<<

BRIEF REVIEW www.jasn.org

Effect of Immunosuppressive Drugs on Humoral Allosensitization after Kidney Transplant

† † Olivier Thaunat,* Alice Koenig,* Claire Leibler, and Philippe Grimbert

*Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, INSERM UMR1111, Université de Lyon, Lyon, France; and †Service de Néphrologie et Transplantation, Hôpital Henri Mondor, Centre de référence maladie rare Syndrome Néphrotique Idiopathique, Institut Francilien de recherche en Néphrologie et Transplantation, INSERM U955, Université Paris Est Créteil, Assistance Publique-Hôpitaux de Paris, Creteil, France

ABSTRACT The negative effect of donor-specific antibodies on the success of solid transplant is Fc segments, resulting in the formation now clearly established. However, the lack of effective treatment to prevent the of ordered antibody hexamers with ex- development of antibody-mediated lesions deepens the need for clinicians to focus quisite ability to recruit and activate on primary prevention of de novo humoral allosensitization. Among the factors C1q.12 The probability that a sufficient associated with the risk of developing de novo donor–specific antibodies, therapeu- number of IgGs binds close enough to tic immunosuppression is the most obvious parameter in which improvement is form such hexameric complexes is possible. Beyond compliance and the overall depth of immunosuppression, it is strongly influenced by antibody titer. likely that the nature of the drugs is also crucial. Here, we provide an overview of In line with this idea is the fact that the molecular effect of the various immunosuppressive drugs on biology. the ability of DSAs to bind C1q or Clinical data related to the effect of these drugs on de novo humoral allosensitiza- C3d in a solid-phase assay correlates tion are also examined, providing a platform from which clinicians can optimize with their MFI.11,13 Another clue that immunosuppression for prevention of de novo donor–specific antibody generation DSA titer is an important factor in at the individual level. their ability to activate complement is in the study by Yell et al.14 which shows J Am Soc Nephrol 27: ccc–ccc, 2016. doi: 10.1681/ASN.2015070781 that most C1q binding DSAs can be con- verted into non–C1q binding DSAs when diluted. However, DSAs with sim- Antibody-mediated rejection (AMR) is HLA molecules represent the most ilar MFI do not always activate the com- widely recognized as the leading cause of documented targets for DSAs, it is clear plement,11 which indicates that, beyond late transplant failure and accounts for that DSAs can also be directed against the quantity, some qualitative features of approximately two thirds of renal allo- other kinds of molecular targets, includ- antibodies also affect their ability to ac- graft losses.1,2 In 2003, AMR was added ing polymorphic minor histocompati- tivate the complement. It is well known to the Banff classification for inter- bility antigens and after a breakdown of pretation of renal allograft biopsy,3 B cell tolerance,8 nonpolymorphic auto- sparking intense interest in the trans- antigens.9 DSAs are massive proteins A.K. and C.L. contributed equally to this work. plant community. Clinical studies over that are largely sequestrated in the circu- the last 10 years have established that lation.10 Binding of circulating DSAs to Published online ahead of print. Publication date antibodies generated de novo post- directly accessible graft endothelial cells available at www.jasn.org. transplantation against donor-specific can trigger the activation of a classic Correspondence: Dr. Olivier Thaunat, Service de antigens (DSAs) are strongly associated complement pathway, a central process Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, 5 Place d’Ar- and may be an important cause of in the pathophysiology of acute AMR sonval, 69003 Lyon, France, or Dr. Philippe Grimbert, allograft loss.1,4,5 (i.e., AMR with acute graft dysfunction).11 Service de Néphrologie et de Transplantation, Hôpital Experimental studies have shed light Complement activation by antibodies Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Creteil, France. Email: 6,7 on the natural history of AMR. The depends on the recruitment and activa- [email protected] or philippe.grimbert@hmn. sequence starts with the generation of tion of the component C1q. After anti- aphp.fr antibodies directed against the graft. Al- gen binding on cells, IgGs establish Copyright © 2016 by the American Society of though highly polymorphic mismatched specific noncovalent interactions between

J Am Soc Nephrol 27: ccc–ccc, 2016 ISSN : 1046-6673/2707-ccc 1 BRIEF REVIEW www.jasn.org that IgG4 is much less effective than INCIDENCE AND RISK FACTORS it is fundamental to take into account IgG1 and IgG3 in triggering the comple- FOR SENSITIZATION drug exposure evaluated by a pretrans- ment cascade. Strikingly, a recent study plant pharmagenomics analysis and reported that DSA subclasses identify The prevalence of de novo donor–specific post–transplant pharmacokinetic moni- distinct phenotypes of kidney allograft antigens (dnDSAs) is generally between toring. Indeed, the incidence of sensitiza- AMR. In this study, IgG3 DSAs were as- 5% and 10% at 1 year postrenal trans- tion is dramatically increased in patients sociated with a shorter rejection time, plantation and slowly increases there- who are nonadherent.2,19 Moreover, strict increased microcirculation injury, and after to 20% at 5 years.18–20 However, these monitoring of DSAs should be performed more C4d capillary deposition; in con- percentages should be interpreted with when clinical situations, such as infectious trast, IgG4 correlated with later allograft caution. During the last 10 years, assays episodes, BK virus viremia,21 diarrhea, or injury.15 for the detection of anti-HLA antibodies ,22 lead to a reduction of immuno- In contrast to acute AMR, activation have evolved from complement-dependent suppression. It is also noteworthy that of the classic complement pathway does cytotoxicity to the highly sensitive death with a functional allograft is cur- not seem to be critical for the develop- microbead–based Luminex assay. There- rently an important cause of graft loss. All ment of chronic AMR lesions. The first fore, it is possible that DSAs identified in recommendations regarding antirejection evidence supporting this concept was in the post-transplant period could, in fact, must, therefore, take into account the study by Colvin and coworkers,16 be preformed DSAs that were not previously the related risk of overimmunosuppression which transplanted immunodeficient detected by the complement–dependent cy- and the benefit-to-risk ratio of all RAG knockout mice with allogenic totoxicity assay. Furthermore, a negative therapeutic decisions.28 Finally, defining heart. Colvin and coworkers16 observed pretransplant serum does not completely the best drug combination for primary that a passive transfer of noncomple- rule out the possibility that DSAs might be prevention requires an understanding of ment binding DSAs was sufficient expressed in the context of B cell memory B cell biology and the molecular mecha- enough to promote allograft vasculop- response. Indeed, using a novel HLA B Cell nisms by which each drug interferes with athy. Innate immune cells, including Enzyme-Linked ImmunoSpot Assay, dnDSA generation. neutrophils, macrophages, and NK cells, Lúcia et al.21 showed that, in 30% of pa- can bind to Fc fragments of antibodies tients, memory B cells could be detected and release lytic enzymes (a mechanism in highly sensitized patients in the ab- IMMUNE MECHANISMS called antibody–dependent, cell–mediated sence of corresponding anti–HLA anti- UNDERLYING DSA GENERATION cytotoxicity), which mediate smoldering bodies in the circulation. endothelial cell damages. In turn, chronic Risk factors for sensitization fall into DSAs are high-affinity IgGs directed vascular inflammation promotes the two categories: either they increase graft against protein antigen. These charac- progressive development of typical vascu- immunogenicity or they increase the teristics imply that DSA generation lar lesions (i.e., transplant glomerulopathy, recipient ’s ability to re- results from a thymo–dependent B cell allograft vasculopathy, and lamination spond to allogeneic epitopes. Graft immu- response. This type of humoral response, of the peritubular capillary basement nogenicity depends on both the quantity which develops within secondary lym- membrane). of allogeneic epitopes expressed by the phoid organs (spleen or lymph nodes), Current treatment protocols for graft (i.e., the number of mismatched involves two distinct events that are sep- AMR use permutations of a multi- amino acid polymorphisms)22 and their arated both spatially and temporally29 pronged approach that include DSA nature.23,24 It is also possible that graft (Figure 1). B cell activation is initiated removal with , B or inflammation (either preexisting in in the B cell area of secondary lymphoid plasma cell depletion with anti-CD20 extended criteria donors or secondary to organs by the binding of cognate antigen and proteasome inhibitors (PIs), respec- ischemia-reperfusion injuries) could to surface B cell receptors (BCRs). This tively, and the blockade of antibody– increase immunogenicity.25 The ability step is strengthened with the help of in- mediated effector functions with of recipients’ immune systems to respond nate immune effectors. Macrophages of intravenous Igs (IVIgs) or terminal to allogeneic epitopes is reflected in their the subcapsular sinus capture lymph- complement pathway inhibitors.7 The pretransplant immunization status (i.e., borne antigens,30 and the binding of 3-year graft survival rate with these pro- responses to previous challenges), acute these antigens to the surface Ig of the tocols is ,50%.17 Given the detrimental cellular rejection episodes, and younger cognate B cells contributes to the estab- effect of DSAs on transplant survival and age, all of which have been consistently lishment of the immunologic synapse the lack of effective treatment to prevent associated with increased risk of sensitiza- between the B cells and the subcapsular the development of antibody-mediated tion.19,26,27 Among the factors associated sinus macrophages30 (Figure 1, top lesions, the priority of clinicians in with post-transplant sensitization, thera- panel). BCR crosslinking leads to the charge of recipients of solid organs peutic immunosuppression represents the phosphorylation of immunoreceptor should be primary prevention of DSA most obvious modifiable risk factor. To tyrosine–based activation motifs by the generation. optimize immunosuppressive strategies, Src family kinase Lyn. This initiates the

2 Journal of the American Society of Nephrology J Am Soc Nephrol 27: ccc–ccc,2016 www.jasn.org BRIEF REVIEW

formation of the signalosome, an assembly of intracellular signaling molecules that, together with adaptor molecules, are re- sponsible for the coordinated regulation of downstream signaling pathways, in- cluding calcineurin and mammalian tar- get of rapamycin (mTOR) pathways, and ultimately, modify the gene expression. Importantly, signaling BCR microclus- ters also associate with molecular motor dynein, which allows for the coordinated movement of antigen into the intracyto- plasmic endosomal compartment of the B cell.29,31 Internalized antigen is pro- cessed and subsequently exposed on the B cell surface in association with MHC molecules.32 B cells that have received this first activation signal migrate toward the B zone-T zone boundary,33 where they pair with a particular subset of cog- nate CD4+ T cells called T follicular helper (Tfh) cells34 (Figure 1, bottom panel). This provides the second activa- tion signal for B cells through cell surface costimulatory ligand interactions (espe- cially CD40L) and directional cytokine production (in particular, IL-21).34 The activated B cells enter the germinal center, where they proliferate. Their pro- genies go through repeated cycles of random somatic hypermutation of Ig variable regions driven by the enzyme activation–induced cytidine deaminase followed by selection of progressively higher–affinity mutants. Differentiation of some of these high-affinity mutants into plasma cells or memory B cells leads to the increased affinity of serum anti- bodies and the high affinity of the recall response.35 Interestingly, recent evidence has shown that plasma cells and memory B cells generated by the germinal center reaction require additional helper signals from innate immune effectors, including eosinophils and basophils, to extend their lifespan in postgerminal center niches (summarized in ref. 36). Recent data suggest that B cell func- tions are not restricted to the deleterious role of secreting dnDSAs.37 In the in- flammatory context, some B cell sub- Figure 1. Key molecular steps of B cell activation and sites of action of immunosuppressive sets have shown regulatory properties drugs. B cell activation is a two-step process. (Top panel) Binding of the cognate antigen to the that rely on either secretion of anti- surface Ig of B cells serves as the first signal of activation. BCR signaling drives the internalization inflammatory cytokines (IL-10) and/ of bound antigens (donor’s HLA molecules) to endosomal compartments, where they are or contact-dependent mechanisms. The

J Am Soc Nephrol 27: ccc–ccc, 2016 Immunosuppression and De Novo DSA 3 BRIEF REVIEW www.jasn.org

fine equilibrium between these two roles onward and exceed baseline levels after 6 dnDSAs, and AMR was not specifically of B cells should also be taken into ac- months.42 Moreover, the composition of addressed).49,50 In a more recent trial, count when discussing immunosup- B cell subsets is altered compared with Brokhof et al.51 reported in a cohort of pressive strategies.38 that found pretransplant; the levels of 114 moderately sensitized recipients of Determining the actions of each im- naïve and transitional B cells increase, deceased donor renal transplants that in- munosuppressive drug on B cell biology whereas the absolute number of mem- duction with ATG is associated with a and their consequent effect on dnDSA ory B cells decreases significantly.42 In- reduction in the occurrence of dnDSAs production represents the first step in terestingly, in , the and AMR compared with . defining practical clinical immunosup- composition of the B cell compartment However, it should be noted that, in pressive strategies. is repeatedly identified as an important this study, (1) the difference became sta- determinant for graft outcome.43,44 tistically different only at 3 years, (2) Heidt et al.42 suggested that lymphocyte only a few patients assigned to basilixi- EFFECT OF INDUCTION THERAPY depletion with may skew mab had a follow-up at 36 months, and ON DNDSA GENERATION the B cell compartment toward a proto- (3) the patients who received ATG also lerogenic profile. This hypothesis has received more plasmapheresis and IVIg Rabbit antithymocyte globulin (ATG) isa been further strengthened by another than the patients who received rituximab. mixture of polyclonal IgGs recovered work that showed that a significant Finally, if ATG should be considered from the sera of rabbits immunized expansion of regulatory–type B cells (de- as the first–line induction therapy in with human thymocytes. The binding fined as CD19+CD5+CD1dhigh) is associ- previously sensitized patients, no data of antibodies to recipient’sTlympho- ated with superior graft function and that are available concerning a potential cytes results in complement– and/or this pattern is more common after alem- benefit of this induction strategy in non- antibody–dependent, cell–mediated cy- tuzumab induction.45 However, al- sensitized recipients. totoxicity and an interaction with though this hypothesis is interesting, it surface antigens, possibly resulting in has been challenged by recent conflicting apoptosis or anergy (Figure 1). Interest- reports from two independent groups EFFECT OF MAINTENANCE ingly, because certain epitopes are shared showing that alemtuzumab–induced IMMUNOSUPPRESSION ON between T and B cells and because pedi- B cell depletion/reconstitution is as- DNDSA GENERATION atric thymi also contain CD20+ B cells sociated with a higher incidence of and CD138+ plasma cells, ATG contain AMR compared with induction with IgGs directed against these latter sub- ATG.46,47 Corticosteroids are the oldest immune sets.39 As a result, in vitro studies have Basiliximab is a chimeric mouse- response–modifying drugs. Their mech- shown that ATG induces apoptosis of human nondepleting mAb targeting anism of action is diverse and includes naive, activated B cells and marrow the a-chain (CD25) of the IL-2 receptor interference with intracellular transcrip- resident plasma cells at clinically rele- (Figure 1). Basiliximab was developed tion factors and signaling pathways of vant concentrations (1–100 ng/ml).39 for induction therapy on the basis of several surface receptors, including the However, the importance of ATG– the concept that it would block IL-2 T cell antigen receptor52 and down- induced plasma and/or B cell depletion binding, thereby inhibiting T cell prolif- stream kinases.53 in vivo has not been clearly shown.40 eration. Interestingly, activated B cells High doses of corticosteroids (.1 An alternative to ATG as a depleting also express CD25,48 and IL-2 has a cru- mg/kg), used for induction therapy in agent for induction is alemtuzumab, a cial role in driving the differentiation of transplantation, also induce lymphocyte humanized mAb directed against CD52, activated B cells toward plasma cells in apoptosis (including B cells)46,54 and can which is a protein widely distributed on vitro.48 prevent in vitro differentiation of B cells the surface of lymphocytes and mono- In practice, ATG should be preferred into plasma cells.55 High concentrations cytes (but with an unknown function). in sensitized patients without DSAs, of corticosteroids are also likely to inter- Alemtuzumab induces rapid and pro- because two randomized clinical trials fere with Fc g–receptor signaling, in- found B (and T) lymphocyte depletion41 showed beneficial short–term effects on cluding the inhibitory Fc g–RIIb, which (Figure 1). A recent study showed that, acute rejection, graft function, and acts as a key negative modulator of B cell after this initial depletion, B cells repo- survival (however, the relationship be- function and controls bone marrow pulate the peripheral blood from 6 weeks tween induction immunosuppression, plasma cell apoptosis.56 The effect of a

processed. Selected peptides are then loaded onto recipient MHC class 2 molecules and presented on the B cell surface. (Bottom panel) B cells interact with cognate Tfh cells that provide the second costimulation signal for the completion of B cell activation. with proven efficiency are shown as red lines. Putative mechanisms of action are dashed red lines with question marks. TCR, T cell receptor.

4 Journal of the American Society of Nephrology J Am Soc Nephrol 27: ccc–ccc,2016 www.jasn.org BRIEF REVIEW standard maintenance dose of cortico- because calcineurin inhibitors (CNIs), in engagement of the BCR activates PI3K, therapy on human B cells was contrast with genetic deletion, do not which beyond activation of NF-kB– studied in a seminal in vitro study by completely suppress calcineurin activity dependent transcription, also initiates a Cupps et al.57 in the mid-1980s. Early in lymphocytes.64 In fact, when Heidt distinct signaling pathway involving events in B cell activation, such as et al.65 tested the effect of CNIs in T and the Akt and mTOR serine/threonine anti-IgM–induced B cell activation and B cell cocultures, they observed that, kinases.70 proliferation, are profoundly suppressed whereas cyclosporin A (CsA) and tacroli- Surprisingly little is known about the by corticosteroids.57 To be effective, the mus were both capable of inhibiting Ig functions of the mTOR complexes in B corticosteroids have to be present in production when B cells were cultured cells. Homozygous knockin mice for the culture during the first 24 hours, with T cells that were not preactivated, hypomorphic mTOR showed decreased suggesting a drug-induced modulation they failed to do so when preactivated T ability to produce antibodies.69 To deter- of an early event.57 cells were used to stimulate B cells. These mine whether these effects were caused Over the last decades, several attempts results indicate that CNIs affect the hu- by a direct mTOR blockade in the B cells have been made to develop steroid- moral immune response of patients with or only reflected T cell deficiency, the sparing strategies to avoid their important transplants by interfering with T helper same group examined the humoral re- deleterious side effects. No increased risk signals and not by targeting B cells di- sponses in mTOR conditional B cell in sensitization has been reported in rectly.65 This hypothesis is supported by knockout mice.71 Mice with mTOR de- patients without corticosteroids in a pro- the study by Struijk et al.,66 which showed leted in their B cell lineage produced spective single–center study on kidney that patients with renal transplants who fewer splenic germinal centers and also, transplantation58 and two retrospective were stable on plus CsA exhibited a decrease in switched high– studies on .59,60 failed to develop antibodies against affinity antibody responses in contrast These encouraging results are also sup- T cell–dependent model antigens but re- to their wild-type littermates.71 The ported by the conclusions in the work by sponded normally to T cell–independent fact that mTOR inhibitors are able to Delgado et al.,61 which randomized 37 antigens. prevent the in vitro production of Ig recipients of kidney transplants with Some studies have reported that the use with equal efficiency when human B ATG induction, , and myco- of CsA is associated with an increased risk cells are cultured with preactivated or phenolic acid (MPA) for of dnDSAs compared with tacrolimus, not preactivated T cells further shows withdrawal 1 week post-transplantation. 59,67 butthisdifferenceisnotalways that mTOR inhibitors, in contrast with Delgado et al.61 did not observe any dif- found.5 The effect of the nature of CNIs CNIs, have a direct effect on B cells.65,72 ference in dnDSA incidence at 5 years. (CsA or tacrolimus) on dnDSA occur- This concept is also validated by the re- However, because early steroid with- rence is made difficult to assess because sults of a recent study that compared the drawal is associated with a higher in- of the specific pharmacokinetic features effects of CNI and mTOR inhibitors on cidence of subclinical rejection and of each molecule (including variance of B cells from healthy volunteers. Only chronic graft injury in recipients with oral bioavailability, action on entero- was able to inhibit the prolifer- high immunologic risk, such as hepatic circulation of MPA observed ation of B cells and their differentiation blacks,62 a steroid-sparing strategy with CsA, and genetic polymorphisms of into plasma cells. Interestingly, the effect should only be considered in selected re- tacrolimus), which are likely to modify of sirolimus seemed more pronounced on cipients with low immunologic risk. drug exposure. Finally, beyond the nature the CD19+CD27+ memory subset.73 of the CNI, compliance19 and trough lev- Several clinical studies have reported Calcineurin Inhibitors els59 have been established as the most successful conversion to an mTOR in- It has been known since the mid-1990s important risk factors for sensitization of hibitor–based regimen.74–77 Although that, similar towhat happens in T cells, the patients on CNI. several of them showed a significant im- engagement of BCR activates NFAT provement in renal function after conver- through a calcineurin-dependent pathway mTOR Inhibitors sion to a CNI-free regimen, long-term in B lymphocytes (Figure 1). The impor- Immunosuppressive drugs targeting the data on the humoral response are limited. tance of this pathway for T cell–dependent mTOR signaling pathway have been de- In the ZEUS Trial,74 recipients of kidney humoral responses has been studied in veloped to avoid or reduce CNI-associated transplants on CsA-based immunosup- mice, in which calcineurin was deleted toxicity. Sirolimus and pression were randomized at 4 months specifically in B cells.63 These mice had engage FK binding proteins, forming a post-transplantation to be either main- larger germinal centers, reduced plasma complex that inhibits mTOR. This in- tained on the same regimen or converted cell development, and lower antigen– hibition prevents the translation of to everolimus-based immunosuppres- specific antibody titers, indicating a block mRNA-encoding proteins needed to en- sion. In a retrospective analysis of a subset in plasma cell differentiation.63 Although ter the cell cycle68 (thus reducing T cell of patients, Liefeldt et al.78 reported that interesting, these experimental results proliferation) and cytokine produc- 23% of those receiving everolimus had cannot be directly translated into patients, tion40,69 (Figure 1). Interestingly, developed dnDSAs compared with only

J Am Soc Nephrol 27: ccc–ccc, 2016 Immunosuppression and De Novo DSA 5 BRIEF REVIEW www.jasn.org

11% of those who were receiving CsA. influences the B cell fate. This mechanism which is crucial for the expansion of This difference was, however, only ob- could account for the recent observation the rare antigen–specific clones and served when steroid withdrawal was al- that the frequency and absolute number therefore, the development of humoral lowed. In contrast, the 4-year report of of CD19+CD24hiCD38hi B cells (a subset response. and mycophe- the CONCEPT Trial79 did not find signif- that has previously been associated with nolate mofetil (MPA) both block cell di- icant difference in dnDSA incidence be- operational tolerance43,44) are increased vision by inhibiting DNA synthesis tween patients switched to sirolimus at 3 in patients on compared with through two distinct mechanisms (Fig- months and the CsA controls (12% versus controls on CNIs.90 ure 1). Azathioprine is a prodrug for 21%; P=0.24). In line with these last reas- Most of the clinical data where belata- 6-mercaptopurine; its metabolites block suring results, a case-control study evalu- cept was used come from two multina- purine synthesis enzymes and impede ating the effect of late conversion from tional phase 3 studies: the BENEFIT Trial later steps of DNA synthesis by being in- CNI to everolimus (median of 22 months and the BENEFIT-EXT Trial. The BENEFIT corporated into newly synthetized DNA. post-transplantation) also fails to show Trial studied patients who received a In contrast, MPA only acts as a selective increased dnDSA incidence.80 Finally, kidney from a living donor or a standard inhibitor of inosine-59-monophosphate the question of whether mTOR inhibitors criteria deceased donor. The BENEFIT- dehydrogenase, a key enzyme in the de favor dnDSA emergence is still controver- EXT Trial, however, studied patients who novo synthesis of purine. Therefore, sial. Available data suggest that conversion receivedakidneyfromanextendedcriteria MPA does not incorporate false purine to mTOR inhibitors after complete CNI deceased donor. In both studies, patients analogs into DNA. withdrawal is a potential risk factor, espe- received corticosteroids and basiliximab Adding MPA to primary human B cially if it is made before the end of the first for induction therapy intraoperatively and cells stimulated in vitro with IL-21, anti- year and/or associated with steroid with- MPA and tapering doses of the steroid CD40, and anti-IgM or CpG-B inhibits drawal. postoperatively. Results from these trials B cell proliferation and the generation of indicate that belatacept may have a para- antibody–producing plasma cells.93 Belatacept doxical effect on T versus B cell–mediated Similar findings have been reported in Belatacept is a human fusion protein alloimmune response. The BENEFIT the T/B coculture model.65,72 As expec- combining the extracellular portion of Trial revealed surprisingly low rates (5%– ted, however, the drug has no effect on cytotoxic T lymphocyte–associated anti- 6%)ofdnDSAformationat3years, terminally differentiated plasma cells,93 gen 4 (CTLA4), which has been mutated whereas the incidence of acute T cell– suggesting that it can only prevent (but to confer greater binding avidity to mediated rejection was markedly in- not cure) AMR. CD80 and CD86, and the constant re- creased.91 Schwarz et al.92 performed a Some clinical studies suggest that MPA gion fragment of human IgG1. The retrospective patient match analysis of 14 might perform better than azathioprine CTLA4 binds surface costimulatory patients on belatacept originally enrolled in preventing dnDSA formation. For ligands (CD80 and CD86) of antigen- in the phase 2 multicenter trial. Fifty-six instance, two retrospective studies in kidney presenting cells, thus preventing their patients treated with CsA were matched transplantation5,26 have reported that interaction with CD28 and thereby, according to age at transplantation, first azathioprine is more frequently used blocking T cell activation81 (Figure 1). transplant/retransplant, and donor type. than MPA among patients who develop The efficiency of belatacept in prevent- None of the patients treated with belatacept dnDSAs. However, this difference, which ing dnDSA generation is generally attrib- had DSAs$10 years post-transplantation was not observed in liver transplanta- uted to its effect on Tfh cells.82 B cells are, compared with 38.5% of tested subjects tion,59 could also be explained by the dos- indeed, known to lack CD28, which has treated with CsA (P=0.05).92 Although age (or the nature) of other concomitant expression that is specifically repressed by extremely promising, these results need medications. In a study designed to deter- the B cell master regulator gene Pax-5.83 to be confirmed through additional ex- mine the effect of immunosuppression However, several experimental studies perimentation. It should also be remem- on both naïve and memory humoral have shown the role of reverse signaling bered that part of the effect could be responses, Struijk et al.66 reported that of CD80/CD86 in CTLA4-mediated coin- related to the fact that, in contrast with all recipients of kidneys on prednisone hibition.84 CTLA4-Ig binds to CD80/ other immunosuppressive drugs, belata- combined with either CsA, MPA, or CD86 molecules on dendritic cells and cept is given intravenously monthly in an everolimus all generated significantly promotes the induction of the immuno- outpatient clinic, which could limit fewer antibodies during primary humoral modulatory enzyme indoleamine 2,3 noncompliance, a major risk factor for responses compared with healthy con- dioxygenase85,86 and the tolerogenic mol- dnDSA generation.2,19 trols. In contrast, when patients were ecule HLA-G.87 Experimental data sug- tested for memory responses against the gest that reverse signaling of CD80/ Purine Inhibitors tetanus toxin, only patients on prednisone CD86 also exists in B cells.88,89 It is, there- After signal 1 (BCR engagement) and plusMPAshowedimpairedsecondary fore, tempting to speculate that, when be- signal 2 (provided bycognate Tfh cells), B humoral responses, suggesting that MPA latacept binds to CD80/CD86, it directly cells undergo massive proliferation, could be of special interest in preventing

6 Journal of the American Society of Nephrology J Am Soc Nephrol 27: ccc–ccc,2016 www.jasn.org BRIEF REVIEW the rise of DSA titers in presensitized re- the scope of this review, because DSAs are (Figure 1). In the context of AMR, PI cipients.66 present before drug administration). An- seems to reduce the titers of immunodo- other problem to determine their effect is minant DSAs by 50%, especially in early In Clinical Practice the fact that these drugs were mostly used AMR.102 A similar but transient reduc- A maintenance immunosuppression in combination with plasma exchanges tion rate has also been observed for strategy should be defined according to and/or IVIg.95 preformed anti–HLA antibodies in a the level of the pretransplant sensitiza- Rituximab is a chimeric depleting prospective trial of PI-based desensitiza- tion status. A conventional strategy using antibody directed against the B cell tion.103 Interestingly, Woodle et al.103 CNI plus MPA and avoiding minimiza- surface molecule CD20 that is not found also reported a low rate of dnDSA for- tion in the absence of specific clinical on mature plasma cells (Figure 1). Thus, mation (12.5%) in patients who were indications should be chosen in sensi- rituximab should not affect serum anti- desensitized. tized recipients. In recipients with low body titers directly. However, some immunologic risk, belatacept can now be studies have reported that prolonged B considered as a promising alternative cell depletion with rituximab (in associ- CONCLUSION strategy in association with MPA and ste- ation with IVIg) could lead to a decrease roids. Regarding the other CNI–sparing in DSA titers in patients diagnosed with The prevention of dnDSA generation is strategies, mTOR inhibitors with MPA chronic AMR.96,97 Arecentstudyalso critical to the success of modern renal should not be considered as the first found significantly less HLA antibody transplant practice. The depth of immu- choice in either highly or moderately rebound in patients treated with rituxi- nosuppression together with the nature of sensitized recipients. In recipients with mab compared with a control cohort de- the drugs used play an important role in low immunologic risk, the effect of sensitized and transplanted without the success of the transplant. The data both mTOR inhibitors (with or without rituximab.98 Compared with controls, provided by clinical trials regarding the low-dose CNI) as steroid-sparing strate- patients treated with rituximab had a effect of therapeutic immunosuppression gies has to be clearly defined by addi- significantly greater mean reduction in on the risk of developing dnDSAs are still tional analysis. DSA MFI but a similar rate of DSA per- limited, relatively recent, and considerably Maintenance immunosuppression sistence.98 Some patients sensitized to varied. This because of the rapid evolution should also be adapted to the results of HLA antigens do not have antibody pre- of methods used to detect anti-HLA clinical, pharmacologic, and immuno- sent in serum before transplanta- antibodies as well as the heterogeneity of logic monitoring. DSA monitoring tion.21,97 Such patients are at risk for monitoring and follow-up across centers. should be performed at least once be- an anamnestic response resulting Nevertheless, these data together with the tween 3 and 12 months after transplan- from a proinflammatory response to basic advances in our understanding of B tation. An evaluation of DSAs should also the trauma of transplant . Elim- cellbiology providea platformfromwhich be performed when a change of immu- ination of HLA–specific memory B cells clinicians in charge of patients with trans- nosuppressive regimen is considered and with rituximab in these patients success- plants can now design immunosuppres- when nonadherence or graft rejection is fully abrogated anamnestic response.99 sive strategies tailored to the specific suspected.94 Detection of a significant Regarding prevention of dnDSA gener- immunologic risk of each recipient. level of dnDSAs should prompt verifica- ation with rituximab, a Japanese group tion of drug compliance, and pharmaco- has recently reported that dnDSA in- fi – kinetic analyses should be performed to cidenceissigni cantly lower in ABO DISCLOSURES ensure that drug exposure is within ther- incompatible renal transplantations None. apeutic ranges (in particular, in patients induced with rituximab.100 Incidence with therapeutic minimization). of chronic AMR was also significantly reduced in the group of ABO-incompati- REFERENCES ble recipients who received induction THE EFFECT OF B OR PLASMA therapy with rituximab or splenectomy 1. Einecke G, Sis B, Reeve J, Mengel M, CELL–TARGETED THERAPY ON compared with a control group of ABO- Campbell PM, Hidalgo LG, Kaplan B, DSAS compatible recipients (without rituxi- Halloran PF: Antibody-mediated microcir- culation injury is the major cause of late mab nor splenectomy). However, this – fi kidney transplant failure. Am J Transplant 9: Data on the effect of B or plasma cell bene cial effect of rituximab should be 2520–2531, 2009 targeted therapy on the prevention de carefully interpreted, because it was not 2. Sellarés J, de Freitas DG, Mengel M, Reeve novo humoral allosensitization after kidney observed in a randomized, double–blind J, Einecke G, Sis B, Hidalgo LG, Famulski K, transplantation are scarce. These drugs trial of rituximab induction for ABO– Matas A, Halloran PF: Understanding the 101 causes of kidney transplant failure: The have, indeed, mainly been used in the con- compatible renal transplantations. dominant role of antibody-mediated re- text of desensitization protocols or AMR PIs deplete plasma cells in human jection and nonadherence. Am J Transplant treatment (two situations that are beyond transplant recipients and animal models 12: 388–399, 2012

J Am Soc Nephrol 27: ccc–ccc, 2016 Immunosuppression and De Novo DSA 7 BRIEF REVIEW www.jasn.org

3. Racusen LC, Colvin RB, Solez K, Mihatsch N, Suberbielle C, Frémeaux-Bacchi V, 23. Kosmoliaptsis V, Chaudhry AN, Sharples MJ, Halloran PF, Campbell PM, Cecka MJ, Méjean A, Desgrandchamps F, Anglicheau LD, Halsall DJ, Dafforn TR, Bradley JA, Cosyns J-P, Demetris AJ, Fishbein MC, D, Nochy D, Charron D, Empana J-P, Taylor CJ: Predicting HLA class I alloantigen Fogo A, Furness P, Gibson IW, Glotz D, Delahousse M, Legendre C, Glotz D, Hill immunogenicity from the number and Hayry P, Hunsickern L, Kashgarian M, GS, Zeevi A, Jouven X: Complement- physiochemical properties of amino acid Kerman R, Magil AJ, Montgomery R, binding anti-HLA antibodies and kidney- polymorphisms. Transplantation 88: 791– Morozumi K, Nickeleit V, Randhawa P, allograft survival. NEnglJMed369: 798, 2009 Regele H, Seron D, Seshan S, Sund S, 1215–1226, 2013 24. Kosmoliaptsis V, Sharples LD, Chaudhry Trpkov K: Antibody-mediated rejection 14. Yell M, Muth BL, Kaufman DB, Djamali A, AN, Halsall DJ, Bradley JA, Taylor CJ: Pre- criteria - an addition to the Banff 97 classifi- Ellis TM: C1q binding activity of de novo dicting HLA class II alloantigen immunoge- cation of renal allograft rejection. Am J donor-specific HLA antibodies in renal nicity from the number and physiochemical Transplant 3: 708–714, 2003 transplant recipients with and without anti- properties of amino acid polymorphisms. 4. Terasaki PI, Cai J: body-mediated rejection. Transplantation Transplantation 91: 183–190, 2011 antibodies and chronic rejection: From as- 99: 1151–1155, 2015 25. Fuquay R, Renner B, Kulik L, McCullough sociation to causation. Transplantation 86: 15. Lefaucheur C, Viglietti D, Bentlejewski C, JW, Amura C, Strassheim D, Pelanda R, 377–383, 2008 Duong van Huyen JP, Vernerey D, Aubert Torres R, Thurman JM: Renal ischemia- 5. Terasaki PI, Ozawa M: Predicting kidney O, Verine J, Jouven X, Legendre C, Glotz D, reperfusion injury amplifies the humoral im- graft failure by HLA antibodies: A prospective Loupy A, Zeevi A: IgG donor-specific mune response. J Am Soc Nephrol 24: trial. Am J Transplant 4: 438–443, 2004 anti-human HLA antibody subclasses and 1063–1072, 2013 6. Smith RN, Kawai T, Boskovic S, Nadazdin O, kidney allograft antibody-mediated in- 26. Hourmant M, Cesbron-Gautier A, Terasaki Sachs DH, Cosimi AB, Colvin RB: Four jury. J Am Soc Nephrol 27: 293–304, PI, Mizutani K, Moreau A, Meurette A, stages and lack of stable accommodation in 2016 Dantal J, Giral M, Blancho G, Cantarovich D, chronic alloantibody-mediated renal allo- 16. Hirohashi T, Uehara S, Chase CM, Karam G, Follea G, Soulillou JP, Bignon JD: graft rejection in Cynomolgus monkeys. Am DellaPelle P, Madsen JC, Russell PS, Colvin Frequency and clinical implications of J Transplant 8: 1662–1672, 2008 RB: Complement independent antibody- development of donor-specific and non- 7. Pouliquen E, Koenig A, Chen CC, Sicard A, mediated endarteritis and transplant arte- donor-specific HLA antibodies after kidney Rabeyrin M, Morelon E, Dubois V, Thaunat riopathy in mice. Am J Transplant 10: transplantation. JAmSocNephrol16: O: Recent advances in renal transplantation: 510–517, 2010 2804–2812, 2005 Antibody-mediated rejection takes center 17. Gupta G, Abu Jawdeh BG, Racusen LC, 27. Chemouny J-M, Suberbielle C, Rabant M, stage. F1000Prime Rep 7: 51, 2015 Bhasin B, Arend LJ, Trollinger B, Kraus E, Zuber J, Alyanakian M-A, Lebreton X, 8. Thaunat O, Graff-Dubois S, Fabien N, Rabb H, Zachary AA, Montgomery RA, Carmagnat M, Pinheiro N, Loupy A, Van Duthey A, Attuil-Audenis V, Nicoletti A, Alachkar N: Late antibody-mediated re- Huyen J-P, Timsit M-O, Charron D, Patey N, Morelon E: A stepwise breakdown jection in renal allografts: Outcome after Legendre C, Anglicheau D: De novo donor- of B-cell tolerance occurs within renal allo- conventional and novel therapies. Trans- specific human leukocyte antigen anti- grafts during chronic rejection. Kidney Int plantation 97: 1240–1246, 2014 bodies in nonsensitized kidney transplant 81: 207–219, 2012 18. Everly MJ, Rebellato LM, Haisch CE, Ozawa recipients after T cell-mediated rejection. 9. Li L, Wadia P, Chen R, Kambham N, M, Parker K, Briley KP, Catrou PG, Bolin P, Transplantation 99: 965–972, 2015 Naesens M, Sigdel TK, Miklos DB, Sarwal Kendrick WT, Kendrick SA, Harland RC, 28. Thaunat O: Finding the safe place between MM, Butte AJ: Identifying compartment- Terasaki PI: Incidence and impact of de the hammer and the anvil: Sounding the specific non-HLA targets after renal trans- novo donor-specific alloantibody in primary depth of therapeutic immunosuppression. plantation by integrating transcriptome and renal allografts. Transplantation 95: 410– Kidney Int 88: 1226–1228, 2015 “antibodyome” measures. Proc Natl Acad 417, 2013 29. Thaunat O, Granja AG, Barral P, Filby A, Sci U S A 106: 4148–4153, 2009 19. Wiebe C, Gibson IW, Blydt-Hansen TD, Montaner B, Collinson L, Martinez-Martin 10. Olszewski WL, Engeset A, Lukasiewicz H: Karpinski M, Ho J, Storsley LJ, Goldberg A, N, Harwood NE, Bruckbauer A, Batista FD: Immunoglobulins, complement and lyso- Birk PE, Rush DN, Nickerson PW: Evolution Asymmetric segregation of polarized anti- zyme in leg lymph of normal men. Scand J and clinical pathologic correlations of de gen on B cell division shapes presentation Clin Lab Invest 37: 669–674, 1977 novo donor-specific HLA antibody post capacity. Science 335: 475–479, 2012 11. Sicard A, Ducreux S, Rabeyrin M, Couzi L, kidney transplant. Am J Transplant 12: 30. Harwood NE, Batista FD: Early events in B McGregor B, Badet L, Scoazec JY, Bachelet 1157–1167, 2012 cell activation. Annu Rev Immunol 28: 185– T, Lepreux S, Visentin J, Merville P, 20. Rebellato LM, Everly MJ, Haisch CE, Ozawa 210, 2010 Fremeaux-Bacchi V, Morelon E, Taupin J-L, M, Briley KP, Parker K, Catrou PG, Bolin P, 31. Schnyder T, Castello A, Feest C, Harwood Dubois V, Thaunat O: Detection of C3d- Kendrick WT, Kendrick SA, Harland RC: A NE, Oellerich T, Urlaub H, Engelke M, binding donor-specific anti-HLA antibodies report of the epidemiology of de novo do- Wienands J, Bruckbauer A, Batista FD: B cell at diagnosis of humoral rejection predicts renal nor-specific anti-HLA antibodies (DSA) in receptor-mediated antigen gathering re- graft loss. J Am Soc Nephrol 26: 457–467, 2015 “low-risk” renal transplant recipients. Clin quires ubiquitin ligase Cbl and adaptors Grb2 12. Diebolder CA, Beurskens FJ, de Jong RN, Transplant 2011: 337–340, 2011 and Dok-3 to recruit dynein to the signaling Koning RI, Strumane K, Lindorfer MA, 21. Lúcia M, Luque S, Crespo E, Melilli E, microcluster. 34: 905–918, 2011 Voorhorst M, Ugurlar D, Rosati S, Heck AJR, Cruzado JM, Martorell J, Jarque M, 32. Lanzavecchia A: Antigen-specificinteraction van de Winkel JGJ, Wilson IA, Koster AJ, Gil-Vernet S, Manonelles A, Grinyó JM, between T and B cells. Nature 314: 537–539, Taylor RP, Saphire EO, Burton DR, Bestard O: Preformed circulating HLA- 1985 Schuurman J, Gros P, Parren PWHI: Com- specificmemoryBcellspredicthighriskof 33. Okada T, Miller MJ, Parker I, Krummel MF, plement is activated by IgG hexamers as- humoral rejection in . Neighbors M, Hartley SB, O’Garra A, sembled at the cell surface. Science 343: Kidney Int 88: 874–887, 2015 Cahalan MD, Cyster JG: Antigen-engaged 1260–1263, 2014 22. Duquesnoy RJ: HLA epitope based match- B cells undergo chemotaxis toward the T 13. Loupy A, Lefaucheur C, Vernerey D, ing for transplantation. Transpl Immunol 31: zone and form motile conjugates with Prugger C, Duong van Huyen J-P, Mooney 1–6, 2014 helper T cells. PLoS Biol 3: e150, 2005

8 Journal of the American Society of Nephrology J Am Soc Nephrol 27: ccc–ccc,2016 www.jasn.org BRIEF REVIEW

34. Crotty S: A brief history of T cell help to B 45. Cherukuri A, Salama AD, Carter C, Smalle early B cells from human bone marrow. Exp cells. Nat Rev Immunol 15: 185–189, 2015 N, McCurtin R, Hewitt EW, Hernandez- Biol Med (Maywood) 227: 763–770, 2002 35. Victora GD: SnapShot: The germinal center Fuentes M, Clark B, Baker RJ: An analysis of 55. Haneda M, Owaki M, Kuzuya T, Iwasaki K, reaction. Cell 159: 700–700.e1, 2014 lymphocyte phenotype after steroid avoid- Miwa Y, Kobayashi T: Comparative analysis 36. Cerutti A, Puga I, Cols M: New helping ance with either alemtuzumab or basilix- of drug action on B-cell proliferation and friends for B cells. Eur J Immunol 42: 1956– imab induction in renal transplantation. Am differentiation for , ever- 1968, 2012 J Transplant 12: 919–931, 2012 olimus, and prednisolone. Transplantation 37. Stolp J, Turka LA, Wood KJ: B cells 46. Todeschini M, Cortinovis M, Perico N, Poli 97: 405–412, 2014 with immune-regulating function in trans- F, Innocente A, Cavinato RA, Gotti E, 56. Xiang Z, Cutler AJ, Brownlie RJ, Fairfax K, plantation. Nat Rev Nephrol 10: 389–397, Ruggenenti P, Gaspari F, Noris M, Remuzzi Lawlor KE, Severinson E, Walker EU, Manz 2014 G, Casiraghi F: In kidney transplant patients, RA, Tarlinton DM, Smith KGC: FcgammaRIIb 38. Thaunat O, Morelon E, Defrance T: alemtuzumab but not basiliximab/low-dose controls bone marrow plasma cell persistence Am“B”valent: Anti-CD20 antibodies un- rabbit anti-thymocyte globulin induces B and apoptosis. Nat Immunol 8: 419–429, ravel the dual role of B cells in im- cell depletion and regeneration, which as- 2007 munopathogenesis. Blood 116: 515–521, sociates with a high incidence of de novo 57. Cupps TR, Gerrard TL, Falkoff RJ, Whalen G, 2010 donor-specific anti-HLA antibody develop- Fauci AS: Effects of in vitro corticosteroids 39. Zand MS, Vo T, Huggins J, Felgar R, ment. JImmunol191: 2818–2828, 2013 on B cell activation, proliferation, and dif- Liesveld J, Pellegrin T, Bozorgzadeh A, Sanz 47. Noureldeen T, Albekioni Z, Machado L, ferentiation. JClinInvest75: 754–761, 1985 I, Briggs BJ: Polyclonal rabbit antithymo- Muddana N, Marcus RJ, Hussain SM, 58. Lachmann N, Terasaki PI, Schönemann C: cyte globulin triggers B-cell and plasma cell Sureshkumar KK: Alemtuzumab induction Donor-specific HLA antibodies in chronic apoptosis by multiple pathways. Trans- and antibody-mediated rejection in kidney renal allograft rejection: A prospective trial plantation 79: 1507–1515, 2005 transplantation. Transplant Proc 46: 3405– with a four-year follow-up. Clin Transpl 40. Morelon E, Lefrançois N, Besson C, 3407, 2014 2006: 171–199, 2006 Prévautel J, Brunet M, Touraine J-L, Badet 48. Le Gallou S, Caron G, Delaloy C, Rossille D, 59. Kaneku H, O’Leary JG, Banuelos N, L, Touraine-Moulin F, Thaunat O, Malcus C: Tarte K, Fest T: IL-2 requirement for human Jennings LW, Susskind BM, Klintmalm GB, Preferential increase in memory and regu- plasma cell generation: Coupling differen- Terasaki PI: De novo donor-specificHLA latory subsets during T-lymphocyte im- tiation and proliferation by enhancing antibodies decrease patient and graft sur- mune reconstitution after Thymoglobulin MAPK-ERK signaling. JImmunol189: 161– vival in liver transplant recipients. Am J induction therapy with maintenance siroli- 173, 2012 Transplant 13: 1541–1548, 2013 mus vs cyclosporine. Transpl Immunol 23: 49. Thibaudin D, Alamartine E, de Filippis JP, 60. Del Bello A, Congy-Jolivet N, Muscari F, 53–58, 2010 Diab N, Laurent B, Berthoux F: Advantage Lavayssière L, Esposito L, Cardeau- 41. Lee F, Luevano M, Veys P, Yong K, Madrigal of antithymocyte globulin induction in Desangles I, Guitard J, Dörr G, Suc B, Duffas A, Shaw BE, Saudemont A: The effects of sensitized kidney recipients: A randomized JP, Alric L, Bureau C, Danjoux M, CAMPATH-1H on cell viability do not cor- prospective study comparing induction Guilbeau-Frugier C, Blancher A, Rostaing L, relate to the CD52 density on the cell sur- with and without antithymocyte globulin. Kamar N: Prevalence, incidence and risk factors face. PLoS One 9: e103254, 2014 Nephrol Dial Transplant 13: 711–715, 1998 for donor-specific anti-HLA antibodies in 42. Heidt S, Hester J, Shankar S, Friend PJ, 50. Noël C, Abramowicz D, Durand D, Mourad maintenance liver transplant patients. Am J Wood KJ: B cell repopulation after alemtu- G, Lang P, Kessler M, Charpentier B, Transplant 14: 867–875, 2014 zumab induction-transient increase in tran- Touchard G, Berthoux F, Merville P, Ouali 61. Delgado JC, Fuller A, Ozawa M, Smith L, sitional B cells and long-term dominance of N, Squifflet J-P, Bayle F, Wissing KM, Terasaki PI, Shihab FS, Eckels DD: No oc- naïve B cells. Am J Transplant 12: 1784– Hazzan M: versus antithymo- currence of de novo HLA antibodies in pa- 1792, 2012 cyte globulin in high-immunological-risk tients with early corticosteroid withdrawal 43. Sagoo P, Perucha E, Sawitzki B, Tomiuk S, renal transplant recipients. JAmSoc in a 5-year prospective randomized study. Stephens DA, Miqueu P, Chapman S, Nephrol 20: 1385–1392, 2009 Transplantation 87: 546–548, 2009 Craciun L, Sergeant R, Brouard S, Rovis F, 51. Brokhof MM, Sollinger HW, Hager DR, 62. Anil Kumar MS, Irfan Saeed M, Ranganna K, Jimenez E, Ballow A, Giral M, Rebollo-Mesa Muth BL, Pirsch JD, Fernandez LA, Malat G, Sustento-Reodica N, Kumar I, Le Moine A, Braudeau C, Hilton R, Bellingham JM, Mezrich JD, Foley DP, AMS, Meyers WC: Comparison of four differ- Gerstmayer B, Bourcier K, Sharif A, D’Alessandro AM, Odorico JS, Mohamed ent immunosuppression protocols without Krajewska M, Lord GM, Roberts I, Goldman MA, Vidyasagar V, Ellis TM, Kaufman DB, long-term steroid therapy in kidney recipients M, Wood KJ, Newell K, Seyfert-Margolis V, Djamali A: Antithymocyte globulin is asso- monitored by surveillance biopsy: Five-year Warrens AN, Janssen U, Volk H-D, Soulillou ciated with a lower incidence of de novo outcomes. Transpl Immunol 20: 32–42, 2008 J-P, Hernandez-Fuentes MP, Lechler RI: donor-specific antibodies in moderately 63. Winslow MM, Gallo EM, Neilson JR, Development of a cross-platform biomarker sensitized renal transplant recipients. Trans- Crabtree GR: The calcineurin phosphatase signature to detect renal transplant toler- plantation 97: 612–617, 2014 complex modulates immunogenic B cell ance in humans. J Clin Invest 120: 1848– 52. Van Laethem F, Baus E, Smyth LA, Andris F, responses. Immunity 24: 141–152, 2006 1861, 2010 BexF,UrbainJ,KioussisD,LeoO: 64. Jørgensen KA, Koefoed-Nielsen PB, 44. Pallier A, Hillion S, Danger R, Giral M, attenuate T cell receptor Karamperis N: Calcineurin phosphatase ac- Racapé M, Degauque N, Dugast E, signaling. J Exp Med 193: 803–814, 2001 tivity and immunosuppression. A review Ashton-Chess J, Pettré S, Lozano JJ, 53. Löwenberg M, Tuynman J, Bilderbeek J, on the role of calcineurin phosphatase ac- Bataille R, Devys A, Cesbron-Gautier A, Gaber T, Buttgereit F, van Deventer S, tivity and the immunosuppressive effect of Braudeau C, Larrose C, Soulillou J-P, Peppelenbosch M, Hommes D: Rapid im- cyclosporin A and tacrolimus. Scand J Im- Brouard S: Patients with drug-free long- munosuppressive effects of glucocorticoids munol 57: 93–98, 2003 term graft function display increased mediated through Lck and Fyn. Blood 106: 65. Heidt S, Roelen DL, Eijsink C, Eikmans M, numbers of peripheral B cells with a 1703–1710, 2005 van Kooten C, Claas FHJ, Mulder A: Cal- memory and inhibitory phenotype. Kid- 54. Lill-Elghanian D, Schwartz K, King L, Fraker cineurin inhibitors affect B cell antibody ney Int 78: 503–513, 2010 P: -induced apoptosis in responses indirectly by interfering with T

J Am Soc Nephrol 27: ccc–ccc, 2016 Immunosuppression and De Novo DSA 9 BRIEF REVIEW www.jasn.org

cell help. Clin Exp Immunol 159: 199–207, 76. Weir MR, Mulgaonkar S, Chan L, Shidban H, 85. Grohmann U, Orabona C, Fallarino F, Vacca 2010 Waid TH, Preston D, Kalil RN, Pearson C, Calcinaro F, Falorni A, Candeloro P, 66. Struijk GH, Minnee RC, Koch SD, TC: Mycophenolate mofetil-based im- Belladonna ML, Bianchi R, Fioretti MC, Zwinderman AH, van Donselaar-van der munosuppression with sirolimus in renal Puccetti P: CTLA-4-Ig regulates tryptophan Pant KAMI, Idu MM, ten Berge IJM, transplantation: A randomized, controlled catabolism in vivo. Nat Immunol 3: 1097– Bemelman FJ: Maintenance immunosup- Spare-the-Nephron trial. Kidney Int 79: 897– 1101, 2002 pressive therapy with everolimus preserves 907, 2011 86. Munn DH, Sharma MD, Mellor AL: Ligation humoral immune responses. Kidney Int 78: 77. Guba M, Pratschke J, Hugo C, Krämer BK, of B7-1/B7-2 by human CD4+ T cells trig- 934–940, 2010 Nohr-Westphal C, Brockmann J, Andrassy gers indoleamine 2,3-dioxygenase activity 67. Huang Y, Ramon D, Luan FL, Sung R, J, Reinke P, Pressmar K, Hakenberg O, in dendritic cells. J Immunol 172: 4100– Samaniego M: Incidences of preformed and Fischereder M, Pascher A, Illner W-D, Banas 4110, 2004 de novo donor-specific HLA antibodies and B, Jauch K-W; SMART-Study Group: Renal 87. Bahri R, Naji A, Menier C, Charpentier B, their clinicohistological correlates in the function, efficacy, and safety of sirolimus Carosella ED, Rouas-Freiss N, Durrbach A: early course of kidney transplantation. Clin and mycophenolate mofetil after short-term Dendritic cells secrete the immunosup- Transpl 2012: 247–256, 2012 calcineurin inhibitor-based quadruple ther- pressive HLA-G molecule upon CTLA4-Ig 68. Kahan BD, Chang JY, Sehgal SN: Preclinical apy in de novo renal transplant patients: treatment: Implication in human renal evaluation of a new potent immunosup- One-year analysis of a randomized multi- transplant acceptance. JImmunol183: pressive agent, rapamycin. Transplantation center trial. Transplantation 90: 175–183, 7054–7062, 2009 52: 185–191, 1991 2010 88. Rau FC, Dieter J, Luo Z, Priest SO, 69. Zhang S, Readinger JA, DuBois W, Janka- 78. Liefeldt L, Brakemeier S, Glander P, Waiser Baumgarth N: B7-1/2 (CD80/CD86) direct Junttila M, Robinson R, Pruitt M, Bliskovsky J, Lachmann N, Schönemann C, Zukunft B, signaling to B cells enhances IgG secretion. V, Wu JZ, Sakakibara K, Patel J, Parent CA, Illigens P, Schmidt D, Wu K, Rudolph B, J Immunol 183: 7661–7671, 2009 Tessarollo L, Schwartzberg PL, Mock BA: Neumayer H-H, Budde K: Donor-specific 89. Kin NW, Sanders VM: CD86 regulates IgG1 Constitutive reductions in mTOR alter cell HLA antibodies in a cohort comparing ev- production via a CD19-dependent mecha- size, immune cell development, and anti- erolimus with cyclosporine after kidney nism. J Immunol 179: 1516–1523, 2007 body production. Blood 117: 1228–1238, transplantation. Am J Transplant 12: 1192– 90. Leibler C, Matignon M, Pilon C, Montespan 2011 1198, 2012 F, Bigot J, Lang P, Carosella ED, Cohen J, 70. Limon JJ, Fruman DA: Akt and mTOR in B 79. Lebranchu Y, Thierry A, Thervet E, Büchler Rouas-Freiss N, Grimbert P, Menier C: cell activation and differentiation. Front M, Etienne I, Westeel PF, Hurault de Ligny Kidney transplant recipients treated with Immunol 3: 228, 2012 B, Moulin B, Rérolle JP, Frouget T, Girardot- belatacept exhibit increased naïve and 71. Zhang S, Pruitt M, Tran D, Du Bois W, Zhang Seguin S, Toupance O: Efficacy and safety transitional B cells. Am J Transplant 14: K, Patel R, Hoover S, Simpson RM, Simmons of early cyclosporine conversion to siroli- 1173–1182, 2014 J, Gary J, Snapper CM, Casellas R, Mock mus with continued MMF-four-year results 91. Vincenti F, Tedesco Silva H, Busque S, BA: B cell-specificdeficiencies in mTOR of the Postconcept study. Am J Transplant O’Connell P, Friedewald J, Cibrik D, Budde limit humoral immune responses. JIm- 11: 1665–1675, 2011 K,YoshidaA,CohneyS,WeimarW,Kim munol 191: 1692–1703, 2013 80. Kamar N, Del Bello A, Congy-Jolivet N, YS,LawendyN,LanS-P,KudlaczE, 72. Matz M, Lehnert M, Lorkowski C, Fabritius Guilbeau-Frugier C, Cardeau-Desangles I, Krishnaswami S, Chan G: Randomized K, Unterwalder N, Doueiri S, Weber UA, Fort M, Esposito L, Guitard J, Gamé X, phase 2b trial of (CP-690,550) in Mashreghi M-F, Neumayer H-H, Budde K: Rostaing L: Incidence of donor-specific an- de novo kidney transplant patients: Efficacy, Effects of sotrastaurin, mycophenolic acid tibodies in kidney transplant patients fol- renal function and safety at 1 year. Am J and everolimus on human B-lymphocyte lowing conversion to an everolimus-based Transplant 12: 2446–2456, 2012 function and activation. Transpl Int 25: calcineurin inhibitor-free regimen. Clin 92. Schwarz C, Mayerhoffer S, Berlakovich GA, 1106–1116, 2012 Transplant 27: 455–462, 2013 Steininger R, Soliman T, Watschinger B, 73. Traitanon O, Mathew JM, La Monica G, Xu 81. Vincenti F, Larsen C, Durrbach A, Wekerle T, Böhmig GA, Eskandary F, König F, L, Mas V, Gallon L: Differential effects of Nashan B, Blancho G, Lang P, Grinyo J, Mühlbacher F, Wekerle T: Long-term out- tacrolimus versus sirolimus on the pro- Halloran PF, Solez K, Hagerty D, Levy E, come of belatacept therapy in de novo liferation, activation and differentiation of Zhou W, Natarajan K, Charpentier B; kidney transplant recipients - a case-match human B cells. PLoS One 10: e0129658, Belatacept Study Group: Costimulation analysis. Transpl Int 28: 820–827, 2015 2015 blockade with belatacept in renal trans- 93. Karnell JL, Karnell FG 3rd, Stephens GL, 74. Budde K, Becker T, Arns W, Sommerer C, plantation. NEnglJMed353: 770–781, Rajan B, Morehouse C, Li Y, Swerdlow B, Reinke P, Eisenberger U, Kramer S, Fischer 2005 Wilson M, Goldbach-Mansky R, Groves C, W, Gschaidmeier H, Pietruck F; ZEUS 82. Kim EJ, Kwun J, Gibby AC, Hong JJ, Farris Coyle AJ, Herbst R, Ettinger R: Mycophe- Study Investigators: Everolimus-based, AB 3rd, Iwakoshi NN, Villinger F, Kirk AD, nolic acid differentially impacts B cell calcineurin-inhibitor-free regimen in recip- Knechtle SJ: Costimulation blockade alters function depending on the stage of dif- ients of de-novo kidney transplants: An germinal center responses and prevents ferentiation. JImmunol187: 3603–3612, open-label, randomised, controlled trial. antibody-mediated rejection. Am J Trans- 2011 Lancet 377: 837–847, 2011 plant 14: 59–69, 2014 94. Tait BD, Süsal C, Gebel HM, Nickerson PW, 75. Lebranchu Y, Thierry A, Toupance O, 83. Delogu A, Schebesta A, Sun Q, Zachary AA, Claas FHJ, Reed EF, Bray RA, Westeel PF, Etienne I, Thervet E, Moulin B, Aschenbrenner K, Perlot T, Busslinger M: Campbell P, Chapman JR, Coates PT, Frouget T, Le Meur Y, Glotz D, Heng A-E, Gene repression by Pax5 in B cells is es- Colvin RB, Cozzi E, Doxiadis IIN, Fuggle SV, Onno C, Buchler M, Girardot-Seguin S, sential for blood cell homeostasis and is Gill J, Glotz D, Lachmann N, Mohanakumar Hurault de Ligny B: Efficacy on renal func- reversed in plasma cells. Immunity 24: 269– T, Suciu-Foca N, Sumitran-Holgersson S, tion of early conversion from cyclosporine 281, 2006 Tanabe K, Taylor CJ, Tyan DB, Webster A, to sirolimus 3 months after renal trans- 84. Chen L: Co-inhibitory molecules of the B7- Zeevi A, Opelz G: Consensus guidelines plantation: Concept study. Am J Transplant CD28 family in the control of T-cell immu- on the testing and clinical management 9: 1115–1123, 2009 nity. Nat Rev Immunol 4: 336–347, 2004 issues associated with HLA and non-HLA

10 Journal of the American Society of Nephrology J Am Soc Nephrol 27: ccc–ccc,2016 www.jasn.org BRIEF REVIEW

antibodies in transplantation. Trans- 98. Jackson AM, Kraus ES, Orandi BJ, Segev 101. Tydén G, Ekberg H, Tufveson G, Mjörnstedt plantation 95: 19–47, 2013 DL,MontgomeryRA,ZacharyAA:Acloser L: A randomized, double-blind, placebo- 95. Vo AA, Lukovsky M, Toyoda M, Wang J, look at rituximab induction on HLA anti- controlled study of single dose rituximab as Reinsmoen NL, Lai C-H, Peng A, Villicana R, body rebound following HLA-incompatible induction in renal transplantation: A 3-year Jordan SC: Rituximab and intravenous im- kidney transplantation. Kidney Int 87: 409– follow-up. Transplantation 94: e21–e22, 2012 mune globulin for desensitization during 416, 2015 102. Walsh RC, Alloway RR, Girnita AL, Woodle renal transplantation. NEnglJMed359: 99. Zachary AA, Lucas DP, Montgomery RA, ES: Proteasome inhibitor-based therapy for 242–251, 2008 Leffell MS: Rituximab prevents an anam- antibody-mediated rejection. Kidney Int 96. Clatworthy MR: B-cell regulation and its nestic response in patients with cryptic 81: 1067–1074, 2012 application to transplantation. Transpl Int sensitization to HLA. Transplantation 95: 103. Woodle ES, Shields AR, Ejaz NS, Sadaka B, 27: 117–128, 2014 701–704, 2013 Girnita A, Walsh RC, Alloway RR, Brailey P, 97. Fehr T, Rüsi B, Fischer A, Hopfer H, 100. Kohei N, Hirai T, Omoto K, Ishida H, Tanabe Cardi MA, Abu Jawdeh BG, Roy-Chaudhury Wüthrich RP, Gaspert A: Rituximab and in- K: Chronic antibody-mediated rejection is P, Govil A, Mogilishetty G: Prospective it- travenous immunoglobulin treatment of reduced by targeting B-cell immunity dur- erative trial of proteasome inhibitor-based chronic antibody-mediated kidney allograft ing an introductory period. Am J Transplant desensitization. Am J Transplant 15: 101– rejection. Transplantation 87: 1837–1841, 2009 12: 469–476, 2012 118, 2015

J Am Soc Nephrol 27: ccc–ccc, 2016 Immunosuppression and De Novo DSA 11