Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Apr 1.
Published in final edited form as: Nat Rev Rheumatol. 2017 Mar 9;13(4):217–233. doi: 10.1038/nrrheum.2017.22

Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases

Michael Croft 1, Richard M Siegel 2
PMCID: PMC5486401  NIHMSID: NIHMS868295  PMID: 28275260

Abstract

TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.


Over 30 years have passed since the molecular identification of TNF as a mediator of fever and cachexia1, and approximately 20 years since the first introduction of TNF inhibitors into clinical practice for the treatment of rheumatoid arthritis (RA)2. During this time, much has been learned about the basic biology of the 19 structurally related cytokines of the TNF superfamily (TNFSF), their receptors (TNF receptor superfamily, TNFRSF), the intracellular signalling pathways activated by these receptors, as well as the unique and overlapping roles of TNFSF cytokines in a number of inflammatory and autoimmune diseases. TNFSF proteins organize lymphoid tissue development, co-stimulate lymphocyte activation and can either increase lymphocyte survival and function or induce cell death36. Outside the immune system, TNFSF cytokines can promote the development and survival of osteoclasts, as well as cells in the mammary glands, hair follicles and sweat glands. TNFSF cytokines can also regulate neuronal activity and drive inflammatory responses in a range of tissue structural cells, including epithelial cells and fibroblasts. These insights have led to intensive efforts to treat other inflammatory diseases through TNF neutralization, and multiple TNF-blocking agents (such as adalimumab, certolizumab pegol, etanercept, golimumab and infliximab) are now approved for diseases such as juvenile idiopathic arthritis, psoriasis, psoriatic arthritis, spondylarthropathies, inflammatory bowel disease and uveitis7,8 (TABLE 1). Investigations into the targeting of other TNFSF members have led to a number of clinical trials in different diseases and resulted in the successful development of belimumab, an antibody against B cell activating factor (BAFF, also known as TNFSF13B), and denosumab, an antibody targeting receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL, also known as TNFSF11), for the treatment of systemic lupus erythematosus (SLE) and osteoporosis, respectively911.

Table 1.

Clinical trials of TNF and TNF receptor superfamilies

TNF family ligand TNF family receptor Biologic agent targeting receptor or ligand Name of biologic agent Stage of drug development for targeted disease(s)
TNF TNFR1, TNFR2 Chimeric anti-TNF mAb Infliximab Approved: AS, CD, PsA, psoriasis, RA, UC
Human TNFR2-Fc fusion protein Etanercept Approved: AS, JIA, PsA, psoriasis, RA
Human anti-TNF mAb Adalimumab
  • Approved: AS, Crohn disease, JIA, PsA, psoriasis, RA, UV

  • Phase III (recruiting): UC

  • Phase III (recruiting): Behcet disease

Human PEGylated Fab anti-TNF mAb Certolizumab pegol
  • Approved: CD, RA

  • Phase III (completed): AS, PsA

  • Phase III (ongoing): psoriasis

  • Phase II (recruiting): UC

Human anti-TNF mAb Golimumab
  • Approved: AS, PsA, RA

  • Phase IV: UC

  • Phase II (completed): asthma

Recombinant human TNF conjujugated to KLH TNF-Kinoid Phase II (completed): CD, RA
LTα3 TNFR1, TNFR2 Human TNFR2-Fc fusion protein Etanercept Approved: AS, JIA, PsA, psoriasis, RA
Human anti-LTα mAb Pateclizumab (MLTA3698A) Phase II (completed): RA
LTα1β2 LTβR Human LTβR-Ig fusion protein Baminercept (BG9924) Phase II (terminated due to lack of activity): RA, Sjögren syndrome
OX40 ligand OX40 Human anti-OX40L mAb Oxelumab Phase II (discontinued owing to lack of activity): asthma
Human anti-OX40 mAb KHK4083 Phase II (recruiting): UC
Human anti-OX40 mAb GBR830 Phase II, (recruiting): AD
CD40L CD40 Humanized anti-CD40L mAb Ruplizumab (BG9588) Phase II (discontinued owing to safety issues): lupus nephritis
Humanized anti-CD40L mAb Toralizumab (IDEC-131) Phase II (discontinued owing to safety issues): CD, MS
Anti-CD40L-Tn3 fusion protein MEDI4920 Phase I (recruiting): RA
Chimeric anti-CD40 mAb FFP104 (PG102) Phase I (recruiting): CD, primary biliary cirrhosis
Human anti-CD40 Fc-silent mAb CFZ533 Phase I–II (recruiting): Grave disease, MG, RA, SS, transplantation
Human anti-CD40 mAb ASKP1240 (4D11)
  • Phase II (completed): psoriasis

  • Phase II (ongoing): transplantation

RANKL RANK Human anti-RANKL mAb Denosumab
  • Approved: osteoporosis

  • Phase III (ongoing): RA

  • Phase II (recruiting): OA

  • Phase I–II (recruiting): CD

TWEAK Fn14 Humanized anti-TWEAK mAab BIIB023
  • Phase II (terminated due to lack of activity): lupus nephritis

  • Phase I (completed): RA

APRIL TACI, BCMA Human TACI-Ig fusion protein Atacicept
  • Phase II (completed): RA

  • Phase II (ongoing): SLE

  • Phase II (terminated due to safety issues): lupus nephritis

  • Phase II (terminated due to increased disease): MS

BAFF BAFFR, BCMA, TACI Human anti-BAFF mAb Belimumab
  • Approved: SLE

  • Phase II (completed): MG, RA, Sjogren syndrome

  • Phase II (ongoing): SSc

Human anti-BAFF mAb Tabalumab (LY2127399)
  • Phase III (completed): RA, SLE

  • Phase II (completed): MS

Human TACI-Ig fusion protein Atacicept
  • Phase II (completed): RA

  • Phase II (ongoing): SLE

  • Phase II (terminated due to safety issues): lupus nephritis

  • Phase II (terminated due to increased disease): MS

Human BAFF-binding peptibody Blisibimod (AMG623) Phase III (ongoing or recruiting): SLE
LIGHT HVEM, LTβR Human LTβR-Ig fusion protein Baminercept (BG9924)
  • Phase II (completed and terminated due to lack of activity): RA

  • Phase II (terminated due to unavailability of biologic): Sjögren syndrome

Human anti-LIGHT mAb KHK252067 Phase I (completed): CD, UC
NGF* NGFR Humanized anti-NGF mAb Tanezumab (RN624) Phase III (recruiting): chronic back pain, osteoarthritis
Human anti-NGF mAb Fulranumab (AMG-403) Phase III (ongoing): osteoarthritis
Human anti-NGF mAb Fasinumab (REGN475) Phase III (ongoing): osteoarthritis
Phase III (recruiting): chronic back pain

AD, atopic dermatitis; APRIL, A proliferation-inducing ligand; AS, ankylosing spondylitis; BAFF, B-cell-activating factor; BAFFR, BAFF receptor; BCMA, B-cell maturation antigen; CD, Crohn’s disease; Fn14, Fibroblast growth factor-inducible protein 14; HVEM, Herpes virus entry mediator; JIA, juvenile idiopathic arthritis; LT, lymphotoxin; LTβR, LTβ receptor; NGF, nerve growth factor; NGFR, NGF receptor; mAb, monoclonal antibody; MG, myasthenia gravis; PsA, psoriatic arthritis; RA, rheumatoid arthritis; RANK, receptor activator of nuclear factor kappa-B (NF-κB); RANKL, RANK ligand; SSc, systemic sclerosis; TACI, transmembrane activator and CAML interactor; TNFR, TNF receptor; TWEAK, TNF-related weak inducer of apoptosis; UC, ulcerative colitis.

*

NGF is not a canonical TNF family ligand on the basis of structure, although NGFR is part of the TNFR superfamily.

Depleting and/or antagonist biologics (all other biologic agents displayed are antagonists).

Clinical targeting of TNF, BAFF and RANKL has been reviewed elsewhere710,1217, as has the targeting of all the TNF and TNFRSF members in both immune and nonimmune disorders11. In this Review, we focus on TNF family proteins that are produced by the immune system but are not yet targets of approved drugs. These molecules might be crucial to the immune response underlying rheumatic diseases and are promising future targets for intervention and therapy in diseases such as RA and SLE (FIG. 1). Although blocking nerve growth factor binding to its receptor TNFRSF16 (also known as nerve growth factor receptor) is of primary interest for the treatment of pain associated with osteoarthritis, TNFRSF16 is not an immune-system-related molecule and so we do not present a discussion here but refer readers to several other published articles11,1821.

Figure 1. Select members of the TNF and TNFR superfamily implicated in rheumatic diseases.

Figure 1

TNF superfamily ligands (TNFSF; top) are active primarily as non-covalently associated homotrimers and can be soluble or membrane-expressed. TNF superfamily receptors (TNFRSF; bottom) contain variable numbers of cysteine-rich domains in their ligand-binding extracellular regions. TNFRSF are mainly membrane-expressed, but can form soluble receptors via enzymatic cleavage of the ectodomains. Also depicted are the primary cell targets that respond to TNFSF through TNFRSF signalling, although this list is not comprehensive in terms of the expression characteristics of each molecule. TNFRSF molecules whose main function is to promote apoptotic cell death (TNFR1, Fas, TNF-related apoptosis-inducing ligands 1 (TRAIL1) and TRAIL2) can recruit a death-inducing signalling complex to their cytoplasmic domains via a death domain. 4-1BBL, 4-1BB ligand; APRIL, a proliferation-inducing ligand; BAFF, B-cell-activating factor; BAFFR, BAFF receptor; BCMA, B-cell maturation antigen; CD40L, CD40 Ligand; DR3, death receptor 3; FasL, Fas ligand; Fn14, fibroblast growth factor-inducible immediate-early response protein 14; GITRL, glucocorticoid-induced TNF receptor-related (GITR) ligand; HVEM, herpes virus entry mediator; LT, lymphotoxin; OX40L, OX40 ligand; RANKL, receptor activator of nuclear factor-κB (RANK) ligand; TACI, transmembrane activator and CAML interactor; TL1A, TNF-like ligand 1; TWEAK, TNF-related weak inducer of apoptosis.

TNF superfamily

Multiple functional polymorphisms in the genes encoding TNFSF cytokines, their receptors and their signalling proteins are associated with susceptibility to autoimmune diseases11,22. Yet, many functions of TNFSF proteins remain poorly understood. TNFSF and TNFRSF proteins have many structural and biological similarities (FIG. 1). TNFSF molecules are trimeric type II transmembrane proteins characterized by C-terminal TNF homology domains that can be cleaved from cells to form soluble ‘cytokine-like’ molecules23. Their receptors are type I transmembrane proteins that have varying numbers of extracellular ligand-binding cysteine-rich domains23. The extracellular domains of the TNFRSF can also be cleaved to form soluble molecules, which might be useful as biomarkers for inflammation, although their exact function is not clear. Engagement of receptors by their cognate ligands is thought to primarily lead to trimerization of the receptors, which can further form higher-order oligomers on a cell’s surface. Although overall sequence similarity between TNFRSF molecules is low (20–30%), once engaged by their cognate ligands they can drive common or overlapping signalling pathways2426 (FIG. 2). Moreover, membrane-bound TNF family ligands can also signal through themselves when engaged to their cognate receptors (a process known as reverse signalling), which might contribute to their function. When crosslinked on the surface of cells, various consequences of reverse signalling have been described, such as proinflammatory cytokine production (for example IL-1 and IL-6) and cell maturation, which depend on the cell type that receives the TNFSF ligand signal27.

Figure 2. General TNFSF receptor signalling.

Figure 2

TNF receptor superfamily (TNFRSF) proteins recruit one or several adaptor proteins (TNFR associated factors 1 to 6 (TRAFs), TNFR associated death domain protein (TRADD) and Fas associated death domain protein (FADD)) after ligand binding. As a generalization, TNFRSF proteins that utilize TRAFs (left) can be regarded as proinflammatory and induce proliferation (cell cycle proteins), survival (anti-apoptotic proteins), differentiation and production of inflammatory mediators such as cytokines and chemokines, according to the responding cell type. These processes can be induced via activation of one or both nuclear factor κB (NF-κB) signalling pathways (canonical and non-canonical) as well as via MAP kinase cascades. The canonical NF-κB signalling pathway is IKKβ-dependent and involves phosphorylation of inhibitor of κB (IkBα) and nuclear translocation of NF-κB subunit p50 and transcription factor p65 (RelA); the non-canonical NF-κB signalling pathway is IKKα-dependent and involves activation of NF-κB-inducing kinase (NIK), processing of p100 to p52, and nuclear translocation of p52 and RelB. The MAP kinase cascades involve c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), p38 or other kinases such as serine/threonine-protein kinase (AKT). TNFRSF members that contain a ‘death domain’ (right) and recruit the death domain-containing adaptor protein FAS-associated death domain protein (FADD), such as Fas, TNF-related apoptosis-inducing ligands 1 (TRAIL1) and TRAIL2, are often regarded as anti-inflammatory as they generally lead to cell death (apoptosis or necroptosis) through activation of cysteine-aspartic proteases (caspases) and receptor-interacting serine/threonine-protein (RIP) kinases (not shown). TNFR1 and death receptor 3 (DR3), the receptors that recruit the TRADD adaptor proteins, can activate inflammatory responses as TRADD can recruit TRAF proteins, but additionally activate death pathways through secondary complexes containing TRADD, FAS-associated death domain protein (FADD) and caspase 8. Adapted from Nat. Rev. Drug Discov. 12, 147–68 (2013) © Macmillan Publishers Limited11.

The expression of TNF family proteins is quite broad and dynamically regulated (FIG. 1). Many ligand–receptor pairs are constitutive or inducible on lymphocytes, including antigen presenting cells (APCs; such as dendritic cells, macrophages and B cells) and T cells, and normally participate in promoting T and B cell responses, which are central to most autoimmune and rheumatic diseases. Similarly, death-inducing molecules can also be expressed by lymphocytes, and participate in maintaining self-tolerance and limiting adaptive immune responses. Additionally, a number of TNF family ligands and/or receptors are constitutive or inducible in non-lymphoid cells including epithelial cells, fibroblasts, smooth muscle cells, and endothelial cells. These molecules participate in the proinflammatory and anti-inflammatory crosstalk that occurs between tissue structural cells and the immune system, which might either contribute to autoimmune tissue pathology or limit damage.

Below, we discuss the biological activities of TNFSF members and their potential involvement in rheumatic diseases. For simplicity, we have grouped TNFSF proteins, as described above, into immune cell activators, tissue inflammatory proteins and molecules that induce cell death or immune suppression. This classification is not absolute and the reader should be aware that molecules such as TNF, CD40 ligand (CD40L, also known as TNFSF5), LIGHT (also known as TNFSF14), TNF-like ligand 1A (TL1A, also known as TNFSF15), and TNF-related apoptosis inducing ligand (TRAIL, also known as TNFSF10) can exert functions on both immune cells and tissue cells (FIG. 1). Moreover, a number of proteins, including TNF and Fas ligand (FasL, also known as TNFSF6), are able to promote cell death as well as being proinflammatory, depending on the target cell type and the context in which they are active.

Immune cell activation

CD40L

CD40 (also known as TNFRSF5) is a stimulatory receptor expressed on dendritic cells, macrophages and B cells, whose signals drive activation, maturation, survival and inflammatory cytokine production28,29 (FIG. 3). CD40 is crucial in both inducing IgG autoantibodies and driving immunoglobulin class switching30,31, and is also a primary driver of T cell immunity. Its ligand, CD40L, is induced in T cells shortly after activation and, via ligation of CD40 on professional APCs, can lead to an increase in antigen presentation and activation of T cells by upregulating MHC molecules and inducing expression of stimulatory ligands such as CD86 and those belonging to the TNF superfamily, which are described below (for example OX40 ligand (OX40L))32,33.

Figure 3. TNFSF activities enhancing immune cell activation.

Figure 3

The simplified diagram highlights the possible interactions between TNF superfamily (TNFSF) ligands and TNF receptor superfamily (TNFRSF) proteins expressed on several cells in the immune system (antigen-presenting cells (APCs), B cells, and T cells). Driven by the appropriate antigen, T cells can receive TNFRSF signals through OX40, glucocorticoid-induced TNF receptor-related protein (GITR), death receptor 3 (DR3), CD27, and 4-1BB. These signals enhance their activation, promote division and survival to augment the size of the autoreactive pool, induce differentiation of follicular helper T (TFH) cells that control antibody responses and induce the expression of cytokines that drive tissue pathology. APCs (dendritic cells and macrophages), via CD40, can upregulate MHC molecules, co-stimulatory ligands (including TNFSF molecules) and inflammatory cytokines, which aid the T-cell response. B cells can receive signals from CD40, CD27, GITR, B-cell-activating factor (BAFF) receptor (BAFFR), B-cell maturation antigen (BCMA) and transmembrane activator and CAML interactor (TACI). These signals drive activation, division and survival, class switching, and plasma cell differentiation, resulting in production of pathogenic autoantibodies. Reverse signalling through membrane-expressed TNFSF ligands such as OX40 ligand (OX40L), CD70 and 4-1BBL, expressed on dendritic cells, macrophages and B cells, can also augment production of inflammatory cytokines and help B cell differentiation. Other reported activities of TNFRSF signalling on immune cells such as mast cells, eosinophils, neutrophils, basophils, Natural killer T cells and innate lymphoid cells are not shown, but these can further result in production of inflammatory mediators that contribute to tissue pathology and amplify the T-cell and B-cell response.

Studies have long linked the interaction between CD40L and CD40 to rheumatic disease pathogenesis. In the early 1990s, studies of multiple autoimmune models, including collagen-induced arthritis and lupus-like disease in NZB/SWR or NZB/NZW F1 mice3436, demonstrated markedly reduced signs of inflammation in mice lacking either CD40 or CD40L, or in wildtype mice treated with CD40L blocking reagents. Similar to other molecules discussed below, the idea that the CD40L–CD40 axis is also active in human disease largely derives from expression studies in patients. The caveat with expression studies is that detection of the molecules in serum or tissues does not automatically imply they are functional or important, but could simply reflect the presence of activated immune cells. However, such data, particularly for conventional cytokines such as IL-5, IL-13, and IL-17, has aided their clinical targeting and enabled patient stratification into those most likely to respond to biologic agents. Therefore, with TNFSF molecules the expression data are highly useful regardless of the caveats, especially if linked to either other disease markers or the magnitude of clinical symptoms. Soluble CD40L in serum, or CD40L expression in inflamed tissue, epithelial cells, endothelium or T cells, is upregulated in patients with RA, psoriatic arthritis, ankylosing spondylitis, SLE, Sjögren syndrome and systemic sclerosis (Ssc), often correlating with disease severity or levels of autoantibodies28,29. Additionally, polymorphisms near the genes encoding CD40L or CD40, which are thought to lead to elevated or prolonged expression, have been associated with susceptibility to SLE, RA and other rheumatic disorders (such as Behçet disease)3744.

Animal studies have shown that the neutralization of CD40L has a strong suppressive effect on pathogenic T cell development and antibody responses. These results, together with data from human expression and association studies, made CD40L an attractive therapeutic target for rheumatic diseases, particularly SLE and RA. As reviewed elsewhere11,28,29, phase I–II trials in several patient groups, including patients with lupus nephritis, demonstrated some beneficial activity of antibodies against CD40L (such as ruplizumab, ab1793 and toralizumab)4547. Unfortunately, the thromboembolic activity of these antibodies, linked to crosslinking of CD40L expressed by platelets, led to discontinuation of their further development (TABLE 1). To circumvent the thromboembolic effect, preclinical studies in mice or nonhuman primates are assessing new biologic agents that block CD40L without causing aggregation of the molecule; these biologic agents either lack an Fc region or are mutated to prevent their binding to Fc receptors. Results suggest that they can be as efficacious as the parent (Fc intact) antibody — without the thromboembolic effect — in scenarios such as animal models of lupus4850. However, in certain settings Fc effector function might be necessary for therapeutic activity, as shown by the lack of activity of an aglycosylated anti-CD40L antibody in nonhuman primate transplantation studies48. MEDI4920, a Tn3-fusion protein with reactivity to CD40L, is currently in phase I safety trials. Additionally, antagonist and/or depleting antibodies against CD40 have been produced (ch5D12, chi220–BMS-224819, ASKP1240, FFP104, CFZ533), with encouraging preclinical results51, and some of them are being tested in phase I–II trials in Sjögren syndrome52, RA53 and other autoimmune conditions (TABLE 1). If these strategies can overcome the adverse effects associated with agents that block CD40-C40L interactions, such agents are an attractive avenue, and the possibility for clinical benefit in rheumatic diseases is high.

OX40L

OX40L (also known as TNFSF4) is an inducible molecule expressed on several cell types, although arguably most importantly, on APCs. OX40 (also known as TNFRSF4) is largely found on activated T cells as well as natural killer T cells and innate lymphoid cells such as natural killer cells5,54 (FIG. 3). OX40L can trigger signalling through its receptor OX40, resulting in a range of activities including expansion and accumulation of effector T cells (such as type 1 T helper cells (TH1), type 2 T helper cells (TH2), type 17 T helper cells (TH17) and cytotoxic T lymphocytes) and their cytokine production5,6,11,54. Additionally, reverse signalling through OX40L can promote expression of inflammatory cytokines (such as IL-12 or TNF) in APCs5,54, although the importance of this activity as compared with that driven by OX40 is not clear at present.

Data from human and mouse studies suggest that the OX40–OX40L axis has an important role in rheumatic diseases. Blockade of OX40L reduces bone and cartilage destruction in mouse models of collagen-induced arthritis55,56 or autoimmune arthritis57, with results from the former model being attributed to reduced numbers of collagen-specific T cells. Synovial fluid samples of patients with RA contain elevated numbers OX40-expressing T cells, suggesting OX40 signalling controls T cell numbers in human RA56,58. Targeting of OX40 with cytotoxic drugs to deplete T cells has also shown some therapeutic benefit in an animal model of adjuvant arthritis59. Surprisingly, signalling via OX40L antagonizes the activity of RANK in promoting osteoclast development from macrophage progenitors. OX40L-deficient mice are accordingly osteopenic56, although the implication of this finding in the context of therapeutic inhibition of OX40–OX40L interactions in arthritis is not clear.

In patients with SLE who have proliferative glomerulonephritis, OX40L is upregulated in glomeruli, most likely on endothelial cells60 and/or dendritic cells61. Similarly, studies have shown that in peripheral blood and renal biopsy samples from patients with lupus nephritis, OX40 expression by CD4+ T cells correlates with disease activity, urine proteinuria and serum creatinine6265. Furthermore, on the basis of an initial report66, many studies have confirmed an association between susceptibility to developing SLE and polymorphisms upstream of the OX40L gene (also known as TNFSF4), which probably leads to its increased expression. The OX40–OX40L axis is also involved in kidney disease, as patients with Henoch–Schönlein purpura with nephritis have elevated levels of serum OX40L and OX40+ T cell numbers compared with patients without nephritis67. Surprisingly, no reports have yet demonstrated a functional role for these molecules in mouse models of nephritis, even though human studies imply that OX40–OX40L crosstalk between T cells and endothelial or dendritic cells might contribute to disease.

As well as controlling the accumulation and/or activity of pathogenic effector T cells, OX40–OX40L interactions have been associated with production of pathogenic antibodies. Transgenic mice overexpressing OX40L display elevated levels of anti-DNA antibodies68. Furthermore, soluble OX40 and/or OX40L are increased in the plasma of patients with early-stage RA compared with healthy individuals, and correlate with levels of anti-citrullinated protein antibodies and IgM rheumatoid factor69. Similarly, an association between OX40L expression on myeloid APCs (dendritic cells and monocytes), SLE disease activity and anti-ribonucleoprotein (RNP) antibodies has been described61. As activated B cells express OX40L, this ligand could directly signal and contribute to autoantibody production. However, the primary rationale for the association with anti-RNP antibodies is that OX40L on dendritic cells can signal via OX40 expressed by T cells and might aid formation of follicular T helper cells that drive B cell differentiation61. An association between polymorphisms in the OX40L locus and Sjögren syndrome or SSc has also been confirmed in multiple studies70,71, with levels of soluble serum OX40 being elevated in patients with early-stage SSc72. Lastly, biopsy samples from patients with Wegener granulomatosis, another rheumatic disease that is associated with elevated levels of anti-neutrophil cytoplasmic antibodies (ANCA) and glomerulonephritis73, contain OX40-expressing T cells73. Although clinical grade drugs that neutralize OX40L (such as oxelumab and KY1005) or OX40 (such as KHK4083) exist, at present no trials have attempted to target the OX40–OX40L interaction in rheumatic diseases (TABLE 1). However, such interventions have strong therapeutic potential and might be beneficial, particularly in RA and SLE.

TL1A

Death receptor 3 (DR3, also known as TNFRSF25) is another stimulatory receptor expressed by T cells (FIG. 3) that can regulate effector cell accumulation and/or reactivity regardless of T helper phenotype7476. Its ligand, TNF-like ligand 1A (TL1A, also known as TNFSF15), can be induced in APCs such as dendritic cells and macrophages, as well as in endothelial cells5,6,11,77,78. TL1A–DR3 interactions might drive many inflammatory responses, especially mucosal inflammation77,79, and increasing evidence suggests a role in rheumatic disease.

Levels of TL1A are elevated in the synovial fluid and serum of patients with RA, and are associated with both autoantibody levels and atherosclerotic lesion development8086. Interestingly, human synovial fibroblasts are capable of expressing TL1A after stimulation with TNF or IL-1β, suggesting a potential local source of TL1A in addition to professional APCs82. In line with the notion that soluble TL1A could be pathogenic in RA, injection of recombinant TL1A into mice with already-developed collagen-induced or bovine serum albumin (BSA)-induced arthritis leads to an increase in the severity of disease, including increased cartilage damage, bone destruction and increased levels of autoantibodies82,87. In these arthritis models, DR3 and TL1A deficiency, or TL1A inhibition in wild-type mice, resulted in reduced swelling and bone erosions, and/or increased kinetics of disease resolution, demonstrating the therapeutic potential of targeting TL1A8789. The reason for reduced disease activity is not clear but could be due to a combination of lower T-cell activity and reduced infiltration of destructive cells such as neutrophils, which is possibly linked to defective chemokine expression. An alternative explanation is that TL1A could have a role in enhancing RANKL-triggered osteoclast differentiation in macrophage precursors that express DR3 (REF. 87). This process could cooperate with the immune-mediated inflammatory effects of TL1A and contribute to bone dysregulation. Although genome-wide association studies have not identified TL1A or DR3 as susceptibility loci for inflammatory arthritides, a duplication in the gene encoding DR3 (TNFRSF25) has been linked to RA90; in addition, an association study indicated that several SNPs downstream of the gene encoding TL1A (TNFSF15) were linked to the development of spondyloarthritis (SpA)91. SpA, a disease closely related to RA, can be characterized by gut inflammatory phenotypes and TH17 cells are thought to be involved in SpA pathogenesis; both of these features are known to be connected with TL1A activity7779. Lastly, DR3 and/or TL1A were found to be upregulated in lesional skin plaques and serum from patients with psoriasis, another disease with a TH17 component that can be directly associated with arthritis92,93. Although the implications of these observations regarding the pathogenesis of SpA and psoriasis are not clear, these data suggest that DR3 and TL1A are involved in bone and joint disorders and manifestations that arise from these inflammatory diseases.

Data directly implicating TL1A involvement in SLE pathogenesis are currently lacking, except for one report describing a weak correlation between elevated TL1A levels in serum and SLE disease activity94. However, during acute kidney allograft rejection, renal tubular epithelial cells express DR3 (REF. 95), and renal vascular endothelial cells express TL1A96. DR3 activity might be protective against nephrotoxicity in some settings96,97, but whether these molecules contribute to nephritis as seen in SLE is an open question. Overall, the data presented above indicate that inhibition of TL1A–DR3 activity might be beneficial for patients with arthritis, and possibly for those with other autoimmune conditions such as SLE.

GITRL

Glucocorticoid-induced TNF receptor-related ligand (GITRL, also known as TNFSF18) is an inducible molecule expressed in professional APCs, and other cell types such as endothelial cells. Its receptor, glucocorticoid-induced TNF receptor-related protein (GITR, also known as TNFRSF18), can stimulate T cell, dendritic cell and B cell activation (FIG. 3). Studies have implicated these molecules in controlling many immune-inflammatory responses, although functional data relating them to rheumatic disease are largely restricted to arthritis at present6,11,98,99. GITR-deficient mice display reduced joint inflammation in collagen-induced arthritis compared with wild-type mice, including decreased T-cell reactivity and lower levels of inflammatory mediators such as TNF100. Serum from patients with RA have increased levels of GITRL compared with healthy controls, a finding associated with increased IL-17 levels101. Furthermore, GITR and GITRL have been detected in synovial tissue sections from patients with RA (primarily in T cells and macrophages); synovial fluid from these patients has also been found to contain both GITR and GITRL as soluble molecules102,103. In line with the idea that soluble GITRL is pathogenic, injection of recombinant GITRL into mice with collagen-induced arthritis increases disease kinetics and clinical symptoms101, as does treatment with an agonistic GITR antibody104; this treatment also increases production of the T-cell-derived inflammatory cytokines such as IL-17, TNF and IFNγ101,104. Furthermore, stimulation of GITR on synovial fluid macrophages leads to upregulation of several inflammatory proteins including TNF, IL-6 and MMP-9 (REF. 102). Lastly, soluble GITRL and/or GITR might represent useful biomarkers for other rheumatic diseases, as in patients with SLE or Sjögren syndrome the levels of these molecules are increased105,106. Given that their expression correlate with disease severity105,106, GITR–GITRL activity might also contribute to the pathogenesis of these diseases

CD70

CD27 (also known as TNFRSF7) is constitutively expressed on most T cells, and the interaction with its ligand CD70 (also known as TNFSF7) can provide signals to T cells to control their accumulation and reactivity, similarly to that seen with OX40, GITR and DR3 (REFS 35) (FIG. 3). In addition to T cells, CD70 is inducible on dendritic cells and B cells, and can induce reverse signals within these APCs to increase their activation status27, therefore participating in the crosstalk between T cells and B cells and antibody production. Genetic deletion or neutralization of either CD27 or CD70 in mice has revealed a pathogenic role for the CD27–CD70 axis in many inflammatory settings46,107. For example, in mice with collagen-induced arthritis, blocking CD27–CD70 interactions with anti-CD70 antibody reduces bone and cartilage erosion and inflammatory infiltrates in the joints, and decreases collagen-specific antibody production, even when the treatment is initiated after disease onset108. In the synovial fluid of patients with RA, soluble CD27 levels and CD27+ T cell numbers are elevated and correlates with the levels of rheumatoid factor, supporting a role for CD27 in human RA109. Furthermore, in patients with RA, CD70 expression is increased in CD4+ T cells that produce the effector cytokines IFNγ and IL-17 (REFS 110,111). Although the implication of this upregulation is not clear, these CD4+ T cells are probably highly pathogenic, given that ligation of CD27 on B cells by CD70 can promote B cell differentiation. Synovial fluid samples from patients with juvenile idiopathic arthritis are also characterized by increased expression of soluble CD27 (REF. 112).

A correlation between CD27 or CD70 expression and disease activity is also observed in other rheumatic diseases, although functional data are in general lacking at present. Soluble CD27 levels correlate with disease activity in patients with SLE113,114, and the proportion of plasma cells expressing high levels of CD27 additionally correlates with SLE disease indices115. Furthermore, several studies showed that T cells derived from patients with SLE express high levels of CD70 and are capable of driving B cell antibody production via CD27 (REF. 116). Similarly, T cells from MRL/lpr mice with lupus-like disease overexpress CD70 (REF. 117), although no studies to date have shown if CD70 expression is required for disease onset in these mice. Interestingly, plasmacytoid dendritic cells (pDCs), which are thought to be central to SLE pathogenesis via their type I interferon production, can strongly express CD70 (REF. 118). These pDCs can drive antibody secretion by B cells via CD27 without the participation of T cells, implicating pDCs as another important source of CD70. CD4+ T cells from patients with SSc and Sjögren syndrome have also been found to express high levels of CD70 (REFS 119,120). Thus, neutralizing the interaction between CD27 and CD70 could potentially dampen disease activity in RA and/or other diseases such as SLE. A clinical-grade antibody to CD70 (SGN-75) has been developed and conjugated to a toxin for targeting CD70+ B cell cancers11. This reagent could be used, with or without toxin, for treatment of rheumatic disease, although no trials have so far been initiated.

4-1BBL

4-1BB (also known as TNFRSF9) is an inducible stimulatory receptor expressed on T cells and innate lymphoid cells that can promote their accumulation and/or activity; expression of its ligand, 4-1BBL (also known as TNFSF9), is also inducible on professional APCs5,6,121. 4-1BB is similar to the molecules described above in terms of intrinsic activity (FIG. 3); As with OX40, GITR and CD27, 4-1BB is currently being targeted with receptor agonists to promote antitumour T-cell responses in the context of clinical cancer immunotherapy 122. However, only a few studies have shown 4-1BB and 4-1BBL involvement in inflammatory disease pathogenesis5,6,121. As such, little data has been generated with regard to rheumatic disease. Serum samples of patients with RA contain elevated levels of soluble 4-1BB and 4-1BBL, which correlate with disease severity123,124. Nevertheless, in collagen-induced arthritis in mice, a reagent that blocks the interaction between these two molecules had only a moderate effect in suppressing disease symptoms such as T-cell reactivity and inflammatory cytokines125. Although this finding does not exclude a role for 4-1BBL-4-1BB interactions in promoting RA in humans, it is in contrast to the much more robust data obtained when other TNF family molecules (such as OX40L, CD70, GITRL and TL1A) were targeted in the same arthritis model. On the other hand, stimulation of 4-1BB with receptor agonists results in strong suppression of joint inflammation and bone destruction in mouse models of RA125,126. This finding is not consistent with the idea that endogenous 4-1BB-4-1BBL interactions promote development or activity of pathogenic T cells in RA. A similar conclusion might be true for SLE; indeed, 4-1BB-deficiency in lupus-prone MRL/lpr mice exacerbates rather than ameliorates disease127, in line with a regulatory rather than pathogenic role. Similar to mouse models of arthritis, 4-1BB agonists also fully inhibit lupus-like disease in MRL/lpr and NZB/NZW F1 mice, including reduction of skin lesions, lymphadenopathy, autoantibody production and nephritis128130. These results suggest that the neutralization of 4-1BB or 4-1BBL might have little effect in rheumatic disease, whereas stimulation of 4-1BB could dampen inflammation.

Increasing tissue inflammation

Lymphotoxin and LIGHT

Lymphotoxin and LIGHT (also known as CD258 and TNFSF14, respectively) are TNFSF cytokines with interrelated functions that are similar to those of TNF. They can control T cell and APC responsiveness, and importantly, have marked effects on both development and homeostasis of lymphoid tissue and structural cell responses of non-haematopoietic tissue 131134 (FIG. 4). Soluble lymphotoxin (also known as LTα or TNFSF1) is a homotrimer that binds TNF receptors (TNFR1 and TNFR2), but might often be redundant with TNF. In RA, anti-TNF antibodies have been found to be as clinically effective as etanercept, a TNFR2-Fc fusion protein that blocks both LTα and TNF135, and in a clinical trial of RA, pateclizumab, a specific blocker of LTα, showed much reduced efficacy compared with the TNF blocker adalimumab136. These findings do not rule out an important role for LTα in some inflammatory diseases, but suggest that its role is secondary to that of TNF when TNF is present in abundance. By contrast, the other version of lymphotoxin, LTαβ might exert distinct and unique functions compared with TNF and LTα. LTαβ is membrane-bound heterotrimer composed of LTα and a distinct β subunit, and exclusively binds to the LTβ receptor (LTβR, also known as TNFRSF3)137. LTαβ is constitutively expressed on resting B cells and can also be induced in activated T cells. LTβR is expressed on some haematopoietic cells, such as dendritic cells and macrophages, but importantly, is expressed on tissue stromal cells such as fibroblasts, adipocytes, hepatocytes, endothelial cells, fibroblastic reticular cells, smooth muscle cells and epithelial cells137. Studies of gene-knockout mice have shown a non-redundant role for LTαβ–LTβR interactions in controlling the development of lymph nodes and Peyer patch structures, which is due to the absence of LTβ-dependent RANKL production137. RANKL acts on stromal cells to induce chemokine expression, which is critical for recruitment and proper positioning of lymphocytes within these structures137. In mature lymphoid tissue, LTαβ signals through LTβR in follicular dendritic cells, controlling the expression of adhesion molecules (vascular cell adhesion protein 1 (VCAM1) and mucosal addressin cell adhesion molecule 1 (MADCAM1)), as well as chemokines, which maintain B cell organization in follicles137. These mechanisms have also been implicated in controlling the arrangement of immune cells in tertiary lymphoid structures, which occur in tissues undergoing chronic inflammatory responses137.

Figure 4. TNFSF inflammatory activities in tissue cells.

Figure 4

The simplified diagram shows the possible interactions between TNF superfamily (TNFSF) ligands and their receptors expressed on tissue cells (epithelium, endothelium, fibroblasts and smooth muscle cells) that can affect tissue homeostasis and inflammatory activity. The TNFSF molecules lymphotoxin (LT) αβ, LIGHT and TNF-related weak inducer of apoptosis (TWEAK), together with TNF, are likely to be produced primarily by cells of the immune system, including T cells, B cells, dendritic cells, macrophages, as well as neutrophils, mast cells and innate lymphoid cells. Amplification loops from tissue structural cells, including endothelial and epithelial cells, might further induce production of these molecules. Signals from TNFR1, lymphotoxin-β receptor (LTβR), herpes virus entry mediator (HVEM) and fibroblast growth factor-inducible protein 14 (Fn14) can directly promote tissue pathology through multiple processes, including differentiation events such as epithelial mesenchymal transition and myofibroblast transformation, hyperplasia and hypertrophy of epithelial cells, fibroblasts, and smooth muscle cells, expression of extracellular matrix proteins and proteinases that contribute to tissue remodelling, production of chemokines and adhesion molecules that attract and maintain inflammatory immune cells within the inflamed tissue. CD40 and death receptor 3 (DR3) are also expressed on some tissue cells such as fibroblasts and could further amplify their inflammatory activity (not shown). Furthermore, receptor activator of nuclear factor-κB ligand (RANKL) and TWEAK are regulators of osteoclast activation and differentiation (also not shown). TNFSF might additionally synergize with proinflammatory T-cell-derived cytokines such as IFNγ, IL-17 and IL-22, which also have receptors on tissue structural cells.

LIGHT binds to LTβR and also to a receptor termed herpes virus entry mediator (HVEM, also known as TNFRSF14). LIGHT can be expressed by activated T cells and other lymphoid cells, and HVEM is expressed on many haematopoietic cells in addition to the same structural cells that express LTβR (such as fibroblasts, epithelial cells and smooth muscle cells)132134. Whereas LIGHT does not participate in controlling lymphoid organogenesis, growing evidence suggests that its activity in tissue cells, via both LTβR and HVEM, might be a strong component of the remodelling processes characteristic of many chronic inflammatory and autoimmune diseases, including epithelial–mesenchymal transition and myofibroblast differentiation134 (FIG. 4). The physiological role of LTβR and HVEM might be to protect the epithelium and other tissues against injury or infection138,139. However, their reported activities in epithelial cells, fibroblasts, osteoclasts, adipocytes and hepatocytes suggest that if LIGHT or LTαβ are produced in excess these receptors directly or indirectly induce the production of inflammatory cytokines, chemokines, extracellular matrix proteins and proteinases. These effects are similar to that seen with TNF–TNFR1 activity, implying that these molecules cooperate in orchestrating tissue inflammation134,140144.

An Fc fusion protein of LTβR, which can neutralize both LTαβ and LIGHT, can block disease symptoms in many mouse models of rheumatic disease, including collagen-induced and adjuvant arthritis, several models of SLE and the Sjögren-syndrome-like salivary gland inflammation of non-obese diabetic mice131133,145148. Additionally, genetic deletion of LIGHT protects mice from lung and skin inflammation and tissue remodelling in models of SSc144,149. Despite these results, targeting the LTαβ–LIGHT axis with baminercept, a soluble LTβR-Fc fusion protein, did not demonstrate clinical efficacy in RA and Sjögren’s syndrome (TABLE 1), although some modulation of immune reactivity was noted150. A caveat of these trials was the recruitment of difficult-to-treat patient populations that had previously shown inadequate responses to TNF inhibitors or other DMARDs. More specific reagents targeting LIGHT, LTβ or their receptors still have potential for the treatment of rheumatic diseases that involve tissue remodelling and inflammation, although they are more likely to be efficacious in patients who are also responsive to TNF-directed therapy. A fully human LIGHT blocking antibody has been generated and has successfully completed phase I safety trials (TABLE 1); this antibody is currently entering phase II studies of paediatric inflammatory bowel disease but hasn’t yet entered any trials for rheumatic disease.

TWEAK

TNF-related weak inducer of apoptosis (TWEAK, also known as TNFSF15) has high degree of homology with TNF and is thought to primarily act on tissue cells151,152. TWEAK is produced by a large range of myeloid and immune cells, but its receptor, fibroblast growth factor-inducible 14 (Fn14, also known as TNFRSF12A), is more highly expressed on non-haematopoietic cells than on lymphoid cells. Fn14 is upregulated by fibroblast-like growth factor153,154, as well as by other factors associated with injury and inflammation151. TWEAK has pleiotropic effects in stromal cell types, including regenerative-like activities in hepatocytes, endothelial cells, myocytes and epithelial cells152 (FIG. 4). Arguably, the physiological role of the TWEAK–Fn14 axis is to protect against tissue injury, but like the LIGHT–LTαβ axis, if TWEAK or Fn14 are excessively produced they could drive and orchestrate inflammation, fibrosis and tissue remodelling.

TWEAK and Fn14 are elevated in the synovium and serum of patients with RA and/or psoriatic arthritis, with levels correlating with disease severity in some instances, although their levels in joints are not affected by TNF inhibitor treatment155158. In normal fibroblasts or fibroblast-like synoviocytes, TWEAK can induce proliferation and upregulate the production of inflammatory cytokines such as IL-6, chemokines, adhesion molecules and proteinases159162. As such, blocking TWEAK reduces disease severity in collagen-induced arthritis in mice without affecting titres of anti-collagen antibodies163,164, suggesting that TWEAK largely contributes to inflammation and bone destruction locally in the joint. Osteoclasts express Fn14, and consequently TWEAK can promote osteoclastogenesis, which is relevant to RA pathogenesis165. These data suggest that neutralizing TWEAK has the potential to dampen disease activity in RA. Phase I trials of a blocking antibody against TWEAK (BIIB023) have been conducted in patients with RA166, but further trials in RA have not yet been pursued (TABLE 1).

TWEAK has also been implicated in kidney disease. Fn14 deficiency or TWEAK blockade reduces a variety of renal pathologies in several mouse models of disease, including fibrosis after ureteral obstruction162, folate-induced interstitial nephritis167, nephrotoxic serum-induced immune complex glomerulonephritis168 and nephritis associated with chronic graft-versus-host disease169. Additionally, in Fn14-deficient mice, renal, neuropsychiatric and dermatological manifestations were considerably reduced in the MRL/lpr model of spontaneous lupus-like autoimmunity170172. As with collagen-induced arthritis, titres of systemic auto-antibodies were not affected in these studies, further suggesting that Fn14 mediates local effects in target tissues. Which cell types receive Fn14 signals in the context of lupus nephritis or other kidney disease is an unresolved question. However, TWEAK can stimulate inflammatory mediator production (cytokines and/or chemokines) in vitro by a variety of different kidney cell types, including renal tubular epithelial cells, podocytes and mesangial cells167,173,174. In human SLE, TWEAK can serve as a urinary biomarker for nephritis175. Despite these promising results, a trial investigating the efficacy of anti-TWEAK antibodies in SLE was terminated following failure to increase rates of renal remission in patients with nephritis already being treated with mycophenolate176 (TABLE 1).

Cell death and immunosuppression

FasL and TRAIL

FasL and TRAIL have a potent ability to induce apoptosis. FasL can promote apoptosis in activated primary B cells, T cells and dendritic cells through Fas (also known as TNFRSF6)174,175, and TRAIL has been shown to induce apoptosis in activated mouse CD8+ T cells via TRAIL receptor 1 (TRAILR1, also known as TNFRSF10A) or TRAILR2 (also known as TNFRSF10B)177179. Defective activity of the FasL–Fas or TRAIL–TRAILR axis might increase the susceptibility to autoimmune disease. Genetic defects in FasL, or more commonly in Fas, result in spontaneous autoimmunity in mice and in auto-immune lymphoproliferative syndrome in humans180184. TRAIL-deficient mice are hypersensitive to diseases such as collagen-induced arthritis185. Less is known about the role of TRAIL and its receptors in human cells. Although activated human T cells express TRAILR1 and TRAIL2, unlike FasL, TRAIL does not generally induce apoptosis in these cells186. Dendritic cells might be more relevant targets for TRAIL in the human immune system as a deficiency in caspase 10, which is activated by TRAIL, underlies a variant of autoimmune lymphoproliferative syndrome, which is marked by accumulation of these cells187. For these reasons, the function of FasL and TRAIL is mainly to restrain persistent immune responses to curb autoimmunity. Fas and TRAILRs can also be expressed outside the immune system; crosslinking of these molecules on cells such as synovial fibroblasts or dermal fibroblasts, which are associated with RA and SSc, respectively, might induce apoptosis188,189. However, either an elevated activation state or increased proliferative activity of such cells might make them more resistant to the effects of the naturally produced death-inducing ligands189,190, which could be another contributing factor to diseases such as RA.

The apoptotic potential of FasL and TRAIL, either to dampen activity of autoreactive T cells or to kill highly proliferative tissue cells, has led to the hypothesis that recombinant FasL or TRAIL, or biologic agents acting as receptor agonists, could be candidate therapeutics for rheumatic diseases191. Results from experimental studies on the injection of various forms of FasL or TRAIL into rodents have reinforced this idea192196. However, several factors might hinder this therapeutic strategy. Fas engagement has the potential to cause off-target effects, as exemplified by induction of hepatocyte cell death and acute liver failure in mice injected with Fas agonists197. Although all activated T cells express Fas, stimulation with this molecule fails to induce efficient apoptosis of memory T cells or T cells in the early stages of activation, which are the likely T cells that would be active in rheumatic diseases178,198. Rather, Fas might stimulate T-cell activation in some scenarios199201. Some data suggest that fibroblast-like synoviocytes can be induced to proliferate when treated with soluble FasL or with low doses of agonist Fas antibody, whereas only oligomeric FasL or high doses of anti-Fas agonists induce apoptosis202,203. These factors further complicate the development of biologic agents to stimulate Fas or TRAILRs that might be therapeutically useful in rheumatic diseases.

Challenges and limitations

Is targeting one TNFSF member enough?

Several potential challenges exist when looking at modulating the activity of TNF family members other than TNF in rheumatic diseases. Blockade of TNF is highly efficacious in treating patients with a wide range of inflammatory arthritides including RA, psoriatic arthritis, ankylosing spondylitis and juvenile idiopathic arthritis, and also in other inflammatory diseases such as plaque psoriasis, Crohn’s disease and ulcerative colitis11,204. However, whether neutralizing another TNFSF protein in isolation will produce the same strong and broad benefit is not clear. Blocking TNF might have a potent therapeutic effect for two main reasons. Firstly, TNF is a primary end-stage inflammatory mediator in tissues, as it is produced at high levels by multiple cell types (both immune and non-immune) and induced by many different stimuli. Secondly, TNF has two receptors that are both expressed on immune cells as well as stromal non-haematopoietic cells, broadening its activity from tissues to the immune system. In comparison, the majority of other TNFSF molecules are produced at lower levels, triggered by fewer stimuli, act on a smaller number of cell types and primarily control immune cells and not tissue cells. Hence, a number of TNFSF proteins, particularly the immune modulators, might have a narrower range of action compared with TNF, limiting the therapeutic effects of biologic agents that target them.

A possible example in this regard is BAFF, a molecule that primarily, although not exclusively, controls B cell activity. Preclinical data, particularly in mouse models, suggest that BAFF and B cells are central to lupus-like autoimmunity10,17. However, belimumab, a BAFF inhibitor, although approved for SLE treatment and having considerable effects on human B cells, has been found to be only moderately efficacious in a small number of patients with SLE205,206. This unexpected outcome could reflect differences between human SLE and the disease that manifests in animal models. As suggested above, the fact that BAFF primarily controls only one immune cell type and does not play an important role within the affected tissues of patients with SLE might also explain this outcome. Belimumab has only a moderate effect in patients with RA, although slightly more promising results have been observed in Sjögren’s syndrome207,208. Again, suppression of the B cell arm of the immune response might not be sufficient for a notable disease modification, given the activity of other immune cell types in these diseases and the strong tissue component, which is dependent on crosstalk between multiple immune cells and tissue structural cells. Thus, when considering other molecules such as CD40L, OX40L, GITRL, TL1A and CD70, which arguably exert the majority of their activity on T cells, B cells, dendritic cells and macrophages, and are possibly not functional within the affected tissues during the active phase of disease, we have to consider whether neutralizing only one of their interactions will produce a pronounced therapeutic effect.

Another obstacle for successfully targeting TNFSF proteins, particularly those that primarily control immune cell activation, is that an alteration in activity of cells such as T cells and B cells might take a long time to manifest in terms of disease symptoms. As current trials are typically short term and largely designed to compare to an already approved drug (such as a TNF inhibitor) whose target or mechanism of action could be different, future success in this area might require careful trial designs and end points based around modulation of the perceived primary target cell or cells.

Towards immunological tolerance

Despite the caveats of targeting some TNFSF members discussed above, inhibiting molecules such as CD40L, OX40L, GITRL, CD70 and TL1A, which control the accumulation and activity of pathogenic T cells and B cells, might be a good strategy to re-establish immunological tolerance. Such targeting could prevent the formation of these disease-causing T cell and B cell populations, lead to their deletion and/or reset immune homeostasis in favour of regulatory T cells and B cells; such regulatory cells are now acknowledged to be critical for limiting autoimmunity.

Abatacept (a CTLA4–Ig fusion protein) is a drug already approved for RA therapy and used either as first-line treatment or in patients not responding to conventional therapy. This reagent is primarily thought to act by disrupting CD28 stimulatory signals in T cells. As CD28 can cooperate with TNFRSF proteins in driving T-cell activation4,209, blocking one or more of these TNF family members might have therapeutic effects similar to those of abatacept in RA and possibly other rheumatic diseases. However, given the apparent overlap in the activities of several TNFRSF molecules on T cells and B cells, and the idea that TNFRSF and CD28 cooperate in driving T cell and B cell responses4,5,210, we still have to consider that combination therapies that neutralize two or more interactions might be required to see marked and broad-reaching activity in many patients, regardless of the disease. Furthermore, as discussed above, therapeutic effects might take time to manifest in terms of disease control. Mouse transplantation models using fully MHC-mismatched allografts have shown that neutralization of CD40L with OX40L, or CD40L with CD70, with or without concomitant inhibition of CD28, can help to establish immune tolerance in situations where targeting the individual interactions is ineffective5. However, the best combined therapy for any given rheumatic disease is not obvious at present. Information regarding the timing of action of TNF family molecules during disease development will also be critical to any therapeutic success. Immune monitoring of levels of TNFSF ligands and receptors in fluids or tissues of patients with rheumatic disease will probably help, although this approach still assumes that their presence signifies their activity. Immune monitoring might also lead to an improved understanding of molecules that can be targeted simultaneously. Furthermore, translational studies in animal models that more realistically mimic the active phases of human rheumatic disease should aid the formulation of effective combination therapies.

Blocking tissue inflammation

Although the TNFSF members that primarily control T cells and APCs (FIG. 3) are probably good targets for restoring tolerance in rheumatic diseases, the molecules that regulate tissue cell responses (FIG. 4) similarly to TNF might be more attractive targets for therapy. For example, several structural cell types express LTβR, HVEM and Fn14. A few reports have shown that molecules such as CD40 and DR3 are expressed and active in mouse and human fibroblasts in disease settings as diverse as RA, SSc and inflammatory bowel disease211213. What is not clear is how much synergy or overlap occurs between these receptors on structural cells in terms of function, and again whether blocking a single molecule in humans is likely to have a profound effect on any given disease phenotype. The failure of TWEAK–Fn14 blockade to achieve its end point in lupus nephritis might reflect the challenges inherent in nephritis trials214, and trials in other diseases will be necessary to assess its full potential. However, TWEAK blockade could be an example of where combining treatment with a biologic agent targeting another protein is necessary, as TWEAK has a functional activity similar to that of other TNFSF molecules such as TNF and LIGHT.

Related to this discussion is the observation that anti-TNF treatment is ineffective in about one-third of patients with RA204. The reasons for this lack of response are not clear, but an open question is whether some patients do not respond to anti-TNF monotherapy because several other TNFSF molecules, such as TWEAK and LIGHT, are also active. Would these anti-TNF nonresponders (in any rheumatic disease) be the preferred population to treat with biologic agents targeting other tissue-acting TNFSF members? To test this theory, a clinical trial investigated the use of baminercept, which inhibits LIGHT and LTαβ, in patients with RA who were unresponsive to TNF blockers. Although some effect on biological activity was noted150, this monotherapy was abandoned as it did not achieve the therapeutic end point. However, in this case redundancy or cooperative action between multiple TNFSF members, including LIGHT, TNF and TWEAK, could explain this lack of activity. Combination therapy might then be more efficacious than targeting molecules separately. This might apply to patients that do respond to TNF inhibitors as well as those who do not respond to anti-TNF therapy alone.

Conclusions

At present, our knowledge of the TNF family members is quite advanced and, at least in some cases, has translated well into the clinic. However, there have been notable failures despite preclinical data suggesting important roles for many of these molecules in rheumatic or other inflammatory diseases. As discussed above, the potential overlap in expression and activities of TNFRSF might hinder therapeutic approaches that only neutralize a single interaction. However, these setbacks should not discourage the enthusiasm for attempting to modulate these molecules alone or in combination. Historically, combination treatment of TNF inhibitors with other biological drugs (such as abatacept and anakinra) has not improved efficacy in the treatment of rheumatic disease and only increased adverse events such as infections215,216. These findings might be specific to TNF, as the evolutionary role of this cytokine is arguably to limit replication of infectious pathogens. Therefore, neutralizing two TNFSF members other than TNF might not result in a similar increase in such deleterious effects. Regardless, any combination will probably require extensive safety data before being introduced in the clinic. Advances in technology might enable two or more proteins to be targeted with a single biologic agent (such as with a bi-specific antibody), potentially making the path to inhibiting multiple interactions more feasible.

An alternative therapeutic strategy is to stimulate the death receptors Fas and TRAILR1/2, and attempt to induce apoptosis of immune or structural cells that contribute to disease pathology. The difficulty in this approach is being able to effectively induce death in the relevant cell types without having off-target effects; given the broad expression of death receptors, more direct approaches (such as bi-specific molecules) that focus the activity of an agonist reagent on individual cell types are probably needed. Another strategy might be to activate stimulatory receptors. In SLE (and multiple sclerosis), TNF inhibitors have not performed well217,218, and in some cases promote lupus-like disease219. Although the reason for this outcome is unclear, studies suggest that inhibition of TNF binding to TNFR2 can impair the expansion of suppressive CD4+ Foxp3+ Treg cells, which maintain immune tolerance in some settings220222. In this regard, similar functional observations have also been drawn for OX40, 4-1BB, CD27, DR3 and GITR. In particular, studies in mouse models of RA and SLE, as well as asthma, graft-versus-host disease and multiple sclerosis, have revealed that 4-1BB agonists are strongly suppressive, as they selectively expand both CD8+ Treg cells that can inhibit effector CD4 T cells and/or CD4+ Foxp3+ Treg cells5,125,223. Similarly, stimulation of DR3, GITR or OX40 in some settings can expand Treg cells, and in several mouse models results in suppression of asthma symptoms, allograft rejection, diabetes and multiple sclerosis-like disease224230. However, owing to the possibility of expanding pathogenic self-reactive T cells, agonist targeting might not be a first-line strategy; neutralization of these molecules is instead the logical choice for therapy. If clinical trials reveal contraindications for certain inhibitory reagents, drugs that stimulate TNFSF receptors might represent an alternative treatment option. Agonist antibodies to 4-1BB, OX40, CD27 and GITR are currently in clinical trials for the treatment of cancer to expand tumour-reactive T cells11, and apart from some hepatotoxicity observed with anti-4-1BB at high doses, they have shown a relatively good safety profile, and could be tested in patients with rheumatic disease.

Key points.

  • TNF inhibitors are among the most effective protein-based drugs for reducing inflammation associated with several rheumatic diseases

  • In addition to TNF, the TNF superfamily (TNFSF) comprises other ligand–receptor combinations that might participate in the pathogenesis of rheumatic disease

  • TNFSF members initiate several processes, including immune activation, tissue inflammatory responses and cell death or suppression

  • Many TNFSF proteins other than TNF are being evaluated in preclinical mouse or human studies as possible therapeutic targets in rheumatic diseases

  • TNFSF members can be targeted to either restore tolerance in rheumatic diseases or to regulate tissue cell responses

Acknowledgments

M.C. is supported by NIH grants AI070535, AI103021, AI110929 and AI123134. R.S. is supported by the NIAMS intramural research program.

Footnotes

Author contributions

Both authors researched data for the article and made a substantial contribution to discussion of content, writing, reviewing and editing of the manuscript before submission.

Competing interests statement

M.C. has licensed patents on several TNF superfamily molecules. R.S. has issued patents on antibodies against the TNF superfamily molecule TL1A.

References

  • 1.Beutler B, et al. Identity of tumour necrosis factor and the macrophage-secreted factor cachectin. Nature. 1985;316:552–554. doi: 10.1038/316552a0. [DOI] [PubMed] [Google Scholar]
  • 2.Feldman M, Taylor P, Paleolog E, Brennan FM, Maini RN. Anti-TNF alpha therapy is useful in rheumatoid arthritis and Crohn’s disease: analysis of the mechanism of action predicts utility in other diseases. Transplant Proc. 1998;30:4126–4127. doi: 10.1016/s0041-1345(98)01365-7. [DOI] [PubMed] [Google Scholar]
  • 3.Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell. 2001;104:487–501. doi: 10.1016/s0092-8674(01)00237-9. [DOI] [PubMed] [Google Scholar]
  • 4.Croft M. Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat Rev Immunol. 2003;3:609–620. doi: 10.1038/nri1148. [DOI] [PubMed] [Google Scholar]
  • 5.Croft M. The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol. 2009;9:271–285. doi: 10.1038/nri2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Croft M, et al. TNF superfamily in inflammatory disease: translating basic insights. Trends Immunol. 2012;33:144–152. doi: 10.1016/j.it.2011.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol. 2016;12:49–62. doi: 10.1038/nrrheum.2015.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cessak G, et al. TNF inhibitors — mechanisms of action, approved and off-label indications. Pharmacol Rep. 2014;66:836–844. doi: 10.1016/j.pharep.2014.05.004. [DOI] [PubMed] [Google Scholar]
  • 9.Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond. Front Immunol. 2014;5:511. doi: 10.3389/fimmu.2014.00511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stohl W. Therapeutic targeting of the BAFF/APRIL axis in systemic lupus erythematosus. Expert Opin Ther Targets. 2014;18:473–489. doi: 10.1517/14728222.2014.888415. [DOI] [PubMed] [Google Scholar]
  • 11.Croft M, Benedict CA, Ware CF. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov. 2013;12:147–168. doi: 10.1038/nrd3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Caso F, Costa L, Del Puente A, Scarpa R. Psoriatic arthritis and TNF inhibitors: advances on effectiveness and toxicity. Expert Opin Biol Ther. 2015;15:1–2. doi: 10.1517/14712598.2015.973398. [DOI] [PubMed] [Google Scholar]
  • 13.Chen X, Oppenheim JJ. Therapy: paradoxical effects of targeting TNF signalling in the treatment of autoimmunity. Nat Rev Rheumatol. 2016;12:625–626. doi: 10.1038/nrrheum.2016.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zaheer S, LeBoff M, Lewiecki EM. Denosumab for the treatment of osteoporosis. Expert Opin Drug Metab Toxicol. 2015;11:461–470. doi: 10.1517/17425255.2015.1000860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nagy V, Penninger JM. The RANKL-RANK story. Gerontology. 2015;61:534–542. doi: 10.1159/000371845. [DOI] [PubMed] [Google Scholar]
  • 16.Wei F, Chang Y, Wei W. The role of BAFF in the progression of rheumatoid arthritis. Cytokine. 2015;76:537–544. doi: 10.1016/j.cyto.2015.07.014. [DOI] [PubMed] [Google Scholar]
  • 17.Vincent FB, Morand EF, Schneider P, Mackay F. The BAFF/APRIL system in SLE pathogenesis. Nat Rev Rheumatol. 2014;10:365–373. doi: 10.1038/nrrheum.2014.33. [DOI] [PubMed] [Google Scholar]
  • 18.Schnitzer TJ, Marks JA. A systematic review of the efficacy and general safety of antibodies to NGF in the treatment of OA of the hip or knee. Osteoarthritis Cartilage. 2015;23(Suppl 1):S8–S17. doi: 10.1016/j.joca.2014.10.003. [DOI] [PubMed] [Google Scholar]
  • 19.Sanga P, et al. Long-term safety and efficacy of fulranumab in patients with moderate-to-severe osteoarthritis pain: a randomized, double-blind, placebo-controlled study. Arthritis Rheumatol. 2016 doi: 10.1002/art.39943. http://dx.doi.org/10.1002/art.39943. [DOI] [PubMed]
  • 20.Kan SL, et al. Tanezumab for patients with osteoarthritis of the knee: a meta-analysis. PLoS ONE. 2016;11:e0157105. doi: 10.1371/journal.pone.0157105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rosenthal A, Lin JC. Modulation of neurotrophin signaling by monoclonal antibodies. Handb Exp Pharmacol. 2014;220:497–512. doi: 10.1007/978-3-642-45106-5_19. [DOI] [PubMed] [Google Scholar]
  • 22.Richard AC, et al. Targeted genomic analysis reveals widespread autoimmune disease association with variants that regulate gene expression in the TNF superfamily cytokine signalling network. Genome Med. 2016;8:76. doi: 10.1186/s13073-016-0329-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bodmer JL, Schneider P, Tschopp J. The molecular architecture of the TNF superfamily. Trends Biochem Sci. 2002;27:19–26. doi: 10.1016/s0968-0004(01)01995-8. [DOI] [PubMed] [Google Scholar]
  • 24.Silke J, Brink R. Regulation of TNFRSF and innate immune signalling complexes by TRAFs and cIAPs. Cell Death Differ. 2010;17:35–45. doi: 10.1038/cdd.2009.114. [DOI] [PubMed] [Google Scholar]
  • 25.Karin M, Gallagher E. TNFR signaling: ubiquitin-conjugated TRAFfic signals control stop-and-go for MAPK signaling complexes. Immunol Rev. 2009;228:225–240. doi: 10.1111/j.1600-065X.2008.00755.x. [DOI] [PubMed] [Google Scholar]
  • 26.Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114:181–190. doi: 10.1016/s0092-8674(03)00521-x. [DOI] [PubMed] [Google Scholar]
  • 27.Eissner G. Ligands working as receptors: reverse signaling by members of the TNF superfamily enhance the plasticity of the immune system. Cytokine Growth Factor Rev. 2004;15:353–366. doi: 10.1016/j.cytogfr.2004.03.011. [DOI] [PubMed] [Google Scholar]
  • 28.Peters AL, Stunz LL, Bishop GA. CD40 and autoimmunity: the dark side of a great activator. Semin Immunol. 2009;21:293–300. doi: 10.1016/j.smim.2009.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Law CL, Grewal IS. Therapeutic interventions targeting CD40L (CD154) and CD40: the opportunities and challenges. Adv Exp Med Biol. 2009;647:8–36. doi: 10.1007/978-0-387-89520-8_2. [DOI] [PubMed] [Google Scholar]
  • 30.Allen RC, et al. CD40 ligand gene defects responsible for X-linked hyper-IgM syndrome. Science. 1993;259:990–993. doi: 10.1126/science.7679801. [DOI] [PubMed] [Google Scholar]
  • 31.Splawski JB, Lipsky PE. CD40-mediated regulation of human B-cell responses. Res Immunol. 1994;145:226–234. doi: 10.1016/s0923-2494(94)80189-4. [DOI] [PubMed] [Google Scholar]
  • 32.Grewal IS, Flavell RA. The role of CD40 ligand in costimulation and T-cell activation. Immunol Rev. 1996;153:85–106. doi: 10.1111/j.1600-065x.1996.tb00921.x. [DOI] [PubMed] [Google Scholar]
  • 33.Blotta MH, Marshall JD, DeKruyff RH, Umetsu DT. Cross-linking of the CD40 ligand on human CD4+ T lymphocytes generates a costimulatory signal that up-regulates IL-4 synthesis. J Immunol. 1996;156:3133–3140. [PubMed] [Google Scholar]
  • 34.Durie FH, et al. Prevention of collagen-induced arthritis with an antibody to gp39, the ligand for CD40. Science. 1993;261:1328–1330. doi: 10.1126/science.7689748. [DOI] [PubMed] [Google Scholar]
  • 35.Mohan C, Shi Y, Laman JD, Datta SK. Interaction between CD40 and its ligand gp39 in the development of murine lupus nephritis. J Immunol. 1995;154:1470–1480. [PubMed] [Google Scholar]
  • 36.Early GS, Zhao W, Burns CM. Anti-CD40 ligand antibody treatment prevents the development of lupus-like nephritis in a subset of New Zealand black x New Zealand white mice. Response correlates with the absence of an anti-antibody response. J Immunol. 1996;157:3159–3164. [PubMed] [Google Scholar]
  • 37.Citores MJ, et al. The dinucleotide repeat polymorphism in the 3′UTR of the CD154 gene has a functional role on protein expression and is associated with systemic lupus erythematosus. Ann Rheum Dis. 2004;63:310–317. doi: 10.1136/ard.2003.006148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Raychaudhuri S, et al. Common variants at CD40 and other loci confer risk of rheumatoid arthritis. Nat Genet. 2008;40:1216–1223. doi: 10.1038/ng.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.van der Linden MP, et al. Association of a single-nucleotide polymorphism in CD40 with the rate of joint destruction in rheumatoid arthritis. Arthritis Rheum. 2009;60:2242–2247. doi: 10.1002/art.24721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Orozco G, et al. Association of CD40 with rheumatoid arthritis confirmed in a large UK case-control study. Ann Rheum Dis. 2010;69:813–816. doi: 10.1136/ard.2009.109579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen F, et al. CD40 gene polymorphisms confer risk of Behcet’s disease but not of Vogt-Koyanagi-Harada syndrome in a Han Chinese population. Rheumatology (Oxford) 2012;51:47–51. doi: 10.1093/rheumatology/ker345. [DOI] [PubMed] [Google Scholar]
  • 42.Joo YB, et al. Association of genetic polymorphisms in CD40 with susceptibility to SLE in the Korean population. Rheumatology. 2013;52:623–630. doi: 10.1093/rheumatology/kes339. [DOI] [PubMed] [Google Scholar]
  • 43.Chen JM, et al. The association of CD40 polymorphisms with CD40 serum levels and risk of systemic lupus erythematosus. BMC Genet. 2015;16:121. doi: 10.1186/s12863-015-0279-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lee YH, Bae SC, Choi SJ, Ji JD, Song GG. Associations between the functional CD40 rs4810485 G/T polymorphism and susceptibility to rheumatoid arthritis and systemic lupus erythematosus: a meta-analysis. Lupus. 2015;24:1177–1183. doi: 10.1177/0961203315583543. [DOI] [PubMed] [Google Scholar]
  • 45.Boumpas DT, et al. A short course of BG9588 (anti-CD40 ligand antibody) improves serologic activity and decreases hematuria in patients with proliferative lupus glomerulonephritis. Arthritis Rheum. 2003;48:719–727. doi: 10.1002/art.10856. [DOI] [PubMed] [Google Scholar]
  • 46.Davis JC, et al. Phase I clinical trial of a monoclonal antibody against CD40-ligand (IDEC-131) in patients with systemic lupus erythematosus. J Rheumatol. 2001;28:95–101. [PubMed] [Google Scholar]
  • 47.Kalunian KC, et al. Treatment of systemic lupus erythematosus by inhibition of T cell costimulation with anti-CD154: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2002;46:3251–3258. doi: 10.1002/art.10681. [DOI] [PubMed] [Google Scholar]
  • 48.Ferrant JL, et al. The contribution of Fc effector mechanisms in the efficacy of anti-CD154 immunotherapy depends on the nature of the immune challenge. Int Immunol. 2004;16:1583–1594. doi: 10.1093/intimm/dxh162. [DOI] [PubMed] [Google Scholar]
  • 49.Xie JH, et al. Engineering of a novel anti-CD40L domain antibody for treatment of autoimmune diseases. J Immunol. 2014;192:4083–4092. doi: 10.4049/jimmunol.1303239. [DOI] [PubMed] [Google Scholar]
  • 50.Shock A, et al. CDP7657, an anti-CD40L antibody lacking an Fc domain, inhibits CD40L-dependent immune responses without thrombotic complications: an in vivo study. Arthritis Res Ther. 2015;17:234. doi: 10.1186/s13075-015-0757-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cordoba F, et al. A novel, blocking, Fc-silent anti-CD40 monoclonal antibody prolongs nonhuman primate renal allograft survival in the absence of B cell depletion. Am J Transplant. 2015;15:2825–2836. doi: 10.1111/ajt.13377. [DOI] [PubMed] [Google Scholar]
  • 52.US National Library of Medicine. ClinicalTrials.gov. 2016 https://clinicaltrials.gov/ct2/show/NCT02291029.
  • 53.US National Library of Medicine. ClinicalTrials.gov. 2017 https://clinicaltrials.gov/ct2/show/NCT02089087.
  • 54.Croft M. Control of immunity by the TNFR-related molecule OX40 (CD134) Annu Rev Immunol. 2010;28:57–78. doi: 10.1146/annurev-immunol-030409-101243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yoshioka T, et al. Contribution of OX40/OX40 ligand interaction to the pathogenesis of rheumatoid arthritis. Eur J Immunol. 2000;30:2815–2823. doi: 10.1002/1521-4141(200010)30:10<2815::AID-IMMU2815>3.0.CO;2-#. [DOI] [PubMed] [Google Scholar]
  • 56.Gwyer Findlay E, et al. OX40L blockade is therapeutic in arthritis, despite promoting osteoclastogenesis. Proc Natl Acad Sci USA. 2014;111:2289–2294. doi: 10.1073/pnas.1321071111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Horai R, et al. TNF-alpha is crucial for the development of autoimmune arthritis in IL-1 receptor antagonist-deficient mice. J Clin Invest. 2004;114:1603–1611. doi: 10.1172/JCI20742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Giacomelli R, et al. T lymphocytes in the synovial fluid of patients with active rheumatoid arthritis display CD134-OX40 surface antigen. Clin Exp Rheumatol. 2001;19:317–320. [PubMed] [Google Scholar]
  • 59.Boot EP, et al. CD134 as target for specific drug delivery to auto-aggressive CD4+ T cells in adjuvant arthritis. Arthritis Res Ther. 2005;7:R604–R615. doi: 10.1186/ar1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Aten J, et al. Strong and selective glomerular localization of CD134 ligand and TNF receptor-1 in proliferative lupus nephritis. J Am Soc Nephrol. 2000;11:1426–1438. doi: 10.1681/ASN.V1181426. [DOI] [PubMed] [Google Scholar]
  • 61.Jacquemin C, et al. OX40 ligand contributes to human lupus pathogenesis by promoting T follicular helper response. Immunity. 2015;42:1159–1170. doi: 10.1016/j.immuni.2015.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Patschan S, et al. CD134 expression on CD4+ T cells is associated with nephritis and disease activity in patients with systemic lupus erythematosus. Clin Exp Immunol. 2006;145:235–242. doi: 10.1111/j.1365-2249.2006.03141.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Abo-Elenein A, Shaaban D, Gheith O. Flowcytometric study of expression of perforin and CD134 in patients with systemic lupus erythematosus. Egypt J Immunol. 2008;15:135–143. [PubMed] [Google Scholar]
  • 64.Dolff S, et al. Increased expression of costimulatory markers CD134 and CD80 on interleukin-17 producing T cells in patients with systemic lupus erythematosus. Arthritis Res Ther. 2010;12:R150. doi: 10.1186/ar3100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Farres MN, Al-Zifzaf DS, Aly AA, Abd Raboh NM. OX40/OX40L in systemic lupus erythematosus: association with disease activity and lupus nephritis. Ann Saudi Med. 2011;31:29–34. doi: 10.4103/0256-4947.75775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cunninghame Graham DS, et al. Polymorphism at the TNF superfamily gene TNFSF4 confers susceptibility to systemic lupus erythematosus. Nat Genet. 2008;40:83–89. doi: 10.1038/ng.2007.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Qin W, et al. Increased OX40 and soluble OX40 ligands in children with Henoch-Schonlein purpura: association with renal involvement. Pediatr Allergy Immunol. 2011;22:54–59. doi: 10.1111/j.1399-3038.2010.01111.x. [DOI] [PubMed] [Google Scholar]
  • 68.Murata K, et al. Constitutive OX40/OX40 ligand interaction induces autoimmune-like diseases. J Immunol. 2002;169:4628–4636. doi: 10.4049/jimmunol.169.8.4628. [DOI] [PubMed] [Google Scholar]
  • 69.Laustsen JK, et al. Soluble OX40L is associated with presence of autoantibodies in early rheumatoid arthritis. Arthritis Res Ther. 2014;16:474. doi: 10.1186/s13075-014-0474-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nordmark G, et al. Association of EBF1, FAM167A(C8orf13)-BLK and TNFSF4 gene variants with primary Sjogren’s syndrome. Genes Immun. 2011;12:100–109. doi: 10.1038/gene.2010.44. [DOI] [PubMed] [Google Scholar]
  • 71.Gourh P, et al. Association of TNFSF4 (OX40L) polymorphisms with susceptibility to systemic sclerosis. Ann Rheum Dis. 2010;69:550–555. doi: 10.1136/ard.2009.116434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Komura K, et al. Increased serum soluble OX40 in patients with systemic sclerosis. J Rheumatol. 2008;35:2359–2362. [PubMed] [Google Scholar]
  • 73.Wilde B, et al. CD4+CD25+ T-cell populations expressing CD134 and GITR are associated with disease activity in patients with Wegener’s granulomatosis. Nephrol Dial Transplant. 2009;24:161–171. doi: 10.1093/ndt/gfn461. [DOI] [PubMed] [Google Scholar]
  • 74.Meylan F, et al. The TNF-family receptor DR3 is essential for diverse T cell-mediated inflammatory diseases. Immunity. 2008;29:79–89. doi: 10.1016/j.immuni.2008.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pappu BP, et al. TL1A-DR3 interaction regulates Th17 cell function and Th17-mediated autoimmune disease. J Exp Med. 2008;205:1049–1062. doi: 10.1084/jem.20071364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fang L, Adkins B, Deyev V, Podack ER. Essential role of TNF receptor superfamily 25 (TNFRSF25) in the development of allergic lung inflammation. J Exp Med. 2008;205:1037–1048. doi: 10.1084/jem.20072528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Meylan F, Richard AC, Siegel RM. TL1A and DR3, a TNF family ligand-receptor pair that promotes lymphocyte costimulation, mucosal hyperplasia, and autoimmune inflammation. Immunol Rev. 2011;244:188–196. doi: 10.1111/j.1600-065X.2011.01068.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Richard AC, et al. The TNF-family cytokine TL1A: from lymphocyte costimulator to disease co-conspirator. J Leukoc Biol. 2015;98:333–345. doi: 10.1189/jlb.3RI0315-095R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shih DQ, et al. Insights into TL1A and IBD pathogenesis. Adv Exp Med Biol. 2011;691:279–288. doi: 10.1007/978-1-4419-6612-4_29. [DOI] [PubMed] [Google Scholar]
  • 80.Cassatella MA, et al. Soluble TNF-like cytokine (TL1A) production by immune complexes stimulated monocytes in rheumatoid arthritis. J Immunol. 2007;178:7325–7333. doi: 10.4049/jimmunol.178.11.7325. [DOI] [PubMed] [Google Scholar]
  • 81.Bamias G, et al. Circulating levels of TNF-like cytokine 1A (TL1A) and its decoy receptor 3 (DcR3) in rheumatoid arthritis. Clin Immunol. 2008;129:249–255. doi: 10.1016/j.clim.2008.07.014. [DOI] [PubMed] [Google Scholar]
  • 82.Zhang J, et al. Role of TL1A in the pathogenesis of rheumatoid arthritis. J Immunol. 2009;183:5350–5357. doi: 10.4049/jimmunol.0802645. [DOI] [PubMed] [Google Scholar]
  • 83.Bamias G, et al. Circulating levels of TNF-like cytokine 1A correlate with the progression of atheromatous lesions in patients with rheumatoid arthritis. Clin Immunol. 2013;147:144–150. doi: 10.1016/j.clim.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • 84.Sun X, et al. Elevated serum and synovial fluid TNF-like ligand 1A (TL1A) is associated with autoantibody production in patients with rheumatoid arthritis. Scand J Rheumatol. 2013;42:97–101. doi: 10.3109/03009742.2012.727026. [DOI] [PubMed] [Google Scholar]
  • 85.Cao L, Xu T, Huang C, Li J. Elevated serum and synovial fluid TNF-like ligand 1A (TL1A) is associated with autoantibody production in patients with rheumatoid arthritis: comments on the article by Sun et al. Scand J Rheumatol. 2014;43:175. doi: 10.3109/03009742.2013.848931. [DOI] [PubMed] [Google Scholar]
  • 86.Xiu Z, Shen H, Tian Y, Xia L, Lu J. Serum and synovial fluid levels of tumor necrosis factor-like ligand 1A and decoy receptor 3 in rheumatoid arthritis. Cytokine. 2015;72:185–189. doi: 10.1016/j.cyto.2014.12.026. [DOI] [PubMed] [Google Scholar]
  • 87.Bull MJ, et al. The death receptor 3-TNF-like protein 1A pathway drives adverse bone pathology in inflammatory arthritis. J Exp Med. 2008;205:2457–2464. doi: 10.1084/jem.20072378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang X, et al. TNF-like ligand 1A (TL1A) gene knockout leads to ameliorated collagen-induced arthritis in mice: implication of TL1A in humoral immune responses. J Immunol. 2013;191:5420–5429. doi: 10.4049/jimmunol.1301475. [DOI] [PubMed] [Google Scholar]
  • 89.Wang EC, et al. Regulation of early cartilage destruction in inflammatory arthritis by death receptor 3. Arthritis Rheumatol. 2014;66:2762–2772. doi: 10.1002/art.38770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Osawa K, Takami N, Shiozawa K, Hashiramoto A, Shiozawa S. Death receptor 3 (DR3) gene duplication in a chromosome region 1p36.3: gene duplication is more prevalent in rheumatoid arthritis. Genes Immun. 2004;5:439–443. doi: 10.1038/sj.gene.6364097. [DOI] [PubMed] [Google Scholar]
  • 91.Zinovieva E, et al. Comprehensive linkage and association analyses identify haplotype, near to the TNFSF15 gene, significantly associated with spondyloarthritis. PLoS Genet. 2009;5:e1000528. doi: 10.1371/journal.pgen.1000528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bamias G, et al. Upregulation and nuclear localization of TNF-like cytokine 1A (TL1A) and its receptors DR3 and DcR3 in psoriatic skin lesions. Exp Dermatol. 2011;20:725–731. doi: 10.1111/j.1600-0625.2011.01304.x. [DOI] [PubMed] [Google Scholar]
  • 93.Li L, et al. TNF-like ligand 1A is associated with the pathogenesis of psoriasis vulgaris and contributes to IL-17 production in PBMCs. Arch Dermatol Res. 2014;306:927–932. doi: 10.1007/s00403-014-1497-z. [DOI] [PubMed] [Google Scholar]
  • 94.Xu WD, Chen DJ, Li R, Ren CX, Ye DQ. Elevated plasma levels of TL1A in newly diagnosed systemic lupus erythematosus patients. Rheumatol Int. 2015;35:1435–1437. doi: 10.1007/s00296-015-3277-2. [DOI] [PubMed] [Google Scholar]
  • 95.Al-Lamki RS, et al. Expression of silencer of death domains and death-receptor-3 in normal human kidney and in rejecting renal transplants. Am J Pathol. 2003;163:401–411. doi: 10.1016/S0002-9440(10)63670-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Al-Lamki RS, et al. TL1A both promotes and protects from renal inflammation and injury. J Am Soc Nephrol. 2008;19:953–960. doi: 10.1681/ASN.2007060706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Al-Lamki RS, et al. DR3 signaling protects against cisplatin nephrotoxicity mediated by tumor necrosis factor. Am J Pathol. 2012;180:1454–1464. doi: 10.1016/j.ajpath.2012.01.003. [DOI] [PubMed] [Google Scholar]
  • 98.Placke T, Kopp HG, Salih HR. Glucocorticoid-induced TNFR-related (GITR) protein and its ligand in antitumor immunity: functional role and therapeutic modulation. Clin Dev Immunol. 2010;2010:239083. doi: 10.1155/2010/239083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nocentini G, Ronchetti S, Petrillo MG, Riccardi C. Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol. 2012;165:2089–2099. doi: 10.1111/j.1476-5381.2011.01753.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cuzzocrea S, et al. Role of glucocorticoid-induced TNF receptor family gene (GITR) in collagen-induced arthritis. FASEB J. 2005;19:1253–1265. doi: 10.1096/fj.04-3556com. [DOI] [PubMed] [Google Scholar]
  • 101.Wang S, et al. Glucocorticoid-induced tumor necrosis factor receptor family-related protein exacerbates collagen-induced arthritis by enhancing the expansion of Th17 cells. Am J Pathol. 2012;180:1059–1067. doi: 10.1016/j.ajpath.2011.11.018. [DOI] [PubMed] [Google Scholar]
  • 102.Bae E, et al. Glucocorticoid-induced tumour necrosis factor receptor-related protein-mediated macrophage stimulation may induce cellular adhesion and cytokine expression in rheumatoid arthritis. Clin Exp Immunol. 2007;148:410–418. doi: 10.1111/j.1365-2249.2007.03363.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bae EM, et al. Reverse signaling initiated from GITRL induces NF-kappaB activation through ERK in the inflammatory activation of macrophages. Mol Immunol. 2008;45:523–533. doi: 10.1016/j.molimm.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 104.Patel M, et al. Glucocorticoid-induced TNFR family-related protein (GITR) activation exacerbates murine asthma and collagen-induced arthritis. Eur J Immunol. 2005;35:3581–3590. doi: 10.1002/eji.200535421. [DOI] [PubMed] [Google Scholar]
  • 105.Gu L, et al. Correlation of circulating glucocorticoid-induced TNFR-related protein ligand levels with disease activity in patients with systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:265868. doi: 10.1155/2012/265868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gan X, et al. Correlation of increased blood levels of GITR and GITRL with disease severity in patients with primary Sjogren’s syndrome. Clin Dev Immunol. 2013;2013:340751. doi: 10.1155/2013/340751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Nolte MA, van Olffen RW, van Gisbergen KP, van Lier RA. Timing and tuning of CD27-CD70 interactions: the impact of signal strength in setting the balance between adaptive responses and immunopathology. Immunol Rev. 2009;229:216–231. doi: 10.1111/j.1600-065X.2009.00774.x. [DOI] [PubMed] [Google Scholar]
  • 108.Oflazoglu E, et al. Blocking of CD27-CD70 pathway by anti-CD70 antibody ameliorates joint disease in murine collagen-induced arthritis. J Immunol. 2009;183:3770–3777. doi: 10.4049/jimmunol.0901637. [DOI] [PubMed] [Google Scholar]
  • 109.Tak PP, et al. Expression of the activation antigen CD27 in rheumatoid arthritis. Clin Immunol Immunopathol. 1996;80:129–138. doi: 10.1006/clin.1996.0106. [DOI] [PubMed] [Google Scholar]
  • 110.Lee WW, Yang ZZ, Li G, Weyand CM, Goronzy JJ. Unchecked CD70 expression on T cells lowers threshold for T cell activation in rheumatoid arthritis. J Immunol. 2007;179:2609–2615. doi: 10.4049/jimmunol.179.4.2609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Park JK, et al. CD70-expressing CD4 T cells produce IFN-gamma and IL-17 in rheumatoid arthritis. Rheumatology. 2014;53:1896–1900. doi: 10.1093/rheumatology/keu171. [DOI] [PubMed] [Google Scholar]
  • 112.Gattorno M, et al. Levels of soluble CD27 in sera and synovial fluid and its expression on memory T cells in patients with juvenile idiopathic arthritides. Clin Exp Rheumatol. 2002;20:863–866. [PubMed] [Google Scholar]
  • 113.Swaak AJ, Hintzen RQ, Huysen V, van den Brink HG, Smeenk JT. Serum levels of soluble forms of T cell activation antigens CD27 and CD25 in systemic lupus erythematosus in relation with lymphocytes count and disease course. Clin Rheumatol. 1995;14:293–300. doi: 10.1007/BF02208342. [DOI] [PubMed] [Google Scholar]
  • 114.Font J, et al. Elevated soluble CD27 levels in serum of patients with systemic lupus erythematosus. Clin Immunol Immunopathol. 1996;81:239–243. doi: 10.1006/clin.1996.0184. [DOI] [PubMed] [Google Scholar]
  • 115.Jacobi AM, et al. HLA-DRhigh/CD27high plasmablasts indicate active disease in patients with systemic lupus erythematosus. Ann Rheum Dis. 2010;69:305–308. doi: 10.1136/ard.2008.096495. [DOI] [PubMed] [Google Scholar]
  • 116.Oelke K, et al. Overexpression of CD70 and overstimulation of IgG synthesis by lupus T cells and T cells treated with DNA methylation inhibitors. Arthritis Rheum. 2004;50:1850–1860. doi: 10.1002/art.20255. [DOI] [PubMed] [Google Scholar]
  • 117.Sawalha AH, Jeffries M. Defective DNA methylation and CD70 overexpression in CD4+ T cells in MRL/lpr lupus-prone mice. Eur J Immunol. 2007;37:1407–1413. doi: 10.1002/eji.200636872. [DOI] [PubMed] [Google Scholar]
  • 118.Shaw J, Wang YH, Ito T, Arima K, Liu YJ. Plasmacytoid dendritic cells regulate B-cell growth and differentiation via CD70. Blood. 2010;115:3051–3057. doi: 10.1182/blood-2009-08-239145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Jiang H, et al. Demethylation of TNFSF7 contributes to CD70 overexpression in CD4+ T cells from patients with systemic sclerosis. Clin Immunol. 2012;143:39–44. doi: 10.1016/j.clim.2012.01.005. [DOI] [PubMed] [Google Scholar]
  • 120.Yin H, et al. Hypomethylation and overexpression of CD70 (TNFSF7) in CD4+ T cells of patients with primary Sjogren’s syndrome. J Dermatol Sci. 2010;59:198–203. doi: 10.1016/j.jdermsci.2010.06.011. [DOI] [PubMed] [Google Scholar]
  • 121.Snell LM, Lin GH, McPherson AJ, Moraes TJ, Watts TH. T-Cell intrinsic effects of GITR and 4-1BB during viral infection and cancer immunotherapy. Immunol Rev. 2011;244:197–217. doi: 10.1111/j.1600-065X.2011.01063.x. [DOI] [PubMed] [Google Scholar]
  • 122.Sanchez-Paulete AR, et al. Deciphering CD137 (4-1BB) signaling in T-cell costimulation for translation into successful cancer immunotherapy. Eur J Immunol. 2016;46:513–522. doi: 10.1002/eji.201445388. [DOI] [PubMed] [Google Scholar]
  • 123.Michel J, Langstein J, Hofstadter F, Schwarz H. A soluble form of CD137 (ILA/4-1BB), a member of the TNF receptor family, is released by activated lymphocytes and is detectable in sera of patients with rheumatoid arthritis. Eur J Immunol. 1998;28:290–295. doi: 10.1002/(SICI)1521-4141(199801)28:01<290::AID-IMMU290>3.0.CO;2-S. [DOI] [PubMed] [Google Scholar]
  • 124.Jung HW, Choi SW, Choi JI, Kwon BS. Serum concentrations of soluble 4-1BB and 4-1BB ligand correlated with the disease severity in rheumatoid arthritis. Exp Mol Med. 2004;36:13–22. doi: 10.1038/emm.2004.2. [DOI] [PubMed] [Google Scholar]
  • 125.Seo SK, et al. 4-1BB-mediated immunotherapy of rheumatoid arthritis. Nat Med. 2004;10:1088–1094. doi: 10.1038/nm1107. [DOI] [PubMed] [Google Scholar]
  • 126.Foell JL, et al. Engagement of the CD137 (4-1BB) costimulatory molecule inhibits and reverses the autoimmune process in collagen-induced arthritis and establishes lasting disease resistance. Immunology. 2004;113:89–98. doi: 10.1111/j.1365-2567.2004.01952.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Vinay DS, Choi JH, Kim JD, Choi BK, Kwon BS. Role of endogenous 4-1BB in the development of systemic lupus erythematosus. Immunology. 2007;122:394–400. doi: 10.1111/j.1365-2567.2007.02653.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sun Y, et al. Costimulatory molecule-targeted antibody therapy of a spontaneous autoimmune disease. Nat Med. 2002;8:1405–1413. doi: 10.1038/nm1202-796. [DOI] [PubMed] [Google Scholar]
  • 129.Foell J, et al. CD137-mediated T cell co-stimulation terminates existing autoimmune disease in SLE-prone NZB/NZW F1 mice. Ann NY Acad Sci. 2003;987:230–235. doi: 10.1111/j.1749-6632.2003.tb06052.x. [DOI] [PubMed] [Google Scholar]
  • 130.Foell J, et al. CD137 costimulatory T cell receptor engagement reverses acute disease in lupus-prone NZB x NZW F1 mice. J Clin Invest. 2003;111:1505–1518. doi: 10.1172/JCI17662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Browning JL. Inhibition of the lymphotoxin pathway as a therapy for autoimmune disease. Immunol Rev. 2008;223:202–220. doi: 10.1111/j.1600-065X.2008.00633.x. [DOI] [PubMed] [Google Scholar]
  • 132.Ware CF. Targeting the LIGHT-HVEM pathway. Adv Exp Med Biol. 2009;647:146–155. doi: 10.1007/978-0-387-89520-8_10. [DOI] [PubMed] [Google Scholar]
  • 133.Shui JW, Steinberg MW, Kronenberg M. Regulation of inflammation, autoimmunity, and infection immunity by HVEM-BTLA signaling. J Leukoc Biol. 2011;89:517–523. doi: 10.1189/jlb.0910528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Herro R, Croft M. The control of tissue fibrosis by the inflammatory molecule LIGHT (TNF Superfamily member 14) Pharmacol Res. 2016;104:151–155. doi: 10.1016/j.phrs.2015.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Greenberg JD, et al. A comparative effectiveness study of adalimumab, etanercept and infliximab in biologically naive and switched rheumatoid arthritis patients: results from the US CORRONA registry. Ann Rheum Dis. 2012;71:1134–1142. doi: 10.1136/annrheumdis-2011-150573. [DOI] [PubMed] [Google Scholar]
  • 136.Kennedy WP, et al. Efficacy and safety of pateclizumab (anti-lymphotoxin-alpha) compared to adalimumab in rheumatoid arthritis: a head-to-head phase 2 randomized controlled study (The ALTARA Study) Arthritis Res Ther. 2014;16:467. doi: 10.1186/s13075-014-0467-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Gommerman JL, Browning JL. Lymphotoxin/light, lymphoid microenvironments and autoimmune disease. Nat Rev Immunol. 2003;3:642–655. doi: 10.1038/nri1151. [DOI] [PubMed] [Google Scholar]
  • 138.Shui JW, et al. HVEM signalling at mucosal barriers provides host defence against pathogenic bacteria. Nature. 2012;488:222–225. doi: 10.1038/nature11242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Petreaca ML, et al. Deletion of a tumor necrosis superfamily gene in mice leads to impaired healing that mimics chronic wounds in humans. Wound Repair Regen. 2012;20:353–366. doi: 10.1111/j.1524-475X.2012.00785.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Anders RA, Subudhi SK, Wang J, Pfeffer K, Fu YX. Contribution of the lymphotoxin beta receptor to liver regeneration. J Immunol. 2005;175:1295–1300. doi: 10.4049/jimmunol.175.2.1295. [DOI] [PubMed] [Google Scholar]
  • 141.da Silva Antunes R, Madge L, Soroosh P, Tocker J, Croft M. The TNF family molecules LIGHT and lymphotoxin alphabeta induce a distinct steroid-resistant inflammatory phenotype in human lung epithelial cells. J Immunol. 2015;195:2429–2441. doi: 10.4049/jimmunol.1500356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Edwards JR, et al. LIGHT (TNFSF14), a novel mediator of bone resorption, is elevated in rheumatoid arthritis. Arthritis Rheum. 2006;54:1451–1462. doi: 10.1002/art.21821. [DOI] [PubMed] [Google Scholar]
  • 143.Pierer M, et al. The TNF superfamily member LIGHT contributes to survival and activation of synovial fibroblasts in rheumatoid arthritis. Rheumatology (Oxford) 2007;46:1063–1070. doi: 10.1093/rheumatology/kem063. [DOI] [PubMed] [Google Scholar]
  • 144.Herro R, Antunes Rda S, Aguilera AR, Tamada K, Croft M. The tumor necrosis factor superfamily molecule LIGHT promotes keratinocyte activity and skin fibrosis. J Invest Dermatol. 2015;135:2109–2118. doi: 10.1038/jid.2015.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Fava RA, et al. A role for the lymphotoxin/LIGHT axis in the pathogenesis of murine collagen-induced arthritis. J Immunol. 2003;171:115–126. doi: 10.4049/jimmunol.171.1.115. [DOI] [PubMed] [Google Scholar]
  • 146.Gatumu MK, et al. Blockade of lymphotoxin-beta receptor signaling reduces aspects of Sjogren’s syndrome in salivary glands of non-obese diabetic mice. Arthritis Res Ther. 2009;11:R24. doi: 10.1186/ar2617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Fava RA, Browning JL, Gatumu M, Skarstein K, Bolstad AI. LTBR-pathway in Sjogren’s syndrome: CXCL13 levels and B-cell-enriched ectopic lymphoid aggregates in NOD mouse lacrimal glands are dependent on LTBR. Adv Exp Med Biol. 2011;691:383–390. doi: 10.1007/978-1-4419-6612-4_39. [DOI] [PubMed] [Google Scholar]
  • 148.Seleznik G, et al. The lymphotoxin beta receptor is a potential therapeutic target in renal inflammation. Kidney Int. 2016;89:113–126. doi: 10.1038/ki.2015.280. [DOI] [PubMed] [Google Scholar]
  • 149.Herro R, Da Silva Antunes R, Aguilera AR, Tamada K, Croft M. Tumor necrosis factor superfamily 14 (LIGHT) controls thymic stromal lymphopoietin to drive pulmonary fibrosis. J Allergy Clin Immunol. 2015;136:757–768. doi: 10.1016/j.jaci.2014.12.1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Bienkowska J, et al. Lymphotoxin-LIGHT pathway regulates the interferon signature in rheumatoid arthritis. PLoS ONE. 2014;9:e112545. doi: 10.1371/journal.pone.0112545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Burkly LC. TWEAK/Fn14 axis: the current paradigm of tissue injury-inducible function in the midst of complexities. Semin Immunol. 2014;26:229–236. doi: 10.1016/j.smim.2014.02.006. [DOI] [PubMed] [Google Scholar]
  • 152.Burkly LC, Michaelson JS, Zheng TS. TWEAK/Fn14 pathway: an immunological switch for shaping tissue responses. Immunol Rev. 2011;244:99–114. doi: 10.1111/j.1600-065X.2011.01054.x. [DOI] [PubMed] [Google Scholar]
  • 153.Wiley SR, et al. A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis. Immunity. 2001;15:837–846. doi: 10.1016/s1074-7613(01)00232-1. [DOI] [PubMed] [Google Scholar]
  • 154.Meighan-Mantha RL, et al. The mitogen-inducible Fn14 gene encodes a type I transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem. 1999;274:33166–33176. doi: 10.1074/jbc.274.46.33166. [DOI] [PubMed] [Google Scholar]
  • 155.Park MC, Chung SJ, Park YB, Lee SK. Relationship of serum TWEAK level to cytokine level, disease activity, and response to anti-TNF treatment in patients with rheumatoid arthritis. Scand J Rheumatol. 2008;37:173–178. doi: 10.1080/03009740801898608. [DOI] [PubMed] [Google Scholar]
  • 156.van Kuijk AW, et al. TWEAK and its receptor Fn14 in the synovium of patients with rheumatoid arthritis compared to psoriatic arthritis and its response to tumour necrosis factor blockade. Ann Rheum Dis. 2010;69:301–304. doi: 10.1136/ard.2008.090548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Dharmapatni AA, et al. TWEAK and Fn14 expression in the pathogenesis of joint inflammation and bone erosion in rheumatoid arthritis. Arthritis Res Ther. 2011;13:R51. doi: 10.1186/ar3294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Xia L, Shen H, Xiao W, Lu J. Increased serum TWEAK levels in psoriatic arthritis: relationship with disease activity and matrix metalloproteinase-3 serum levels. Cytokine. 2011;53:289–291. doi: 10.1016/j.cyto.2010.12.003. [DOI] [PubMed] [Google Scholar]
  • 159.Chicheportiche Y, et al. TWEAK, a new secreted ligand in the tumor necrosis factor family that weakly induces apoptosis. J Biol Chem. 1997;272:32401–32410. doi: 10.1074/jbc.272.51.32401. [DOI] [PubMed] [Google Scholar]
  • 160.Kamijo S, et al. Involvement of TWEAK/Fn14 interaction in the synovial inflammation of RA. Rheumatology (Oxford) 2008;47:442–450. doi: 10.1093/rheumatology/ken006. [DOI] [PubMed] [Google Scholar]
  • 161.Novoyatleva T, Sajjad A, Engel FB. TWEAK-Fn14 cytokine-receptor axis: a new player of myocardial remodeling and cardiac failure. Front Immunol. 2014;5:50. doi: 10.3389/fimmu.2014.00050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Ucero AC, et al. TNF-related weak inducer of apoptosis (TWEAK) promotes kidney fibrosis and Ras-dependent proliferation of cultured renal fibroblast. Biochim Biophys Acta. 2013;1832:1744–1755. doi: 10.1016/j.bbadis.2013.05.032. [DOI] [PubMed] [Google Scholar]
  • 163.Perper SJ, et al. TWEAK is a novel arthritogenic mediator. J Immunol. 2006;177:2610–2620. doi: 10.4049/jimmunol.177.4.2610. [DOI] [PubMed] [Google Scholar]
  • 164.Kamata K, et al. Involvement of TNF-like weak inducer of apoptosis in the pathogenesis of collagen-induced arthritis. J Immunol. 2006;177:6433–6439. doi: 10.4049/jimmunol.177.9.6433. [DOI] [PubMed] [Google Scholar]
  • 165.Park JS, et al. TWEAK promotes osteoclastogenesis in rheumatoid arthritis. Am J Pathol. 2013;183:857–867. doi: 10.1016/j.ajpath.2013.05.027. [DOI] [PubMed] [Google Scholar]
  • 166.Wisniacki N, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of anti-TWEAK monoclonal antibody in patients with rheumatoid arthritis. Clin Ther. 2013;35:1137–1149. doi: 10.1016/j.clinthera.2013.06.008. [DOI] [PubMed] [Google Scholar]
  • 167.Sanz AB, et al. The cytokine TWEAK modulates renal tubulointerstitial inflammation. J Am Soc Nephrol. 2008;19:695–703. doi: 10.1681/ASN.2007050577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Xia Y, et al. Inhibition of the TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic serum nephritis. Clin Immunol. 2012;145:108–121. doi: 10.1016/j.clim.2012.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Zhao Z, et al. TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the chronic graft-versus-host model of systemic lupus erythematosus. J Immunol. 2007;179:7949–7958. doi: 10.4049/jimmunol.179.11.7949. [DOI] [PubMed] [Google Scholar]
  • 170.Wen J, et al. Neuropsychiatric disease in murine lupus is dependent on the TWEAK/Fn14 pathway. J Autoimmun. 2013;43:44–54. doi: 10.1016/j.jaut.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Doerner JL, et al. TWEAK/Fn14 signaling involvement in the pathogenesis of cutaneous disease in the MRL/lpr model of spontaneous lupus. J Invest Dermatol. 2015;135:1986–1995. doi: 10.1038/jid.2015.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Xia Y, et al. Deficiency of fibroblast growth factor-inducible 14 (fn14) preserves the filtration barrier and ameliorates lupus nephritis. J Am Soc Nephrol. 2015;26:1053–1070. doi: 10.1681/ASN.2014030233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Campbell S, et al. Proinflammatory effects of TWEAK/Fn14 interactions in glomerular mesangial cells. J Immunol. 2006;176:1889–1898. doi: 10.4049/jimmunol.176.3.1889. [DOI] [PubMed] [Google Scholar]
  • 174.Gao HX, et al. TNF-like weak inducer of apoptosis (TWEAK) induces inflammatory and proliferative effects in human kidney cells. Cytokine. 2009;46:24–35. doi: 10.1016/j.cyto.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 175.Schwartz N, et al. Urinary TWEAK as a biomarker of lupus nephritis: a multicenter cohort study. Arthritis Res Ther. 2009;11:R143. doi: 10.1186/ar2816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.US National Library of Medicine. ClinicalTrials.gov. 2016 https://clinicaltrials.gov/ct2/show/NCT01930890.
  • 177.Peter ME, Krammer PH. The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ. 2003;10:26–35. doi: 10.1038/sj.cdd.4401186. [DOI] [PubMed] [Google Scholar]
  • 178.Ramaswamy M, et al. Specific elimination of effector memory CD4+ T cells due to enhanced Fas signaling complex formation and association with lipid raft microdomains. Cell Death Differ. 2011;18:712–720. doi: 10.1038/cdd.2010.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Janssen EM, et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2005;434:88–93. doi: 10.1038/nature03337. [DOI] [PubMed] [Google Scholar]
  • 180.Ramaswamy M, Siegel RMA. FAScinating receptor in self-tolerance. Immunity. 2007;26:545–547. doi: 10.1016/j.immuni.2007.05.004. [DOI] [PubMed] [Google Scholar]
  • 181.Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 1992;356:314–317. doi: 10.1038/356314a0. [DOI] [PubMed] [Google Scholar]
  • 182.Rieux-Laucat F, et al. Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity. Science. 1995;268:1347–1349. doi: 10.1126/science.7539157. [DOI] [PubMed] [Google Scholar]
  • 183.Fisher GH, et al. Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome. Cell. 1995;81:935–946. doi: 10.1016/0092-8674(95)90013-6. [DOI] [PubMed] [Google Scholar]
  • 184.Drappa J, Vaishnaw AK, Sullivan KE, Chu JL, Elkon KB. Fas gene mutations in the Canale-Smith syndrome, an inherited lymphoproliferative disorder associated with autoimmunity. N Engl J Med. 1996;335:1643–1649. doi: 10.1056/NEJM199611283352204. [DOI] [PubMed] [Google Scholar]
  • 185.Lamhamedi-Cherradi SE, Zheng SJ, Maguschak KA, Peschon J, Chen YH. Defective thymocyte apoptosis and accelerated autoimmune diseases in TRAIL−/− mice. Nat Immunol. 2003;4:255–260. doi: 10.1038/ni894. [DOI] [PubMed] [Google Scholar]
  • 186.LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its death and decoy receptors. Cell Death Differ. 2003;10:66–75. doi: 10.1038/sj.cdd.4401187. [DOI] [PubMed] [Google Scholar]
  • 187.Wang J, et al. Inherited human Caspase 10 mutations underlie defective lymphocyte and dendritic cell apoptosis in autoimmune lymphoproliferative syndrome type II. Cell. 1999;98:47–58. doi: 10.1016/S0092-8674(00)80605-4. [DOI] [PubMed] [Google Scholar]
  • 188.Ichikawa K, et al. TRAIL-R2 (DR5) mediates apoptosis of synovial fibroblasts in rheumatoid arthritis. J Immunol. 2003;171:1061–1069. doi: 10.4049/jimmunol.171.2.1061. [DOI] [PubMed] [Google Scholar]
  • 189.Pundt N, et al. Susceptibility of rheumatoid arthritis synovial fibroblasts to FasL- and TRAIL-induced apoptosis is cell cycle-dependent. Arthritis Res Ther. 2009;11:R16. doi: 10.1186/ar2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Garcia S, Liz M, Gomez-Reino JJ, Conde C. Akt activity protects rheumatoid synovial fibroblasts from Fas-induced apoptosis by inhibition of Bid cleavage. Arthritis Res Ther. 2010;12:R33. doi: 10.1186/ar2941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Neve A, Corrado A, Cantatore FP. TNF-related apoptosis-inducing ligand (TRAIL) in rheumatoid arthritis: what’s new? Clin Exp Med. 2014;14:115–120. doi: 10.1007/s10238-012-0226-1. [DOI] [PubMed] [Google Scholar]
  • 192.Liu Z, et al. CII-DC-AdTRAIL cell gene therapy inhibits infiltration of CII-reactive T cells and CII-induced arthritis. J Clin Invest. 2003;112:1332–1341. doi: 10.1172/JCI19209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Yao Q, Seol DW, Mi Z, Robbins PD. Intra-articular injection of recombinant TRAIL induces synovial apoptosis and reduces inflammation in a rabbit knee model of arthritis. Arthritis Res Ther. 2006;8:R16. doi: 10.1186/ar1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Jin CH, et al. Effect of tumor necrosis factor-related apoptosis-inducing ligand on the reduction of joint inflammation in experimental rheumatoid arthritis. J Pharmacol Exp Ther. 2010;332:858–865. doi: 10.1124/jpet.109.159517. [DOI] [PubMed] [Google Scholar]
  • 195.Shi Q, et al. Anti-arthritic effects of FasL gene transferred intra-articularly by an inducible lentiviral vector containing improved tet-on system. Rheumatol Int. 2014;34:51–57. doi: 10.1007/s00296-011-2002-z. [DOI] [PubMed] [Google Scholar]
  • 196.Zhang W, Wang B, Wang F, Zhang J, Yu J. CTLA4-FasL fusion product suppresses proliferation of fibroblast-like synoviocytes and progression of adjuvant-induced arthritis in rats. Mol Immunol. 2012;50:150–159. doi: 10.1016/j.molimm.2012.01.007. [DOI] [PubMed] [Google Scholar]
  • 197.Ogasawara J, et al. Lethal effect of the anti-Fas antibody in mice. Nature. 1993;364:806–809. doi: 10.1038/364806a0. [DOI] [PubMed] [Google Scholar]
  • 198.Schmitz I, et al. An IL-2-dependent switch between CD95 signaling pathways sensitizes primary human T cells toward CD95-mediated activation-induced cell death. J Immunol. 2003;171:2930–2936. doi: 10.4049/jimmunol.171.6.2930. [DOI] [PubMed] [Google Scholar]
  • 199.Maksimow M, Soderstrom TS, Jalkanen S, Eriksson JE, Hanninen A. Fas costimulation of naive CD4 T cells is controlled by NF-kappaB signaling and caspase activity. J Leukoc Biol. 2006;79:369–377. doi: 10.1189/jlb.0505238. [DOI] [PubMed] [Google Scholar]
  • 200.Puliaeva I, Puliaev R, Shustov A, Haas M, Via CS. Fas expression on antigen-specific T cells has costimulatory, helper, and down-regulatory functions in vivo for cytotoxic T cell responses but not for T cell-dependent B cell responses. J Immunol. 2008;181:5912–5929. doi: 10.4049/jimmunol.181.9.5912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Klebanoff CA, et al. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. J Clin Invest. 2016;126:318–334. doi: 10.1172/JCI81217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Audo R, et al. Distinct effects of soluble and membrane-bound fas ligand on fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Rheumatol. 2014;66:3289–3299. doi: 10.1002/art.38806. [DOI] [PubMed] [Google Scholar]
  • 203.Li X, et al. Effect of CD95 on inflammatory response in rheumatoid arthritis fibroblast-like synoviocytes. Cell Immunol. 2014;290:209–216. doi: 10.1016/j.cellimm.2014.07.004. [DOI] [PubMed] [Google Scholar]
  • 204.Monaco C, Nanchahal J, Taylor P, Feldmann M. Anti-TNF therapy: past, present and future. Int Immunol. 2015;27:55–62. doi: 10.1093/intimm/dxu102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Sanz I, Yasothan U, Kirkpatrick P. Belimumab. Nat Rev Drug Discov. 2011;10:335–336. doi: 10.1038/nrd3436. [DOI] [PubMed] [Google Scholar]
  • 206.Furie R, et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63:3918–3930. doi: 10.1002/art.30613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Mariette X, et al. Efficacy and safety of belimumab in primary Sjogren’s syndrome: results of the BELISS open-label phase II study. Ann Rheum Dis. 2015;74:526–531. doi: 10.1136/annrheumdis-2013-203991. [DOI] [PubMed] [Google Scholar]
  • 208.Stohl W, et al. Efficacy and safety of belimumab in patients with rheumatoid arthritis: a phase II, randomized, double-blind, placebo-controlled, dose-ranging Study. J Rheumatol. 2013;40:579–589. doi: 10.3899/jrheum.120886. [DOI] [PubMed] [Google Scholar]
  • 209.Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005;23:23–68. doi: 10.1146/annurev.immunol.23.021704.115839. [DOI] [PubMed] [Google Scholar]
  • 210.Croft M. The TNF family in T cell differentiation and function — unanswered questions and future directions. Semin Immunol. 2014;26:183–190. doi: 10.1016/j.smim.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Fukasawa C, et al. Increased CD40 expression in skin fibroblasts from patients with systemic sclerosis (SSc): role of CD40-CD154 in the phenotype of SSc fibroblasts. Eur J Immunol. 2003;33:2792–2800. doi: 10.1002/eji.200324088. [DOI] [PubMed] [Google Scholar]
  • 212.Liu MF, Chao SC, Wang CR, Lei HY. Expression of CD40 and CD40 ligand among cell populations within rheumatoid synovial compartment. Autoimmunity. 2001;34:107–113. doi: 10.3109/08916930109001958. [DOI] [PubMed] [Google Scholar]
  • 213.Shih DQ, et al. Inhibition of a novel fibrogenic factor Tl1a reverses established colonic fibrosis. Mucosal Immunol. 2014;7:1492–1503. doi: 10.1038/mi.2014.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Anders HJ, Jayne DR, Rovin BH. Hurdles to the introduction of new therapies for immune-mediated kidney diseases. Nat Rev Nephrol. 2016;12:205–216. doi: 10.1038/nrneph.2015.206. [DOI] [PubMed] [Google Scholar]
  • 215.Genovese MC, et al. Combination therapy with etanercept and anakinra in the treatment of patients with rheumatoid arthritis who have been treated unsuccessfully with methotrexate. Arthritis Rheum. 2004;50:1412–1419. doi: 10.1002/art.20221. [DOI] [PubMed] [Google Scholar]
  • 216.Weinblatt M, et al. Selective costimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomised clinical trial. Ann Rheum Dis. 2007;66:228–234. doi: 10.1136/ard.2006.055111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Aringer M, Smolen JS. Therapeutic blockade of TNF in patients with SLE-promising or crazy? Autoimmun Rev. 2012;11:321–325. doi: 10.1016/j.autrev.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 218.Stohl W. Future prospects in biologic therapy for systemic lupus erythematosus. Nat Rev Rheumatol. 2013;9:705–720. doi: 10.1038/nrrheum.2013.136. [DOI] [PubMed] [Google Scholar]
  • 219.Soforo E, et al. Induction of systemic lupus erythematosus with tumor necrosis factor blockers. J Rheumatol. 2010;37:204–205. doi: 10.3899/jrheum.081312. [DOI] [PubMed] [Google Scholar]
  • 220.Chen X, et al. TNFR2 is critical for the stabilization of the CD4+Foxp3+ regulatory T. cell phenotype in the inflammatory environment. J Immunol. 2013;190:1076–1084. doi: 10.4049/jimmunol.1202659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.McCann FE, et al. Selective tumor necrosis factor receptor I blockade is antiinflammatory and reveals immunoregulatory role of tumor necrosis factor receptor II in collagen-induced arthritis. Arthritis Rheumatol. 2014;66:2728–2738. doi: 10.1002/art.38755. [DOI] [PubMed] [Google Scholar]
  • 222.Tsakiri N, Papadopoulos D, Denis MC, Mitsikostas DD, Kollias G. TNFR2 on non-haematopoietic cells is required for Foxp3+ Treg-cell function and disease suppression in EAE. Eur J Immunol. 2012;42:403–412. doi: 10.1002/eji.201141659. [DOI] [PubMed] [Google Scholar]
  • 223.Vinay DS, Kim CH, Choi BK, Kwon BS. Origins and functional basis of regulatory CD11c+CD8+ T cells. Eur J Immunol. 2009;39:1552–1563. doi: 10.1002/eji.200839057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Schreiber TH, et al. Therapeutic Treg expansion in mice by TNFRSF25 prevents allergic lung inflammation. J Clin Invest. 2010;120:3629–3640. doi: 10.1172/JCI42933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Wolf D, et al. Tregs expanded in vivo by TNFRSF25 agonists promote cardiac allograft survival. Transplantation. 2012;94:569–574. doi: 10.1097/TP.0b013e318264d3ef. [DOI] [PubMed] [Google Scholar]
  • 226.Ruby CE, et al. Cutting Edge: OX40 agonists can drive regulatory T cell expansion if the cytokine milieu is right. J Immunol. 2009;183:4853–4857. doi: 10.4049/jimmunol.0901112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Bresson D, Fousteri G, Manenkova Y, Croft M, von Herrath M. Antigen-specific prevention of type 1 diabetes in NOD mice is ameliorated by OX40 agonist treatment. J Autoimmun. 2011;37:342–351. doi: 10.1016/j.jaut.2011.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Ray A, Basu S, Williams CB, Salzman NH, Dittel BN. A novel IL-10-independent regulatory role for B cells in suppressing autoimmunity by maintenance of regulatory T cells via GITR ligand. J Immunol. 2012;188:3188–3198. doi: 10.4049/jimmunol.1103354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Nowakowska DJ, Kissler S. Ptpn22 modifies regulatory T cell homeostasis via GITR upregulation. J Immunol. 2016;196:2145–2152. doi: 10.4049/jimmunol.1501877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Carrier Y, et al. Enhanced GITR/GITRL interactions augment IL-27 expression and induce IL-10-producing Tr-1 like cells. Eur J Immunol. 2012;42:1393–1404. doi: 10.1002/eji.201142162. [DOI] [PubMed] [Google Scholar]

RESOURCES