/
B cells are multifunctional lymphocytes that contribute to the pathoge B cells are multifunctional lymphocytes that contribute to the pathoge

B cells are multifunctional lymphocytes that contribute to the pathoge - PDF document

ashley
ashley . @ashley
Follow
342 views
Uploaded On 2022-08-16

B cells are multifunctional lymphocytes that contribute to the pathoge - PPT Presentation

Antibody B cell checkpoints in autoimmune SamuelJSRubin MichelleSBloom and WilliamHRobinsonAbstract B cells have important functions in the pathogenesis of autoimmune diseases including ID: 936993

cells cell activation autoimmune cell cells autoimmune activation antibody autoimmunity act xualt xext0 treatment receptor immune arthritis patients bcr

Share:

Link:

Embed:

Download Presentation from below link

Download Pdf The PPT/PDF document "B cells are multifunctional lymphocytes ..." is the property of its rightful owner. Permission is granted to download and print the materials on this web site for personal, non-commercial use only, and to display it on your personal computer provided you do not modify the materials and that you retain all copyright notices contained in the materials. By downloading content from our website, you accept the terms of this agreement.


Presentation Transcript

B cells are multifunctional lymphocytes that contribute to the pathogenesis of autoimmune diseases via B cell-intrinsic, antibody-mediated and Tcell-dependent mechanisms. Although antibody production by B cells promotes both mediated cytotoxicity(ADCC) and dependent cytotoxicity (CDC), B cells can also present antigen and provide Tcell help. B cell activation and effector functions are regulated by immune checkpoints, including activating and Antibody- B cell checkpoints in autoimmune SamuelJ.S.Rubin, MichelleS.Bloom and WilliamH.RobinsonAbstract B cells have important functions in the pathogenesis of autoimmune diseases, including autoimmune rheumatic diseases. In addition to producing autoantibodies, B cells contribute to autoimmunity by serving as professional antigen-presenting cells (APCs), producing cytokines, and through additional mechanisms. B cell activation and effector functions are regulated D[|KOOWPGEJGEMRQKPVUKPENWFKPIDQVJCEVKXCVKPICPFKPJKDKVQT[EJGEMRQKPVTGEGRVQTUVJCVEQPVTKDWVGVQVJGTGIWNCVKQPQH$EGNNVQNGTCPEGCEVKXCVKQPCPVKIGPRTGUGPVCVKQP6|EGNNJGNRENCUU|UYKVEJKPICPVKDQF[RTQFWEVKQPCPFE[VQMKPGRTQFWEVKQP6JGXCTKQWUCEVKXCVKPIEJGEMRQKPV presentation, Tcell help, cytokine secretion and potentially other processes. Further, we discuss the mechanisms of B cell activation by antigen and co-receptor ligands, as well as B cell regulatory networks governed by inhibitory receptors. Finally, we present an overview of current and next-generation therapeutic strategies for targeting B cells and B cell checkpoints for the treatment of autoimmune rheumatic diseases.B cell functions including autoantibody production, antigen presentation, Tcell help and cytokine production all contribute to the pathogenesis of autoimmune diseases . As a result of the growing appreciation of these functions in self-tolerance and the mechanisms by which they contribute to autoimmunity, next-generation therapeutic approaches are focusing on specifically modulating B cell activation and effector mechanisms rather than globally depleting B cells.The classic paradigm of B cell-mediated autoimmune disease centres around production of autoantibodiesThere are multiple mechanisms by which autoantibodies contribute to the pathogenesis of autoimmune disease. First, immune complexes can form or deposit in tissues where they can activate complement and induce CDC to cause tissue damage. For example, in a mouse model of lupus nephritis, blockade of B cell co-stimulation in combination with cyclophosphamide treatment reduced immune complex deposition and glomerulonephritis and preserved renal function. RA is associated with the production of rheumatoid factor, autoantibodies that bind the Fc region of IgG, and anti-citrullinated protein antibodies (ACPAs). In RA, both rheumatoid factor-containing and ACPA-containing immune complexes activate complement pathways in joints, leading to the production of C5a and the generation of the membrane attack complex (MAC), which both contribute to joint damage; IgM rheumatoid factor can also increase complement activation mediated by ACPA-containing immune complexes23. Second, autoantibodies can promote tissue damage via ADCC by co-engagement of antigens on the target tissue and Fc receptors (FcRs) on macrophages, neutrophils, natural killer (NK) cells and other effector cell types. For example, lupus-prone mice with mutations in the IgG FcR that abrogate effector cell engagement had reduced glomerulonephritis and improved renal outcomes. Third, autoantibody-containing immune complexes can also activate immune cells through dual engagement of FcRs and Toll-like receptors (TLRs) (on macrophages and dendritic cells) or dual engagement of the B cell receptor (BCR) and TLRs (on B cells; as discussed in a later section). For example, ACPAs, a hallmark of RA, form immune complexes with citrullinated proteins that can stimulate macrophages via dual engagement of the FcR and TLR4 to produce pro-inflammatory cytokines. IgG rheumatoid factor can crosslink immune complexes to potentiate citrullinated antigen–immune complex-mediated macrophage activation and cytokine production. Finally, immune complexes facilitate antigen loading onto dendritic cells via immune complexes, enabling these cells to efficiently activate Tcells2830. Together, these examples illustrate the diverse mechanisms through which autoantibodies contribute to pathologies seen in autoimmune disease.6|EGNNs$EGNNKPVGTCEVKQPUAlthough autoantibody production is widely implicated in the pathogenesis of autoimmune diseases, pathological interactions between B cells and Tcells can also contribute to autoimmunity. B cells are one of a few cell types that can function as professional APCs through constitutive expression of MHC class II molecules. Although dendritic cells are thought to be the primary initiators of naive CD4 Tcell responses, B cells can also interact with and activate CD4 Tcells via MHC class II-mediated antigen presentation, and CD4 Tcells, in turn, provide help to cognate B cells. During an immune response, naive CD4 Tcells are primed by antigen-presenting dendritic cells and subsequently differentiate into T helper cell subsets including T follicular helper ) cells. In the germinal centre, Tcells interact with cognate B cells to promote isotype switching and somatic hypermutation, as well as B cell differentiation into memory B cells and plasma cellsDysregulated antigen presentation is implicated in the pathogenesis of autoimmunity. For example, some HLA-DRB1 alleles can bind citrullinated peptides33and are associated with the development of RAFurthermore, other HLA class I and class II alleles have been associated with susceptibility to systemic lupus erythematosus (SLE) and ankylosing spondylitisPathological Tcell–B cell interactions are also implicated by the presence of ectopic lymphoid structures in inflamed tissues (for example, in the synovium in RA), as well as by evidence of autoantibody affini

ty maturationFor example, in mouse models of SLE, antinuclear antibodies (ANAs) undergo somatic hypermutation to become high-affinity (and hence, highly pathogenic) autoantibodies. Additionally, ACPAs and rheumatoid factor from patients with RA show signs of somatic hypermutation, implicating the involvement of affinity maturation in RA pathogenesisIn addition to B cell–T cell interactions, interactions between B cells and other cells can be dysregulated in autoimmunity. For example, a distinct Tcell population Key pointsey pointsþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;ECPþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;RTQFWEGRTQHGUUKQPCNRTGUGPVKPIRTQFWEGQFWEGr$þÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;EGNNUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;GZRTGUUTGEGRVQTUGEGRVQTUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;TGEGRVQTUTGIWNCVGTGEGRVQTUTGEGRVQT6QNNTGEGRVQTUGEGRVQTUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;TGEGRVQTUGEGRVQTUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;TGEGRVQTUGEGRVQTUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;TGEGRVQTUGEGRVQTUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;KOOWPQINQDWNKPþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;þÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;(ETGIKQPTGEGRVQT\n(ERTQITCOOGFTGEGRVQTUVTCPUOKVCPUOKVþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;UKIPCNUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;VQþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;$þÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;EGNNUr8CTKQWUVCTIGVKPIUVTCVGIKGUVTGCVOGPVRTQQþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;E[VQMKPGUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;VTKIIGTKPIþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;QHþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;$þÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;EGNNþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;KPJKDKVQT[þÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;EJGEMRQKPVUþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;CPFþÿ &#x/Act;&#xualT;xt0;&#x/Act;&#xualT;xt0;VTCHHKEMKPI Fc regionThe tail region of an antibody, containing two heavy chain constant domains, that interacts with Fc receptors (FcRs) to mediate immune cell effector functions.Ectopic lymphoid structureslso known as tertiary lymphoid structures; organized aggregates of lymphocytes and other cells that possess some features of germinal centres. These structures can develop in chronically inflamed nonlymphoid tissues such asthe synovium in rheumatoid arthritis.Affinity maturation process in the germinal centre by which cells, following interaction and activation by follicular helper Tcells, undergo immunoglobulin gene mutation and subsequent selection to generate cells that express antibodies with increased affinity for the target antigen. that can augment B cell responses in nonlymphoid tissue (referred to as peripheral T helper cells) is expanded in the synovium of patients with RA. Inaddition, in RA, fibroblast-like synoviocytes can interact with and provide pro-survival factors to B cellsTogether, these findings suggest the important involvement of B cell–T cell interactions in autoimmunity. Thus, targeting these interactions could be effective for preventing the development and progression of autoimmune diseases.%[VQMKPGRTQFWEVKQPIn addition to autoantibody production and antigen presentation, B cells can regulate immune responses through the production of cytokines. B cells can produce pro-inflammatory cytokines such as IFN, IL-6 and IL-2 as well as anti-inflammatory cytokines such as IL-10 and IL-4 . Multiple sclerosis, in particular, involves perturbations in cytokine production by B cells, but B cell-mediated cytokine secretion is also perturbed in other autoimmune diseases such as SLE and RAThe production of IL-6 and IFN by B cells is required for spontaneous germinal centre formation and Tcell differentiation in a mouse model of SLE, and this process probably also occurs in other autoimmune diseases (for example, IFN-expressing B cells are expanded in multiple sclerosis). B cell-derived IL-6 can promote Bcell proliferation as well as exert pleiotropic effects on Tcells and other cell types (reviewed elsewhere). Thus, therapeutically targeting B cells could prevent loss of tolerance in both B cells and Tcells as well as reduce inflammatory responses that can promote autoimmunity.In contrast to pro-inflammatory B cell responses, regulatory B (Breg) cells are characterized by the production of anti-inflammatory cytokines such as IL-10, TGF and IL-35. IL-10-producing Breg cells (so-called B10 cells) have important protective functions against the development of autoimmunity, including in mouse models of autoimmunity and in multiple sclerosis, SLE and RA. TGF is produced by some Breg cells and can also modulate Tcell activity. Additionally, in 2014, researchers described a population of IL-35-expressing Breg cells that can suppress autoimmunityHowBreg cells mediate their suppressive functions is not fullyunderstood, and additional work is needed to fully define their developmental paths and regulatory functions in immune responses as well as the mechanisms by which they can suppress autoimmunity (reviewed elsewhere). Understanding these mechanisms could facilitate development of B cell-modulating therapies for the treatment of autoimmune rheumatic disease.B cell activationB cell activation is important for adaptive immune responses and is regulated by key stimulatory and inhibitory checkpoints. B cell inhibitory checkpoints serve to both inhibit activation of autoreactive B cells and dampen overstimulated responses . For non-polyvalent antigens, two signals are required for the activation of B cells: the engagement of the BCR and stimulatory signal. Anti

gen binding to the BCR defines the specificity of the B cell response, whereas stimulatory functions are important for overcoming inhibitory checkpoints.Throughout the development of B cells, both positive and negative selection via the BCR shape the mature Bcell repertoire and reduce reactivity to autoantigens. Early in the development of B cells, autoreactive B cells in the bone marrow undergo the process of negative selection that involves receptor editing and/or cell deletion. The detection of extraneous autoreactive naive Bcells in both SLE and RA indicates that dysregulation of these central tolerance mechanisms contributes to autoimmunity. Once B cells migrate to the periphery, additional inhibitory checkpoint pathways ensure that residual autoreactive B cells are eliminated or silenced, whereas co-stimulatory signals ensure that only B cells with non-autoreactive BCRs are activated. Many of these stimulatory and inhibitory checkpoint pathways function indirectly to control the survival, proliferation and activation of B cells. Ongoing studies by multiple groups aim to further define and characterize the effect of individual stimulatory and inhibitory checkpoints on antigen presentation, antibody production, co-stimulation, reg cell activity and other B cell functions.In addition to extrinsic regulatory signals, conformational changes in the BCR are also required to activate B cells. There are two predominant models for how antigen binding and BCR oligomerization mediate the activation of B cells. The conformation-induced model posits that activation occurs via crosslinking of multiple BCRs on the B cell membrane, which then triggers downstream signalling. The dissociation activation model alternatively suggests that unstimulated BCRs reside in auto-inhibited oligomers on the B cell membrane and that these structures are inaccessible to the kinases involved in B cell activation. Antigen binding promotes the opening of clustered BCR oligomers, enabling access to these kinases and downstream signalling (reviewed elsewhere). Regardless of these models, B cell CD28MHCclass IITCRCD80 or AutoreactiveAutoreactive Autoantibodies Autoantigen Autoantibodyproduction Co-stimulation production Antigenpresentation (KIThe functions of B cells in autoimmune disease. Central B cell functions, KPENWFKPICPVKDQF[RTQFWEVKQPCPVKIGPRTGUGPVCVKQPCPF6|EGNNJGNRXKCEQUVKOWNCVKQPand/or cytokine secretion, can all contribute to the pathogenesis of autoimmune FKUGCUGU$%4$EGNNTGEGRVQT6%46|EGNNTGEGRVQT activation by either model is dependent on non-covalent interactions between the BCR and the invariant Ig and Ig chains (also known as CD79A and CD79B). These chains contain immunoreceptor tyrosine-based activation motifs (ITAMs) that, upon antigen engagement of the BCR, are phosphorylated by the tyrosine kinase LYN. Downstream activation signals are then amplified and relayed by the SYK and BTK kinases (reviewed elsewhere). Thus, BCR activation is mediated by activating co-receptors and their associated kinases, which requires overcoming constitutive inhibitory signals from inhibitory co-receptors.$EGNNUVKOWNCVQT[RCVJYC[UB cell stimulatory checkpoints are important in the regulation of B cell activation. These stimulatory checkpoints consist of numerous cytokines, cytokine receptors, other cell surface receptors and downstream signalling pathways. Cognate Tcells promote B cell and plasma cell differentiation by providing co-stimulatory signals in the form of CD40 ligand (CD40L; also known as CD154). These cognate Tcells are reciprocally activated by engagement of CD40L with CD40 on the surface of Bcells (reviewed elsewhere). CD40, a member of the TNF receptor (TNFR) family, is expressed by a variety of immune cells, including B cells, but also by other cells such as platelets. Without co-stimulation via CD40 or other receptors, BCR triggering can lead to apoptosis of the B cell instead of B cell activation. The inability of B cells to be activated without CD40-mediated co-stimulation serves as a checkpoint to prevent the maturation of autoreactive B cells. However, the breakdown of tolerance to self-antigens in autoimmune diseases can be mediated by B cell–T cell interactions, as exemplified in one study that found that CD40 signalling in B cells is required for the development of SLE-like disease in mice and is also probably an important contributor to 6CDNGApproved and advanced development therapeutics that could be used to target B cells in autoimmunity TargetFormatsclerosisB cell depletionMonoclonal antibodyApprovedlabel uselabel useApprovedOcrelizumabMonoclonal antibodyApprovedMonoclonal antibodyMonoclonal antibodyApprovedApprovedDaratumumabMonoclonal antibodyApprovedIndatuximab ravtansineChimeric monoclonal ApprovedB cell activation or activity modulation%&CPF(EGPIKPGGTGFmonoclonal antibodyPhase I and phase IIPhase IICPF(EDARTPhase IMonoclonal antibodyPhase IPhase IIDapirolizumab pegol2GI[NCVGF(CDHTCIOGPVPhase IIMonoclonal antibodyPhase IMonoclonal antibodyPhase IEpratuzumabMonoclonal antibodySmall moleculeApprovedSmall moleculeApprovedInhibition of cytokines or cytokine signallingMonoclonal antibodyApproved$#((CPF#24+.6#%+CPFJWOCP+I)(Efusion proteinPhase IIIPhase IITocilizumabMonoclonal antibodyApprovedMonoclonal antibodyPhase I,#-CPF,#-TofacitinibSmall moleculeApprovedPhase I and phase II,#-CPF,#-Small moleculeApprovedPhase IIITrafficking blockade+PVGITKPMonoclonal antibodyPhase IIApproved5JQYPCTGCWVQKOOWPGKPFKECVKQPUHQTTJGWOCVQKFCTVJTKVKU\n4# U[UVGOKENWRWUGT[VJGOCVQUWU\n5.' CPFQTOWNVKRNGUENGTQUKUCNVJQWIJUQOGQHVJGUGKPVGTXGPVKQPUJCXGDGGPCRRTQXGFHQTQVJGTFKUGCUGUCUYGNN#RRTQXGFCPFKPXGUVKICVKQPCNUVCVWUGUTGHGT

VQVJG75CPF(&#TGIKUVGTGFENKPKECNVTKCNU#24+.CRTQNKHGTCVKQPKPFWEKPINKICPF$#(($EGNNCEVKXCVKPIHCEVQT%&.%&NKICPF&#46FWCNCHHKPKV[TGVCTIGVKPIOQNGEWNG(E4++DNQYCHHKPKV[KOOWPQINQDWNKP(ETGIKQPTGEGRVQT++D+.4+.TGEGRVQT,#-,CPWUMKPCUG2+-RJQURJQKPQUKVKFGMKPCUG; TACI, transmembrane activator and calcium modulator. autoimmunity in humans. Consistent with this finding, CD40L is upregulated on Tcells in multiple autoimmune diseases including SLE, RA and multiple sclerosis, and soluble CD40L levels correlate with autoantibody titres and disease activityToll-like receptors. As an alternative to CD40-mediated stimulation of BCRs, B cells can be activated independently of Tcells via dual stimulation of the BCR and TLRs. TLRs recognize pathogen-associated molecularpatterns (PAMPs) and damage-associated molecular patterns (DAMPs) that are important for host defence and wound healing (FIG.. Bacterial or viral PAMPs, such as lipopolysaccharide (LPS), double-stranded RNA or virus-like particles, can be detected by both the BCR (on antigen-specific B cells) and TLRs. Cell surface TLRs can engage ligands residing on the surface of the pathogen simultaneously with the BCR, whereas lysosomal TLRs can interact with RNA, DNA and other intracellular ligands upon internalization and digestion of pathogens or pathogen debris following interaction with the BCR. Thus, engagement of BCRs by bacterial or viral structures in an antigen-specific manner and simultaneous engagement of TLRs in a PAMP-specific manner leads to activation of B cellsTLRs are also capable of binding self-ligands or DAMPs, which can promote protective and reparative responses as well as pathogenic autoimmune responses under certain conditions . Intracellular components, such as DNA and RNA, can be exposed during cell death and tissue damage and can then interact with TLRs on B cells. Although these components are normally cleared by macrophages, interactions with antibodies can lead to the accumulation and stabilization of these self-antigens. The resultant immune complexes can simultaneously activate TLRs and BCRs, lowering the receptor engagement threshold needed for B cell activationIn 2002, TLRs were noted to contribute to autoimmune B cell activation in an invitro system, in which dual stimulation of TLR9 and the BCR by self-DNA-containing immune complexes led to autoimmune B cell activation (in this case, the activation of rheumatoid factor-positive Bcells). This observation provided a mechanistic model to explain the production of rheumatoid factor; in this model, engagement of the BCR on rheumatoid factor-positive B cells with the Fc domain of an antibody also bound to a TLR ligand (such as DNA, RNA or fibrinogen) leads to dual stimulation and activation of the B cell. In addition, TLR9, TLR7 and TLR8, which bind viral single-stranded RNA (ssRNA), have also been implicated in the development of autoimmunity. For instance, immune complexes containing ssRNA or RNA-binding proteins (such as small nuclear ribonucleoproteins (snRNPs), Sm, Ro, La and transfer RNA synthetases) can stimulate rheumatoid factor-positive B cells upon dual engagement of TLR7 and the BCR. B cells that directly bind TLR ligands, or RNA-binding proteins that bind TLR ligands via their BCR, might also be activated in a similar fashion. Dual stimulation of TLRs and BCRs probably contributes to autoantibody production in, and to the pathogenesis of SLE, myositis, Sjögren syndrome and other autoimmune rheumatic diseases in which autoreactive B cells and autoantibodies target proteins or molecular complexes containing DNA or RNA In addition to co-stimulation by CD40 or dual stimulation by TLRs, B cell activation can be facilitated by the activation of the co-receptor CD19. CD19 is an immunoglobulin superfamily glycoprotein associated BCR Antigen C3d IgIg BAFFR CD40LCD40 BAFF IL-6RIL-21IL-21R IL-6 PAMP TLR CD19CD21 MHC LAIR1 FcRIIbIgG CD22 a Stimulatory checkpointsB cellOther cellb Inhibitory checkpoints Collagen (KI B cells express a repertoire of stimulatory and KPJKDKVQT[TGEGRVQTUVJGTGNCVKXGNGXGNUQHYJKEJEJCPIGVJTQWIJVJGEQWTUGQH$EGNNFGXGNQROGPV$EGNNHWPEVKQPUCTGmodulated by the balance of expression and colocalization of stimulatory checkpoint receptors (part ) and inhibitory checkpoint receptors (part 5QOG6QNNNKMGTGEGRVQTU\n6.4U CTGCNUQGZRTGUUGFKPVJGGPFQUQOG$#(($EGNNCEVKXCVKPI$#((4$EGNNCEVKXCVKPIHCEVQTTGEGRVQT$%4$EGNNTGEGRVQT%&.%&NKICPF%'#%#/ECTEKPQGODT[QPKETGNCVGFEGNNCFJGUKQPOQNGEWNG&#/2FCOCIGCUUQEKCVGFOQNGEWNCTRCVVGTP(E4++DNQYCHHKPKV[(ETGIKQPTGEGRVQT++D+.4+.TGEGRVQT+.4+.TGEGRVQT.#+4NGWMQE[VGCUUQEKCVGFNKMGTGEGRVQT.+4NGWMQE[VGKOOWPQINQDWNKPNKMGTGEGRVQT2#/2RCVJQIGPCUUQEKCVGFOQNGEWNCTRCVVGTP2&RTQITCOOGFEGNNFGCVJ with the BCR and is expressed on B cells from the pre-B cell stage through to the plasma cell differentiation stage. CD19 signals though the tyrosine kinases LYN and phosphoinositide 3-kinase (PI3K), which amplify signals from the BCR, decreasing the threshold for BCR activation. CD19 is an integral feature of the BCR signalling complex and is important for B cell activation. Both CD19 and the BCR are needed for B cell activa

tion, whereas stimulation of either receptor alone leads to Bcell apoptosisInterestingly, investigators have identified a unique population of CD19 B cells in patients with SLE or pemphigus, although the function of these cells in disease requires further investigation. Thus, CD19 is a potential immunomodulatory therapeutic target, in addition to CD20 and other B cell antigens, for the treatment of autoimmune diseasesBAFF. B cell activating factor (BAFF; also known as TNFSF13B) is a cytokine that belongs to the TNF family and can facilitate B cell activation indirectly by promoting B cell survival, proliferation and/or differentiation. BAFF is produced by various types of immune cells including follicular dendritic cells and monocytes and can influence B cell populations through interactions with three different receptors: transmembrane activator and calcium modulator (TACI; also known as TNFRSF13B), the predominant BAFF receptor on splenic transitional type 2 and marginal zone B cells; B cell maturation antigen (BCMA; also known as TNFRSF17), the predominant BAFF receptor on germinal centre Bcells; and BAFF receptor (BAFFR), the predominant BAFF receptor on peripheral B cellsSimilar to some other cytokines, BAFF facilitates Tcell co-stimulation of B cells by functioning as a pro-survival factor rather than by providing a traditional stimulatory signal. Instead of providing a secondary B cell activating signal, BAFF promotes B cell survival and increases the probability that a B cell will encounter sufficient signals to become activated. Thus, in autoimmunity, BAFF can increase the overall numbers of autoreactive B cells and/or outweigh peripheral tolrance mechanisms by predisposing B cells to receive activation signalsBAFF concentrations are often increased in the serum of patients with SLE or other autoimmune conditionsFor example, serum concentrations of BAFF correlate with anti-double-stranded DNA (dsDNA) antibody titres in patients with SLE. Furthermore, a variant in the BAFF-encoding gene, TNFSF13B, is strongly associated with multiple sclerosis and SLE. This variant encodes a truncated mRNA, which escapes microRNA-mediated inhibition, leading to increased production of the BAFF protein. Belimumab, a human IgG1 monoclonal antibody that targets BAFF, is an effective treatment for patients with SLE who are autoantibody positiveIL-6. The cytokine IL-6 was originally identified as a Bcell growth factor and plasma cell differentiation factor, but IL-6 can also have pleiotropic effects on other immune cell types. IL-6 is produced both intrinsically by B cells and by other cells such as macrophagesmacrophages increase their IL-6 production as a result of feedforward processes in response to FcR engagement by IgG antibodies85. In RA, increased IL-6 serum concentrations correlate with joint damage, probably because of the involvement of IL-6 in promoting osteoclastogenesis. Blockade of IL-6 with tocilizumab, a humanized monoclonal antibody against the IL-6 receptor (IL-6R), leads to improved radiographic outcomes in patients with RA. In SLE, B cell-derived IL-6 can drive autoimmune GC formation by promoting T helper cell differentiation and IL-21 production, which in turn can lead to B cell growth and differentiation. Preliminary studies of IL-6 blockade in patients with SLE, however, have shown minimal beneficial effectsIL-21. The cytokine IL-21 is produced by multiple Thelper cell subsets and has critical functions in Bcell activation, proliferation, differentiation, affinity maturation and antibody production. IL-21 drives pro-inflammatory responses by promoting B cell activation and expansion, and patients with SLE, type 1 diabetes or inflammatory bowel diseases have increased serum concentrations of IL-21 compared with healthy individuals. In germinal centres, Tcell-derived IL-21 regulates class switching to IgG, IgA and IgE in B cells through the activation of activation-induced cytidine deaminase (AID) and promotes differentiation of activated B cells into memory B cells and plasma cellsBlockade of IL-21 consequently results in decreased Tcell-induced B cell proliferation, differentiation and B cellBCRIgIgRBPRNA Internalized NF- NF- MYD88 PAMP Internalized (KIB cell activation via dual stimulation by antigens. During an immune response to an infection, the simultaneous engagement of the B cell receptor (BCR) and 6QNNNKMGTGEGRVQTU\n6.4U QP$EGNNUD[DCEVGTKCNQTXKTCNUVTWEVWTGU\nRCVJQIGPCUUQEKCVGFmolecular patterns (PAMPs)) or by damage-associated molecular patterns (DAMPs) can lead to B cell activation. In some autoimmune diseases, autoantigens can dual UVKOWNCVG$EGNNUVJTQWIJ6.4UCPFVJG$%46JGGZCORNGRTQXKFGFKUHQTU[UVGOKENWRWUGT[VJGOCVQUWU\n5.' KPYJKEJ$EGNNUVJCVTGEQIPK\G40#DKPFKPIRTQVGKPU\n4$2U VJTQWIJVJGKTOGODTCPGDQWPF$%4DKPFCPFEQKPVGTPCNK\G4$2UCNQPIYKVJVJGKTCUUQEKCVGFTKDQPWENGKECEKFUCPFVJGTKDQPWENGKECEKFUVJGPDKPF6.4KPVJGGPFQUQOGU5KOKNCTexamples are found in other autoimmune diseases. antibody secretion. Interestingly, IL-21 also prevents nonspecific B cell activation by promoting apoptosis in B cells that receive signals through the BCR and/or TLRs but that do not receive Tcell co-stimulatory signalsThus, as IL-21 contributes to B cell function through multiple mechanisms, targeting IL-21 might be valuable as a broad treatment for autoimmune diseases that stem from dysregulated B cell function at various stages of development and/or activation.$EGNNKPJKDKVQT[RCVJYC[UDuring B cell development and maturation, inhibitory checkpoints balance activating signals to ensure central tolerance CD22 is important for regulating B cell activation in response to self-antigens. This molecule is a transmembrane receptor and member of the sialic binding immunoglobulin-like lectin family. CD22 binds 2,

6-linked sialic acids found on the surfaces of eukaryotic but not bacterial cells. The receptor is constitutively associated with the BCR and contains three cytoplasmic immunoreceptor tyrosine-based inhibition motifs (ITIMs), which are phosphorylated upon ligand binding in conjunction with BCR-antigen bindingSubsequently, recruitment of the phosphatases SHIP1 and SHP1 inhibits signalling downstream of the BCR. This inhibition is thought to modulate the BCR activation threshold to prevent B cell activation in response toself-antigensRIIb. The low-affinity immunoglobulin- Fc region receptor IIb (FcRIIb) is the only inhibitory FcR and the only FcR expressed by B cells; this molecule is crucial for restricting antibody-mediated immune responsesAfter binding of the IgG Fc region to the extracellular domain of FcRIIb, its single intracellular ITIM domain is phosphorylated by BCR-associated kinases; subsequent recruitment of the SHIP1 phosphatase inhibits signalling downstream of the BCR and destabilizes the BCR complex101. This process is thought to modulate immune responses when antigens saturated with IgG antibodies in immune complexes trigger FcRIIb in conjunction with the BCR. As a result, FcRIIb is thought to help prevent autoimmunity, and mutations at its genetic locus have been associated with RA, SLE and multiple sclerosis in human genome-wide association studies and mouse studiesOther inhibitory receptors. Polymorphisms in genes encoding other inhibitory receptors expressed by Bcells, including CD5, CD72, leukocyte immunoglobulin-like receptor 1 (LIR1; also known as CD85j) and programmed cell death 1 (PD1), have also been associated with autoimmunity96102. B cells express additional inhibitory receptors, including carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) and leukocyte-associated immunoglobulin-like receptor1 (LAIR1). These receptors, along with other, less-well understood molecules, contribute to the maintenance of B cell tolerance during development and in the periphery. These receptors provide additional targets for future Bcell-modulating therapeutics as our understanding of their mechanism(s) continues to improve.Therapeutic approachesB cell depletion with the B cell-depleting anti-CD20 antibody, rituximab, is currently the therapeutic approach most widely used to target B cells. The finding that B cell dysregulation contributes to autoimmune diseases has inspired the development of novel, mechanistically informed treatments. A number of both Bcell-depleting antibodies, such as anti-CD138 and anti-CD38, and small-molecule kinase inhibitors, such as Janus kinase (JAK), SYK and BTK inhibitors, hold promise for the treatment of autoimmune disease (TABLE. However, these nonspecific strategies result in broad immune suppression, which increases the risk of infectious complications and other adverse events. The shortcomings of these therapies have led to new efforts focused on the development of approaches that engage B cell inhibitory or other checkpoint pathways in a specific manner . Ongoing strategies for the development of therapeutics to treat autoimmunity include antibody-based B cell depletion, small-molecule kinase inhibition, antibody-based blockade of B cell activating receptors and antibody-based induction of B cell inhibitory signals$EGNNFGRNGVKPIVJGTCRKGUCell depletion was an initial approach used to target B cells for the treatment of autoimmune disease. The B cell-depleting anti-CD20 antibody rituximab is FDA-approved for the treatment of RA. Although not approved for multiple sclerosis and having failed trials in SLE, rituximab is also used off-label for the treatment of these diseases on the basis of clinician experience. In addition, ocrelizumab, a humanized anti-CD20 monoclonal antibody, was approved in 2017 for the treatment of multiple sclerosis. Clinical studies have shown some efficacy of ocrelizumab in treating SLE114although concerns over adverse events have halted clinical trials for SLE and RA, and future studies will probably demonstrate its utility in other B cell-mediated diseases. Despite depleting only B cells that express CD20, which is downregulated by antibody-secreting cells, these antibody therapies are thought to function by targeting precursors to these antibody-secreting B cells and/or B cells with antigen-presenting or other pathogenic functions.Given that many antibody-secreting B cells, including plasmablasts and plasma cells, lack CD20 but retain CD19 expression, CD19-targeting therapies that are being developed for B cell malignancies are also being explored as therapeutics for autoimmune diseases. Inebilizumab, a humanized monoclonal anti-CD19 antibody that is afucosylated for increased ADCChas shown promising results in multiple mouse models of autoimmunity and early-phase clinical trialsFurthermore, in clinical trials in multiple sclerosis and other diseases, inebilizumab treatment resulted in considerable depletion of plasma cells. However, CD19 expression is absent on a subset of plasma cells in the bone marrow that are known to contribute to autoantibody production. Thus, these CD19-targeting therapies might miss subsets of cells that produce autoantibodies and contribute to the pathogenesis of autoimmune diseases. Further studies regarding the efficacy of CD19-targeting therapies are ongoingAlemtuzumab is an anti-CD52 monoclonal antibody that depletes Tcells, B cells and macrophagesThe efficacy of alemtuzumab, which is FDA-approved for the treatment of multiple sclerosis, is potentially high because of its B cell-depleting properties. However, alemtuzumab is also associated with a high rate of serious adverse events, probably because the treatment results in depletion of such broad leukocyte populations.Antibodies that target other molecules expressed by B cells including CD38 (daratumumab124), CD138 (indatuximab ravtansine), and other surface receptors are undergoing clinical development. Daratumumab and indatuximab ravtansine are currently being investigated for their efficacy in treating patients with multiple myeloma but could have potential in treating autoimmune disease.6CTIGVKPI$EGNNEJGEMRQKPVUThe risks associated with systemic depletion of B cells have led to the developmen

t of therapies that modulate, rather than deplete, B cells.One approach for therapeutically modulating B cell activation is the disruption of CD40–CD40L interactions. Despite initial promising results from clinical trials of the anti-CD40L antibodies toralizumab, BG9588 and ABI793 in SLE, thromboembolic events, caused by engagement of CD40L expressed on platelets, have hampered this approach. Nevertheless, next-generation antagonists for CD40–CD40L are now in the JAKs BTK IL-6RTACIreceptor4 Integrin Tofacitinib andbaricitinib Ibrutinib Atacicept Belimumab NNC114-0005 Tocilizumab AMG557 andb PI3KBCR IgIgCD20 CD22 CD52 CD38CD40 ICOSL CD138 CD19 gp130 IL-21R Disruption of B cell activation or activity+PJKDKVKQPQHRTQKP1COOCVQT[E[VQMKPGU6TCH0EMKPI FcRIIbFcRIIb B cell Natalizumab Rituximab Alemtuzumab Daratumumab ravtansine Inebilizumab MGD010a XmAb5871a Epratuzumab CFZ533 Dapirolizumab pegol Idelalisib (KITherapeutic approaches for modulating B cells in autoimmunity. Disruption of B cell activation or engagement of inhibitory checkpoint receptors (part ), B cell depletion (part ), inhibition of pro-inflammatory soluble factors (part and trafficking blockade (part ) are being used to target B cells in autoimmunity. :O#DCPF/)&HWPEVKQPD[DKPFKPIDQVJVJGNQYCHHKPKV[KOOWPQINQDWNKP(ETGIKQPTGEGRVQT++D\n(ERIIb) and components that are positioned PGCT|VJG$EGNNTGEGRVQT\n$%4 DTKPIKPIVJKUKPJKDKVQT[TGEGRVQTKPENQUGRTQZKOKV[VQVJG$%4VQKPJKDKV$%4UKIPCNNKPI&CRKTQNK\WOCDRGIQNCPF/'&+VCTIGV%&NKICPF\n%&. CPF+%15TGURGEVKXGN[CNVJQWIJVJGUGTGEGRVQTUCTGPQVPQTOCNN[GZRTGUUGFQP$EGNNUVJG[CTGGZRTGUUGFQP6|EGNNUCPFDKPFVQTGEGRVQTUQP$EGNNU\n%&CPF+%15. VQOGFKCVG$EGNNs6EGNNKPVGTCEVKQPU$#((4$EGNNCEVKXCVKPIHCEVQTTGEGRVQT+.4+.TGEGRVQT+.4+.TGEGRVQT,CPWUMKPCUG2+-RJQURJQKPQUKVKFGMKPCUG; TACI, transmembrane activator and calcium modulator. clinic, with promising results for multiple autoimmune diseases(TABLE. In addition, CD40 agonizing antibodies are being pursued for antitumour therapySimilarly, inhibition of interactions between ICOS and ICOSL (also known as ICOSLG) (expressed on Tcells and B cells, respectively) to disrupt Tcell–Bcell interactions is being pursued for the treatment of autoimmune diseases. Antibodies that block ICOSL (AMG557 ) or ICOS (MEDI-570 ) are being developed. Both MEDI-570 and AMG557 can block Tcell–B cell interactions and the production of autoantibodies and have shown promise in clinical trials for SLE treatmentThere is also considerable interest in targeting CD22 for the treatment of autoimmune disease. As CD22 has three ITIMs, promoting the inhibitory function of this receptor could have potent inhibitory effects on Bcells. However, treatment with the anti-CD22 antibody epratuzumab (an antibody that is thought to promote the function of CD22 and thereby inhibit BCR activation134) did not result in improved response rates in patients with SLE in a phase II clinical trialAnother approach being pursued is the co-engagement of a B cell inhibitory receptor with a surface receptor on the same B cells. For example, the anti-CD19 antibody XmAb5871 contains an Fc region that is engineered to have an increased affinity and selectivity for the inhibitory FcRIIb receptor. XmAb5871, hence, co-engages CD19 and FcRIIb on B cells, bringing the inhibitory receptor in close proximity to the BCR to inhibit downstream signalling. In clinical trials for the treatment of SLE and IgG4-related disease, this dual-targeting antibody decreased the production of ACPAs and rheumatoid factor by B cells from patients with RA138139A similar approach for simultaneously targeting a Bcell inhibitory receptor and a B cell surface receptor to modulate B cell function makes use of a bispecific antibody platform termed dual-affinity retargeting molecules (DARTs). DARTs target two molecules by linking together single-chain variable fragments derived from two different monoclonal antibodies. One DART therapy that is currently being developed (MGD010) targets FcRIIb and Ig on B cells in an effort to disrupt BCR signalling. This molecule is in early-phase clinical trials and might be developed for RA or other disease indications for which B cells and/or autoantibodies contribute to disease pathogenesis. Overall, antibody-based constructs are an attractive approach for targeting B cell activity, as they can be highly engineered and customized for bioactivity and biocompatibility using the growing antibody knowledge base.Finally, the inhibition of signalling molecules involved in B cell activation is being pursued as a strategy to abrogate B cell activation in autoimmune disease. Following BCR engagement, the tyrosine kinase BTK initiates intracellular signalling networks that result in B cell activation. Ibrutinib is a small-molecule drug that covalently binds BTK to inhibit its function, resulting in B cell apoptosis. This inhibitor is FDA-approved for the treatment of B cell malignancies, and next-generation BTK inhibitors are being developed for the treatment of multiple autoimmune diseases, including SLE, RA and other autoimmune rheumatic diseases(TABLE. In addition, several companies are pursuing the development of BTK inhibitors in combination with other small-molecule kinase inhibitors for the treatment of B cell malignancy and autoimmunity. Similar efforts are directed towards developing inhibitors of PI3K, another important molecule in the signalling cascade downstream of the BCR6CTIGVKPIE[VQMKPGUAn alternative approach t

o targeting autoimmune B cells themselves is inhibiting the soluble factors that activate B cells. Examples of these strategies include inhibiting the BAFF signalling pathway and cytokines such as IL-6.Several therapeutic strategies that target soluble Bcell activating factors are either FDA-approved or are under development. For example, belimumab, a monoclonal antibody that targets BAFF (and prevents BAFF from binding to its three receptors) is approved for the treatment of SLE. In patients with SLE, treatment with belimumab reduces the number of CD20-positive Bcells and plasma cells in the blood and lowers the titres of anti-dsDNA antibodies. Alternatively, atacicepta fusion protein of the extracellular domain of TACI anda human IgG1 Fc, is currently in clinical trials for the treatment of SLE. Treatment with atacicept leads to a reduction in the total number of B cells and a dose-dependent reduction in antibody titres. In contrast to the ability of belimumab to bind BAFF, atacicept binds and blocks both BAFF and a proliferation-inducing ligand (APRIL, or TNFSF13, another B cell stimulatory molecule), preventing their signalling via BAFFR and TACI, respectively, on B cells, which could potentially lead to greater potency(TABLETargeting IL-6 has shown efficacy in the treatment of several autoimmune diseases. This cytokine is produced by a variety of leukocytes and promotes the differentiation of CD40-activated B cells into plasma cells. The concentration of IL-6 is increased in the serum and synovial tissue of patients with RA as well as in the serum of patients with other inflammatory diseases including Crohn’s disease, SLE and multiple sclerosis. The downstream inflammatory effects of IL-6 can be offset by treatment with conventional therapies including corticosteroids and NSAIDs. However, antibodies that directly block the actions of IL-6 are also available. Both tocilizumab, a humanized anti-IL-6R monoclonal antibody, and sarilumab, a fully human anti-IL-6R monoclonal antibody, are approved for the treatment of RA, whereas additional IL-6-targeting therapies are showing promise in clinical trials for various other autoimmune diseasesJAKs mediate signalling by the IL-6R and many other cytokine receptors. Small-molecule inhibitors of JAKs are available, such as tofacitinib and baricitinib, that block the effects of these pro-inflammatory cytokines. JAK inhibition is an effective strategy in the treatment of RA and other autoimmune diseaseswhich is possibly in part dependent on the inhibition of IL-6-mediated B cell survival, maturation and activation. Indeed, blockade of JAK1-mediated and JAK3-mediated signalling with tofacitinib inhibits B cell activationAdditional JAK inhibitors are being developed to treat chain variable fragmentsingle polypeptide fusion proteins of the variable regions of the heavy and light chains of an antibody connected by a peptide linker. malignancy and/or autoimmunity, many of which are being engineered to have a greater specificity for specific JAK family members.Molecules that target IL-21 are under development for the treatment of autoimmunity owing to its function in driving Tcell-dependent B cell maturation. NNC114-0005 (also known as NNC0114-0006) is an anti-IL-21 neutralizing monoclonal antibody in phaseII clinical trials for the treatment of RA. Moreover, JAK inhibitors such as tofacitinib also interfere with IL-21 signalling, as JAKs mediate signalling events downstream of IL-21–IL-21 receptor binding.Other FDA-approved therapeutics for autoimmune disease might also exert their beneficial effects by inhibiting B cells or their effector functions. For example, natalizumab, a humanized monoclonal antibody against 4 integrin (CD49d), inhibits leukocyte trafficking and is FDA-approved for the treatment of multiple sclerosis and Crohn’s disease. Although the beneficial effects of natalizumab treatment are widely thought to be caused by blockade of Tcell trafficking, this antibody also inhibits B cell trafficking, which might also contribute to its efficacy(WVWTGVJGTCRGWVKERTQURGEVUAlthough several targeted therapies for autoimmune diseases currently exist, therapies that target more specialized subsets of B cells such as activated or autoreactive B cells could have improved efficacy and a lower risk of adverse effects. Several monoclonal antibody therapies that target subsets of B cells, such as targeting of CD138B cells by indatuximab ravtansine, and factors that promote B cell activation, such as IL-21, are in development. Dual-targeting strategies with engineered bispecific proteins also hold promise.Therapies that block inhibitory checkpoint proteins on Tcells, such as PD1 and CTLA4, to promote immune responses are being used to treat patients with cancer. The use of these treatments can result in secondary autoimmune complications, suggesting that these inhibitory checkpoints could potentially be targeted with agonists to treat autoimmunity. Notably, PD1 is expressed on both Tcells and B cells, and although anti-PD1 therapy is known to promote antitumour Tcell responses in patients with cancer, this strategy could potentially also promote antitumour B cell responses. As blockade of these inhibitory receptors has proved revolutionary for the treatment of cancer, activating the same targets might be useful for inducing tolerance in the context of autoimmunity. Many of the proteins targeted by checkpoint immunotherapy on Tcells and NK cells are also expressed on B cells, where they regulate B cell functions that contribute to autoimmunity. Therefore, targeting these inhibitory checkpoints could enable a more fine-tuned approach to modulating B cell responses and reducing their pathogenic cellular activitiesThe field is also working to develop molecules that target specific pathogenic B cell subsets or functions, such as antigen presentation or antibody production, to minimize unnecessary immunosuppressive effects and to maximize their potency in restricting autoimmunity. In the future, autoantigen-specific B cell-tolerizing therapies might also be developed to precisely target pathogenic B cells.ConclusionsMultiple B cell effector functions contribute to the pathogenesis of autoimmune disease, and targeting thecheckpoints that control B cell activation

has therapeutic potential. Compared with anti-CD20 B cell-depleting therapeutics, emerging efforts are using mechanistically informed strategies to target B cells to combat disease in a more precise manner. Targeting B cell activation and effector functions, rather than broadly depleting B cells, has the potential to control autoimmunity without broad immunosuppression. Such approaches might leverage newly developed tools including bispecific or trispecific antibody engineering, cell-targeting therapies or other next-generation biologic therapeutics. In the future, these strategies could be used to target specific pathogenic B cell subpopulations or B cell responses.Chan, O. T., Madaio, M. P. & Shlomchik, M. J. Thecentral and multiple roles of B cells in lupus pathogenesis. Immunol. Rev., 107–121 (1999).Townsend, M. J., Monroe, J. G. & Chan, A. C. B cell targeted therapies in human autoimmune diseases: anupdated perspective. Immunol. Rev.264–283 (2010).Yuseff, M. I., Pierobon, P., Reversat, A. & Dumenil, A. M. How B cells capture, process and present antigens: a crucial role for cell polarity. Nat. Rev. Immunol., 475–486 (2013).Stone, J. H. etal. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. 221–232 (2010).Specks, U. etal. Efficacy of remission-induction regimens for ANCA-associated vasculitis. N. Engl. J.Med., 417–427 (2013).Edwards, J. C. etal. Efficacy of B cell-targeted therapy with rituximab in patients with rheumatoid arthritis. , 2572–2581 (2004).Hauser, S. L. etal. B cell depletion with rituximab in relapsing-remitting multiple sclerosis. Rovin, B. H. etal. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. , 1215–1226 (2012).Cambridge, G. etal. Serologic changes following Blymphocyte depletion therapy for rheumatoid arthritis. , 2146–2154 (2003).Cortazar, F. B. etal. Effect of continuous B cell depletion with rituximab on pathogenic autoantibodies and total IgG levels in antineutrophil cytoplasmic associated vasculitis. Arthritis Rheumatol., 1045–1053 (2017).Du, F. H., Mills, E. A. & Mao-Draayer, Y. Next-generation CD20 monoclonal antibodies in autoimmune disease treatment. Auto Immun. Highlights, 12 (2017).Rehnberg, M., Amu, S., Tarkowski, A., Bokarewa, M. I. CD20 treatment on B cell ontogeny in bone marrow of patients with rheumatoid arthritis. Arthritis Res. Ther.Garrido, I., Carmona, E. M., Specks, U. & Limper, A. H. Pneumocystis pneumonia in patients treated with rituximab. , 258–265 (2013).Elsegeiny, W., Eddens, T., Chen, K. & Kolls, J. K. CD20 antibody therapy and susceptibility to Pneumocystis pneumonia. 2043–2052 (2015).Wei, K. C. etal. Pneumocystis jirovecii pneumonia in HIV-uninfected, rituximab treated non-Hodgkin lymphoma patients. , 8321 (2018).Shah, S., Jaggi, K., Greenberg, K. & Geetha, D. Immunoglobulin levels and infection risk with rituximab induction for anti-neutrophil cytoplasmic associated vasculitis. 470–474 (2017).Bingham, C. O. 3rd etal. Immunization responses in rheumatoid arthritis patients treated with rituximab: results from a controlled clinical trial. , 64–74 (2010).van Assen, S. etal. Humoral responses after influenza vaccination are severely reduced in patients with rheumatoid arthritis treated with rituximab. Arthritis , 75–81 (2010).Kapetanovic, M. C. Rituximab and abatacept but not tocilizumab impair antibody response to pneumococcal conjugate vaccine in patients with rheumatoid arthritis. Arthritis Res. Ther., R171 (2013).Suurmond, J. & Diamond, B. Autoantibodies in systemic autoimmune diseases: specificity and pathogenicity. J. Clin. Invest., 2194–2202 (2015).Schiffer, L. etal. Short term administration of costimulatory blockade and cyclophosphamide induces remission of systemic lupus erythematosus nephritis in NZB/W F1 mice by a mechanism downstream of renal immune complex deposition. 171 Okroj, M., Heinegard, D., Holmdahl, R. & Blom, A. M. Rheumatoid arthritis and the complement system. , 517–530 (2007).Anquetil, F., Clavel, C., Offer, G., Serre, G. & Sebbag, M. IgM and IgA rheumatoid factors purified from rheumatoid arthritis sera boost the Fc receptor- and dependent effector functions of the citrullinated protein autoantibodies. , 3664–3674 (2015).Clynes, R., Dumitru, C. & Ravetch, J. V. Uncoupling of immune complex formation and kidney damage in autoimmune glomerulonephritis. 1052–1054 (1998).Sokolove, J. etal. Rheumatoid factor as a potentiator citrullinated protein antibody-mediated inflammation in rheumatoid arthritis. Arthritis Rheumatol., 813–821 (2014).Daniels, R. H., Williams, B. D. & Morgan, B. P. Human rheumatoid synovial cell stimulation by the membrane attack complex and other pore-forming toxins invitro: the role of calcium in cell activation. 312–316 (1990).Hay, F. C., Jones, M. G., Bond, A. & Soltys, A. J. Rheumatoid factors and complex formation. The role chain framework sequences and glycosylation. Clin Orthop. Relat. Res., 54–62 (1991).Regnault, A. etal. Fc receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization. 371–380 (1999).Hamano, Y., Arase, H., Saisho, H. & Saito, T. Immune complex and Fc receptor-mediated augmentation of antigen presentation for invivo Th cell responses. J.Immunol., 6113–6119 (2000).Schuurhuis, D. H. etal. Antigen-antibody immune complexes empower dendritic cells to efficiently prime CTL responses invivo. Ise, W. etal. T follicular helper cell-germinal center Bcell interaction strength regulates entry into plasma cell or recycling germinal center cell fate. , 702–715 (2018).Stebegg, M. etal. Regulation of the germinal center response. Front. Immunol., 2469 (2018).Ting, Y. T. etal. The interplay between citrullination and HLA-DRB1 polymorphism in shaping peptide binding hierarchies in rheumatoid arthritis. J. Biol. , 3236–3251 (2018).McInnes, I. B. & Schett, G. The pathogenesis of rheumatoid arthritis. , 2205–2219 (2011).Grumet, F. C., Coukell, A., Bodmer, J. G., Bodmer, W. F. & McDevitt, H. O. Histocompatibility (HL-A) antigens associated with systemic lupus erythematosus. Apossible genetic predisposition to disease. N. Engl. , 193–196 (1971).Gaffney, P. M. etal. Genome screening in human sy

stemic lupus erythematosus: results from a second Minnesota cohort and combined analyses of 187 pair families. , 547–556 Brown, M. A. etal. HLA class I associations of ankylosing spondylitis in the white population in the Ann. Rheum. Dis., 268–270 Brown, M. A. etal. Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and theenvironment. , 1823–1828 Shipman, W. D., Dasoveanu, D. C. & Lu, T. T. Tertiary lymphoid organs in systemic autoimmune diseases: pathogenic or protective? F1000Res., 196 (2017).Detanico, T. etal. Somatic mutagenesis in autoimmunity. Autoimmunity, 102–114 (2013).Elliott, S. E. etal. Affinity maturation drives epitope spreading and generation of pro-inflammatory anti-citrullinated protein antibodies in rheumatoid arthritis. Arthritis Rheumatol., 1946–1958 (2018).Lu, D. R. etal. Tcell-dependent affinity maturation and innate immune pathways differentially drive autoreactive B cell responses in rheumatoid arthritis. Arthritis Rheumatol., 1732–1744 (2018).Lindhout, E. etal. Fibroblast-like synoviocytes from rheumatoid arthritis patients have intrinsic properties of follicular dendritic cells. Harris, D. P. Reciprocal regulation of polarized cytokine production by effector B and Tcells. Nat.Immunol.Yanaba, K. etal. A regulatory B cell subset with aunique CD1d phenotype controls Tcell-dependent inflammatory responses. Piancone, F. etal. B lymphocytes in multiple sclerosis: bregs and BTLA/CD272 expressing- lymphocytes modulate disease severity. , 29699 (2016).Grammer, A. C. & Lipsky, P. E. B cell abnormalities in systemic lupus erythematosus. Arthritis Res. Ther.Hirano, T. etal. Excessive production of interleukin 6/B cell stimulatory factor-2 in rheumatoid arthritis. Eur. J. Immunol., 1797–1801 (1988).Arkatkar, T. etal. B cell-derived IL-6 initiates spontaneous germinal center formation during systemic autoimmunity. 214, 3207–3217 (2017).Dienz, O. & Rincon, M. The effects of IL-6 on CD4 Tcell responses. Kalampokis, I., Yoshizaki, A. & Tedder, T. F. IL-10-producing regulatory B cells (B10 cells) in autoimmune disease. Arthritis Res. Ther. (Suppl. 1), 1 (2013).Lee, K. M. etal. TGF--producing regulatory B cells induce regulatory Tcells and promote transplantation tolerance. Eur. J. Immunol., 1728–1736 (2014).Shen, P. etal. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature507, 366–370 (2014).Wang, R. X. etal. Interleukin-35 induces regulatory Bcells that suppress autoimmune disease. , 633–641 (2014).Egwuagu, C. E. & Yu, C. R. Interleukin 35-producing Bcells (i35-Breg): a new mediator of regulatory B-cell functions in CNS autoimmune diseases. Crit. Rev. , 49–57 (2015).Rosser, E. C. & Mauri, C. Regulatory B cells: origin, phenotype, and function. , 607–612 (2015).Meffre, E. & Wardemann, H. B cell tolerance checkpoints in health and autoimmunity. Curr. Opin. Kuokkanen, E., Sustar, V. & Mattila, P. K. Molecular control of B cell activation and immunological synapse formation. Traffic, 311–326 (2015).Yang, J. & Reth, M. in B Cell Receptor Signaling(edsKurosaki, T. & Wienands, J.) 27–43 (Springer International Publishing, 2016).Avalos, A. M., Meyer-Wentrup, F. & Ploegh, H. L. B cell receptor signaling in lymphoid malignancies and autoimmunity. Adv. Immunol., 1–49 (2014).Dal Porto, J. M. etal. B cell antigen receptor signaling 101. , 599–613 (2004).Lane, P. etal. Activated human Tcells express a ligand for the human B cell-associated antigen CD40 which participates in Tcell-dependent activation of Blymphocytes. Bishop, G. A. & Hostager, B. S. The CD40–CD154 interaction in B cell–T cell liaisons. Cytokine Growth Factor Rev.Hokazono, Y. etal. Inhibitory coreceptors activated by antigens but not by Anti-Ig heavy chain antibodies install requirement of costimulation through CD40 for survival and proliferation of B cells. 171Voynova, E. etal. Requirement for CD40/CD40L interactions for development of autoimmunity differs depending on specific checkpoint and costimulatory pathways. ImmunoHorizons, 54–66 (2018).Peters, A. L., Stunz, L. L. & Bishop, G. A. CD40 and autoimmunity: the dark side of a great activator. Toubi, E. & Shoenfeld, Y. The role of CD40-CD154 interactions in autoimmunity and the benefit of disrupting this pathway. Autoimmunity, 457–464 Vinuesa, C. G. & Goodnow, C. C. Immunology: DNA drives autoimmunity. Nature416Pelka, K., Shibata, T., Miyake, K. & Latz, E. Nucleic sensing TLRs and autoimmunity: novel insights from structural and cell biology. Immunol. Rev.60–75 (2016).Hoffmann, M. H. & Steiner, G. A common pathway for all autoimmune diseases? The unholy alliance of environment, cell death and nucleic acids. Curr. Immunol. Rev.Leadbetter, E. A. etal. Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors. Nature416, 603–607 (2002).Lau, C. M. etal. RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement. 1171–1177 (2005).Uckun, F. M., Sun, L., Qazi, S., Ma, H. & Ozer, Z. Recombinant human CD19-ligand protein as a potent leukaemic agent. Br. J. Haematol.15–23 (2011).Liu, Z. etal. Peripheral CD19(hi) B cells exhibit activated phenotype and functionality in promoting IgG and IgM production in human autoimmune diseases. , 13921 (2017).Abraham, P. M., Quan, S. H., Dukala, D. & Soliven, B. CD19 as a therapeutic target in a spontaneous autoimmune polyneuropathy. , 181–191 (2014).Stuve, O. etal. CD19 as a molecular target in CNS autoimmunity. Acta Neuropathol., 177–190 (2014).Ng, L. G. etal. B cell-activating factor belonging to the TNF family (BAFF)-R is the principal BAFF receptor facilitating BAFF costimulation of circulating T and Bcells. , 807–817 (2004).Mackay, F. & Browning, J. L. BAFF: a fundamental survival factor for B cells. Nat. Rev. Immunol.Salazar-Camarena, D. C. etal. Association of BAFF, APRIL serum levels, BAFF-R, TACI and BCMA expression on peripheral B cell subsets with clinical manifestations in systemic lupus erythematosus. , 582–592 (2016).Wallace, D. J. etal. A phase II, randomized, double-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus. , 1168–1178 Steri, M. etal. Overexpression of the cytokine BAFF and autoimmunity risk. 1615–1626 (2017).Touma,

Z. & Gladman, D. D. Current and future therapies for SLE: obstacles and recommendations for the development of novel treatments. , e000239 (2017).Vincent, F. B., Morand, E. F., Schneider, P. & Mackay, F. The BAFF/APRIL system in SLE pathogenesis. Nat. Rev. Rheumatol., 365–373 (2014).Dienz, O. etal. The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by Tcells. Maeda, K., Mehta, H., Drevets, D. A. & Coggeshall, K. M. IL-6 increases B cell IgG production in a feed-forward proinflammatory mechanism to skew hematopoiesis and elevate myeloid production. 115, 4699–4706 (2010).Axmann, R. etal. Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and invivo. , 2747–2756 Wu, Q., Zhou, X., Huang, D., Ji, Y. & Kang, F. IL-6 mediated osteoclastogenesis by promoting JAK2 and RANKL activity invitro. , 1360–1369 (2017).Smolen, J. S. & Maini, R. N. Interleukin-6: a new therapeutic target. Arthritis Res. Ther. (Suppl. 2), Scott, L. J. Tocilizumab: a review in rheumatoid arthritis. , 1865–1879 (2017).Illei, G. G. etal. Tocilizumab in systemic lupus erythematosus: data on safety, preliminary efficacy, and impact on circulating plasma cells from an escalation study. Arthritis , 542–552 (2010).Wallace, D. J. etal. Efficacy and safety of an interleukin 6 monoclonal antibody for the treatment of systemic lupus erythematosus: a phase II dose-ranging randomised controlled trial. Ann. Rheum. Dis., 534–542 (2017).Liu, S. M. & King, C. IL-21-producing Th cells in immunity and autoimmunity. 1913501–3506 (2013).Konforte, D., Simard, N. & Paige, C. J. IL-21: an executor of B cell fate. , 1781–1787 Kuchen, S. etal. Essential role of IL-21 in B cell activation, expansion, and plasma cell generation Tcell-B cell collaboration. , 5886–5896 (2007).Wu, Y. etal. The biological effects of IL-21 signaling cell-mediated responses in organ transplantation. Front. Immunol., 319 (2016).Pritchard, N. R. & Smith, K. G. B cell inhibitory receptors and autoimmunity. 108Gagneux, P. etal. Human-specific regulation of 2-6-linked sialic acids. Tedder, T. F., Poe, J. C. & Haas, K. M. CD22: a multifunctional receptor that regulates B lymphocyte survival and signal transduction. Adv. Immunol.Lumb, S. etal. Engagement of CD22 on B cells withthe monoclonal antibody epratuzumab stimulates thephosphorylation of upstream inhibitory signals of the B cell receptor. 143–151 (2016).100Nimmerjahn, F. & Ravetch, J. V. Fc receptors as regulators of immune responses. Nat. Rev. Immunol.Sohn, H. W., Pierce, S. K. & Tzeng, S. J. Live cell imaging reveals that the inhibitory FcRIIB destabilizes B cell receptor membrane-lipid interactions and blocks immune synapse formation. 102Kleinau, S. The impact of Fc receptors on the development of autoimmune diseases. Curr. Pharm. Des., 1861–1870 (2003).103Meyaard, L. The inhibitory collagen receptor LAIR-1 J. Leukoc. Biol.104Franks, S. E., Getahun, A., Hogarth, P. M. & Cambier, J. C. Targeting B cells in treatment of autoimmunity. Curr. Opin. Immunol.(2016).105Chatenoud, L. Biotherapies targeting T and B cells: from immune suppression to immune tolerance. Curr.Opin. Pharmacol., 92–97 (2015).106Hardy, I. R. etal. Anti-CD79 antibody induces B cell anergy that protects against autoimmunity. , 1641–1650 (2014).Rossi, E. A., Chang, C. H. & Goldenberg, D. M. CD22/CD20 Bispecific antibody with enhanced trogocytosis for treatment of Lupus. e98315 (2014).108Donahue, A. C. & Fruman, D. A. Proliferation and survival of activated B cells requires sustained antigen receptor engagement and phosphoinositide 3-kinase activation. 170, 5851–5860 (2003).109Galinier, A., Delwail, V. & Puyade, M. Ibrutinib is effective in the treatment of autoimmune haemolytic , 127–129 (2017).110Muschen, M. Autoimmunity checkpoints as therapeutic targets in B cell malignancies. Nat. Rev. Cancer103–116 (2018).Ryden-Aulin, M. etal. Off-label use of rituximab for systemic lupus erythematosus in Europe. Lupus Sci. , e000163 (2016).112Hauser, S. L. etal. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N. Engl. J.Med., 221–234 (2017).113Kappos, L. etal. Ocrelizumab in relapsing-remitting multiple sclerosis: a phase 2, randomised, placebo-controlled, multicentre trial. , 1779–1787 (2011).114Mysler, E. F. etal. Efficacy and safety of ocrelizumab in active proliferative lupus nephritis: results from a randomized, double-blind, phase III study. Arthritis , 2368–2379 (2013).115Stashenko, P., Nadler, L. M., Hardy, R, & Schlossman, S. F. Expression of cell surface markers after human B lymphocyte activation. Proc. Natl Acad. Sci. USA, 3848–3852 (1981).116DiLillo, D. J. etal. Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice. , 361–371 (2007).117Forsthuber, T. G., Cimbora, D. M., Ratchford, J. N., Katz, E. & Stuve, O. B cell-based therapies in CNS autoimmunity: differentiating CD19 and CD20 as therapeutic targets. Ther. Adv. Neurol. Disord.1756286418761697 (2018).118Herbst, R. etal. B cell depletion invitro and invivo with an afucosylated anti-CD19 antibody. J. Pharmacol. Exp. Ther., 213–222 (2010).119Chen, D. etal. Single dose of glycoengineered anti-CD19 antibody (MEDI551) disrupts experimental autoimmune encephalomyelitis by inhibiting pathogenic adaptive immune responses in the bone marrow and spinal cord while preserving peripheral regulatory mechanisms. , 4823–4832 (2014).Chen, D. etal. Autoreactive CD19CD20- plasma cells contribute to disease severity of experimental autoimmune encephalomyelitis. 1541–1549 (2016).121Agius, M. A. etal. Safety and tolerability of inebilizumab (MEDI-551), an anti-CD19 monoclonal antibody, in patients with relapsing forms of multiple sclerosis: results from a phase 1 randomised, placebo-controlled, escalating intravenous and subcutaneous dose study. Mult. Scler., 235–245 (2019).Mei, H. E. etal. A unique population of IgG-expressing plasma cells lacking CD19 is enriched in human bone marrow. , 1739–1748 (2015).Ruck, T., Bittner, S., Wiendl, H. & Meuth, S. G. Alemtuzumab in multiple sclerosis: mechanism of action and beyond. , 16414–16439 (2015).Tolbert, V. P. etal. Daratumumab is effective in the treatment of refractory post-transplant autoimmune hemolytic anemia: a pedia

tric case report. 4819–4819 (2016).Jagannath, S. etal. Indatuximab ravtansine (BT062) monotherapy in patients with relapsed and/or refractory multiple myeloma. (2019).Usmani, S. Z. etal. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. 37–44 (2016).Sidiropoulos, P. I. & Boumpas, D. T. Lessons learned from anti-CD40L treatment in systemic lupus erythematosus patients. , 391–397 (2004).Chamberlain, C. etal. Repeated administration of dapirolizumab pegol in a randomised phase I study is well tolerated and accompanied by improvements in several composite measures of systemic lupus erythematosus disease activity and changes in whole blood transcriptomic profiles. Ann. Rheum. Dis.1837–1844 (2017).Fisher, B. etal. The novel anti-CD40 monoclonal antibody CFZ533 shows beneficial effects in patients with primary Sjögren’s syndrome: a phase IIa double-controlled randomized trial. Abstract presented at 2017 ACR/ARHP Annual Meeting (SanDiego, CA).Johnson, P. etal. Clinical and biological effects of an CD40 antibody: a Cancer Research UK phase I study. Clin. Cancer Res., 1321–1328 (2015).131Dahan, R. etal. Therapeutic activity of agonistic, human CD40 monoclonal antibodies requires selective , 820–831 (2016).Cheng, L. E. etal. Brief report: a randomized, double-blind, parallel-group, placebo-controlled, multiple-dose study to evaluate AMG 557 in patients with systemic lupus erythematosus and active lupus arthritis. Arthritis Rheumatol., 1071–1076 (2018).Bluml, S., McKeever, K., Ettinger, R., Smolen, J. & Herbst, R. B cell targeted therapeutics in clinical development. Arthritis Res. Ther. (Suppl. 1), 4 (2013).Clowse, M. E. etal. Efficacy and safety of epratuzumab in moderately to severely active systemic lupus erythematosus: results from two phase III randomized, blind, placebo-controlled trials. Arthritis Rheumatol., 362–375 (2017).Chu, S. Y. etal. Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement engineered antibodies. US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov(2018).Horton, H. M. etal. Antibody-mediated coengagement RIIb and B cell receptor complex suppresses humoral immunity in systemic lupus erythematosus. J.Immunol., 4223–4233 (2011).Chu, S. Y. etal. Suppression of rheumatoid arthritis Bcells by XmAb5871, an anti-CD19 antibody that coengages B cell antigen receptor complex and Fcreceptor IIb inhibitory receptor. Arthritis Rheumatol., 1153–1164 (2014).Veri, M. C. etal. Therapeutic control of B cell activation via recruitment of Fc receptor IIb (CD32B) inhibitory function with a novel bispecific antibody scaffold. , 1933–1943 (2010).141US National Library of Medicine. ClinicalTrials.gov (2017).Pal Singh, S., Dammeijer, F. & Hendriks, R. W. Role of Bruton’s tyrosine kinase in B cells and malignancies. , 57 (2018).Gillooly, K. M. etal. Bruton’s tyrosine kinase inhibitor BMS-986142 in experimental models of rheumatoid arthritis enhances efficacy of agents representing clinical standard-of-care. , e0181782 (2017).Lee, S. K. etal. Safety, pharmacokinetics, and pharmacodynamics of BMS-986142, a novel reversible BTK inhibitor, in healthy participants. Eur. J. Clin. , 689–698 (2017).Crawford, J. J. etal. Discovery of GDC-0853: a potent, selective, and noncovalent Bruton’s tyrosine kinase inhibitor in early clinical development. , 2227–2245 (2018).Akinleye, A., Chen, Y., Mukhi, N., Song, Y. & Liu, D. Ibrutinib and novel BTK inhibitors in clinical development. J. Hematol. Oncol., 59 (2013).Katewa, A. etal. Btk-specific inhibition blocks pathogenic plasma cell signatures and myeloid cell-driven lupus nephritis. , e90111 (2017).Gopal, A. K. etal. PI3Kdelta inhibition by idelalisib in patients with relapsed indolent lymphoma. N. Engl. 370, 1008–1018 (2014).Stark, A. K., Sriskantharajah, S., Hessel, E. M. & Okkenhaug, K. PI3K inhibitors in inflammation, autoimmunity and cancer. Curr. Opin. Pharmacol.82–91 (2015).Baker, K. P. etal. Generation and characterization of LymphoStat-B, a human monoclonal antibody that antagonizes the bioactivities of B lymphocyte stimulator. 151Navarra, S. V. etal. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. , 721–731 (2011).Dall’Era, M. etal. Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-controlled, dose-4142–4150 (2007).Furie, R. etal. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. 3918–3930 (2011).Merrill, J. T. etal. Efficacy and safety of atacicept in patients with systemic lupus erythematosus: results of a twenty-four-week, multicenter, randomized, blind, placebo-controlled, parallel-arm, phase IIb study. Arthritis Rheumatol., 266–276 (2018).Jego, G. etal. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon Yao, X. etal. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol. Ther.141, 125–139 (2014).Wallace, D. J. etal. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. , 222–231 (2018).Ghoreschi, K. etal. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J.Immunol., 4234–4243 (2011).Kubo, S., Nakayamada, S. & Tanaka, Y. Baricitinib for the treatment of rheumatoid arthritis. Expert Rev. , 911–919 (2016).Tanaka, Y. & Yamaoka, K. JAK inhibitor tofacitinib for treating rheumatoid arthritis: from basic to clinical. Mod. Rheumatol., 415–424 (2013).161Wang, S. P. etal. Tofacitinib, a JAK inhibitor, inhibits human B cell activation invitro. Ann. Rheum. Dis.2213–2215 (2014).Rizzi, M. etal. Impact of tofacitinib treatment on human B cells invitro and invivo. J. Autoimmun.55–66 (2017).Ignatenko, S., Skrumsager, B. K., Steensberg, A. & Mouritzen, U. First in human study with recombinant IL-21 monoclonal antibody in healthy subjects and patients with rheumatoid arthritis. Abstract presented at 2012 ACR/ARHP Annual Meeting (Washington, DC).Wa

gner, F., Skrumsager, B. K. & Fitilev, S. Safety andtolerability of NNC0114IL-21 monoclonal antibody, at multiple s.c. dose levels in patients with rheumatoid arthritis. Abstract presented at 2014 ACR/ARHP Annual Meeting (Boston, MA).Ignatenko, S., Skrumsager, B. K. & Mouritzen, U. Safety, PK, and PD of recombinant anti-interleukin-21 monoclonal antibody in a first-Int. J. Clin. Pharmacol. Ther., 243–252 (2016).Yednock, T. A. etal. Prevention of experimental autoimmune encephalomyelitis by antibodies against NatureGuagnozzi, D. & Caprilli, R. Natalizumab in the treatment of Crohn’s disease. , 275–284 Yaldizli, O. & Putzki, N. Natalizumab in the treatment of multiple sclerosis. Ther. Adv. Neurol. Disord. 115–128 (2009). Stuve, O. etal. Immune surveillance in multiple sclerosis patients treated with natalizumab. Ann.Neurol.170Stüve, O. The effects of natalizumab on the innate and adaptive immune system in the central nervous system. J. Neurol. Sci., 39–41 (2008).171Tocheva, A. S. & Mor, A. Checkpoint inhibitors: applications for autoimmunity. Curr. Allergy Asthma , 72 (2017).Agata, Y. etal. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int.Immunol.Okazaki, T., Iwai, Y. & Honjo, T. New regulatory receptors: inducible co-stimulator and PD-1. Curr. Opin. Immunol.Thibult, M. L. etal. PD-1 is a novel regulator of human B cell activation. , 129–137 (2013).Topalian, S. L. etal. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J.Med., 2443–2454 (2012).Hess, K. L. etal. Engineering immunological tolerance using quantum dots to tune the density of self-antigen display. Adv. Funct. Mater., 1700290 (2017).Ortiz, D. F. etal. Elucidating the interplay between Fc valency and Fc activation for the design of immune complex inhibitors. Sci. Transl Med.365ra158 (2016).Karnell, J. L. etal. CD19 and CD32b differentially regulate human B cell responsiveness. , 1480–1490 (2014).Wu, X. & Demarest, S. J. Building blocks for bispecific and trispecific antibodies. , 3–9 (2019).Moore, P. A. etal. Application of dual affinity retargeting molecules to achieve optimal redirected Tcell killing of 117, 4542–4551 (2011).181Huehls, A. M., Coupet, T. A. & Sentman, C. L. Bispecific Tcell engagers for cancer immunotherapy. , 290–296 (2015).US National Library of Medicine. ClinicalTrials.gov (2018).Ikeda, H. etal. The monoclonal antibody nBT062 conjugated to cytotoxic Maytansinoids has selective cytotoxicity against CD138-positive multiple myeloma cells invitro and invivo. Clin. Cancer Res.US National Library of Medicine. ClinicalTrials.gov (2017).US National Library of Medicine. ClinicalTrials.gov (2019).US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov (2019).US National Library of Medicine. ClinicalTrials.gov (2014).US National Library of Medicine. ClinicalTrials.gov (2019).191US National Library of Medicine. ClinicalTrials.gov (2017).US National Library of Medicine. ClinicalTrials.gov (2013).US National Library of Medicine. ClinicalTrials.gov (2015).US National Library of Medicine. ClinicalTrials.gov (2018).Byrd, J. C. etal. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J.Med., 32–42 (2013).US National Library of Medicine. ClinicalTrials.gov (2013).Stohl, W. etal. Efficacy and safety of subcutaneous belimumab in systemic lupus erythematosus: two-week randomized, double-controlled study. Arthritis Rheumatol., 1016–1027 (2017).US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov (2016).US National Library of Medicine. ClinicalTrials.gov (2016).201US National Library of Medicine. ClinicalTrials.gov (2016).US National Library of Medicine. ClinicalTrials.gov (2017).US National Library of Medicine. ClinicalTrials.gov (2019).Khanna, D. etal. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. , 2630–2640 (2016).US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov (2017).207Fleischmann, R. etal. Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, to-head, randomised controlled , 457–468 (2017).US National Library of Medicine. ClinicalTrials.gov (2019).US National Library of Medicine. ClinicalTrials.gov (2018).US National Library of Medicine. ClinicalTrials.gov(2017).211Taylor, P. C. etal. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. , 652–662 (2017).212US National Library of Medicine. ClinicalTrials.gov (2017).213US National Library of Medicine. ClinicalTrials.gov (2019).214US National Library of Medicine. ClinicalTrials.gov (2019).215US National Library of Medicine. ClinicalTrials.gov (2019).216US National Library of Medicine. ClinicalTrials.gov (2016).217US National Library of Medicine. ClinicalTrials.gov (2016).218Polman, C. H. etal. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N.Engl. J. Med., 899–910 (2006).219US National Library of Medicine. ClinicalTrials.gov (2017).AcknowledgementsS.J.S.R. thanks the National Science Foundation Graduate Research Fellowship and the Stanford Graduate Fellowship for their generous support. This Review is based upon work supported by the National Science Foundation Graduate Research Fellowship Program under grant no. DGE–1656518. Any opinions, findings and conclusions or recommendations expressed in this material are those of the author(s) and do not necessarily reflect the views of the National Science Foundation. Research in the laboratory of W.H.R. is supported by US National Institutes of Health (NIH) National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) grants R01 AR063676, U19 AI11049103 and U01 AI101981.Author contributionsThe authors contributed equally to all aspects of the article.Competing interestsThe authors declare no competing interests.Publisher’s noteSpringer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affili