<<

BRIEF REVIEW www.jasn.org

Targeting B Cells and Plasma Cells in Glomerular Diseases: Translational Perspectives

Eva Schrezenmeier,1 David Jayne,2 and Thomas Dörner3

1Divisions of Nephrology and Intensive Care, and 3Rheumatology and Clinical , Department of Medicine, Charité Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany; and 2Department of Medicine, University of Cambridge, Cambridge, United Kingdom

ABSTRACT The unique contributions of memory B cells and plasma cells in kidney diseases idiopathic membranous nephropathy remain unclear. In this review, we evaluate the clinical experience with treatments (IMN). In contrast, SLE did not show directed at B cells, such as , and at plasma cells, such as in- similarly convincing responses to CD20 hibition, to shed on the role of these two B lineage compartments in glomerular targeting. In chronic -mediated diseases. Specifically, analysis of these targeted interventions in diseases such as rejection (ABMR), the addition of PC ANCA-associated vasculitis, SLE, and antibody-mediated per- targeting agents (e.g., the proteasome in- mits insight into the pathogenetic effect of these cells. Notwithstanding the limita- hibitor bortezomib) appears to be bene- tions of preclinical models and clinical studies (heterogeneous populations, among ficial, with less evidence for rituximab.4 others), the data suggest that memory B and plasma cells represent two engines of Currently available data from more se- , with variable involvement in these diseases. Whereas memory B cells lective immune targeting suggests that and plasma cells appear to be key in ANCA-associated vasculitis and antibody- the pathogenic relevance of memory B mediated transplant rejection, respectively, SLE seems likely to be driven by both cells and PCs may vary between autoim- autoimmune compartments. These conclusions have implications for the future de- mune diseases (reviewed recently5), im- velopment of targeted therapeutics in immune-mediated renal disease. proving our understanding of individual diseases. J Am Soc Nephrol 29: 741–758, 2018. doi: https://doi.org/10.1681/ASN.2017040367 Here we will take a reverse translational perspective to learn from the clinical use of Bcell–directed therapies such as anti- CD20 or therapies that target the PC com- In health, memory B cells and plasma recirculate upon re-exposure. partment in renal autoimmunity. cells (PCs) comprise important but Overall, memory B cells and PCs form independently regulated compartments two immune defense lines that allow pres- 1 of our immunologic memory (Figure 1). ervation of previous immune encounters INDUCTION OF MEMORY B CELLS Humoral is key to the patho- by produced by long-lived PCs AND PCS genesis of many autoimmune renal dis- and a dynamic component to adapt hu- eases. Here B cells, when recognizing moral immunity by memory B cells. The Distinct PC subsets can be induced via (auto) and receiving appropriate two cellular subsets ensure stability and different pathways (Figure 1). First, B fl fi help, can differentiate into short- exibility to maintain a suf cient cells from the B1 cell lineage, which lived PCs, memory B or long-lived PCs lineage defense. The extent to which these have been mainly studied in mice and also termed memory PCs2. However, their “two engines” of B cell lineage memory detailed contributions to different kidney contribute to different renal autoimmune diseases are not known. diseases is not fully understood, but is Published online ahead of print. Publication date Long-lived PCs are considered ulti- likely to differ between disorders. available at www.jasn.org. mately differentiated, marrow In this context, clinical experiences Correspondence: Dr. Thomas Dörner, Department (BM) resident cells secreting high-affinity with therapeutics selectively targeting of Medicine/Rheumatology and Clinical Immunol- + 2 ogy, Charité Universitätsmedizin Berlin, Campus antibodies that disseminate through the CD20 memory B cells but not CD20 Mitte, Berlin, Germany. Email: thomas.doerner@ body, whereas memory B cells3 can rap- PCs provided interesting lessons (Figure charite.de idly proliferate in a clonally and antigen 2A), e.g., the response to rituximab in Copyright © 2018 by the American Society of specific manner, and differentiate and ANCA-associated vasculitis (AAV) and Nephrology

J Am Soc Nephrol 29: 741–758, 2018 ISSN : 1046-6673/2903-741 741 BRIEF REVIEW www.jasn.org

Figure 1. Distinct developmental and differentiation pathways of normal B cells. B1 and B2 B cell lineages appear to be independently regulated and undergo tightly controlled differentiation into certain memory B and PC subsets (Adapted from Dörner T et al.187

lack a defined phenotype in humans, switch, first producing memory B cells Most but not all memory B cells have can form short-lived PCs, which pro- and subsequently long-lived PCs13 with undergone class-switch recombination, duce polyreactive IgM for immediate increasing affinity. It has long been sug- typically from IgM to IgG, and carry so- defense. Second, B cells from the B2 gested, but now experimentally proven, matically hypermutated IgV re- cell lineage can form short-lived PCs in that memory B cells can also develop in a arrangements16,17 as one signature of aTcell–independent manner (so-called T cell–dependent, but – previous T cell encounters. Loss of IgD 2 B2 cells), for example by independent pathway.14 Bcelllineage expression (IgD )identifies B cells that stimulation with T cell-independent an- differentiation paths are summarized in have undergone class-switch recombi- tigens such as pneumococcal capsular Figure 1. The bulk of the data reported nation. Switched memory B cells de- polysaccharides, and predominantly se- above has been obtained from preclinical velop during T cell–dependent germinal crete low-affinity IgM antibodies.6,7 models, although it remains to be delin- center responses.13 In addition to Ig sub- Moreover, B2 lineage cells are the main eated which differentiation pathway(s) class B cell (BCR) expression, source of long-lived PCs and memory B and B cell lineages are involved in certain other phenotypic B memory markers cells upon activation by cognate T cells autoimmune conditions. have been identified. Here, expression in germinal centers.8 The generation of of CD27 serves as a universal marker long-lived PCs is the result of a two-step for memory B cells in healthy donors. process. First, an extrafollicular response MEMORY B CELLS The BCR of memory B cells shows a leads to the generation of short-lived ac- high affinity to the immunizing antigen tivated B cells, of which some re-enter the The definition of a com- compared with naïve B cells caused by B cell follicle and become plasmablasts prises an antigen-experienced, nonpro- using affinity-matured and somatically in a T cell–dependent pathway. Subse- liferating and, in the absence of antigen, hypermutated BCR .18 Memory B quently, plasmablasts migrate through persisting cell15 that responds more rap- cells that emerge from the T cell–dependent the stream and reside as long-lived idly and efficiently when re-exposed to but germinal center–independent PCs primarily in BM niches, possibly also antigen. In contrast to sessile PCs, mem- pathway do not show somatic hypermu- in inflamed tissues.9,10 It is currently de- ory B cells recirculate and scan the body tations, and thus are not affinity ma- bated whether a small number of BM continuously. Their phenotype does not tured.19 After a given , memory PCs can be induced indepen- differ between certain lymphoid organs, memory B cells must repress their acti- dently of T cells.11,12 The germinal center and they do not depend on the presence vation program and rest in a quiescent response is a time-regulated developmental of the or .3 state for a long period to maintain their

742 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW

Figure 2. Interventions and their potential to target distinctly B lineage subsets and plasma cells. (A) Principles of direct (anti-CD20, anti- CD22) and indirect targeting of B cells and PCs (anti-BAFF or anti-APRIL strategies) have preferential effects on naïve versus memory B cells and PCs. (B) Principle of unspecific B cell and PC targeting, i.e., by proteasome inhibition or autologous transplantation (ASCT) with or without antithymocyte globulin (ATG) as well as mycophenolate mofetil (MMF) or cyclophosphamide. There appears to be a distinct susceptibility of memory B cell and PC dependent on the pharmacologic mechanisms. memory capacity.20 Upon antigen reacti- is not clear if they result from incomplete high-affinity antibodies and reside preferen- vation, memory B cells become activated germinal center responses, increased extra- tially inthe BM. Onlyafew plasmablasts that and can differentiate either into PCs or follicular activity and/or enhanced T cell– arise from the germinal center response mi- further mature by re-entering the germi- independent B cell activation. Usually, these grate through the blood stream to become nal center for additional affinity matura- cells carry characteristics of B cell memory memory PCs.9 Crucialforthesurvivalof tion.21 The involvement of memory B (e.g., mutated IgV genes), expressed costi- a plasmablast to become an ultimately dif- cells during resolution of an immune re- mulatory molecules, or lacked an active ferentiated, long-lived PC is that they find sponse and the mechanisms by which res- ABCB1 transporter as functional character- appropriate soluble and insoluble (niche) ident memory B cells are prevented from istic of memory B cells. It is currently not survival conditions, whereas no postgerminal uncontrolled activation remain unclear clear if these abnormalities of B cell memory center selection processes of PCs are known. for immunity as well as autoimmunity. are a source of autoimmunity or result of Long-lived PCs, also called memory PCs, Notably, disturbances of circulating overly active immune activation. find optimal survival conditions in the memory B cells in patients with chronic The role of T cell–dependent memory BM in healthy individuals at the end of a immune activation have been reported in B cell induction in SLE, AAV, and ABMR competitive journey starting as an early B chronic autoimmune conditions,22,23 has been concluded from molecular data lineage cell. such as SLE,22,24 of Ig switched, hypermutated autoanti- As direct PC precursors, plasmablasts (RA)25 or in HIV infection26 compared bodies characteristic of these diseases,30–32 express the C-X-C receptor with healthy controls. In this context, and histologic data of germinal center-like type 4 (CXCR4)35 and downregulate 2 2 enhanced CD27 IgD B cells as well as structures in affected tissues.33,34 CXCR5 and CXCR7 when they start to 2 2 CD27 IgD coexpressing CD95+,27 migrate toward the BM. The BM survival CD21low28 or intracellular spleen tyro- niches are composed of stromal cells ex- sine kinase Sykhigh29 were found. There PCS pressing C-X-C motif chemokine ligand is a possibility that these abnormalities re- 12 (CXCL12) (ligand of CXCR4) and vas- sult from an overly active PCs are responsible for maintaining serum cular cell adhesion 1 (VCAM1).36 with enhanced B cell differentiation, but it antibody levels by continuously producing Other cell types such as megakaryocytes,

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 743 BRIEF REVIEW www.jasn.org , and contribute to and relevant autoantibodies50–52 provide nondividing PC memory is not affected by the production of survival factors such the basis that activation of B cells has taken therapies dependent on proliferation.56 as a proliferation-inducing ligand (APRIL), place. However, the particular role of B B cell-activating factor (BAFF or BLyS), or cells independent of PC producing auto- IL-6, as well as adhesion molecules.37–39 antibodies has only been clearly demon- CORTICOSTEROIDS The number of available niches in the strated in a model.52,53 Tables 1 and BM or its capacity appears to be limited 2 summarize key features, advantages, and Corticosteroids interrupt multiple steps of and serves as a regulatory factor for serum disadvantages of the most instructive an- the immune response with genomic and Ig levels. Recently, it has been debated imal models in which B cell and PC con- nongenomiceffects,but are mainly related if there is a physical or rather cell type– tributions have been studied within AAV, to the inhibition of transcription dependent functional BM niche.40 lupus nephritis, and ABMR. via blocking transcription factors. This Besides the BM, other tissues can also leads to the inhibition of certain ILs, offer survival niches in states of chronic thereby depleting T cells (IL-2) and eosin- inflammation. These niches disappear TARGETING MEMORY B CELLS ophils (IL-5), interrupting when the inflammation is resolved.41–43 VERSUS TARGETING MEMORY function (IL-1, TNF-a), and increasing Once it was thought that survival niches PCS IN HUMAN DISEASE release from the BM and neu- harbor almost exclusively highly-affinity trophil migration to the sites of inflamma- matured PCs, but other studies revealed Because expression of certain surface tion.57 Less is known about the detailed that immature PCs can access survival markers on memory B cells and PCs are effect of corticosteroids on B cells. It has niches.44,45 Recent data suggest that distinct, selective targeting by using spe- been suggested that B cells are not signif- there is heterogeneity of PCs in human cific agents is a clinical possibility. Mem- icantly inhibited by steroids and Ig levels BM. Here, a stable and highly differenti- ory B cells show the phenotype of mature are only slightly decreased.58 However, 2 ated CD19 PC fraction with a distinct B cells expressing CD19, CD20, and CD22 high-dose affects PCs phenotype (CD56+,HLA-DRnegative, besides the above discussed surface mark- and has been widely used in myeloma, es- CD44negative etc.)andalessmature ers. PCs differ on the basis of their larger pecially dexamethasone. Recent data from CD19+ PC fraction exist in human sizeandgranularity,aswellaslossofCD20 children with steroid-responsive ne- BM, which suggests that further differen- and CD22 expression, downregulation of phrotic syndrome show that B cell num- tiation even within the BM PCs is a pos- CD19, and expression of CD38 and, to bers decline under corticosteroid therapy sibility.46 These human PC subsets in the large extent, CD138.8 Memory B cells ex- and that especially peripheral memory B BM have been confirmed subse- press three B cell cytokine receptors, cells further decline after corticosteroid 2 quently.47,48 Because CD19 PCs usually BAFF receptor (BAFF-R), transmem- cessations.59 In patients with autoim- enriched in human BM were also found brane activator and CAML interactor mune hemolytic anemia, splenic germinal in autoimmune tissues including kidney (TACI), and B cell maturation antigen center B cells and circulating plasmablasts transplant rejection,46 it needs to be fur- (BCMA), whereas PCs have very dimin- were largely suppressed in patients under ther delineated what factors are involved ished BAFF-R and TACI expression but long-term corticosteroid therapy.60 in their induction and/or maintenance in receive survival signals through BCMA. autoimmunity. Notably, a potential role There is differential signaling of BLyS/ of T cells within the BM has been sugges- BAFF and APRIL through these recep- ANTI-CD20 MONOCLONAL ted recently. Here, regulatory T cells tors. We will briefly summarize certain ANTIBODIES (Treg) cells share a particular niche with agents that have an effect on PCs or PCs in CD11c+ BM cells and appear to (memory) B cells. Anti-CD20 monoclonal antibodies, such as regulate PC survival via CTLA-4.49 This the type 1 antibodies rituximab, ofatumu- suggeststhatTcellshavedistinctbut mab, and , largely deplete pe- largely unknown functions determining CONVENTIONAL ripheral B cells including memory B cells the lifestyle of PCs and mandates further IMMUNOSUPPRESSANTS from the peripheral blood but not always in studies. the BM,61 nodes,62 or synovium.63 2 Although a number of agents have an effect CD20 pro-B cells and PCs are not targe- on B cell subsets, nonproliferating memory ted by these antibodies (Figure 2A), but ANIMAL MODELS Bcellshavebeenshowntobemoreresistant they lead to impaired generation of plas- to conventional immunosuppressants mablasts and short-lived PCs. These Different animal models for SLE, the two such as cyclophosphamide,54 as well as to monoclonals act by several mechanisms, AAV conditions (myeloperoxidase mycophenolate mofetil55 (Figure 2B). Fur- including antibody-dependent cellular cy- [MPO] or proteinase 3), and ABMR ther, generation of plasmablasts and short- totoxicity, complement-dependent cyto- could provide evidence for the role of lived PCs are sufficiently inhibited by toxicity, and apoptosis induction.64 Novel B lineage cells. Here analyses of T cells conventional immunosuppressants, and type 2 monoclonal anti-CD20 antibodies,

744 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 mScNephrol Soc Am J

29: 2 Table 1. Indicators of the presumed pathogenic role of CD20+ B cells and CD20 PCs on the basis of available data of therapeutic responses to different B lineage 741 cell directed agents – 5,21 el n lsaClsi lmrlrDiseases Glomerular in Cells Plasma and Cells B 2018 758, Glomerular Disease AAV SLE ABMR Response to... Anti-CD20 (rituximab) Experience from RCTs leading to approval Experience from two RCTs and Experience from one RCT and several and several observational studies. several observational studies. cohort studies. Effective in remission induction in and RCTs failed to reach primary end points No difference in RCT of acute ABMR.189 noninferior to cyclophosphamide.106,107 in renal140 and nonrenal SLE.188 Superior to in maintenance Meta-analysis of published data and EULAR Partial success in retrospective of remission.108 and ERA-EDTA recommendations warrant cohort studies of acute ABMR.171 the use of rituximab.142,144 Meta-analysis of uncontrolled studies support No effect of rituximab in chronic ABMR.171 data from RCTs. Rituximab might be superior in relapsing disease.190 Proteasome inhibition Very limited experience. Limited experience from small case series. Limited experience from small cohort studies. (bortezomib) One patient with AAV refractory to rituximab Report of five patients with lupus nephritis, Bortezomib is able to reduce DSA successfully treated with bortezomib.119 three had complete remission after 6–12 mo.156 in most patients but graft survival remains very poor.181 Report of 12 patients with SLE, six with lupus nephritis. All patients responded to therapy.155 Pathogenic relevance of ... (Memory) B cells Regulatory B cells, but not memory B cells Antibody-independent mechanisms Memory B cells can be reactivated in predict relapse after rituximab therapy.104,105 e.g., autoantigen secondary transplantation upon , costimulation of T cells, re-exposure.191 production of regulatory .192 Reconstitution of memory B cell subset can predict disease flares after rituximab therapy.136,137 Long-lived PCs/ Materno-fetal transfer of anti-MPO antibodies Involved in the formation of immune complexes Responsible for the maintenance of serum led to vasculitis with pulmonary renal syndrome1 that lead to nephritis.192 DSA that cause transplant glomerulopathy.164 www.jasn.org 12 in neonates and can cause murine necrotizing crescentic GN.193 Pathogenicity of anti-PR3 antibodies is not entirely proven.194

RCT, randomized, controlled trial; EULAR, European League Against Rheumatism; ERA-EDTA, European Renal Association/European Dialysis and Transplant Association. REVIEW BRIEF 745 746 RE REVIEW BRIEF ora fteAeia oit fNephrology of Society American the of Journal

Table 2. Summary of the most relevant animal models of AAV, lupus nephritis, and ABMR with regard to B cell and PC function www.jasn.org Animal Model Kind of Intervention Key Feature Advantages and Disadvantages Reference AAV 2 2 Mpo / mice/ Transfer of anti-MPO antibodies or Induces necrotizing pauci-immune Mainly B cell but not Xiao et al.193 2 2 (Rag2 / ) mice splenocytes, generated by MPO GN and, in some cases, Tcell–dependent, of MPO deficient mice. pulmonary capillaritis. Transfer of splenocytes: more severe are required. disease with deposition in the glomeruli. Transfer of antibody alone: mild Only mild disease without granuloma disease but pauci-immune. formation. Disease induction strongly depends on genetic background.195 Depends on the passive transfer of antibodies. No cellular immunity. Wistar–Kyoto rats Immunization of Wistar–Kyoto rats with Leads to crescentic nephritis and Stable and severe disease induction. Little et al.196 human MPO and adjuvants. hemorrhage in all immunized rats. Depends on the passive transfer Chavelel et al.197 of antibodies. No cellular immunity. Disease induction strongly depends on genetic background. Humanized NOD- Transfer of patients’ PR3-ANCA IgG into Induction of hematuria, glomerular Very mild disease. Little et al.198 2 2 SCID-IL-2Rg / mice endotoxin-primed human-hematopoietic hypercellularity, and pulmonary Technically challenging. stem cell (HSC) mice, created using healthy capillaritis in a some of mice. Low level neutrophil reconstitution and donor (PR3) HSC. absence of human T cells due to mixed chimerism. Lupus nephritis New Zealand Black Spontaneous polygenic. Develop severe GN at 5–6 mo of age Difficult to use as a genetically Dixon et al.199 (NZB)/New Zealand and kidney failure at 10–12 mo modified model. Alleles have to Andrews et al.200

mScNephrol Soc Am J white (NZW) mice of age. Animals develop anti-nuclear be matched to NZB and F1 intercross antibodies without anti-Sm antibodies. NZW backgrounds. New Zealand mixes Spontaneous polygenic. Severe GN with an earlier onset No sex difference. Rudofsky et al.201 mice (NZM2328 compared with NZB/NZW F1. Morel202 and NZM2410) 29: 741 – 5,2018 758, mScNephrol Soc Am J

29: Table 2. Continued 741 –

5,21 el n lsaClsi lmrlrDiseases Glomerular in Cells Plasma and Cells B 2018 758, Animal Model Kind of Intervention Key Feature Advantages and Disadvantages Reference MRL/MpJ-FASlpr MRL is a polygenetic mouse model. Production of class-switched autoantibodies Model meets all of the ACR Dixon et al.199 (MRL/lpr) mice FASlpr mutation accelerates the (anti-DNA, Sm), end-organ disease including criteria for SLE. time course of murine disease. dermatitis, GN cardiovascular and lung disease. Mice that expressed a mutant transgene encoding Rapid and stable onset of Cohen et al.203 surface Ig but are unable to secrete Ig developed disease. nephritis in the absence of soluble autoantibodies. B cells with cognate BCR are essential for lupus nephritis via antigen-presentation to cognate T cells or other functions (cytokine, chemokine production, formation of germinal center). Andrews et al.200 Herlands et al.204 Chan et al.53 ABMR Mouse heterotopic Repeated passive transfer Animals develop chronic Depends on the passive transfer Uehara et al.205 cardiac allografts of alloantibodies. transplant arteriopathy. of antibodies. In B cell–deficient mice it was shown that No cellular immunity. chronic allograft vasculopathy was only observed in the presence of alloantibodies. No insights into the mechanism of antibody formation. Humanized CD52Tg Transgenic mouse model in which CD52 Animals show cardiac allograft Offers the possibility to study outcomes Kwun et al.206 (H-2K) mice is only expressed on T cells. vasculopathy, whereas animals of animals with continuous de novo. T cells are depleted by without DSA show normal grafts.206 (anti-CD52) and de novo allospecific B cells and DSA but not alloreactive T cells arise in some animals. www.jasn.org RE REVIEW BRIEF 747 BRIEF REVIEW www.jasn.org such as (GA 101), are pri- terminal unfolded protein response causing extrinsic and intrinsic characteristics of marily inducing cell death with a more pro- apoptosis81 and prevention of the activation memory B cell maintenance are far less found and lasting B cell depletion and are of NF-k light chain enhancer of activated B understood. currently under investigation in autoim- cells (NF-kB), which is crucial for PC sur- mune diseases.65 vival.82 Initial data were obtained for mul- tiple myeloma but proteasome inhibitors CLINICAL EXPERIENCES SUGGEST are toxic to non-neoplastic cells as well.83 DISTINCT ROLE OF MEMORY B TARGETING B CELL SURVIVAL Further, cells with high protein production, AND PCS IN DIFFERENT FACTORS such as PCs, are susceptible to proteasome GLOMERULAR DISEASES inhibition.84 Bortezomib is the most widely There are several approaches indirectly used proteasome inhibitor approved for the AAV targeting B cell and PC survival via cyto- treatment of mantle cell and Small-vessel vasculitis, which is associated kine targeting. One is blocking BAFF or myeloma.85 The main limitation of borte- with autoantibodies against neutrophil cy- the related cytokine APRIL (Figure 2B). zomib is severe neurotoxicity that occurs in toplasmic antigens, manifests as a necrotiz- These cytokines bind to three different re- up to 30% of all patients, resulting in ther- ing inflammation of vessels with little or no ceptors expressed on B cells: BAFF-R, apy discontinuation,86 which is less with the Igandcomplementdepositioninthevessel TACI, and BCMA. BAFF-R exclusively new generation compounds carfilzomib87 wall. This lack of Ig deposit distin- binds BAFF,66 whereas both cytokines and delanzomib.88 guishes ANCA-associated GN (directed can bind to TACI and BCMA.67 APRIL, against MPO or proteinase 3) from immune binding to BCMA is crucial for the survival complex-mediated GN, e.g., found in lupus of PCs,68 whereas memory B cells do not ANTI-CD38 nephritis.98 In human kidney specimens of 2 depend on these survival factors.69,70 Sev- patients with AAV, B cell clusters with IgD eral biologic drugs targeting the BAFF/ The monoclonal targets B cells forming germinal center-like BLyS and APRIL axis have been studied CD38, which is highly expressed on the sur- structures were found, whereas CD138- over the last years, including , face of short-lived PCs, long-lived PCs, and positive plasma was almost absent.99 T , , and . Be- myeloma cells,89 butonlyexpressedatlow cells that are present in the inflamed kid- limumabbindstosolubleBAFFandhas levels on lymphoid or myeloid cells and ney tissue mostly belong to a senescent been approved for the treatment of SLE.71 some other tissues.90 Daratumumab mon- population, whereas the Tabalumab is a human mAb whereas bli- otherapy was recently approved for the number of memory T cells in circulation sibimod is a fusion polypeptide protein. treatment of refractory is decreased, indicating their migration Both block biologically active BAFF72–74 on the basis of two studies.91–93 Also, the as effectors into the kidney.100,101 but development programs of tabalumab addition of daratumumab to standard Disturbances of peripheral B cell sub- in SLE have been cancelled after poor effi- care has shown favorable results,94,95 with sets have been described in patients with cacy results in clinical trials. Atacicept is a an overall acceptable safety profile.96 AAV. Here memory B cells are reduced in 2 TACI Ig fusion protein that blocks both Nonetheless, a high number (71% of patients with active disease,102 possibly BAFF and APRIL.75 So far, the effect of patients) of adverse reactions of cough, by recruiting B cells to the sites of in- these compounds on PCs becomes evident bronchospasm, and dyspnea were observed flammation or their differentiation into by the substantial reduction of all Ig classes upon infusion of daratumumab.93 Patients PCs. During remission memory B cell to different degrees, whereas the effect on with autoimmune diseases have not yet levels are restored.102 An imbalance of memory B cells is largely confined to the been treated with this antibody but it regulatory B cells, a B cell fraction that initial increase in peripheral blood likely appears to lead to a profound depletion of is commonly defined by the production related to their mobilization.76,77 plasma cells. of IL 10, a cytokine able to suppress T cells, has been reported.103 These regu- latory B cells are decreased in patients PROTEASOME INHIBITORS TARGETING PC HOMING AND with AAV. Diminished regulatory B cells SURVIVAL were found to predict relapse after ritux- Proteasome inhibitors are prototypic for imab therapy.103,104 The overall memory their effect on highly differentiated PCs Anumberofrecenteffortshavebeenmade B cell population did not correlate with with substantial to target certain factors involved in PC disease activity or the time to flare after B stress. They selectively target the 26S homing/migration or interfere with their cell depletion,104 whereas CD38++ pe- proteasome,78,79 which is needed for the survival in order to affect long-lived PCs ripheral PC and B cell activation have degradation of ubiquinated .80 otherwise refractory to treatment (re- been reported to correlate with active Their cytotoxic effect causes accumulation viewed recently2), including autologous ANCA vasculitis.105 of misfolded proteins in the endoplasmic stem cell transplantation.97 Clinical expe- Anti-CD20 therapy with rituximab reticulum, leading to the activation of the riences are very limited. In contrast, the has been approved on the basis of clinical

748 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW studies for remission induction,106,107 surge seen after rituximab, due to the re- SLE and the expression of programmed and is also a valuable maintenance ther- duction of B cell BAFF-R clearance, pro- death 1 correlates with circulating plasma- apy,108 especially in refractory disease or motes autoreactivity in the reconstituting blasts and anti-dsDNA antibodies.127,128 patients not responding to cyclophos- B cell repertoire. BLyS/BAFF inhibition is The presence of these tertiary lymphoid phamide.109 Because the ANCA titers also effective in IMN, where it signifi- structures is associated with the local for- typically, but not always correlate with cantly reduces anti-PLA2R antibodies ti- mation of autoantibodies and tubular disease activity and are reliably reduced ters within 28 weeks (Clinicaltrials.gov basement membrane immune complexes by rituximab to a greater extent than identifier NCT01610492). in more active disease.124 seen with cyclophosphamide, they ap- There is little experience with agents All these data show that the interac- pear to be the product of short-lived targeting PCs in AAV. Bontscho et al.118 tion of B and T cells is critical in lupus PCs,110 which provides the basis for the reported that bortezomib is able to in- nephritis. In addition to the footprints of key role of autoreactive B cells in AAV. hibit anti-MPO–mediated necrotizing B cell abnormalities (autoantibodies and However, rituximab has also been effec- crescentic GN in mice by depleting immune complexes), continuous activa- tive in ANCA-negative patients with MPO-specific PCs in the spleen and tion of innate and the adaptive immunity granulomatosis and polyangiitis,111 and BM. A favorable effect of proteasome is operative. Here, T cells provide help to the presence of B cells in close proximity inhibition was also reported in one pa- B cells or are cytotoxic to glomeruli or to T cells and at sites of in- tient with AAV,119 but precludes any tubular cells.129 A key characteristic of flammation indicates alternative roles, firm conclusion in the absence of larger SLE T cells is their reduced IL-2 produc- such as antigen presentation, which sup- experiences. tion, which inversely correlates with port the disease process. The hypothesis enhanced IL-17 regulated by the tran- of the pathogenicity of ANCA is also SLE scription factor CREM-a.Moreover, supported by a case report in which fe- Lupus nephritis is the most common severe IL-2 as an essential growth and survival tomaternal transfer of anti-MPO auto- organ manifestation in SLE, seen in up to factor for Treg cells has been found to be antibodies led to vasculitis with pulmo- 50%ofcases.120 Histologically, it can pre- functionally deficient in patients with nary renal syndrome112 in the offspring. sent with a spectrum of glomerular, vascu- SLE.130 A number of genetic risk alleles Another case, however, did not develop lar, and tubulointerstitial lesions, including and epigenetic modifications altering GN or vasculitis upon transplacental germinal center-like structures in the inter- the and the function of transfer of MPO-ANCA.113 Amore stitium. Cytokine-mediated activation of certain T cell subsets in SLE appear to be direct correlation of titers glomerular mesangial cells and in situ tu- involved.131 These abnormal T cell re- and response to therapy is present bulointerstitial macrophages promotes sponses can result in disturbances of B in IMN, where a fall in phospholipase the deposition of anti-dsDNA, anti-C1q, cellsinSLE.BecauseSLEpathology A2 receptor (PLA2R) antibody titers anti-nucleosome, and anti-glomerular apparently comprises pathogenic contri- exactly predicts reductions in protein- antibodies recognizing glomerular butions of memory B cells and PCs de- uria and correlates with the extent of structures—the formation of immune pendent on T helper cells, abnormal T proteinuria.114 complexes with complement deposition cell activation may represent a critical Circulating BAFF levels correlate with initiating renal damage.121,122 Circulating link.Proofforthisideaisverysmall, disease activity in AAVand therefore this neutrophil activation with neutrophil ex- but a recent phase 2 trial is using voclo- cytokine has been suggested as potential tracellular trap deposition in the glomeruli sporine in lupus nephritis.132 Alterna- treatment target.115–117 An ongoing trial may also be an initiating event in renal in- tively, therapies with an effect on T and is addressing the role of BAFF inhibition flammation. Anti-dsDNA antibodies B cell interaction (Figure 3) are attrac- in patients with AAV: BREVAS is evalu- represent a risk factor for lupus nephritis, tive. In addition, low-dose IL-2 has been ating belimumab in combination with with negative predictive value for renal out- studied in non-renal SLE133 and demon- azathioprine as maintenance of remission come in some studies.123 In addition to the strated some clinical efficacy, leading to after a standard induction regimen (Clin- typical presence of autoantibodies, anti- an increase of Treg cells in vitro134 and in icaltrials.gov identifier NCT01663623). body-independent mechanisms contribute vivo.135 This approach follows the hypothesis to to lupus pathogenesis. Lupus nephritis bi- The contribution of CD20+ and espe- selectively block enhanced BAFF and opsies contain interstitial B cell cially memory B cells became apparent reduce ANCA producing peripheral infiltrates that are organized along with by experience with B cell–depleting PCs and thereby affect AAV activity. A T cells and dendritic cells,124–126 and un- agents, such as rituximab. In several re- further study, COMBIVAS, is address- dergo further differentiation as in germinal ports the reconstitution of the memory ing the combination of BLyS/BAFF centers. Within these aggregates, T follicu- B cell subset after rituximab predicted blockade and anti-CD20 on the basis larhelpercellsareimportantforestablish- recurrence of active disease.136,137 This of two hypotheses: that local BLyS/ ing germinal centers to promote activation phenomenon has not exclusively been BAFF impairs B cell depletion at inflam- and differentiation of B cells.126 T follicular observedinSLEbutalsoinpatients matory sites and that the BLyS/BAFF helper cells are expanded in patients with with rheumatoid arthritis25,138 and

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 749 BRIEF REVIEW www.jasn.org

IMN, and suggests that memory B cells However, there are conflicting data about delanzomib.88 Clinical experiences with contribute to the pathogenesis of these the interrelation of the cytokine levels of proteasome inhibition in lupus nephritis diseases. A reduction in autoantibody ti- BAFF and APRIL with lupus activity.148 rely only on bortezomib. Alexander ters, in contrast to AAV, did not always Notably, these studies found an associa- et al.155 reported 12 patients with predict the effectiveness of rituximab tion between response to belimumab refractory SLE including six patients therapy.139 Rituximab did not meet the and reductions in autoantibody titers, with nephritis. Musculoskeletal and mu- primary efficacy end point in a lupus ne- suggesting successful targeting of PCs. Al- cocutaneous manifestations improved phritis trial (LUNAR),140 partly due to though other compounds targeting BAFF, in all patients, and proteinuria signifi- aspects of the trial design. Clinical expe- such as tabalumab149,150 or atacicept151 cantly decreased in patients with nephri- riences141,142 in otherwise refractory pa- cotargeting APRIL, did not provide con- tis. A recently published study reports a tients support a clinical value that has led vincing clinical data, a more profound ef- beneficial effect with a decrease in creat- to recommendations as a second line fect on PCs reflected by Ig reductions inine in three of five patients with lupus agent by the American College of Rheu- was noted. Selective inhibition of the nephritis.156 A phase 2 study is currently matology143 and European League PC-relevant cytokines without sufficient investigating the safety and efficacy of Against Rheumatism-European Renal treatment of CD20+ memory B cells may bortezomib in patients with SLE (Clini- Association/European Dialysis and hold more promise. In this context, a se- caltrials.gov identifier NCT02102594). Transplant Association.144 Rituximab, quential approach of B cell depletion A phase 1 study in lupus nephritis with in combination with mycophenolate and BLyS/BAFF inhibition to influence the oral proteasome inhibitor ixazomib mofetil, has been reported in newly di- PC activity is being evaluated in lupus is underway (Clinicaltrials.gov identifier agnosed patients with lupus nephritis nephritis, the Rituximab and Belimumab NCT02176486). Although proteasome in- in a UK center,145 with a substantial for Lupus Nephritis (CALIBRATE) study hibition is often considered a PC-targeted rate of patients achieving complete re- (clinicaltrials.gov identifier NCT02260934) therapy, it has the advantage to cotarget PCs mission without use of oral glucocorti- and in extrarenal lupus, the BEAT-LUPUS but also affect other immune cells.155 Apo- coids. This study requires confirmation study.152 tential difference of lupus nephritis versus in treatment-naïve patients, whereas the Efforts to preferentially target the PC AAVisthateffectiveBlineagetherapyre- currently available data suggest that anti- compartment have been undertaken. In a quires cotargeting of memory B cells CD20 depletion alone may not be suffi- murine model of lupus-like nephritis, and PCs. cient in patients with established disease. short- and long-lived PCs were depleted In this population, the additional role of by bortezomib and lupus-like nephritis Kidney Transplantation PCs likely requires cotargeting. improved remarkably.83 These results In the transplant setting, donor-specific Belimumab has been approved for gen- were confirmed by Ichikawa et al.153 antibodies (DSA) are likely the main bi- eralSLEonthebasisofthesuccessfulphase and Seavey et al.,154 using the less toxic ologic reason for allograft injury and 3 studies BLISS-52146 and BLISS-76.147 proteasome inhibitors carfilzomib87 and loss.157 Their detection either pre- or post-transplant increases the risk of graft loss.158,159 DSA directly bind to the grafts and cause local inflammation and tissue damage through complement activation and human Fc gamma-mediated cytotoxicity, and also serve as that facilitate the activation of alloreactive T cells.160–162 These mechanisms lead to the development of transplant glomerulop- athy, the histologic feature of chronic ABMR.163 Long-lived PCs are responsible for the maintenance of long-term circulat- ing DSA, which are detectable in solid- organ transplant recipients and are associated with worse transplant outcomes.164 Quies- cent memory B cells appear to be reactiva- ted in cases of secondary transplantation in presensitized individuals. The best risk Figure 3. Immune intervention at key nodes of immune interaction in SLE. Targeting key management to avoid transplant glomerul- nodes of T cell and B cellinteraction holds promise to improve abnormal activity opathy comprises transplantation without or enhanced activity (i.e., using low-dose IL-2) that may also have the preexisting DSA159 and avoidance of trans- potential to normalize B lineage cell abnormalities. plantation with HLA mismatches, especially

750 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW

NCT02201576), and the Effect of BM- MSCs on Chronic AMR after Kidney Transplantation study (Clinicaltrials. gov identifier NCT02563340), will eval- uate the clinical value of bortezomib on chronic ABMR. Notably, levels of BAFF were found to be associated with the occurrence of trans- plant glomerulopathy183 and enhanced ABMR,184 thus BAFF targeting appeared an attractive option. However, a study of tabalumab for desensitization did not lead to clinically significant reduction of DSA,185 which may be taken as another indicator that outstanding long-lived PCs are a key challenge in this clinical Figure 4. Distinct role of B lineage and functions in glomerular diseases. The setting. Studies of belimumab for pre- sites of induction and residence, however, remain to be delineated. transplant desensitization and at the time of transplantation are ongoing (clin- icaltrials.gov identifier NCT01025193). as class 2 HLA mismatches are a strong pre- Kidney transplantation permits com- An innovative approach to reduce HLA dictive biomarker for DSA.165 The induc- prehensive corresponding insights in tis- antibodies in the setting of desensitiza- tion of DSA requires interaction between B sue resident immunology. Just as in other tion of highly sensitized patients is the cells and antigen-specific CD4 T helper glomerular diseases, tertiary lymphoid use of the endopeptidase derived from cells. The presence of memory T cells before structures have been identified in kidney Streptococcus pyogenes (IdeS), which transplantation is associated with a poor grafts.173 Within the tertiary lymphoid cleaves IgG into F(ab9)2 and Fc frag- transplant outcome.166,167 ABMR and the structures an increase of activated mem- ments. IdeS leads to a complete IgG de- underlying importance of pathogenically ory CD4+ T cells and a decrease in T ficiency what makes it less attractive for relevant PCs producing DSA are a key target regulatory subsets (IL-10–producing long-term use in chronic ABMR.186 of to eradicate DSA. These Tr1 cells and Foxp3-positive Tregs) has However, early clinical experiences with strategies include plasmapheresis and im- been observed.174 Also PC-rich infil- IdeS in renal transplants provide addi- munoadsorption for the direct removal of trates have been found in renal allografts tional proof that targeting autoreactive DSA, intravenous Igs for immunomodula- and their occurrence was associated with PCs is key in targeting ABMR. tion,168 rituximab to deplete PC precursors, an increased risk of allograft failure in and targeting PCs by proteasome inhibi- the pediatric population.175 However, tion.169 Combined renal and allogeneic data on PC-rich infiltrates in the adult CONCLUDING REMARKS BM transplantation have also aimed to de- population are limited.176–178 To target pletePCsandinducetolerancewithoutthe the PC compartment, proteasome inhi- A number of preclinical models have need for antirejection therapy.170 bition has been included in the treat- supported the role of B lineage cells in B cell–directed therapy by rituximab ment repertoire. In a model of chronic glomerulopathies, but do not reflect the seems to be partially effective in acute allograft nephropathy, bortezomib re- complexity of human diseases. Interven- ABMRbutineffectiveinchronic duced antibody titers and depleted PCs tions in human disease have inspired a re- ABMR.171 Jackson et al.172 provided in- in the BM.179 The reduction of PCs was vision of the role of B cells and long-lived teresting evidence about long-lived PCs also observed in BM aspirates of two pa- (memory) PCs in these entities. Here, cur- in humans and the capacity of rituximab tients treated with bortezomib for hu- rently available data support a key role of B to interfere with relevant B cells as PC moral rejection.180 Several small case cells (likely memory and activated B cells) precursors when they compared DSA series report the effect of bortezomib in AAVand IMN, and long-lived PCs as a levels in patients who have been desen- treatment on graft survival, kidney func- pathogenic driver in chronic ABMR, sitized with or without rituximab. They tion, and DSA levels. Overall, patients whereas lupus nephritis appears to be un- found a greater reduction of DSA but a responded together with a reduction in der the influence of both B cells and PCs similar rate of DSA persistence, most DSA levels and a stabilization of kidney (Figure 4). As a consequence, the idea that likely explained by refractory long-lived function in refractory patients.181 The distinct immunopathogenic contribu- PCs, whereas the difference in DSA titers BORTEJECT (Clinicaltrials.gov identi- tions by memory B cells and PCs is also can be ascribed to memory B cells un- fier NCT01873157),182 the TRIBUTE probably related to distinct T helper cell dergoing differentiation into PCs. study (Clinicaltrials.gov identifier abnormalities requires consideration.

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 751 BRIEF REVIEW www.jasn.org

ACKNOWLEDGMENTS 11. Bortnick A, Chernova I, Quinn WJ, Mugnier low frequency of . JIm- M, Cancro MP, Allman D: Long-lived bone munol 177: 3728–3736, 2006 marrow plasma cells are induced early in 24. Anolik JH, Barnard J, Cappione A, E.S. is supported by the Charité Junior Clinical response to T cell-independent or T cell- Pugh-Bernard AE, Felgar RE, Looney RJ, Scientist Program (Charité-Universitätsmedizin dependent antigens. JImmunol188: 5389– Sanz I: Rituximab improves peripheral B cell Berlin and the Berlin Institute of Health). Re- 5396, 2012 abnormalities in human systemic lupus er- lated support has been provided by projects 12. Bortnick A, Allman D: What is and what ythematosus. Arthritis Rheum 50: 3580– funded by the German Research Foundation should always have been: Long-lived plasma 3590, 2004 cells induced by T cell-independent antigens. 25. Roll P, Dörner T, Tony H-P: Anti-CD20 (SFB650, 633, CRC “Immunobone,” TR130, JImmunol190: 5913–5918, 2013 therapy in patients with rheumatoid arthri- and individual DFG projects Do491/7-4, 8-1,2, 13. Weisel FJ, Zuccarino-Catania GV, Chikina tis: Predictors of response and B cell subset 10-1, all granted to T.D.). M, Shlomchik MJ: A temporal switch in the regeneration after repeated treatment. germinal center determines differential Arthritis Rheum 58: 1566–1575, 2008 output of memory B and plasma cells. Im- 26. Cagigi A, Du L, Dang LVP, Grutzmeier S, munity 44: 116–130, 2016 Atlas A, Chiodi F, Pan-Hammarström Q, DISCLOSURES 14. Kaji T, Ishige A, Hikida M, Taka J, Hijikata A, Nilsson A: CD27(-) B-cells produce class None. Kubo M, Nagashima T, Takahashi Y, switched and somatically hyper-mutated Kurosaki T, Okada M, Ohara O, Rajewsky K, antibodies during chronic HIV-1 infection. Takemori T: Distinct cellular pathways se- PLoS One 4: e5427, 2009 REFERENCES lect germline-encoded and somatically 27. Jacobi AM, Reiter K, Mackay M, Aranow C, mutated antibodies into immunological Hiepe F, Radbruch A, Hansen A, Burmester memory. JExpMed209: 2079–2097, G-R, Diamond B, Lipsky PE, Dörner T: Acti- 1. Ahuja A, Anderson SM, Khalil A, Shlomchik 2012 vated memory B cell subsets correlate with MJ: Maintenance of the plasma cell pool is 15. Maruyama M, Lam KP, Rajewsky K: Memory disease activity in systemic lupus eryth- independent of memory B cells. Proc Natl B-cell persistence is independent of per- ematosus: Delineation by expression of – Acad Sci U S A 105: 4802 4807, 2008 sisting immunizing antigen. Nature 407: CD27, IgD, and CD95. Arthritis Rheum 58: 2. Hiepe F, Radbruch A: Plasma cells as an in- 636–642, 2000 1762–1773, 2008 novative target in with 16. Klein U, Küppers R, Rajewsky K: Evidence 28. Wehr C, Eibel H, Masilamani M, Illges H, renal manifestations. Nat Rev Nephrol 12: for a large compartment of IgM-expressing Schlesier M, Peter H-H, Warnatz K: A new – 232 240, 2016 memory B cells in humans. Blood 89: 1288– CD21low B cell population in the peripheral 3. Giesecke C, Frölich D, Reiter K, Mei HE, 1298, 1997 blood of patients with SLE. Clin Immunol Wirries I, Kuhly R, Killig M, Glatzer T, Stölzel 17. Klein U, Rajewsky K, Küppers R: Human 113: 161–171, 2004 K, Perka C, Lipsky PE, Dörner T: Tissue dis- immunoglobulin (Ig)M+IgD+ peripheral 29. Fleischer SJ, Daridon C, Fleischer V, Lipsky tribution and dependence of responsiveness of blood B cells expressing the CD27 cell sur- PE, Dörner T: Enhanced tyrosine phospha- fi human antigen-speci cmemoryBcells.J face antigen carry somatically mutated tase activity underlies dysregulated B cell – Immunol 192: 3091 3100, 2014 variable region genes: CD27 as a general receptor signaling and promotes survival of 4. Waiser J, Budde K, Schütz M, Liefeldt L, marker for somatically mutated (memory) B human lupus B cells. Arthritis Rheumatol 68: Rudolph B, Schönemann C, Neumayer H-H, cells. JExpMed188: 1679–1689, 1998 1210–1221, 2016 Lachmann N: Comparison between borte- 18. Rajewsky K: and learning in 30. Schroeder K, Herrmann M, Winkler TH: The zomib and rituximab in the treatment of the antibody system. Nature 381: 751–758, role of in the gen- antibody-mediated renal allograft rejection. 1996 eration of pathogenic antibodies in SLE. – Nephrol Dial Transplant 27: 1246 1251, 19. Takemori T, Kaji T, Takahashi Y, Shimoda M, Autoimmunity 46: 121–127, 2013 2012 Rajewsky K: Generation of memory B cells 31. Jethwa HS, Clarke SH, Itoh-Lindstrom Y, 5. Dörner T, Lipsky PE: Beyond pan-B-cell-directed inside and outside germinal centers. Eur J Falk RJ, Jennette JC, Nachman PH: Re- therapy - new avenues and insights into the Immunol 44: 1258–1264, 2014 striction in V kappa gene use and antigen pathogenesis of SLE. Nat Rev Rheumatol 12: 20. Good KL, Avery DT, Tangye SG: Resting selection in anti-myeloperoxidase response – 645 657, 2016 human memory B cells are intrinsically pro- in mice. JImmunol165: 3890–3897, 2000 6. Baumgarth N: Innate-like B cells and their grammed for enhanced survival and re- 32. Winkler TH, Fehr H, Kalden JR: Analysis of rules of engagement. Adv Exp Med Biol sponsiveness to diverse stimuli compared immunoglobulin variable region genes – 785: 57 66, 2013 to naive B cells. JImmunol182: 890–901, from human IgG anti-DNA hybridomas. Eur 7. Defrance T, Taillardet M, Genestier L: T cell- 2009 J Immunol 22: 1719–1728, 1992 independent B cell memory. Curr Opin Im- 21. McHeyzer-Williams LJ, Milpied PJ, Okitsu 33. Ferdman J, Porcheray F, Gao B, Moore C, munol 23: 330–336, 2011 SL, McHeyzer-Williams MG: Class-switched DeVito J, Dougherty S, Thomas MV, 8. Radbruch A, Muehlinghaus G, Luger EO, memory B cells remodel BCRs within sec- Farkash EA, Elias N, Kawai T, Malek SK, Inamine A, Smith KGC, Dörner T, Hiepe F: ondary germinal centers. Nat Immunol 16: Tullius SG, Wong W, Zorn E: Expansion and Competence and competition: The chal- 296–305, 2015 somatic hypermutation of B-cell clones in lenge of becoming a long-lived plasma cell. 22. Wei C, Anolik J, Cappione A, Zheng B, Pugh- rejected human kidney grafts. Trans- Nat Rev Immunol 6: 741–750, 2006 Bernard A, Brooks J, Lee E-H, Milner ECB, plantation 98: 766–772, 2014 9. Tokoyoda K, Zehentmeier S, Chang H-D, Sanz I: A new population of cells lacking ex- 34. Cheng J, Torkamani A, Grover RK, Jones Radbruch A: Organization and maintenance pression of CD27 represents a notable com- TM, Ruiz DI, Schork NJ, Quigley MM, Hall of by stroma niches. ponent of the B cell memory compartment in FW, Salomon DR, Lerner RA: Ectopic B-cell Eur J Immunol 39: 2095–2099, 2009 systemic lupus erythematosus. JImmunol clusters that infiltrate transplanted human 10. Tokoyoda K, Hauser AE, Nakayama T, 178: 6624–6633, 2007 kidneys are clonal. Proc Natl Acad Sci U S A Radbruch A: Organization of immunological 23. Fecteau JF, Côté G, Néron S: A new mem- 108: 5560–5565, 2011 memory by stroma. Nat Rev ory CD27-IgG+ B cell population in pe- 35. Odendahl M, Mei H, Hoyer BF, Jacobi AM, Immunol 10: 193–200, 2010 ripheral blood expressing VH genes with Hansen A, Muehlinghaus G, Berek C, Hiepe F,

752 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW

Manz R, Radbruch A, Dörner T: Generation of enriched in human bone marrow. Blood and clinical considerations. AnnInternMed migratory antigen-specific plasma blasts and 125: 1739–1748, 2015 84: 304–315, 1976 mobilization of resident plasma cells in a 47. Halliley JL, Tipton CM, Liesveld J, Rosenberg 58. Matas AJ: Minimization of steroids in kidney secondary immune response. Blood 105: AF, Darce J, Gregoretti IV, Popova L, transplantation. Transpl Int 22: 38–48, 2009 1614–1621, 2005 Kaminiski D, Fucile CF, Albizua I, Kyu S, 59. Baris HE, Baris S, Karakoc-Aydiner E, Gokce 36. Hargreaves DC, Hyman PL, Lu TT, Ngo VN, Chiang K-Y, Bradley KT, Burack R, Slifka M, I, Yildiz N, Cicekkoku D, Ogulur I, Ozen A, Bidgol A, Suzuki G, Zou YR, Littman DR, Hammarlund E, Wu H, Zhao L, Walsh EE, Alpay H, Barlan I: The effect of systemic Cyster JG: A coordinated change in che- Falsey AR, Randall TD, Cheung WC, Sanz I, corticosteroids on the innate and adaptive mokine responsiveness guides plasma cell Lee FE-H: Long-lived plasma cells are con- immune system in children with steroid re- movements. JExpMed194: 45–56, 2001 tained within the CD19(-)CD38(hi)CD138(+) sponsive nephrotic syndrome. Eur J Pediatr 37. Chu VT, Fröhlich A, Steinhauser G, Scheel T, subset in human bone marrow. Immunity 43: 175: 685– 693, 2016 Roch T, Fillatreau S, Lee JJ, Löhning M, 132–145, 2015 60. Mahévas M, Michel M, Vingert B, Moroch J, Berek C: Eosinophils are required for the 48. Bhoj VG, Arhontoulis D, Wertheim G, Boutboul D, Audia S, Cagnard N, Ripa J, maintenance of plasma cells in the bone Capobianchi J, Callahan CA, Ellebrecht Menard C, Tarte K, Mégret J, Le Gallou S, marrow. Nat Immunol 12: 151–159, 2011 CT, Obstfeld AE, Lacey SF, Melenhorst JJ, Patin P, Thai L, Galicier L, Bonnotte B, 38. Gomez MR, Talke Y, Goebel N, Hermann F, Nazimuddin F, Hwang W-T, Maude SL, Godeau B, Noizat-Pirenne F, Weill J-C, Reich B, Mack M: Basophils support the Wasik MA, Bagg A, Schuster S, Feldman Reynaud C-A: Emergence of long-lived au- survival of plasma cells in mice. J Immunol MD, Porter DL, Grupp SA, June CH, toreactive plasma cells in the spleen of pri- 185: 7180–7185, 2010 Milone MC: Persistence of long-lived mary warm auto-immune hemolytic anemia 39. Winter O, Moser K, Mohr E, Zotos D, Kaminski plasma cells and in in- patients treated with rituximab. J Auto- H, Szyska M, Roth K, Wong DM, Dame C, dividuals responding to CD19-directed immun 62: 22–30, 2015 Tarlinton DM, Schulze H, MacLennan ICM, CAR T-cell therapy. Blood 128: 360–370, 61. Leandro MJ, Cooper N, Cambridge G, Manz RA: Megakaryocytes constitute a func- 2016 Ehrenstein MR, Edwards JCW: Bone mar- tional component of a plasma cell niche in the 49. Glatman Zaretsky A, Konradt C, Dépis F, row B-lineage cells in patients with rheu- bone marrow. Blood 116: 1867–1875, 2010 Wing JB, Goenka R, Atria DG, Silver JS, Cho matoid arthritis following rituximab therapy. 40. Wilmore JR, Allman D: Here, there, and S, Wolf AI, Quinn WJ, Engiles JB, Brown Rheumatology (Oxford) 46: 29–36, 2007 anywhere? Arguments for and against the DC, Beiting D, Erikson J, Allman D, Cancro 62. Kamburova EG, Koenen HJPM, Borgman physical plasma cell survival niche. JIm- MP, Sakaguchi S, Lu L-F, Benoist CO, KJE, ten Berge IJ, Joosten I, Hilbrands LB: A munol 199: 839–845, 2017 Hunter CA: T regulatory cells support single dose of rituximab does not deplete B 41. Cassese G, Lindenau S, de Boer B, Arce S, plasma cell populations in the bone mar- cells in secondary lymphoid organs but al- Hauser A, Riemekasten G, Berek C, Hiepe F, row. Cell Reports 18: 1906–1916, 2017 ters phenotype and function. Am J Trans- Krenn V, Radbruch A, Manz RA: Inflamed 50. Salama AD, Little MA: Animal models of an- plant 13: 1503–1511, 2013 kidneys of NZB / W mice are a major site for tineutrophil antibody-associated 63. Thurlings RM, Vos K, Wijbrandts CA, the homeostasis of plasma cells. Eur J Im- vasculitis. Curr Opin Rheumatol 24: 1–7, 2012 Zwinderman AH, Gerlag DM, Tak PP: munol 31: 2726–2732, 2001 51. Kenney LL, Shultz LD, Greiner DL, Brehm Synovial tissue response to rituximab: 42. Hutloff A, Büchner K, Reiter K, Baelde HJ, MA: models for trans- Mechanism of action and identification of Odendahl M, Jacobi A, Dörner T, Kroczek plant immunology. Am J Transplant 16: biomarkers of response. Ann Rheum Dis 67: RA: Involvement of inducible costimulator 389–397, 2016 917–925, 2008 in the exaggerated memory B cell and 52. Shlomchik MJ, Craft JE, Mamula MJ: From T 64. Pescovitz MD: Rituximab, an anti- mono- plasma cell generation in systemic lupus to B and back again: Positive feedback in clonal antibody: History and mechanism of erythematosus. Arthritis Rheum 50: 3211– systemic autoimmune disease. Nat Rev action. Am J Transplant 6: 859–866, 2006 3220, 2004 Immunol 1: 147–153, 2001 65. Reddy V, Dahal LN, Cragg MS, Leandro M: 43. Schröder AE, Greiner A, Seyfert C, Berek C: 53. Chan OT, Madaio MP, Shlomchik MJ: B Optimising B-cell depletion in autoimmune Differentiation of B cells in the nonlymphoid cells are required for lupus nephritis in the disease: Is obinutuzumab the answer? Drug tissue of the of patients polygenic, Fas-intact MRL model of sys- Discov Today 21: 1330–1338, 2016 with rheumatoid arthritis. Proc Natl Acad Sci temic autoimmunity. JImmunol163: 3592– 66. Thompson JS, Bixler SA, Qian F, Vora K, USA93: 221–225, 1996 3596, 1999 Scott ML, Cachero TG, Hession C, 44. Slocombe T, Brown S, Miles K, Gray M, Barr 54. Dörner T, Jacobi AM, Lipsky PE: B cells in Schneider P, Sizing ID, Mullen C, Strauch TA, Gray D: Plasma cell homeostasis: The autoimmunity. Arthritis Res Ther 11: 247, K, Zafari M, Benjamin CD, Tschopp J, effects of chronic antigen stimulation and 2009 Browning JL, Ambrose C: BAFF-R, a newly inflammation. JImmunol191: 3128–3138, 55. Fassbinder T, Saunders U, Mickholz E, Jung identified TNF receptor that specifically 2013 E, Becker H, Schlüter B, Jacobi AM: Differ- interacts with BAFF. Science 293: 2108– 45. Chernova I, Jones DD, Wilmore JR, Bortnick ential effects of cyclophosphamide and 2111, 2001 A, Yucel M, Hershberg U, Allman D: Lasting mycophenolate mofetil on cellular and se- 67. Schneider P: The role of APRIL and BAFF in antibody responses are mediated by a rological parameters in patients with sys- lymphocyte activation. Curr Opin Immunol combination of newly formed and estab- temic lupus erythematosus. Arthritis Res 17: 282–289, 2005 lished bone marrow plasma cells drawn Ther 17: 92, 2015 68. O’Connor BP, Raman VS, Erickson LD, Cook from clonally distinct precursors. J Immunol 56. Hoyer BF, Moser K, Hauser AE, Peddinghaus WJ, Weaver LK, Ahonen C, Lin L-L, Mantchev 193: 4971–4979, 2014 A, Voigt C, Eilat D, Radbruch A, Hiepe F, GT, Bram RJ, Noelle RJ: BCMA is essential 46. Mei HE, Wirries I, Frölich D, Brisslert M, Manz RA: Short-lived plasmablasts and long- for the survival of long-lived bone marrow Giesecke C, Grün JR, Alexander T, Schmidt lived plasma cells contribute to chronic hu- plasma cells. JExpMed199: 91–98, 2004 S, Luda K, Kühl AA, Engelmann R, Dürr M, moral autoimmunity in NZB/W mice. JExp 69. Benson MJ, Dillon SR, Castigli E, Geha RS, Scheel T, Bokarewa M, Perka C, Radbruch Med 199: 1577–1584, 2004 Xu S, Lam KP, Noelle RJ: Cutting edge: The A, Dörner T: A unique population of IgG- 57. Fauci AS, Dale DC, Balow JE: Glucocorti- dependence of plasma cells and in- expressing plasma cells lacking CD19 is costeroid therapy: Mechanisms of action dependence of memory B cells on BAFF

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 753 BRIEF REVIEW www.jasn.org

and APRIL. JImmunol180: 3655–3659, M, King R, Chabot-Fletcher MC, Breton JJ, CD38/CD19: A lipid raft-dependent sig- 2008 Allsop D, Rivett AJ: Characterization of naling complex in human B cells. Blood 109: 70. Scholz JL, Crowley JE, Tomayko MM, peptidyl boronic acid inhibitors of mam- 5390–5398, 2007 Steinel N, O’Neill PJ, Quinn WJ 3rd, malian 20 S and 26 S and their 91. McKeage K: Daratumumab: First global Goenka R, Miller JP, Cho YH, Long V, Ward inhibition of proteasomes in cultured cells. approval. Drugs 76: 275–281, 2016 C, Migone T-S, Shlomchik MJ, Cancro MP: Biochem J 346: 447–454, 2000 92. Lonial S, Weiss BM, Usmani SZ, Singhal S, BLyS inhibition eliminates primary B cells 80. Ciechanover A: The ubiquitin-proteasome Chari A, Bahlis NJ, Belch A, Krishnan A, but leaves natural and acquired humoral proteolytic pathway. Cell 79: 13–21, 1994 Vescio RA, Mateos MV, Mazumder A, immunity intact. Proc Natl Acad Sci U S A 81. Obeng EA, Carlson LM, Gutman DM, Orlowski RZ, Sutherland HJ, Bladé J, Scott 105: 15517–15522, 2008 Harrington WJ Jr., Lee KP, Boise LH: EC, Oriol A, Berdeja J, Gharibo M, Stevens 71. Stohl W: Therapeutic targeting of the BAFF/ Proteasome inhibitors induce a terminal DA, LeBlanc R, Sebag M, Callander N, APRIL axis in systemic lupus erythematosus. unfolded protein response in multiple my- Jakubowiak A, White D, de la Rubia J, Expert Opin Ther Targets 18: 473–489, eloma cells. Blood 107: 4907–4916, 2006 Richardson PG, Lisby S, Feng H, Uhlar 2014 82. Hideshima T, Chauhan D, Richardson P, CM, Khan I, Ahmadi T, Voorhees PM: 72. Hsu H, Khare SD, Lee F, Miner K, Hu Y-L, Mitsiades C, Mitsiades N, Hayashi T, Munshi Daratumumab monotherapy in patients Stolina M, Hawkins N, Chen Q, Ho S-YJ, Min N, Dang L, Castro A, Palombella V, Adams with treatment-refractory multiple mye- H, Xiong F, Boone T, Zack DJ: A novel mo- J, Anderson KC: NF-kappa B as a thera- loma (SIRIUS): An open-label, randomised, dality of BAFF-specific inhibitor AMG623 peutic target in multiple myeloma. JBiol phase 2 trial. Lancet 387: 1551–1560, 2016 peptibody reduces B-cell number and im- Chem 277: 16639–16647, 2002 93. Lokhorst HM, Plesner T, Laubach JP, Nahi proves outcomes in murine models of au- 83. Neubert K, Meister S, Moser K, Weisel F, H, Gimsing P, Hansson M, Minnema MC, toimmune disease. Clin Exp Rheumatol 30: Maseda D, Amann K, Wiethe C, Winkler Lassen U, Krejcik J, Palumbo A, van de Donk 197–201, 2012 TH, Kalden JR, Manz RA, Voll RE: The NWCJ, Ahmadi T, Khan I, Uhlar CM, Wang 73. Genovese MC, Fleischmann RM, proteasome inhibitor bortezomib depletes J, Sasser AK, Losic N, Lisby S, Basse L, Brun Greenwald M, Satterwhite J, Veenhuizen M, plasma cells and protects mice with lupus- N, Richardson PG: Targeting CD38 with Xie L, Berclaz P-Y, Myers S, Benichou O: like disease from nephritis. Nat Med 14: daratumumab monotherapy in multiple Tabalumab, an anti-BAFF monoclonal anti- 748–755, 2008 myeloma. N Engl J Med 373: 1207–1219, body, in patients with active rheumatoid 84. Meister S, Schubert U, Neubert K, 2015 arthritis with an inadequate response to Herrmann K, Burger R, Gramatzki M, Hahn 94. Palumbo A, Chanan-Khan A, Weisel K, TNF inhibitors. Ann Rheum Dis 72: 1461– S, Schreiber S, Wilhelm S, Herrmann M, Nooka AK, Masszi T, Beksac M, Spicka I, 1468, 2013 Jäck H-M, Voll RE: Extensive immunoglob- Hungria V, Munder M, Mateos MV, Mark 74. Manetta J, Bina H, Ryan P, Fox N, Witcher ulin production sensitizes myeloma cells for TM, Qi M, Schecter J, Amin H, Qin X, DR, Kikly K: Generation and characteriza- proteasome inhibition. Res 67: Deraedt W, Ahmadi T, Spencer A, tion of tabalumab, a human monoclonal 1783–1792, 2007 Sonneveld P; CASTOR Investigators: Dar- antibody that neutralizes both soluble and 85. Kane RC, Farrell AT, Sridhara R, Pazdur R: atumumab, bortezomib, and dexametha- membrane-bound B-cell activating factor. J United States Food and Drug Administra- sone for multiple myeloma. N Engl J Med Inflamm Res 7: 121–131, 2014 tion approval summary: Bortezomib for the 375: 754–766, 2016 75. Gatto B: Atacicept, a homodimeric fu- treatment of progressive multiple myeloma 95. Dimopoulos MA, Oriol A, Nahi H, San- sion protein for the potential treatment after one prior therapy. Clin Cancer Res 12: Miguel J, Bahlis NJ, Usmani SZ, Rabin N, of diseases triggered by plasma cells. 2955–2960, 2006 Orlowski RZ, Komarnicki M, Suzuki K, Curr Opin Investig Drugs 9: 1216–1227, 86. Schiff D, Wen PY, van den Bent MJ: Neu- Plesner T, Yoon S-S, Ben Yehuda D, 2008 rological adverse effects caused by cyto- Richardson PG, Goldschmidt H, Reece D, 76. Nestorov I, Papasouliotis O, Pena Rossi C, toxic and targeted therapies. Nat Rev Clin Lisby S, Khokhar NZ, O’Rourke L, Chiu C, Munafo A: Pharmacokinetics and immuno- Oncol 6: 596–603, 2009 Qin X, Guckert M, Ahmadi T, Moreau P; globulin response of subcutaneous and in- 87. O’Connor OA, Stewart AK, Vallone M, POLLUX Investigators: Daratumumab, le- travenous atacicept in patients with Molineaux CJ, Kunkel LA, Gerecitano JF, nalidomide, and dexamethasone for multi- systemic lupus erythematosus. J Pharm Sci Orlowski RZ: A phase 1 dose escalation ple myeloma. N Engl J Med 375: 1319– 99: 524–538, 2010 study of the safety and pharmacokinetics of 1331, 2016 77. Stohl W, Hiepe F, Latinis KM, Thomas M, the novel proteasome inhibitor carfilzomib 96. Oliva S, Palumbo A: Monoclonal antibodies Scheinberg MA, Clarke A, Aranow C, (PR-171) in patients with hematologic ma- for treating multiple myeloma - a new era, Wellborne FR, Abud-Mendoza C, Hough lignancies. Clin Cancer Res 15: 7085–7091, new safety considerations? Expert Opin DR, Pineda L, Migone T-S, Zhong ZJ, 2009 Drug Saf 15: 1295–1300, 2016 Freimuth WW, Chatham WW; BLISS-52 88. Gallerani E, Zucchetti M, Brunelli D, 97. Alexander T, Schneider S, Hoyer B, Cheng Study Group; BLISS-76 Study Group: Beli- Marangon E, Noberasco C, Hess D, Q, Thiel A, Ziemer S, Burmester G-R, Arnold mumab reduces autoantibodies, normal- Delmonte A, Martinelli G, Böhm S, Driessen R, Radbruch A, Hiepe F: Development izes low complement levels, and reduces C, De Braud F, Marsoni S, Cereda R, Sala F, and resolution of secondary autoimmunity select B cell populations in patients with D’Incalci M, Sessa C: A first in human phase I after autologous haematopoietic stem cell systemic lupus erythematosus. Arthritis study of the proteasome inhibitor CEP- transplantation for systemic lupus eryth- Rheum 64: 2328–2337, 2012 18770 in patients with advanced solid tu- ematosus: Competition of plasma cells for 78. Adams J, Behnke M, Chen S, Cruickshank mours and multiple myeloma. Eur J Cancer survival niches? Ann Rheum Dis 72: 1102– AA, Dick LR, Grenier L, Klunder JM, Ma YT, 49: 290–296, 2013 1104, 2013 Plamondon L, Stein RL: Potent and selective 89. Lin P, Owens R, Tricot G, Wilson CS: Flow 98. Jennette JC: Overview of the 2012 revised inhibitors of the proteasome: Dipeptidyl cytometric immunophenotypic analysis of International Chapel Hill Consensus Con- boronic acids. Bioorg Med Chem Lett 8: 306 cases of multiple myeloma. Am J Clin ference nomenclature of vasculitides. Clin 333–338, 1998 Pathol 121: 482–488, 2004 Exp Nephrol 17: 603–606, 2013 79. Gardner RC, Assinder SJ, Christie G, Mason 90. Deaglio S, Vaisitti T, Billington R, Bergui L, 99. Steinmetz OM, Velden J, Kneissler U, Marx GG, Markwell R, Wadsworth H, McLaughlin Omede’ P, Genazzani AA, Malavasi F: M, Klein A, Helmchen U, Stahl RAK, Panzer

754 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW

U: Analysis and classification of B-cell infil- X, Carron P-L, Hatron P-Y, Limal N, 119. Novikov P, Moiseev S, Bulanov N, trates in lupus and ANCA-associated ne- HamidouM,DucretM,DaugasE,PapoT, Shchegoleva E: Bortezomib in refractory phritis. Kidney Int 74: 448–457, 2008 Bonnotte B, Mahr A, Ravaud P, Mouthon L; ANCA-associated vasculitis: A new option? 100. Abdulahad WH, van der Geld YM, French Vasculitis Study Group: Rituximab Ann Rheum Dis 75: e9, 2016 Stegeman CA, Kallenberg CGM: Persistent versus azathioprine for maintenance in 120. Almaani S, Meara A, Rovin BH: Update on expansion of CD4+ effector memory T cells ANCA-associated vasculitis. NEnglJMed lupus nephritis. Clin J Am Soc Nephrol 12: in Wegener’s granulomatosis. Kidney Int 371: 1771–1780, 2014 825–835, 2016 70: 938–947, 2006 109. Jones RB, Ferraro AJ, Chaudhry AN, 121. Sterner RM, Hartono SP, Grande JP: The 101. Voswinkel J, Müller A, Lamprecht P: Is PR3- Brogan P, Salama AD, Smith KGC, Savage Pathogenesis of lupus nephritis. JClinCell ANCA formation initiated in Wegener’s COS, Jayne DRW: A multicenter survey of Immunol 5: 1–7, 2014 granulomatosis lesions? Granulomas as rituximab therapy for refractory antineutrophil 122. Davidson A: What is damaging the kidney in potential lymphoid tissue maintaining au- cytoplasmic antibody-associated vasculitis. lupus nephritis? Nat Rev Rheumatol 12: toantibody production. Ann N Y Acad Sci Arthritis Rheum 60: 2156–2168, 2009 143–153, 2016 1051: 12–19, 2005 110. Fussner LA, Specks U: Can antineutrophil 123. de Leeuw K, Bungener L, Roozendaal C, 102. Lepse N, Abdulahad WH, Rutgers A, cytoplasmic antibody levels be used to in- Bootsma H, Stegeman CA: Auto-antibodies Kallenberg CGM, Stegeman CA, Heeringa form treatment of pauci-immune vasculitis? to double-stranded DNA as biomarker in P: Altered B cell balance, but unaffected B cell Curr Opin Rheumatol 27: 231–240, 2015 systemic lupus erythematosus: Comparison capacity to limit activation in anti- 111. Shah S, Hruskova Z, Segelmark M, Morgan of different assays during quiescent and neutrophil cytoplasmic antibody-associated MD, Hogan J, Lee SK, Dale J, Harper L, active disease. Rheumatology (Oxford) 56: vasculitis in remission. Rheumatology (Oxford) Tesar V, Jayne DRW, Geetha D: Treatment 698–703, 2017 53: 1683–1692, 2014 of severe renal disease in ANCA positive 124. Chang A, Henderson SG, Brandt D, Liu N, 103. Todd SK, Pepper RJ, Draibe J, Tanna A, and negative small vessel vasculitis with Guttikonda R, Hsieh C, Kaverina N, Utset Pusey CD, Mauri C, Salama AD: Regulatory rituximab. Am J Nephrol 41: 296–301, 2015 TO, Meehan SM, Quigg RJ, Meffre E, Clark B cells are numerically but not functionally 112. Bansal PJ, Tobin MC: Neonatal microscopic MR: In situ B cell-mediated immune re- deficient in anti-neutrophil cytoplasm anti- polyangiitis secondary to transfer of ma- sponses and tubulointerstitial inflammation body-associated vasculitis. Rheumatology ternal myeloperoxidase-antineutrophil cy- in human lupus nephritis. JImmunol186: (Oxford) 53: 1693–1703, 2014 toplasmic antibody resulting in neonatal 1849–1860, 2011 104. Bunch DO, McGregor JG, Khandoobhai pulmonary hemorrhage and renal in- 125. Espeli M, Bökers S, Giannico G, Dickinson NB,AybarLT,BurkartME,HuY,HoganSL, volvement. Ann Asthma Immunol HA, Bardsley V, Fogo AB, Smith KGC: Local Poulton CJ, Berg EA, Falk RJ, Nachman PH: 93: 398–401, 2004 renal autoantibody production in lupus ne- Decreased CD5+ B cells in active ANCA 113. SilvaF,SpecksU,SethiS,IrazabalMV,Fervenza phritis. JAmSocNephrol22: 296–305, vasculitis and relapse after rituximab. Clin J FC: Successful pregnancy and delivery of a 2011 Am Soc Nephrol 8: 382–391, 2013 healthy newborn despite transplacental trans- 126. Liarski VM, Kaverina N, Chang A, Brandt D, 105. Popa ER, Stegeman CA, Bos NA, fer of antimyeloperoxidase antibodies from a Yanez D, Talasnik L, Carlesso G, Herbst R, Kallenberg CG, Tervaert JW: Differential B- mother with microscopic polyangiitis. Am J Utset TO, Labno C, Peng Y, Jiang Y, Giger and T-cell activation in Wegener’s gran- Kidney Dis 54: 542–545, 2009 ML, Clark MR: Cell distance mapping ulomatosis. J Allergy Clin Immunol 103: 114. Hofstra JM, Beck LH Jr., Beck DM, Wetzels identifies functional T follicular helper cells

885–894, 1999 JF, Salant DJ: Anti-phospholipase A2 re- in inflamed human renal tissue. Sci Transl 106. Specks U, Merkel PA, Seo P, Spiera R, ceptor antibodies correlate with clinical Med 6: 230ra46, 2014 Langford CA, Hoffman GS, Kallenberg status in idiopathic membranous nephrop- 127. Choi J-Y, Ho JH-E, Pasoto SG, Bunin V, Kim CGM, St Clair EW, Fessler BJ, Ding L, athy. Clin J Am Soc Nephrol 6: 1286–1291, ST, Carrasco S, Borba EF, Gonçalves CR, Viviano L, Tchao NK, Phippard DJ, Asare 2011 Costa PR, Kallas EG, Bonfa E, Craft J: Cir- AL, Lim N, Ikle D, Jepson B, Brunetta P, 115. Holden NJ, Williams JM, Morgan MD, culating follicular helper-like T cells in sys- Allen NB, Fervenza FC, Geetha D, Keogh K, Challa A, Gordon J, Pepper RJ, Salama AD, temic lupus erythematosus: Association Kissin EY, Monach PA, Peikert T, Stegeman Harper L, Savage COS: ANCA-stimulated with disease activity. Arthritis Rheumatol C, Ytterberg SR, Mueller M, Sejismundo LP, neutrophils release BLyS and promote B 67: 988–999, 2015 Mieras K, Stone JH; RAVE-ITN Research cell survival: A clinically relevant cellular 128. Le Coz C, Joublin A, Pasquali J-L, Korganow Group: Efficacy of remission-induction process. Ann Rheum Dis 70: 2229–2233, A-S, Dumortier H, Monneaux F: Circulating regimens for ANCA-associated vasculitis. N 2011 TFH subset distribution is strongly affected Engl J Med 369: 417–427, 2013 116. Nagai M, Hirayama K, Ebihara I, Shimohata in lupus patients with an active disease. 107. Stone JH, Merkel PA, Spiera R, Seo P, H, Kobayashi M, Koyama A: Serum levels PLoS One 8: e75319, 2013 Langford CA, Hoffman GS, Kallenberg of BAFF and APRIL in myeloperoxidase 129. Suárez-Fueyo A, Bradley SJ, Klatzmann D, CGM, St Clair EW, Turkiewicz A, Tchao NK, anti-neutrophil cytoplasmic autoantibody- Tsokos GC: T cells and autoimmune kidney Webber L, Ding L, Sejismundo LP, Mieras K, associated renal vasculitis: Association with disease. Nat Rev Nephrol 13: 329–343, Weitzenkamp D, Ikle D, Seyfert-Margolis V, disease activity. Nephron Clin Pract 118: 2017 Mueller M, Brunetta P, Allen NB, Fervenza c339–c345, 2011 130. Lyssuk EY, Torgashina AV, Soloviev SK, FC, Geetha D, Keogh KA, Kissin EY, 117. Krumbholz M, Specks U, Wick M, Kalled SL, Nassonov EL, Bykovskaia SN: Reduced Monach PA, Peikert T, Stegeman C, Jenne D, Meinl E: BAFF is elevated in serum number and function of CD4+CD25high- Ytterberg SR, Specks U; RAVE-ITN Research of patients with Wegener’s granulomatosis. FoxP3+ regulatory T cells in patients with Group: Rituximab versus cyclophosphamide J Autoimmun 25: 298–302, 2005 systemic lupus erythematosus. Adv Exp for ANCA-associated vasculitis. NEnglJMed 118. Bontscho J, Schreiber A, Manz RA, Schneider W, Med Biol 601: 113–119, 2007 363: 221–232, 2010 Luft FC, Kettritz R: Myeloperoxidase-specific 131. Suárez-Fueyo A, Bradley SJ, Tsokos GC: T 108. Guillevin L, Pagnoux C, Karras A, Khouatra plasma cell depletion by bortezomib protects cells in systemic lupus erythematosus. Curr C, Aumaître O, Cohen P, Maurier F, Decaux from anti-neutrophil cytoplasmic autoanti- Opin Immunol 43: 32–38, 2016 O, Ninet J, Gobert P, Quémeneur T, bodies-induced glomerulonephritis. JAm 132. Parikh SV, Pendergraft WF, Tumlin JA, Blanchard-Delaunay C, Godmer P, Puéchal Soc Nephrol 22: 336–348, 2011 Saxena R, Solomons N, Huizinga RB:

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 755 BRIEF REVIEW www.jasn.org

Treatment of active lupus nephritis with Metzler C, Zettl U, Westphal J, Heitmann S, systemic lupus erythematosus. Arthritis voclosporin: 48 week data from the Aura-LV Herzog AL, Wiendl H, Jakob W, Schmidt E, Rheum 63: 3918–3930, 2011 study. Am J Kidney Dis 69: A2, 2017 Freivogel K, Dörner T; GRAID investigators: 148. Vincent FB, Morand EF, Mackay F: BAFF 133. Humrich JY, von Spee-Mayer C, Siegert E, Safety and clinical outcomes of rituximab and innate immunity: New therapeutic tar- Alexander T, Hiepe F, Radbruch A, therapy in patients with different autoim- gets for systemic lupus erythematosus. Im- Burmester GR, Riemekasten G: Rapid in- mune diseases: Experience from a national munol Cell Biol 90: 293–303, 2012 duction of clinical remission by low-dose registry (GRAID). Arthritis Res Ther 13: R75, 149. Merrill JT, van Vollenhoven RF, Buyon JP, -2 in a patient with refractory 2011 Furie RA, Stohl W, Morgan-Cox M, Dickson SLE. Ann Rheum Dis 74: 791–792, 2015 142. Duxbury B, Combescure C, Chizzolini C: C, Anderson PW, Lee C, Berclaz P-Y, Dörner 134. von Spee-Mayer C, Siegert E, Abdirama D, Rituximab in systemic lupus erythematosus: T: Efficacy and safety of subcutaneous taba- Rose A, Klaus A, Alexander T, Enghard P, An updated systematic review and meta- lumab, a to B-cell acti- Sawitzki B, Hiepe F, Radbruch A, Burmester analysis. Lupus 22: 1489–1503, 2013 vating factor, in patients with systemic lupus GR, Riemekasten G, Humrich JY: Low-dose 143. Hahn BH, McMahon MA, Wilkinson A, erythematosus: Results from ILLUMINATE-2, interleukin-2 selectively corrects regulatory Wallace WD, Daikh DI, Fitzgerald JD, a 52-week, phase III, multicentre, randomised, T cell defects in patients with systemic lupus Karpouzas GA, Merrill JT, Wallace double-blind, placebo-controlled study. Ann erythematosus. Ann Rheum Dis 75: 1407– DJ, Yazdany J, Ramsey-Goldman R, Singh Rheum Dis 75: 332–340, 2016 1415, 2016 K, Khalighi M, Choi S-I, Gogia M, Kafaja 150. Isenberg DA, Petri M, Kalunian K, Tanaka Y, 135. He J, Zhang X, Wei Y, Sun X, Chen Y, Deng S, Kamgar M, Lau C, Martin WJ, Parikh S, Urowitz MB, Hoffman RW, Morgan-Cox M, J, Jin Y, Gan Y, Hu X, Jia R, Xu C, Hou Z, Peng J, Rastogi A, Chen W, Grossman JM; Iikuni N, Silk M, Wallace DJ: Efficacy and LeongYA,ZhuL,FengJ,AnY,JiaY,LiC, American College of Rheumatology: Ameri- safety of subcutaneous tabalumab in patients Liu X, Ye H, Ren L, Li R, Yao H, Li Y, Chen S, can College of Rheumatology guidelines for with systemic lupus erythematosus: Results Zhang X, Su Y, Guo J, Shen N, Morand EF, screening, treatment, and management of from ILLUMINATE-1, a 52-week, phase III, Yu D, Li Z: Low-dose interleukin-2 treatment lupus nephritis. Arthritis Care Res (Hoboken) multicentre, randomised, double-blind, pla- selectively modulates CD4(+) T cell subsets 64: 797–808, 2012 cebo-controlled study. Ann Rheum Dis 75: in patients with systemic lupus eryth- 144. Bertsias GK, Tektonidou M, Amoura Z, 323–331, 2016 ematosus. Nat Med 22: 991–993, 2016 Aringer M, Bajema I, Berden JHM, Boletis J, 151. Isenberg D, Gordon C, Licu D, Copt S, Rossi 136. Vital EM, Dass S, Buch MH, Henshaw K, Cervera R, Dörner T, Doria A, Ferrario F, CP, Wofsy D: Efficacy and safety of ataci- Pease CT, Martin MF, Ponchel F, Rawstron Floege J, Houssiau FA, Ioannidis JPA, cept for prevention of flares in patients with AC, Emery P: B cell biomarkers of rituximab Isenberg DA, Kallenberg CGM, Lightstone moderate-to-severe systemic lupus eryth- responses in systemic lupus erythematosus. L, Marks SD, Martini A, Moroni G, Neumann ematosus (SLE): 52-week data (APRIL-SLE Arthritis Rheum 63: 3038–3047, 2011 I, Praga M, Schneider M, Starra A, Tesar V, randomised trial). Ann Rheum Dis 74: 2006– 137. Iwata S, Tanaka Y: B-cell subsets, signaling Vasconcelos C, van Vollenhoven RF, 2015, 2015 and their roles in secretion of autoanti- Zakharova H, Haubitz M, Gordon C, Jayne 152. Ehrenstein MR, Wing C: The BAFFling ef- bodies. Lupus 25: 850–856, 2016 D, Boumpas DT: Joint European League fects of rituximab in lupus: Danger ahead? 138. Sellam J, Rouanet S, Hendel-Chavez H, Against Rheumatism and European Renal Nat Rev Rheumatol 12: 367–372, 2016 Abbed K, Sibilia J, Tebib J, Le Loët X, Combe Association-European Dialysis and Trans- 153. Ichikawa HT, Conley T, Muchamuel T, Jiang B, Dougados M, Mariette X, TaoufikY:Blood plant Association (EULAR/ERA-EDTA) rec- J, Lee S, Owen T, Barnard J, Nevarez S, memory B cells are disturbed and predict the ommendations for the management of Goldman BI, Kirk CJ, Looney RJ, Anolik JH: response to rituximab in patients with rheu- adult and paediatric lupus nephritis. Ann Beneficial effect of novel proteasome inhibi- matoid arthritis. Arthritis Rheum 63: 3692– Rheum Dis 71: 1771–1782, 2012 tors in murine lupus via dual inhibition of type 3701, 2011 145. Condon MB, Ashby D, Pepper RJ, Cook HT, I interferon and autoantibody-secreting cells. 139. Leandro MJ, Edwards JC, Cambridge G, Levy JB, Griffith M, Cairns TD, Lightstone L: Arthritis Rheum 64: 493–503, 2012 Ehrenstein MR, Isenberg DA: An open Prospective observational single-centre 154. Seavey MM, Lu LD, Stump KL, Wallace NH, study of B lymphocyte depletion in systemic cohort study to evaluate the effectiveness Ruggeri BA: Novel, orally active, proteasome lupus erythematosus. Arthritis Rheum 46: of treating lupus nephritis with rituximab inhibitor, delanzomib (CEP-18770), amelio- 2673–2677, 2002 and mycophenolate mofetil but no oral rates disease symptoms and glomerulone- 140. Rovin BH, Furie R, Latinis K, Looney RJ, steroids. Ann Rheum Dis 72: 1280–1286, phritis in two preclinical mouse models of SLE. Fervenza FC, Sanchez-Guerrero J, Maciuca 2013 Int Immunopharmacol 12: 257–270, 2012 R, Zhang D, Garg JP, Brunetta P, Appel G; 146. Navarra SV, Guzmán RM, Gallacher AE, Hall 155. Alexander T, Sarfert R, Klotsche J, Kühl AA, LUNAR Investigator Group: Efficacy and S, Levy RA, Jimenez RE, Li EK-M, Thomas M, Rubbert-Roth A, Lorenz H-M, Rech J, Hoyer safety of rituximab in patients with active Kim H-Y, León MG, Tanasescu C, Nasonov BF, Cheng Q, Waka A, Taddeo A, Wiesener proliferative lupus nephritis: The Lupus E, Lan J-L, Pineda L, Zhong ZJ, Freimuth W, M, Schett G, Burmester G-R, Radbruch A, Nephritis Assessment with Rituximab study. Petri MA; BLISS-52 Study Group: Efficacy Hiepe F, Voll RE: The proteasome inhibitior Arthritis Rheum 64: 1215–1226, 2012 and safety of belimumab in patients with bortezomib depletes plasma cells and 141. Tony H-P, Burmester G, Schulze-Koops H, active systemic lupus erythematosus: A ameliorates clinical manifestations of re- Grunke M, Henes J, Kötter I, Haas J, Unger randomised, placebo-controlled, phase 3 fractory systemic lupus erythematosus. Ann L, Lovric S, Haubitz M, Fischer-Betz R, trial. Lancet 377: 721–731, 2011 Rheum Dis 74: 1474–1478, 2015 Chehab G, Rubbert-Roth A, Specker C, 147. Furie R, Petri M, Zamani O, Cervera R, 156. Zhang H, Liu Z, Huang L, Hou J, Zhou M, Weinerth J, Holle J, Müller-Ladner U, König Wallace DJ, Tegzová D, Sanchez-Guerrero Huang X, Hu W, Liu Z: The short-term effi- R, Fiehn C, Burgwinkel P, Budde K, J, Schwarting A, Merrill JT, Chatham WW, cacy of bortezomib combined with gluco- Sörensen H, Meurer M, Aringer M, Kieseier Stohl W, Ginzler EM, Hough DR, Zhong ZJ, corticoids for the treatment of refractory B, Erfurt-Berge C, Sticherling M, Veelken R, Freimuth W, van Vollenhoven RF; BLISS-76 lupus nephritis. Lupus 26: 952–958, 2017 Ziemann U, Strutz F, von Wussow P, Meier Study Group: A phase III, randomized, pla- 157. Loupy A, Jordan SC: Transplantation: FM, Hunzelmann N, Schmidt E, Bergner R, cebo-controlled study of belimumab, a Donor-specific HLA antibodies and renal Schwarting A, Eming R, Hertl M, Stadler R, monoclonal antibody that inhibits B lym- allograft failure. Nat Rev Nephrol 9: 130– Schwarz-Eywill M, Wassenberg S, Fleck M, phocyte stimulator, in patients with 131, 2013

756 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018 www.jasn.org BRIEF REVIEW

158. Mohan S, Palanisamy A, Tsapepas D, 170. Chen Y-B, Kawai T, Spitzer TR: Combined alloantibody production. Am J Transplant Tanriover B, Crew RJ, Dube G, Ratner LE, bone marrow and kidney transplantation for 9: 201–209, 2009 Cohen DJ, Radhakrishnan J: Donor-specific the induction of specific tolerance. Adv 181. Ejaz NS, Alloway RR, Halleck F, Dürr M, antibodies adversely affect kidney allograft Hematol 2016: 6471901–6471908, 2016 Budde K, Woodle ES: Review of bortezomib outcomes. J Am Soc Nephrol 23: 2061– 171. Macklin PS, Morris PJ, Knight SR: A sys- treatment of antibody-mediated rejection 2071, 2012 tematic review of the use of rituximab for in renal transplantation. Antioxid Redox 159. Loupy A, Lefaucheur C, Vernerey D, Prugger the treatment of antibody-mediated renal Signal 21: 2401– 2418, 2014 C, Duong van Huyen J-P, Mooney N, transplant rejection. Transplant Rev (Or- 182. Eskandary F, Bond G, Schwaiger E, Kikic Z, Suberbielle C, Frémeaux-Bacchi V, Méjean A, lando) 31: 87–95, 2017 Winzer C, Wahrmann M, Marinova L, Desgrandchamps F, Anglicheau D, Nochy D, 172. Jackson AM, Kraus ES, Orandi BJ, Segev Haslacher H, Regele H, Oberbauer R, Charron D, Empana J-P, Delahousse M, DL, Montgomery RA, Zachary AA: A closer Böhmig GA: Bortezomib in late antibody- Legendre C, Glotz D, Hill GS, Zeevi A, Jouven look at rituximab induction on HLA anti- mediated kidney transplant rejection X: Complement-binding anti-HLA antibodies body rebound following HLA-incompatible (BORTEJECT Study): Study protocol for a and kidney-allograft survival. N Engl J Med kidney transplantation. Kidney Int 87: 409– randomized controlled trial. Trials 15: 107, 369: 1215–1226, 2013 416, 2015 2014 160. Carroll MC, Isenman DE: Regulation of hu- 173. Koenig A, Thaunat O: Lymphoid neo- 183. Sango C, Merino D, San Segundo D, moral immunity by complement. Immunity genesis and tertiary lymphoid organs in Rodrigo E, Lopez-Hoyos M, Benito A, 37: 199–207, 2012 transplanted organs. Front Immunol 7: 646, Ángeles Ramos M, Gómez-Román J, Arias 161. Burns AM, Chong AS: Alloantibodies pre- 2016 M: B-cell-activating factor levels are asso- vent the induction of transplantation toler- 174. Thaunat O, Graff-Dubois S, Brouard S, ciated with antibody-mediated histological ance by enhancing alloreactive T cell Gautreau C, Varthaman A, Fabien N, Field damage in kidney transplantation. Trans- . J Immunol 186: 214–221, 2011 A-C, Louedec L, Dai J, Joly E, Morelon E, plant Proc 48: 2910–2912, 2016 162. Burns AM, Ma L, Li Y, Yin D, Shen J, Xu J, Soulillou J-P, Michel J-B, Nicoletti A: Im- 184. Pongpirul W, Chancharoenthana W, Pongpirul K, Chong AS: Memory alloreactive B cells and mune responses elicited in tertiary lym- Leelahavanichakul A, Kittikowit W, Jutivorakool K, alloantibodies prevent anti-CD154-mediated phoid tissues display distinctive features. Nonthasoot B, Avihingsanon Y, Eiam-Ong S, allograft acceptance. JImmunol182: 1314– PLoS One 5: e11398, 2010 Praditpornsilpa K, Townamchai N: B-cell 1324, 2009 175. Dufek S, Khalil A, Mamode N, Sebire NJ, activating factor (BAFF), a predictor of anti- 163. Schinstock CA, Stegall M, Cosio F: New in- Marks SD: Plasma-cell-rich infiltrates in body mediated rejection in kidney trans- sights regarding chronic antibody-mediated paediatric renal transplant biopsies are as- plantation recipients [published online rejection and its progression to transplant glo- sociated with increased risk of renal allo- ahead of print Nov 26, 2016]. Nephrology merulopathy. Curr Opin Nephrol Hypertens graft failure. Pediatr Nephrol 32: 679–684, (Carlton) 10.1111/nep.12972 23: 611–618, 2014 2017 185. Mujtaba MA, Komocsar WJ, Nantz E, 164. Perry DK, Pollinger HS, Burns JM, Rea D, 176. Hasegawa J, Honda K, Wakai S, Shirakawa Samaniego MD, Henson SL, Hague JA, Ramos E, Platt JL, Gloor JM, Stegall MD: H, Omoto K, Okumi M, Ishida H, Tanabe K: Lobashevsky AL, Higgins NG, Czader M, Two novel assays of alloantibody-secreting Plasma cell-rich rejection after kidney Book BK, Anderson MD, Pescovitz MD, cells demonstrating resistance to de- transplantation and the role of donor-spe- Taber TE: Effect of treatment with sensitization with IVIG and rATG. Am J cific antibodies: A case report and review of tabalumab, a B cell-activating factor inhibitor, Transplant 8: 133–143, 2008 the literature. Transplant Proc 47: 2533– on highly sensitized patients with end-stage 165. Wiebe C, Pochinco D, Blydt-Hansen TD, Ho 2536, 2015 renal disease awaiting transplantation. Am J J, Birk PE, Karpinski M, Goldberg A, 177. Katsuma A, Yamamoto I, Komatsuzaki Y, Transplant 16: 1266–1275, 2016 Storsley LJ, Gibson IW, Rush DN, Nickerson Niikura T, Kawabe M, Okabayashi Y, 186. Jordan SC, Lorant T, Choi J, Kjellman C, PW: Class II HLA matching-A strat- Yamakawa T, Katsumata H, Nakada Y, Winstedt L, Bengtsson M, Zhang X, Eich T, egy to minimize de novo donor-specifican- Kobayashi A, Tanno Y, Miki J, Yamada H, Toyoda M, Eriksson B-M, Ge S, Peng A, tibody development and improve outcomes. Ohkido I, Tsuboi N, Yamamoto H, Yokoo T: Järnum S, Wood KJ, Lundgren T, Am J Transplant 13: 3114–3122, 2013 Subclinical antibody-mediated rejection Wennberg L, Bäckman L, Larsson E, 166. Heeger PS, Greenspan NS, Kuhlenschmidt due to anti-human-leukocyte-antigen- Villicana R, Kahwaji J, Louie S, Kang A, Haas S, Dejelo C, Hricik DE, Schulak JA, Tary- DR53 antibody accompanied by plasma M, Nast C, Vo A, Tufveson G: IgG endo- Lehmann M: Pretransplant frequency of cell-rich acute rejection in a patient with peptidase in highly sensitized patients un- donor-specific, IFN-gamma-producing lym- cadaveric kidney transplantation. Nephrol- dergoing transplantation. NEnglJMed phocytes is a manifestation of immunologic ogy (Carlton) 21[Suppl 1]: 31–34, 2016 377: 442–453, 2017 memory and correlates with the risk of post- 178. Uppin MS, Gudithi S, Taduri G, Prayaga AK, 187. Dörner T, Radbruch A, Burmester GR: B- transplant rejection episodes. J Immunol 163: Raju SB: Expanding the antibody-mediated cell-directed therapies for autoimmune 2267–2275, 1999 component of plasma cell-rich acute re- disease. Nat Rev Rheumatol 5: 433–441, 167. Gorbacheva V, Fan R, Fairchild RL, Baldwin WM jection: A case series. Indian J Nephrol 26: 2009 3rd, Valujskikh A: Memory CD4 T cells induce 176–181, 2016 188. Merrill JT, Neuwelt CM, Wallace DJ, antibody-mediated rejection of renal allografts. 179. Vogelbacher R, Meister S, Gückel E, Starke Shanahan JC, Latinis KM, Oates JC, Utset JAmSocNephrol27: 3299–3307, 2016 C, Wittmann S, Stief A, Voll R, Daniel C, TO, Gordon C, Isenberg DA, Hsieh H-J, 168. Kazatchkine MD, Kaveri SV: Immunomodulation Hugo C: Bortezomib and inhibit the Zhang D, Brunetta PG: Efficacy and safety of autoimmune and inflammatory diseases with chronic active antibody-mediated rejection in of rituximab in moderately-to-severely intravenous immune globulin. NEnglJMed experimental renal transplantation in the rat. active systemic lupus erythematosus: The 345: 747–755, 2001 Nephrol Dial Transplant 25: 3764–3773, randomized, double-blind, phase II/III sys- 169. Abu Jawdeh BG, Cuffy MC, Alloway RR, 2010 temic lupus erythematosus evaluation of rit- Shields AR, Woodle ES: Desensitization in 180. Perry DK, Burns JM, Pollinger HS, Amiot uximab trial. Arthritis Rheum 62: 222–233, kidney transplantation: Review and future BP,GloorJM,GoresGJ,StegallMD: 2010 perspectives. Clin Transplant 28: 494–507, Proteasome inhibition causes apoptosis of 189. Sautenet B, Blancho G, Büchler M, Morelon 2014 normal human plasma cells preventing E, Toupance O, Barrou B, Ducloux D,

J Am Soc Nephrol 29: 741–758, 2018 B Cells and Plasma Cells in Glomerular Diseases 757 BRIEF REVIEW www.jasn.org

Chatelet V, Moulin B, Freguin C, Hazzan M, neutrophils in the induction of glomerulo- expression of lupus nephritis in New Zea- Lang P, Legendre C, Merville P, Mourad G, nephritis by anti-myeloperoxidase anti- land mixed H-2z homozygous inbred strains Mousson C, Pouteil-Noble C, Purgus R, bodies. Am J Pathol 167: 39–45, 2005 of mice derived from New Zealand black Rerolle J-P, Sayegh J, Westeel P-F, Zaoui P, 196. Little MA, Smyth L, Salama AD, Mukherjee and New Zealand white mice. Origins and Boivin H, Le Gouge A, Lebranchu Y: One- S, Smith J, Haskard D, Nourshargh S, Cook initial characterization. Lab Invest 68: 419– year results of the effects of rituximab on acute HT, Pusey CD: Experimental autoimmune 426, 1993 antibody-mediated rejection in renal trans- vasculitis: An animal model of anti-neutrophil 202. Morel L: Mapping lupus susceptibility plantation: RITUX ERAH, a multicenter double- cytoplasmic autoantibody-associated sys- genes in the NZM2410 mouse model. Adv blind randomized placebo-controlled trial. temic vasculitis. Am J Pathol 174: 1212– Immunol 115: 113–139, 2012 Transplantation 100: 391–399, 2016 1220, 2009 203. Cohen MG, Pollard KM, Schrieber L: Re- 190. Silva-Fernández L, Loza E, Martínez- 197. Chavele K-M, Shukla D, Keteepe-Arachi T, lationship of age and sex to autoantibody Taboada VM, Blanco R, Rúa-Figueroa I, Seidel JA, Fuchs D, Pusey CD, Salama AD: expression in MRL-+/+ and MRL-lpr/lpr Pego-Reigosa JM, Muñoz-Fernández S; Regulation of myeloperoxidase-specificTcell mice: Demonstration of an association Systemic Autoimmune Diseases Study responses during disease remission in anti- between the expression of antibodies Group of the Spanish Society for Rheuma- neutrophil cytoplasmic antibody-associated to histones, denatured DNA and Sm in tology (EAS-SER): Biological therapy for vasculitis: The role of Treg cells and trypto- MRL-+/+ mice. Clin Exp Immunol 72: 50– systemic vasculitis: A systematic review. phan degradation. Arthritis Rheum 62: 1539– 54, 1988 Semin Arthritis Rheum 43: 542–557, 2014 1548, 2010 204. Herlands RA, William J, Hershberg U, 191. Chong AS, Sciammas R: Memory B cells in 198. Little MA, Al-Ani B, Ren S, Al-Nuaimi H, Shlomchik MJ: Anti-chromatin anti- transplantation. Transplantation 99: 21–28, Leite M Jr., Alpers CE, Savage CO, Duffield bodies drive in vivo antigen-specificac- 2015 JS: Anti-proteinase 3 anti-neutrophil cyto- tivation and somatic hypermutation of 192. Jacob N, Stohl W: Autoantibody-dependent plasm autoantibodies recapitulate systemic rheumatoid factor B cells at extra- and autoantibody-independent roles for B vasculitis in mice with a humanized immune follicular sites. Eur J Immunol 37: 3339– cells in systemic lupus erythematosus: Past, system. PLoS One 7: e28626, 2012 3351, 2007 present, and future. Autoimmunity 43: 84– 199. Dixon FJ, Andrews BS, Eisenberg RA, 205. Uehara S, Chase CM, Cornell LD, Madsen 97, 2010 McConahey PJ, Theofilopoulos AN, Wilson JC, Russell PS, Colvin RB: Chronic cardiac 193. Xiao H, Heeringa P, Hu P, Liu Z, Zhao M, CB: Etiology and pathogenesis of a spon- transplant arteriopathy in mice: Relation- Aratani Y, Maeda N, Falk RJ, Jennette JC: taneous lupus-like syndrome in mice. Ar- ship of alloantibody, C4d deposition and Antineutrophil cytoplasmic autoantibodies thritis Rheum 21[5 Suppl]: S64–S67, 1978 neointimal fibrosis. Am J Transplant 7: 57– specific for myeloperoxidase cause glo- 200. Andrews BS, Eisenberg RA, 65, 2007 merulonephritis and vasculitis in mice. J TheofilopoulosAN,IzuiS,WilsonCB, 206. Kwun J, Oh BC, Gibby AC, Ruhil R, Lu VT, Clin Invest 110: 955–963, 2002 McConahey PJ, Murphy ED, Roths JB, Kim DW, Page EK, Bulut OP, Song MQ, 194. Dumoitier N, Terrier B, London J, Lofek S, Dixon FJ: Spontaneous murine lupus-like Farris AB, Kirk AD, Knechtle SJ, Iwakoshi Mouthon L: Implication of B in syndromes. Clinical and immunopathological NN: Patterns of de novo allo B cells and the pathogenesis of ANCA-associated vascu- manifestations in several strains. JExpMed antibody formation in chronic cardiac allo- litides. Autoimmun Rev 14: 996–1004, 2015 148: 1198– 1215, 1978 graft rejection after alemtuzumab treat- 195. Xiao H, Heeringa P, Liu Z, Huugen D, Hu P, 201. Rudofsky UH, Evans BD, Balaban SL, ment. Am J Transplant 12: 2641–2651, Maeda N, Falk RJ, Jennette JC: The role of Mottironi VD, Gabrielsen AE: Differences in 2012

758 Journal of the American Society of Nephrology J Am Soc Nephrol 29: 741–758, 2018