reactions, but does of only 5% in the control group. Although sotrastaurin not exhibit hematological cytotoxic effects. sotrastaurin was associ ated with benefits to renal function and had blocks alloantigen­stimulated proliferation of effector acceptable tolerability, this result led to the early termina­ T cells, while leaving intact the regulatory functions and tion of the study. Treatment was discontinued in 12 of phenotype of CD4+CD25+FOXP3+CD127+ cells. This 81 patients who received sotrastaurin compared with two finding suggests that sotrastaurin does not abrogate the of 44 patients who received . The main reason + functions of tolerogenic CD4 TReG cells in patients after for discontinuation was graft rejection. 26% of patients in organ transplantation. the sotrastaurin­treatment group met the composite effi­ Investigation of the and exposure– cacy endpoint of biopsy­proven acute rejection, death, or efficacy relationship in de novo kidney transplant recipi­ graft loss at 3 months, versus 5% in the control group. ents showed that 1 week after transplantation, sotrastaurin exposure (defined by the area under the curve) after Inhibition of JAK‑3 daily treatment was stable in all patients (n = 152) and The development of has focused on the inhibi­ remained similar in the presence of tacrolimus or myco­ tion of ubiquitously expressed serine–threonine kinases phenolic acid.47 In addition, in contrast to the effects to prevent immunological responses driven by IL­2,51,52 of myco phenolic acid, the pharmacokinetic interactions of as cytokines have an important role in acute and chronic sotrastaurin generally increases tacrolimus blood levels by rejection, favoring clonal expansion and survival of naive less than twofold.47 sotrastaurin, is primarily metabolized and memory alloreactive T cells.53,54 Blockade of IL­2 alone by the liver, with metabolites being excreted via the bile,47 is not sufficient to impair the allogenic response owing to bound to the acute phase protein α1­acid glycoprotein. the involvement and redundancy of cytokines that share Levels of the drug in plasma should, therefore, be carefully some common poly peptide chains in the structure of their monitored after the transplantation. receptors.55 For example, the γ­chain of the IL­2 receptor Oral sotrastaurin is efficacious in rodents and non­ (IL­2R) is shared by IL­2, IL­7, and IL­15.56 The specificity primate humans in preventing graft rejection. In a rat of the different IL interactions with the receptor is, there­ heart transplantation model and a monkey kidney fore, achieved through another associated chain, which transplantation model, 30–50 mg/kg sotrastaurin mono­ is frequently the α­chain.56,57 In most cases, intracellular therapy or combined subtherapeutic doses of sotrastaurin trans duction is achieved through the activation of Janus (10–20 mg/kg) with ciclosporin or the sirolimus deriva­ kinase 3 (JAK­3). This tyrosine protein kinase is one of the tive resulted in significantly prolonged graft four known members of the Janus kinase family (JAK­1, survival compared with that in control animals. JAK­2, JAK­3, and TYK2), and has a key role in mediating A phase II study of sotrastaurin has been started in signals from the γc receptors, including IL­2R. JAK­3 is recipients of kidney allografts. Patients in this study activated via several cytokines (IL­2, IL­4, IL­7, IL­9, IL­15 receive 200 mg or 300 mg sotrastaurin twice daily in and IL­21).58–60 This protein has restricted and regulated combination with tacrolimus (0.1 mg/kg daily) or expression in lymphoid and myeloid cells; it is expressed enteric­coated mycophenolic acid (1,440 mg daily).48 at high levels in natural killer cells and thymocytes, In combination with tacrolimus, sotrastaurin has so far and is inducible in T cells, B cells and myeloid cells.61–63 shown excellent efficacy, whereas, when combined with Receptor oligomerization by IL­2 binding induces cross mycophenolic acid or everolimus, efficacy seems lower phosphoryla tion and trans activation of JAK­3 and JAK­1, than that achieved with the control regimen (tacrolimus which in turn results in tyrosine phosphorylation of and mycophenolic acid). Overall, the clinical tolerability multiple target molecules. As a consequence, the latent

NATURe RevIeWs | nepHrology vOLUMe 6 | MARCH 2010 | 163 © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 163 8/2/10 15:29:15 revIewS

transcription factors, signal transducer and trans cription Antibody-mediated rejection activator (sTAT) 3 and 5, bind to IL­2R via newly created Over the past 15 years, important progress has been docking sites, where they are tyrosine phosphorylated. made to prevent acute rejection. However, the rate of sTAT dimers are trans located to the nucleus where severe and/or steroid­resistant rejection has increased. they bind to specific regulatory sequences within target The reported incidence of antibody­mediated rejection is genes.58–60,64–66 An indispensable role for JAK­3 in cytokine variable, ranging from 5.6% to 23% in unselected popula­ receptor signal transduction is reinforced by observations tions,81–83 and from 30% to 60% in patients undergoing of Jak3 knockout mice64–66 and of humans with autosomal preconditioning for ABO­incompatible transplantation severe combined immunodeficiency syndrome caused by or transplantation across a positive donor­specific cross­ mutations in JAK3.64–68 JAK­3 deficiency is associated with match.84–88 Over the past 5 years, the development of a severe deficit in the immune system that leads to sub­ sensi tive methods to detect antibodies and to find markers stantial alterations in lymphocyte development, functional for antibody injury, such as C4d­A staining, made on the incompetence and susceptibility to apoptosis in peripheral basis of a careful analysis of biopsy samples, has empha­ T and B cells, and a reduced number or a lack of natural sized the role of HLA and non­HLA allo antibodies as killer cells.60 effectors of acute and chronic immune allograft injury.89,90 several inhibitors of JAK­3 have been developed. This action has become the predominant focus of the AG­490 has been developed as a JAK­2 inhibitor but also complement pathway in the rejection process. impairs JAK­3 signaling. This compound blocks the effect The current strategies to cure antibody­mediated rejec­ of γc cytokines, which prevents activation of JAK­3 in tion are based on the use of plasma exchange, intravenous T cells.69–71 By contrast, AG­490 does not affect antigen­ immunoglobulin, and agents that deplete B and T cells, induced activation of toxoid­responsive human including antilymphocyte globulins or . The T­cell clones, stimulation of Zap70 or p56Lck tyrosine overall outcomes, however, have not been as good as phosphorylation by monoclonal antibodies to CD3, was expected. or expression of α­chains, β­chains, or γ­chains in the The persistence of donor­specific antibodies can arise IL­2R.70–73 In a rat study, AG­490 was shown to substan­ from memory B cells or long­lived plasma cells,91,92 such tially prolong allograft survival when acting synergisti­ as CD20– and CD138+ cells. These cells are not targeted by cally with ciclosporin, a first­signal inhibitor, but not with a rituximab regimen and, therefore, new strategies need the third­signal inhibitor sirolimus.67 Graft infiltration to be explored to inhibit their functions. by mononuclear cells was minimal, and ex vivo stat5a/b DNA­binding activity stimulated by IL­2 was inhibited, plasma‑cell‑directed therapy with bortezomib but failed to affect the expression of IL­2Rα.74 The Rel/NFκB pathway is critical for the survival of CP­690,550 is an orally administered JAK­3 inhibitor memory B cells and long­lived plasma cells.93 For with nanomolar potency against JAK­3. Both in vitro and example, mice lacking transcription factors belonging in vivo, CP­690,550 causes a marked reduction of IL­2 to the Rel/NFκB family exhibit defects in their ability to and cytokine production by T cells, and reduces T­cell develop effective B­cell responses in terms of prolifera tion proliferation.75 In animal models of transplantation, and differentiation. In addition, mice deficient in NFκB CP­690,550 significantly reduces numbers of natural have defects in the formation of their germinative centers killer cells and T cells, but not CD8­effector memory involved in the development of memory B cells.94–96 T­cell populations.75,76 Furthermore, NFκB has an important role in controlling In nonhuman primate models of transplantation, cell cycle progression, the action of cell­adhesion mol­ CP­690,550, alone or in combination with myco phenolic ecules, and cytokine secretion.97 Activation of NFκB is acid, caused prominent delays in allogenic kidney rejec­ controlled by the degradation of its inhibitor, IκB via the tion and reduced intimal hyperplasia in an allogenic proteasome complex.98,99 Proteasome inhibitors are impli­ rat aorta transplantation model.77–79 In addition, the cated in the inhibition of NFκB100 by maintaining a high researchers of those studies observed a sizeable reduc­ level of IκB, and are linked with direct growth inhibition tion in the production of donor­specific IgG. A phase II and apoptosis of plasma cells. trial included 61 renal transplant recipients who were ran­ The proteasome inhibitor bortezomib exerts numerous domly assigned to receive CP­690,550 at doses of either biological effects, including blockade of T­cell cycling, 15 mg or 30 mg twice daily, or tacrolimus in combination inhibition of the upregulation of CD25 or adhesion mol­ with an IL­2R antagonist, mycophenolic acid, and ster­ ecules, decreasing T­helper­1 responses101–103 and the oids. The incidence of biopsy­proven acute rejection and subsequent apoptosis of activated T cells. Proteosomal renal function at 6 months was similar in all three groups, inhibition also affects the function of dendritic cells (by and no graft loss, deaths, or malig nancies were reported.78 reducing co­stimulatory molecule expression and cytokine However, patients receiving 30 mg CP­690,550 twice daily production and by inducing apoptosis).98,104 Proteosomal developed more infections, including cytomegalovirus inhibitors also affect B­cell survival by reducing the , and BK­virus­associated nephropathy, than production of IL­6 by bone marrow stromal cells.105 patients in the other groups. Bortezomib is a boronic acid dipeptide derivative Finally, a novel JAK­3 and sYK inhibitor, R348, that effectively treats multiple myeloma.106 Owing to reduces allogenic heart rejection in animal models of its effect on plasma tumor cells, bortezomib has been transplantation.80 tested in vitro on CD138+ cells from patients sensitized

164 | MARCH 2010 | voluMe 6 www.nature.com/nrneph © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 164 8/2/10 15:29:15 revIewS

to HLA.93 In contrast to rituximab and antilymphocyte without inhibiting the forma tion of complement C3b globulins, bortezomib induces apoptosis of CD138+ cells which participates in the opsonization process. and in vitro substantially reduces the production of class I The role of eculizumab in acute or chronic antibody­ and II antibodies. Bortezomib has also been successfully mediated rejection has yet to be defined. Prevention used to treat six kidney transplant patients with antibody­ has been reported in patients who had undergone renal mediated rejection who were refractory to standard transplantation with a B­cell­positive or T­cell­positive therapies.107 Treatment with bortezomib is associated flow cytometry cross­matched donor organ, in whom with a decrease in donor alloantibodies.108 Investigation donor­specific antibody levels remained low. No cases into its use to control the production of donor­specific of antibody­mediated rejection developed, whereas anti bodies could, therefore, be of interest, especially in in historical control groups, frequency ranged from patients who are highly sensitized, and this drug could 61% to 92%.115 eculizumab has been reported to cure have a role in controlling the allogenic response, although steroid­ resistant and thymoglobulin­resistant antibody­ a sustained response needs to be obtained to reduce the mediated rejection.115 In another patient, eculizumab risk of antibody­mediated rejection. therapy, in combination with plasmapheresis and intra­ venous immunoglobulin, led to a substantial reduction eculizumab for complement‑associated rejection of MAC deposition and salvage of a kidney allograft in During antibody­mediated rejection, complement one patient.107 The use of this antibody may, therefore, activa tion is observed via the classical pathway, which modulate antibody­mediated rejection and might have leads to capillary deposition of C4d­A.88 The presence roles in the preventive and therapeutic management of of this complement protein has been used as a marker this complication. for antibody­ mediated rejection. In addition, it is a marker for production of C3a and b, C5a and b, and, Conclusions consequently, the membrane­attack complex associated The current aim of treatment development in renal with endo thelial cell injury, which leads to the release of transplantation is to enable the reduction or avoidance of chemo attractants, procoagulant factors and/or to cell drugs with renal toxic effects while lessening the risk death.109,110 The activation of C3 convertase is associated of allograft rejection. New molecules have been devel­ with the production of C5a anaphylatoxin, which favors oped to inhibit specific pathways activated during the poly nuclear and macrophage recruitment, and of comple­ allogenic response or with roles in antibody production. ment C5b, which lead to synthesis of the membrane­ attack These compounds have been evaluated to complement complex. Different grades of comple ment activation are current validated strategies. The capacity of these agents present in the various stages of antibody­mediated rejec­ to prevent acute rejection, however, needs to be defined to tion. In addition, protocol biopsies of ABO­incompatible determine the target populations (that is, immunized or allografts have shown that, despite the reappearance of new patients). Compounds that can impair long­ living ABO antibodies, accommoda tion occurs, mostly enabled B cells or plasma cells, and inhibit the complement­ by cellular mechanisms that control activation of the activating pathway, offer new opportunities for the treat­ complement pathway.109 In a xenogenic model of cardiac ment of antibody­mediated rejection to prevent rejection trans plantation (pig to baboon), accommodation corre­ and prolong allograft survival. Important aspects of post­ lated with the overexpression in endothelial cells of CD59, transplantation therapy are maintaining the patient’s a complement regulatory molecule that prevents the ability to deal with infections and avoiding tumor synthesis of C5a anaphylatoxin and complement C5b.111 develop ment. An increase in immuno suppression needs Modulation of the complement cascade is, therefore, to be integrated sequentially and for a short period of becoming of interest as a treatment target in association time, after which standard maintenance therapy should with current available therapies, such as plasma exchange, be employed. Thus, understanding the impact of these intravenous immunoglobulin infusion, and T­cell and agents on the immune system and on patient survival B­cell depletion with antithymoglobulin or rituximab. needs to be a major objective for the future. eculizumab is a humanized antibody in which the constant domains of the parental IgG1 have been Review criteria replaced by the constant domain of human IgG4 and IgG2 to avoid complement and Fcγ receptor binding, Most articles were selected by searching the PubMed respectively.112 The drug acts against complement database using the search terms “alefacept”, C5 and was developed to treat paroxysmal nocturnal “LFA-3”, “CD2”, “BTI”, “apoptosis”, “transplantation”, hemoglobinuria, which is related to the deficiency of “belatacept”, “CTLA4-Ig”, “PKC”, “sotrastaurin”, “AEB”, “T-cell activation”, “CD80/86”, “Jak3 inhibitors”, “IL-2 glycosylphosphatidylinositol­linked surface proteins, receptor”, “IL-15 receptor”, “mTOR pathway”, “T-cell 112,113 including the terminal complement inhibitor CD59. regulation”, “bortezomib”, “plasma cell”, “memory B cell”, Recurrent hemolytic uremic syndrome is associated with “NFκB inhibitor”, “eculizumab”, “complement activation”, a heterozygous missense mutation of the gene encod­ “complement regulation”, “anti-C5 antibody”, “humoral ing the regulatory complement factor H.114 eculizumab rejection”, “C4d deposition”. The PubMed search was blocks the proinflammatory and cytolytic effects of ter­ performed between 1995 and 2009. Abstracts from minal complement activation.113 This effect reduces the the most recent ESOT and AST congresses were also searched for information on very new agents. chronic complement­mediated intravascular hemolysis

NATURe RevIeWs | nepHrology vOLUMe 6 | MARCH 2010 | 165 © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 165 8/2/10 15:29:16 revIewS

1. Hibberd, A. D. et al. Cancer risk associated with 18. Bromley, S. K. et al. The immunological synapse. critical for export of Il-2 from T cells. J. Immunol. ATG/OKT3 in renal transplantation. Transplant. Annu. Rev. Immunol. 19, 375–396 (2001). 167, 636–640 (2001). Proc. 31, 1271–1272 (1999). 19. Greenwald, R. J., Freeman, G. J. & Sharpe, A. H. 40. Pfeifhofer, C. et al. Defective IgG2a/2b class 2. Danielian, S., Fagard, R., Alcover, A., Acuto, O. & The B7 family revisited. Annu. Rev. Immunol. 23, switching in PKC alpha-/- mice. J. Immunol. 176, Fischer, S. The tyrosine kinase activity of p56lck 515–548 (2005). 6004–6011 (2006). is increased in human T cells activated via CD2. 20. vincenti, F. et al. Costimulation blockade with 41. Pfeifhofer, C. et al. Protein kinase C theta affects Eur. J. Immunol. 21, 1967–1970 (1991). belatacept in renal transplantation. N. Engl. J. Ca2+ mobilization and NFAT cell activation in 3. June, C. H., Fletcher, M. C., Ledbetter, J. A. & Med. 353, 770–781 (2005). primary mouse T cells. J. Exp. Med. 197, Samelson, L. E. Increases in tyrosine 21. Linsley, P. S. et al. Human B7–1 (CD80) and B7–2 1525–1535 (2003). phosphorylation are detectable before (CD86) bind with similar avidities but distinct 42. Sun, Z. et al. PKC-theta is required for TCR- phospholipase C activation after T cell receptor kinetics to CD28 and CTLA-4 receptors. Immunity induced NF-kappaB activation in mature but not stimulation. J. Immunol. 144, 1591–1599 1, 793–801 (1994). immature T lymphocytes. Nature 404, 402–407 (1990). 22. Finger, E. B. & Bluestone, J. A. When ligand (2000). 4. Déas, O. et al. Caspase-independent cell death becomes receptor--tolerance via B7 signaling on 43. volkov, Y., Long, A., McGrath, S., Ni Eidhin, D. & induced by anti-CD2 or staurosporine in activated DCs. Nat. Immunol. 3, 1056–1057 (2002). Kelleher, D. Crucial importance of PKC-beta(I) in human peripheral T lymphocytes. J. Immunol. 23. Grohmann, U. et al. CTLA-4-Ig regulates LFA-1-mediated locomotion of activated T cells. 161, 3375–3383 (1998). tryptophan catabolism in vivo. Nat. Immunol. 3, Nat. Immunol. 2, 508–514 (2001). 5. Dumont, C. et al. Targeting additional 1097–1101 (2002). 44. Marsland, B. J. & Kopf, M. T-cell fate and costimulatory pathways: a subtle role for CD2. 24. Chavez, H. et al. Absence of CD4CD25 regulatory function: PKC-theta and beyond. Trends Immunol. Transplant. Proc. 33, 199–200 (2001). T cell expansion in renal transplanted patients 29, 179–185 (2008). 6. Mollereau, B., Deas, O., Dumont, C., treated in vivo with belatacept mediated 45. Monks, C. R., Kupfer, H., Tamir, I., Barlow, A. & Charpentier, B. & Senik, A. Effects of anti-CD2 CD28-CD80/86 blockade. Transpl. Immunol. 17, Kupfer, A. Selective modulation of protein kinase : CD2- and CD95-mediated 243–248 (2007). C-theta during T-cell activation. Nature 385, apoptosis of human peripheral T cells. 25. Akalin, E. et al. CD28-B7 T cell costimulatory 83–86 (1997). Transplant. Proc. 31, 1245 (1999). blockade by CTLA4Ig in the rat renal allograft 46. Anderson, K. et al. Mice deficient in PKC theta 7. Mollereau, B. et al. CD2-induced apoptosis in model: inhibition of cell-mediated and humoral demonstrate impaired in vivo T cell activation and activated human peripheral T cells: a Fas- immune responses in vivo. Transplantation 62, protection from T cell-mediated inflammatory independent pathway that requires early protein 1942–1945 (1996). diseases. Autoimmunity 39, 469–478 (2006). tyrosine phosphorylation. J. Immunol. 156, 26. Larsen, C. P. et al. Long-term acceptance of skin 47. Kovarik, J. M., Huang, H. L., Slade, A., Sfikas, N. 3184–3190 (1996). and cardiac allografts after blocking CD40 and & Chandler, P. A. The effect on sotrastaurin 8. Rouleau, M. et al. Mitogenic CD2 monoclonal CD28 pathways. Nature 381, 434–438 (1996). pharmacokinetics of strong CYP3A inhibition by antibody pairs predispose peripheral T cells to 27. Lenschow, D. J. et al. Long-term survival of ketoconazole. Br. J. Clin. Pharmacol. 68, 381–385 undergo apoptosis on interaction with a third xenogeneic pancreatic islet grafts induced by (2009). CD2 monoclonal antibody. J. Immunol. 152, CTLA4lg. Science 257, 789–792 (1992). 48. vincenti, F. & Kirk, A. D. What’s next in the 4861–4872 (1994). 28. Pearson, T. C. et al. Transplantation tolerance pipeline. Am. J. Transplant. 8, 1972–1981 9. Snanoudj, R. et al. A role for CD2 antibodies induced by CTLA4-Ig. Transplantation 57, (2008). (BTI-322 and its humanized form) in the in vivo 1701–1706 (1994). 49. Budde, K. et al. AEB071, a novel protein kinase elimination of human T lymphocytes infiltrating 29. Turka, L. A. et al. T-cell activation by the CD28 C-inhibitor: first clinical results of an AEB071 an allogeneic human skin graft in SCID mice: ligand B7 is required for cardiac allograft (sotrastaurin) plus tacrolimus regimen in renal an Fcgamma receptor-related mechanism rejection in vivo. Proc. Natl Acad. Sci. USA 89, transplant recipients [abstract]. AST 391 (2009). involving co-injected human NK cells. 11102–11105 (1992). 50. Friman, S. et al. AEB071 (sotrastaurin) a novel Transplantation 78, 50–58 (2004). 30. Kirk, A. D. et al. CTLA4-Ig and anti-CD40 ligand protein kinase C-inhibitor: evaluation of an 10. Kaplon, R. J. et al. Short course single agent prevent renal allograft rejection in primates. Proc. AEB071 plus mycophenolate regiman in renal therapy with an LFA-3-IgG1 fusion protein Natl Acad. Sci. USA 94, 8789–8794 (1997). transplant recipients [abstract]. AST 458 prolongs primate cardiac allograft survival. 31. Kirk, A. D. et al. Induction therapy with (2009). Transplantation 61, 356–363 (1996). monoclonal antibodies specific for CD80 and 51. Abraham, R. T. Mammalian target of rapamycin: 11. Majeau, G. R., Meier, W., Jimmo, B., Kioussis, D. CD86 delays the onset of acute renal allograft immunosuppressive drugs uncover a novel & Hochman, P. S. Mechanism of lymphocyte rejection in non-human primates. Transplantation pathway of cytokine receptor signaling. Curr. function-associated molecule 3-Ig fusion 72, 377–384 (2001). Opin. Immunol. 10, 330–336 (1998). proteins inhibition of T cell responses. 32. Durrbach, A. et al. A Phase III Study of Belatacept 52. Denton, M. D., Magee, C. C. & Sayegh, M. H. Structure/function analysis in vitro and in human vs Cyclosporine in Kidney Transplants from Immunosuppressive strategies in CD2 transgenic mice. J. Immunol. 152, Extended Criteria Donors (BENEFIT-EXT Study). transplantation. Lancet 353, 1083–1091 2753–2767 (1994). Am. J. Transplant. (in press). (1999). 12. Miller, G. T. et al. Specific interaction of 33. vincenti, F. et al. A Phase III Study of Belatacept- 53. Schorle, H., Holtschke, T., Hünig, T., Schimpl, A. & lymphocyte function-associated antigen 3 with based Immunosuppression Regimens vs Horak, I. Development and function of T cells in CD2 can inhibit T cell responses. J. Exp. Med. Cyclosporine in Renal Transplant Recipients mice rendered interleukin-2 deficient by gene 178, 211–222 (1993). (BENEFIT Study). Am. J. Transplant. (in press). targeting. Nature 352, 621–624 (1991). 13. Krueger, G. G. et al. A randomized, double-blind, 34. Evenou, J. P. et al. The potent protein kinase C 54. Steiger, J., Nickerson, P. W., Steurer, W., placebo-controlled phase III study evaluating selective inhibitor AEB071 (sotrastaurin) Moscovitch-Lopatin, M. & Strom, T. B. IL-2 efficacy and tolerability of 2 courses of alefacept represents a new class of immunosuppressive knockout recipient mice reject islet cell in patients with chronic plaque psoriasis. J. Am. agents affecting early T-cell activation. allografts. J. Immunol. 155, 489–498 (1995). Acad. Dermatol. 47, 821–833 (2002). J. Pharmacol. Exp. Ther. 330, 792–801 (2009). 55. Thèze, J., Alzari, P. M. & Bertoglio, J. 14. Lebwohl, M. et al. An international, randomized, 35. Takai, Y., Kishimoto, A., Inoue, M. & Nishizuka, Y. and its receptors: recent advances and new double-blind, placebo-controlled phase 3 trial of Studies on a cyclic nucleotide-independent immunological functions. Immunol. Today 17, intramuscular alefacept in patients with chronic protein kinase and its proenzyme in mammalian 481–486 (1996). plaque psoriasis. Arch. Dermatol. 139, 719–727 tissues. I. Purification and characterization of an 56. Giri, J. G. et al. Utilization of the beta and gamma (2003). active enzyme from bovine cerebellum. J. Biol. chains of the IL-2 receptor by the novel cytokine 15. Weaver, T. A. et al. Alefacept promotes co- Chem. 252, 7603–7609 (1977). IL-15. EMBO J. 13, 2822–2830 (1994). stimulation blockade based allograft survival in 36. Baier, G. The PKC gene module: 57. Willerford, D. M. et al. Interleukin-2 receptor nonhuman primates. Nat. Med. 15, 746–749 molecular biosystematics to resolve its T cell alpha chain regulates the size and content of the (2009). functions. Immunol. Rev. 192, 64–79 (2003). peripheral lymphoid compartment. Immunity 3, 16. Shapira, M. Y. et al. Alefacept treatment for 37. Spitaler, M. & Cantrell, D. A. Protein kinase C and 521–530 (1995). refractory chronic extensive GvHD. Bone Marrow beyond. Nat. Immunol. 5, 785–790 (2004). 58. Liu, K. D., Gaffen, S. L. & Goldsmith, M. A. JAK/ Transplant. 43, 339–343 (2009). 38. Tan, S. L. & Parker, P. J. Emerging and diverse STAT signaling by cytokine receptors. Curr. Opin. 17. Stotler, C. J., Eghtesad, B., Hsi, E. & Silver, B. roles of protein kinase C in immune cell Immunol. 10, 271–278 (1998). Rapid resolution of GvHD after orthotopic liver signalling. Biochem. J. 376, 545–552 (2003). 59. Nelson, B. H. & Willerford, D. M. Biology of the transplantation in a patient treated with 39. Long, A., Kelleher, D., Lynch, S. & volkov, Y. interleukin-2 receptor. Adv. Immunol. 70, 1–81 alefacept. Blood 113, 5365–5366 (2009). Cutting edge: protein kinase C beta expression is (1998).

166 | MARCH 2010 | voluMe 6 www.nature.com/nrneph © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 166 8/2/10 15:29:16 revIewS

60. Thomis, D. C. & Berg, L. J. The role of Jak3 in 78. Borie, D. C. et al. Combined use of the JAK3 98. Baldwin, A. S. Jr. The NF-kappa B and I kappa B lymphoid development, activation, and signaling. inhibitor CP-690,550 with mycophenolate mofetil proteins: new discoveries and insights. Annu. Rev. Curr. Opin. Immunol. 9, 541–547 (1997). to prevent kidney allograft rejection in nonhuman Immunol. 14, 649–683 (1996). 61. Hofmann, S. R. et al. Cytokines and their role in primates. Transplantation 80, 1756–1764 (2005). 99. Thanos, D. & Maniatis, T. NF-kappa B: a lesson in lymphoid development, differentiation and 79. Rousvoal, G. et al. Janus kinase 3 inhibition with family values. Cell 80, 529–532 (1995). homeostasis. Curr. Opin. Allergy Clin. Immunol. 2, CP-690,550 prevents allograft vasculopathy. 100. Sunwoo, J. B. et al. Novel proteasome inhibitor 495–506 (2002). Transpl. Int. 19, 1014–1021 (2006). PS-341 inhibits activation of nuclear factor- 62. Leonard, W. J. & O’Shea, J. J. Jaks and STATs: 80. velotta, J. B. et al. A novel JAK3 inhibitor, R348, kappa B, cell survival, tumor growth, and biological implications. Annu. Rev. Immunol. 16, attenuates chronic airway allograft rejection. angiogenesis in squamous cell carcinoma. Clin. 293–322 (1998). Transplantation 87, 653–659 (2009). Cancer Res. 7, 1419–1428 (2001). 63. O’Shea, J. J., Pesu, M., Borie, D. C. & 81. Crespo, M. et al. Acute humoral rejection in renal 101. Ghosh, S., May, M. J. & Kopp, E. B. NF-kappa B Changelian, P. S. A new modality for allograft recipients: I. Incidence, serology and and Rel proteins: evolutionarily conserved immunosuppression: targeting the JAK/STAT clinical characteristics. Transplantation 71, mediators of immune responses. Annu. Rev. pathway. Nat. Rev. Drug Discov. 3, 555–564 652–658 (2001). Immunol. 16, 225–260 (1998). (2004). 82. Rocha, P. N. et al. Beneficial effect of 102. McCarthy, P. L. Jr et al. Inhibition of interleukin-1 by 64. Nosaka, T. et al. Defective lymphoid development plasmapheresis and intravenous immunoglobulin an interleukin-1 receptor antagonist prevents in mice lacking Jak3. Science 270, 800–802 on renal allograft survival of patients with acute graft-versus-host disease. Blood 78, 1915–1918 (1995). humoral rejection. Transplantation 75, (1991). 65. Park, S. Y. et al. Developmental defects of 1490–1495 (2003). 103. Xun, C. Q., Thompson, J. S., Jennings, C. D., lymphoid cells in Jak3 kinase-deficient mice. 83. Shah, A. et al. Treatment of C4d-positive acute Brown, S. A. & Widmer, M. B. Effect of total body Immunity 3, 771–782 (1995). humoral rejection with plasmapheresis and rabbit irradiation, busulfan-cyclophosphamide, or 66. Thomis, D. C., Gurniak, C. B., Tivol, E., polyclonal antithymocyte globulin. Transplantation cyclophosphamide conditioning on inflammatory Sharpe, A. H. & Berg, L. J. Defects in B 77, 1399–1405 (2004). cytokine release and development of acute and lymphocyte maturation and T lymphocyte 84. Gloor, J. M. et al. Overcoming a positive chronic graft-versus-host disease in activation in mice lacking Jak3. Science 270, crossmatch in living-donor kidney transplantation. H-2-incompatible transplanted SCID mice. Blood 794–797 (1995). Am. J. Transplant. 3, 1017–1023 (2003). 83, 2360–2367 (1994). 67. Macchi, P. et al. Mutations of Jak-3 gene in 85. Jordan, S. C. et al. Intravenous immune globulin 104. Wang, X., Luo, H., Chen, H., Duguid, W. & Wu, J. patients with autosomal severe combined treatment inhibits crossmatch positivity and Role of proteasomes in T cell activation and immune deficiency (SCID). Nature 377, 65–68 allows for successful transplantation of proliferation. J. Immunol. 160, 788–801 (1998). (1995). incompatible organs in living-donor and cadaver 105. Finn, P. W., Stone, J. R., Boothby, M. R. & 68. Russell, S. M. et al. Mutation of Jak3 in a patient recipients. Transplantation 76, 631–636 (2003). Perkins, D. L. Inhibition of NF-kappaB-dependent with SCID: essential role of Jak3 in lymphoid 86. Montgomery, R. A. et al. Plasmapheresis and T cell activation abrogates acute allograft development. Science 270, 797–800 (1995). intravenous immune globulin provides effective rejection. J. Immunol. 167, 5994–6001 (2001). 69. Gazit, A. et al. Tyrphostins. 2. Heterocyclic and rescue therapy for refractory humoral rejection 106. Richardson, P. G. et al. A phase 2 study of alpha-substituted benzylidenemalononitrile and allows kidneys to be successfully bortezomib in relapsed, refractory myeloma. tyrphostins as potent inhibitors of EGF receptor transplanted into cross-match-positive recipients. N. Engl. J. Med. 348, 2609–2617 (2003). and ErbB2/neu tyrosine kinases. J. Med. Chem. Transplantation 70, 887–895 (2000). 107. Everly, M. J. et al. Bortezomib provides effective 34, 1896–1907 (1991). 87. Park, W. D. et al. Accommodation in ABO- therapy for antibody- and cell-mediated acute 70. Kirken, R. A. et al. Tyrphostin AG-490 inhibits incompatible kidney allografts, a novel mechanism rejection. Transplantation 86, 1754–1761 (2008). cytokine-mediated JAK3/STAT5a/b signal of self-protection against antibody-mediated injury. 108. Trivedi, H. L. et al. Abrogation of anti-HLA transduction and cellular proliferation of antigen- Am. J. Transplant. 3, 952–960 (2003). antibodies via proteasome inhibition. activated human T cells. J. Leukoc. Biol. 65, 88. Tanabe, K. et al. Long-term results of ABO- Transplantation 87, 1555–1561 (2009). 891–899 (1999). incompatible living kidney transplantation: 109. Colvin, R. B. Antibody-mediated renal allograft 71. Meydan, N. et al. Inhibition of acute a single-center experience. Transplantation 65, rejection: diagnosis and pathogenesis. J. Am. Soc. lymphoblastic leukaemia by a Jak-2 inhibitor. 224–228 (1998). Nephrol. 18, 1046–1056 (2007). Nature 379, 645–648 (1996). 89. Racusen, L. C. & Haas, M. Antibody-mediated 110. Regele, H. et al. Capillary deposition of 72. Säemann, M. D. et al. Suppression of primary rejection in renal allografts: lessons from complement split product C4d in renal allografts T-cell responses and induction of alloantigen- pathology. Clin. J. Am. Soc. Nephrol. 1, 415–420 is associated with basement membrane injury in specific hyporesponsiveness in vitro by the Janus (2006). peritubular and glomerular capillaries: kinase inhibitor tyrphostin AG490. 90. Terasaki, P. I. Humoral theory of transplantation. a contribution of humoral immunity to chronic Transplantation 70, 1215–1225 (2000). Am. J. Transplant. 3, 665–673 (2003). allograft rejection. J. Am. Soc. Nephrol. 13, 73. Wang, L., Chen, J. J., Gelman, B. B., Konig, R. & 91. McHeyzer-Williams, M. G. & Ahmed, R. B cell 2371–2380 (2002). Cloyd, M. W. A novel mechanism of CD4 memory and the long-lived plasma cell. Curr. Opin. 111. Williams, J. M. et al. Acute vascular rejection and lymphocyte depletion involves effects of HIv on Immunol. 11, 172–179 (1999). accommodation: divergent outcomes of the resting lymphocytes: induction of lymph node 92. Slifka, M. K., Antia, R., Whitmire, J. K. & Ahmed, R. humoral response to organ transplantation. homing and apoptosis upon secondary signaling Humoral immunity due to long-lived plasma cells. Transplantation 78, 1471–1478 (2004). through homing receptors. J. Immunol. 162, Immunity 8, 363–372 (1998). 112. Brodsky, R. A. How I treat paroxysmal nocturnal 268–276 (1999). 93. Perry, D. K. et al. Proteasome inhibition causes hemoglobinuria. Blood 113, 6522–6527 (2009). 74. Behbod, F. et al. Concomitant inhibition of Janus apoptosis of normal human plasma cells 113. Rother, R. P., Rollins, S. A., Mojcik, C. F., kinase 3 and calcineurin-dependent signaling preventing alloantibody production. Am. J. Brodsky, R. A. & Bell, L. Discovery and pathways synergistically prolongs the survival of Transplant. 9, 201–209 (2009). development of the complement inhibitor rat heart allografts. J. Immunol. 166, 3724–3732 94. Caamaño, J. H. et al. Nuclear factor (NF)-kappa B2 eculizumab for the treatment of paroxysmal (2001). (p100/p52) is required for normal splenic nocturnal hemoglobinuria. Nat. Biotechnol. 25, 75. Paniagua, R. et al. Effects of JAK3 inhibition with microarchitecture and B cell-mediated immune 1256–1264 (2007). CP-690,550 on immune cell populations and responses. J. Exp. Med. 187, 185–196 (1998). 114. Mache, C. J. et al. Complement inhibitor their functions in nonhuman primate recipients of 95. Franzoso, G. et al. Mice deficient in nuclear factor eculizumab in atypical hemolytic uremic kidney allografts. Transplantation 80, (NF)-kappa B/p52 present with defects in humoral syndrome. Clin. J. Am. Soc. Nephrol. 4, 1312–1316 1283–1292 (2005). responses, germinal center reactions, and splenic (2009). 76. Conklyn, M., Andresen, C., Changelian, P. & microarchitecture. J. Exp. Med. 187, 147–159 115. Locke, J. E. et al. The use of antibody to Kudlacz, E. The JAK3 inhibitor CP-690550 (1998). complement protein C5 for salvage treatment of selectively reduces NK and CD8+ cell numbers in 96. Ranger, A. M. et al. Delayed lymphoid repopulation severe antibody-mediated rejection. Am. J. cynomolgus monkey blood following chronic oral with defects in IL-4-driven responses produced by Transplant. 9, 231–235 (2009). dosing. J. Leukoc. Biol. 76, 1248–1255 (2004). inactivation of NF-ATc. Immunity 8, 125–134 77. Borie, D. C. et al. Immunosuppression by the (1998). Acknowledgments JAK3 inhibitor CP-690,550 delays rejection and 97. Barnes, P. J. & Karin, M. Nuclear factor-kappaB: The authors thank Professor Lionel Rostaing (Head of significantly prolongs kidney allograft survival in a pivotal transcription factor in chronic the Department of Transplantation, Rangueil Teaching nonhuman primates. Transplantation 79, inflammatory diseases. N. Engl. J. Med. 336, Hospital, Toulouse, France) for his constructive 791–801 (2005). 1066–1071 (1997). discussion and advice.

NATURe RevIeWs | nepHrology vOLUMe 6 | MARCH 2010 | 167 © 2010 Macmillan Publishers Limited. All rights reserved

nrneph_233_MAR10.indd 167 8/2/10 15:29:16