Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: J Autoimmun. 2018 Aug 31;95:77–99. doi: 10.1016/j.jaut.2018.08.007

A Comprehensive Review on the Role of Co-signaling Receptors and Treg Homeostasis in Autoimmunity and Tumor Immunity

Prabhakaran Kumar a, Palash Bhattacharya a, Bellur S Prabhakar a
PMCID: PMC6289740  NIHMSID: NIHMS1505742  PMID: 30174217

Abstract

The immune system ensures optimum T-effector (Teff) immune responses against invading microbes and tumor antigens while preventing inappropriate autoimmune responses against self-antigens with the help of T-regulatory (Treg) cells. Thus, Treg and Teff cells help maintain immune homeostasis through mutual regulation. While Tregs can contribute to tumor immune evasion by suppressing anti-tumor Teff response, loss of Treg function can result in Teff responses against self-antigens leading to autoimmune disease. Thus, loss of homeostatic balance between Teff/Treg cells is often associated with both cancer and autoimmunity. Co-stimulatory and co-inhibitory receptors, collectively known as co-signaling receptors, play an indispensable role in the regulation of Teff and Treg cell expansion and function and thus play critical roles in modulating autoimmune and anti-tumor immune responses. Over the past three decades, considerable efforts have been made to understand the biology of co-signaling receptors and their role in immune homeostasis. Mutations in co-inhibitory receptors such as CTLA4 and PD1 are associated with Treg dysfunction, and autoimmune diseases in mice and humans. On the other hand, growing tumors evade immune surveillance by exploiting co-inhibitory signaling through expression of CTLA4, PD1 and PDL-1. Immune checkpoint blockade (ICB) using anti-CTLA4 and anti-PD1 has drawn considerable attention towards co-signaling receptors in tumor immunology and created renewed interest in studying other co-signaling receptors, which until recently have not been as well studied. In addition to co-inhibitory receptors, co-stimulatory receptors like OX40, GITR and 4-1BB have also been widely implicated in immune homeostasis and T-cell stimulation, and use of agonistic antibodies against OX40, GITR and 4-1BB has been effective in causing tumor regression. Although ICB has seen unprecedented success in cancer treatment, autoimmune adverse events arising from ICB due to loss of Treg homeostasis poses a major obstacle. Herein, we comprehensively review the role of various costimulatory and co-inhibitory receptors in Treg biology and immune homeostasis, autoimmunity, and anti-tumor immunity. Furthermore, we discuss the autoimmune adverse events arising upon targeting these co-signaling receptors to augment anti-tumor immune responses.

Keywords: Tregs, Co-stimulatory receptors, Co-inhibitory receptors, Autoimmunity, Cancer

1. Introduction

Immune surveillance is an essential component of the defense mechanism against infections and tumor progression. However, aberrant immune responses against self-antigens can result in autoimmune disease[1, 2] and failure to recognize and/or mount an effective immune response against tumor antigens can allow cancer to grow[3, 4]. Immune system comprises of many lymphoid and myeloid cell types which coordinately function to maintain immune homeostasis and ensure mounting of protective responses against infections while attenuating deleterious responses against self-antigens. T-cells are the most important effector cell types that mediate various immune responses and therefore, have been preferred targets for immunomodulation. T cells can be broadly classified as T-effector (Teff) cells and T-regulatory (Treg) cells based on the paradoxical nature of their function. Foxp3 is the lineage-specific transcription factor exclusively expressed in Tregs and not in Teff cells at least in mice[5]. The major discriminating factor between Treg and Teff cells is their affinity for self-antigens[6]. During thymic selection, T-cell clones expressing high-affinity T-cell receptors (TCRs) for self-antigens are either deleted by negative selection or rendered anergic. However, thymic negative selection is imperfect in that self-reactive T-cell clones often escape negative selection, migrate to the periphery and contribute to autoimmunity. However, T-cells expressing TCRs with an intermediate affinity for self-antigens gain Foxp3 expression and become Tregs are positively selected and migrate to the periphery where they help maintain peripheral self-tolerance[6]. In addition, CD4+CD25Foxp3- Tconv cells can differentiate into induced Treg (piTregs) cells in the periphery and suppress excessive inflammatory response directed against microbial antigens[7]. Physiologically, the antigen-specific Teff cells can eliminate infectious agents[8] and kill tumor cells[9]. However, uncontrolled or excessive Teff response can result in tissue destruction as seen in inflammatory diseases.[10] Similarly, inappropriate Teff response against self-antigens can result in autoimmune diseases[11]. In contrast, Treg cells can prevent or attenuate pro-inflammatory and autoimmune responses of Teff cells and help maintain immune homeostasis [12, 13]. In addition, Tregs can also suppress tumor-specific immune responses and thus facilitate immune evasion by tumor-cells resulting in tumor growth[14]. Therefore, understanding the molecular underpinnings regulating T-cell responses is not only of immense interest but also of clinical importance.

T-cell activation and expansion are regulated through a bi-signaling mechanism [15, 16]. Binding of MHC bound antigenic peptides displayed on antigen presenting cells (APCs) to TCRs of antigen-specific T cells activates the primary (first) TCR signal. This primary TCR signal when combined with secondary co-signals resulting from interactions between co-stimulatory/co-inhibitory ligands expressed on APCs and their cognate receptors expressed on T-cells determine the fate of T-cell response [17, 18]. Therefore, the co-signaling molecules play a critical role in regulating the immune response. Co-signaling molecules can be divided into two superfamilies based on their structure; 1) Immunoglobulin superfamily (Ig SF), 2) Tumor Necrosis Factor Superfamily (TNFSF)[19]. Ig SF co-signaling receptors are grouped based on their extracellular domain architecture[19] and consist of CD28, PVR-like, T-cell Immunoglobulin and Mucin (TIM) and CD2/Signaling Lymphocytic Activation Molecule (SLAM) families. TNF receptors share a conserved cysteine-rich signature containing domain[20] and include Glucocorticoid-Induced TNFR-related protein (GITR), OX40, 4-1BB, CD27, TNF-RI & II, Herpes Virus Entry Mediator (HVEM), etc.

Based on their function, co-signaling molecules can be divided into co-stimulatory and co-inhibitory molecules. Major co-stimulatory molecules include CD28, ICOS, TNFR-II, GITR, OX40, and 4-1BB, etc, and these co-stimulatory interactions play an essential role in many facets of the immune response such as T cell-antigen priming, expansion, survival, and differentiation and effector functions[19]. On the other hand, co-inhibitory molecules counteract costimulatory signaling and antagonize afore-mentioned facets of an immune response. Major co-inhibitory molecules include CTLA4, PD1, LAG3, TIM3, and TIGIT[19]. In general, co-stimulatory interactions are involved in the activation and expansion of naïve T-cells while co-inhibitory interactions provide feedback inhibitory signals to activated T cells to prevent excessive immune response [21]. Thus, the outcome of an immune response can be modulated through sequential co-signaling in a context-dependent manner to maintain immune homeostasis.

In general, co-signaling receptors, other than CD28 which is constitutively expressed, are predominantly expressed on Teff cells largely upon TCR-activation, but not on naïve T-cells. In contrast, Tregs constitutively express several co-signaling receptors like CTLA4, GITR, and OX40. In addition, many co-signaling receptors are preferentially over-expressed on Tregs which include PD1, ICOS, TIGIT, LAG3, TIM3, TNF-RII, and 4-1BB[22, 23]. This preferential overexpression is due to relatively higher TCR-signal strength experienced by Tregs compared to Teff cells during thymic selection. Such preferential overexpression of co-signaling receptors on Tregs might allow selective targeting for therapeutic utility. Overexpression of co-inhibitory receptors by Tregs is one of the prominent mechanisms by which they subvert APC-T-cell communication and suppress Teff cell response in autoimmunity[24]. Agonistic antibodies to co-inhibitory receptors and antagonistic antibodies to co-stimulatory receptors have shown promising results in treating autoimmune diseases[21]. However, tumor cells and tumor-infiltrating myeloid cells such as Myeloid-Derived Suppressor Cells (MDSCs)[25] and Tumor-Associated Macrophages (TAMs)[26] hijack this mechanism by expressing co-inhibitory receptors to maintain an immune suppressive microenvironment to evade immune surveillance[27]. In addition, tumor-infiltrating Tregs also significantly contribute to immune evasion by tumors through co-inhibitory receptor signaling[28] (Fig-1).

Figure-1: Regulation of Treg/Teff functions by co-signaling receptors in the tumor microenvironment.

Figure-1:

A) Ligation of co-inhibitory receptors expressed by Tregs such as CTLA4, PD1, TIGIT, LAG3 and TIM3 and their respective cognate ligands CD80/CD86, PDL1/L2, CD155/CD112, MHCII and galectin-9/phosphatidyl serine(PS) expressed by tumor tissues, Tumor-Associated Macrophages (TAMs) and Myeloid-Derived Suppressor Cells (MDSCs) leads to enhanced Treg function and diminished anti-tumor responses resulting in tumor progression. B) Ligation of corresponding agonists to GITR, OX40, 4-1BB, CD30 and CD27 receptors expressed on activated Teff cells leads to increased expansion, effector function through enhanced inflammatory cytokine production resulting in tumor regression.

Activation of co-stimulatory receptors such as CD28, OX40, and GITR promote Teff cell proliferation and effector functions. Agonistic antibodies to TNFRSFs such as OX40, GITR, 4-1BB, CD27, and CD30 have been used for inducing antitumor Teff cell responses in many cancers[29] (Fig-1). Therefore, agonistic signaling through these receptors is generally thought to regulate Teff cell response and contribute to immune enhancement. However, that notion was called into question upon discovering reduced Tregs in mice deficient in co-stimulatory receptor CD28 and accelerated autoimmune diabetes in CD28−/− NOD mice.[30] The second breakthrough occurred when it was found that co-stimulation through TNF-RII, GITR, and OX40 coupled to TCR signal strength facilitated thymic Treg selection and differentiation, and combined inhibition of these signaling pathways significantly impaired Treg generation in the thymus[31]. Thus, it has become evident that these co-stimulatory receptors play a key role in Treg differentiation and functions as well.

Immune Checkpoint Blockade (ICB) using anti-CTLA4 and anti-PD-1 antibodies have been successfully used to overcome immune evasion by tumor cells and facilitate an effective anti-tumor immune response[28]. Although modulating tumor micro-environment with ICB has resulted in better therapeutic efficacy than other interventions, still many patients are refractory to this approach, attributed to either compensatory mechanisms mediated through other co-inhibitory receptors or due to yet unknown inhibitory mechanisms. Moreover, autoimmune adverse events observed in patients who received ICB[32, 33] have revealed the challenges in targeting co-signaling receptors for selective immunomodulation.

Here, we critically review the role of important co-stimulatory and co-inhibitory molecules such as CD28, CTLA4, PD1, TIGIT, TIM3, LAG3, TNF-RII, GITR, OX40 and 4-1BB in Treg homeostasis and autoimmunity, and their involvement in tumor immune evasion and autoimmune complications arising upon targeting those molecules in cancer immunotherapy.

2. Immunoglobulin superfamily co-signaling receptors

2.1. CD28 family

Initially, the co-signaling was thought to be unidirectional due to the expression of ligands and their cognate receptors on different cell types. However, a growing body of evidence shows not only the expression of co-signaling ligands and their cognate receptors on the same cell type but also bidirectional signaling activated by the resultant interactions[19]. Therefore, we will use the term counter-receptor to denote such complex signaling partners which are expressed on both T-cells and APCs. Co-signaling receptors and ligands of IgSF are summarized in Table-1. CD28 was the first co-stimulatory molecule to be discovered and led to the conceptual evolution of co-signaling and description of the two-signal model [16, 34] of T cell activation. CD28 is constitutively expressed on both naive and activated CD4+/CD8+ T-cells and Tregs. B7.1 (CD80) and B7.2 (CD86) are expressed on APCs and their interaction with CD28 acts as a costimulatory signal for T-cell activation [35-37]. Although efforts were made to block CD28 co-signaling using soluble CD28-Ig, a major breakthrough in modulating CD28 co-signaling was attained upon discovering CTLA4, another IgSF member which is structurally similar to CD28[38, 39]. While CTLA4 is expressed on activated CD4+/CD8+ T-cells, it is constitutively expressed on Tregs.

Table-1:

Features of IgSF co-signaling receptors

Molecule Name Ligand/Counter receptor Cell types expressed Fate Functions
CD28 B7.1 (CD80), B7.2 (CD86), B7H2 (ICOSL) (Human) Constitutive expression on naïve and activated CD4+/CD8+ T-cells, Tregs Co-stimulation Antigen priming, Survival, Expansion, Memory differentiation, Effector functions[44, 67]
CTLA4 (CD252) B7.1 (CD80), B7.2 (CD86), B7H2 (ICOSL) (Human) Activated CD4+/CD8+ T-cells, Tregs Co-inhibition Survival, Expansion, Memory differentiation Effector functions[44, 67]
ICOS (CD278) B7H2 (ICOSL) Activated CD4+/CD8+ T-cells, Tregs Co-stimulation Survival, expansion, Th cell differentiation, effector functions, Memory differentiation[68]
PD-1 (CD279) PD-L1 (CD274, B7H1), PD-L2 (CD273, B7DC) Activated CD4+/CD8+ T-cells, B-cells, myeloid DCs, monocytes, exhausted T-cells and basal mesenchymal stem cells, subset of Tregs. Co-inhibition Antigen priming, T-cell exhaustion, effector functions[58]
CD226 (DNAM1) CD112, CD155 NK cells, platelets, monocytes and activated CD4+/CD8+ T cells, Subset of Tregs Co-stimulation Expansion, Th cell differentiation, effector functions[69]
TIGIT (VSIG9, VSTM3) CD112, CD155 NK cells, activated CD4+/CD8+ T cells, Subset of Tregs Co-inhibition Expansion and effector functions[69]
CD2 (LFA2) CD48 All Hematopoietic cells Co-stimulation Antigen priming, expansion, Effector function and memory differentiation[70, 71]
SLAMF1 (CD150) SLAM Hematopoietic stem cells, B cells, DCs, activated CD4+/CD8+ T cells, T follicular helper (Tfh) cells, macrophages, platelets. Co-stimulation Th cell differentiation, effector functions[71]
SLAMF4 (2BE4) CD48 Hematopoietic progenitors , NK cells, activated CD4+/CD8+ T cells, monocytes, basophils, Eosinophils Co-stimulation/co-inhibition Expansion and effector functions[71, 72]
TIM1 (HAVCR1) TIM1, TIM4, Phosphatidyl serine Activated CD4+ T-cells, B cells, Bregs, mast cells, DCs and macrophages Co-stimulation Expansion, Th cell differentiation, Effector functions[73-75]
TIM3 (HAVCR2) Galectin9, Phosphatidyl serine, HMGB1 Activated CD4+/CD8+ T-cells, NK cells Subset of Tregs, DCs and monocytes Co-inhibition Expansion, Th cell differentiation, Effector functions, memory differentiation[3, 75]
LAG3 MHCII Activated CD4+/CD8+ T cells, Subset of NK cells and Tregs. Co-inhibition Expansion and effector functions[23, 72]
CD160 HVEM NK cells, NKT cells, intraepithelial T cells, activated CD4+/CD8+ T cells. Co-inhibition Survival, Expansion, Memory differentiation, Effector functions[19]
BTLA (CD272) HVEM Naïve and activated CD4+/CD8+ T-cells, DCs Co-inhibition Survival, effector functions and memory differentiation[19, 72]

The CTLA4, which has a much higher affinity for B7.1 and B7.2, could effectively compete with CD28 for binding to those ligands and thus minimize or eliminate co-stimulatory effects of CD28, and this finding gave rise to the concept of co-inhibitory signaling [40, 41]. CTLA4 contains an extracellular IgV-like domain with B7.1/B7.2 ligand binding site and a short cytoplasmic domain which lacks neither enzymatic activity or Immunoreceptor tyrosine based inhibitor motifs (ITIM). In contrast to CD28, which is an exclusive cell membrane receptor, CTLA4 is highly endocytic in nature and exists in both membrane and cytoplasmic forms [42, 43]. CTLA4 exists as a dimer in cell membrane and each CTLA4 dimer binds to two B7.1/B7.2 homodimers [44, 45]. Upon B7.1/B7.2 ligand binding, CTLA4 induces trans-endocytosis of B7.1/B7.2 ligands, resulting in reduced expression of those ligands and consequent down-modulation of their co-stimulatory interaction with CD28 on Teff cells [46]. In addition, CTLA4 has been shown to induce cell-intrinsic signaling in T-cells leading to inhibition of T-cell activation/expansion through 1. Altered phosphorylation of CD3ζ chain; 2. Disruption of ZAP70 micro-cluster formation; 3.Intreaction with (Phosphatidyl Inositol 3-Kinase) PI3K and tyrosine phosphatases SHP1 and SHP2 which can dephosphorylate and thus inactivate TCR-signaling proteins [ reviewed in 43]. However, other studies have shown that these individual mechanisms are dispensable for CTLA4 function under varying contexts [43]. While extracellular domain of CTLA4 is essential for its co-inhibitory functions, cytoplasmic domain is likely involved in the regulation of endocytosis and trafficking of CTLA4. Several cytoplasmic motifs contribute to clathrin-mediated endocytosis through interaction with clathrin adaptor activating protein-2 (AP2)[47]. In addition, AP-1 interaction with cytoplasmic domain has also been shown to be associated with CTLA4 degradation[47].

ICOS, which shares structural similarity with CD28 and CTLA4[48], was first identified in humans[49] and later in mice[50]. As the name suggests, it is not expressed in resting T-cells and induced only upon TCR and/or CD28 costimulation [51]. While ICOS specifically interacts with its unique ligand, ICOSL (B7-H2)[52]; B7-H2 can interact with CTLA4 and CD28 as well at least in humans[53]. PD1 has gained greater attention due to its potential role in tumor evasion [54]. Unlike CTLA4 which undergoes endocytosis, membrane expression of PD1 is stable[55] and has two cognate ligands namely, PD-L1[56] and PD-L2[57]. The PD1 expression has been noted in activated CD4+/CD8+ T-cells, a subset of Tregs, B-cells, myeloid DCs, monocytes, exhausted T-cells and basal mesenchymal stem cells[58]. Lower levels of PDL1 expression was reported in unstimulated CD4+/CD8+ T-cells which was increased upon activation. Basal levels of expression of PDL1 was also observed in mesenchymal stem cells and vascular endothelium. In addition, activated B-cells, DCs and monocytes also express both PD-L1 and PD-L2[58]. Binding of these ligands to PD1 during T-cell activation leads to phosphorylation of tyrosine-based switch motif of PD-1, resulting in the recruitment of tyrosine phosphatase SHP-2[59], which in turn dephosphorylates proximal kinase Zeta chain Associated Protein kinase (Zap70) in T cells[60]. Inhibition of Zap70 and consequent suppression of downstream PKC-θ signaling leads to inhibition of T-cell activation [60]. Furthermore, PD-1 signaling inhibits TCR-signaling-induced cell cycle progression through downregulation of casein kinase-2 (CK2), which is an inhibitor of PTEN activation[61]. Thus, in the absence of CK2, increased PTEN activity can cause inhibition of PI3K-ZAP7O-PKC-θ cascade downstream of TCR signaling resulting in reduced T-cell growth and survival [62]. In addition, PD1 has also been shown to down-regulate TCR-expression by inducing TCR-internalization by increasing the expression of E3-ubiquitin ligases like (Casitas B-Lineage Lymphoma) c-CBL CBL-B, and ITCHY [63]. B and T-Lymphocyte Associated (BTLA) is a unique co-inhibitory receptor of CD28 IgSF [64] which binds to TNFRSF member HVEM (TNFRSF14)[65]. BTLA is expressed on T-cells and APCs, whereas its ligand/counter-receptor HVEM is expressed on resting and activated T-cells[66].

2.2. PVR-like family

Polio Virus Receptor (PVR)-like family of co-signaling molecules are newly identified members of IgSF and they share PVR signature motifs within their first Ig variable-like (IgV) domain[69, 76]. This family includes CD226, CD355, T-cell ImmunoGlobulin and Immunoreceptor Tyrosine-based inhibitory motif (TIGIT) and CD96 receptors and CD155 and CD112 serve as ligands [77, 78]. Regulation of co-signaling mechanism of PVR family resembles that of CD28-CTLA4 axis. Binding of CD226 with CD155 and CD112 leads to co-stimulation, whereas ligation of TIGIT with CD155 and CD112 delivers co-inhibitory signal [79]. TIGIT is expressed on effector and memory T-cells[80], a subset of Tregs[81] , Tfh cells[82], and NK cells[83]. CD155 and CD112 are expressed on T cells, APCs, several non-hematopoietic cells and tumor tissues [80, 83, 84]. While TIGIT strongly interacts with CD155, its interaction with CD112 is relatively weaker [85]. CD96 (Tactile), also binds to CD155, but not to CD112[86]. More recently, a strong PVR family receptor for CD112, known as CD112R, has been identified and likely plays a role in tumor evasion [87].

2.3. TIM family

T-cell Immunoglobulin and Mucin (TIM) domain containing co-signaling molecules include TIM1, TIM2, TIM3 and TIM4 of which TIM1, TIM3, and TIM4 are conserved between human and mouse[88-90], whereas TIM2 human analog has not yet been found[91, 92]. TIM1 is the prototypic member of this family which was initially identified as a receptor for hepatitis A virus[93].TIM1 and TIM4 can act as ligands and counter-receptor for each other and are expressed on both T-cells and APCs. Although TIM1 and TIM4 have been shown to deliver a co-stimulatory signal for T-cell activation [94, 95], TIM4 is also capable of inhibiting T-cell activation through interaction with a not yet known receptor [96]. TIM3 is expressed on activated T-cells and subset of Tregs, and binding of TIM3 with its ligand galectin-9 results in co-inhibitory signaling [90, 97]. In addition to galectin-9, high mobility group protein B1 (HMGB1) has been found to bind TIM3 and suppress innate immune responses [98]. Phosphatidyl serine can also act as a ligand for TIM1, TIM3, and TIM4 [23]. The Co-inhibitory function of TIM3 is implicated in tumor evasion and TIM3+ Tregs inhibit anti-tumor responses in many cancers [99, 100].

2.4. CD2/SLAM family

CD2/Signaling Lymphocytic Activation Molecule (SLAM) family members are receptors for morbilliviruses. The SLAM family includes CD2 (LFA2, OX34), SLAM (CD150, SLAMF1), CD48 (SLAMF2), Ly9 (SLAMF3), 2B4 (CD244, SLAMF4), CD84 (SLAMF5), Ly108 (CD352, SLAMF6), CRACC (CD319, SLAMF7)[19, 101]. SLAM family members possess an extracellular domain containing an IgV-like domain and an IgC2-like domain, and an intracellular cytoplasmic domain with multiple tyrosine-based motifs[102]. The only exceptional candidate is SLAMF3 which contains two tandem repeats of IgV-like and IgC2-like domains. They are widely expressed in many immune cell types including T-cells, B-cells, DCs and NK cells[101]. SLAM family members like SLAMF1, SLAMF3, SLAMF5 and SLAMF6 function as self-ligands, whereas CD2, SLAMF2, and SLAMF4 do not[103, 104]. CD2 and SLAMF1 signaling lead to co-stimulatory functions [71, 105], whereas 2BE4 (SLAMF4) and SLAMF6 signaling function as either co-stimulatory or co-inhibitory signal [106, 107].

In addition to the above mentioned IgSFs, two other well-known co-signaling molecules Lymphocyte Activating Gene-3 (LAG-3) and CD160 also have IgV domains and function as co-inhibitory receptors. LAG3 is expressed on activated CD4+ and CD8+ T-cells, a subset of Tregs and NK cells, and co-inhibitory functions of LAG3 have been implicated in autoimmunity [108] and immune evasion by tumors [23]. LAG3 is structurally homologous to CD4 and interacts with MHCII with a higher affinity than CD4[109]. Recently, LSECtin, a member of DC-SIGN family which is expressed in tumors has been characterized as a ligand for LAG3 and shown to be involved in tumor evasion [110]. CD160, another unique IgSF member which can act as a ligand for HVEM in addition to BTLA and exhibit co-inhibitory functions on T-cells [111]. Conversely, CD160 has also been shown to act as a co-stimulatory receptor for NK-cells [112].

3. TNF Superfamily co-signaling molecules

Co-signaling receptors and ligands of the TNF superfamily are summarized in Table-2. TNFSF ligands predominantly assemble as homotrimers, with the exception of mouse GITRL (dimeric) and LTaB2 (heterotrimer). In addition to their membrane-bound form, TNFSF ligands can also be produced in soluble form upon proteolytic cleavage of their ectodomains[20]. TNFRSF co-signaling receptors share a cysteine-rich domain with three disulfide bonds adjoining a core CXXCXXC motif. There are significant differences in the number of cysteine-rich domains between family members, varying from only one in BAFFR to six in CD30[113]. TNFSF receptors can be classified into three subtypes based on their cytosolic signaling domains.1) Death receptors containing death domains -DR3, DR6, TNFRI, TRAILR1, and TRAILR2. 2) TRAF-interacting receptors - TNFRII, LTbR, GITR, OX40, 41BB, CD30, CD40, HVEM, RANK, CD27, and BAFFR. 3) Decoy receptors lacking the cytosolic domain- TRAILR3, TRAILR4, DcR3[114]. High-affinity TNFSF ligand binding to their cognate receptors induces receptor clustering in target cells leading to the recruitment of adaptor proteins and resulting in the initiation of intracellular signal transduction[115]. Depending upon the signaling context, TNFRSF signaling can induce cell death, differentiation, survival or proliferation[20]. However, factors determining the outcome of TNFRSF signaling are not fully known and thus targeting TNFRSF signaling for the desired outcome remains challenging.

Table-2:

Features of TNFRSF co-signaling receptors

Molecule Name Ligand/Counter receptor Cell types expressed Fate Functions
TNFR2 (TNFRSF1B) TNF-α, LT-α3 Activated CD4+/CD8+ T-cells and B cells, a subset of Tregs, macrophages, DCs, endothelial cells and neural cells. Co-stimulation Expansion, Th cell differentiation and effector functions[118, 119]
GITR (TNFRSF18) GITRL (CD) CD4+/CD8+ T-cells and B cells, Tregs, NK cells. Co-inhibition Expansion and effector functions[120]
OX40 (TNFRSF4) OX40L (ICOSL) Activated CD4+/CD8+ T-cells, Tregs, NK cells. Co-stimulation Antigen priming, Survival, Expansion, Th cell differentiation, memory differentiation, Effector functions[121]
4-1BB (TNFRSF9) 4-1BBL Activated CD4+/CD8+ T-cells, Subset of Tregs. Co-stimulation Survival, Expansion, Memory differentiation, Effector functions[122, 123]
CD27 (TNFRSF7) CD70 CD4+/CD8+ T cells, Subset of B-cells and Tregs Co-stimulation Antigen priming, Survival, Expansion, Memory differentiation, Effector functions[123, 124]
CD30 CD30L (CD153) Activated CD4+/CD8+ T cells, B-cells, Tregs Co-stimulation Expansion and Th cell differentiation[125, 126]
HVEM LIGHT, LT-α3, BTLA, CD160 CD4+/CD8+ T-cells, DCs, Tregs, monocytes, NK cells, and neutrophils, Co-stimulation Antigen priming, Survival, Expansion, Memory differentiation Effector function [111, 125]

Unlike IgSF which constitute both co-inhibitory and co-stimulatory molecules, TNFRSF predominantly consists of co-stimulatory molecules. TNF signaling has been implicated in the development and functions of many cell types. TNFRI and II bind to TNF-α and lymphotoxin-α (LTα)[116]. Binding of TNF-α to TNFRI can promote cell death through activation of the death domain-containing adaptor proteins such as Fas-associated death domain (FADD) and TNFR1-associated death domain (TRADD). In contrast, binding of TNF-α to TNFR-II can lead to the activation of TRAF-mediated NF-kB activation resulting in cell survival and proliferation[117].

The TNFR-II expression is mainly restricted to immune cell types, neural and endothelial cells, whereas TNFR-I is widely expressed on many other cell types. TNFRII is expressed on activated, but not naive T-cells and delivers a costimulatory signal for their expansion and differentiation. It is highly expressed on a subset of naive Tregs and can promote their survival and proliferation [119, 127]. GITR (TNFSRF18) is expressed at low levels on naïve T-cells, B cells, and NK-cells, whereas it is constitutively expressed at higher levels on Tregs[119]. Activated T-cells also express higher levels of GITR[128]. Its cognate ligand GITRL is expressed on APCs and medullary thymic epithelial cells [31]. GITR has potent costimulatory functions and can induce a robust proliferation of Teff cells[128] and it is essential for CD4+ and CD8+ T-cell differentiation/expansion during infections[129]. 4-1BB (TNFRSF9) is expressed at low levels on naive Tregs, higher levels on activated T-cells and Tregs, and on several endothelial and epithelial cells. Its ligand 4-1BBL is expressed on professional APCs [119]. 4-1BBL/4-1BB ligation provides a co-stimulatory signal for T-cell expansion, survival, effector functions, and differentiation into memory phenotype [130, 131]. OX40 (CD134), similar to GITR, is expressed on activated T-cells and constitutively expressed on Tregs[121]. OX40 ligand is (OX40L) transiently expressed on activated APCs and highly expressed on Innate Lymphoid Cells (ILC)-2 and ILC-3[132]. OX40 signaling can promote Treg proliferation in the absence of canonical antigen presentation through MHC class-II in an IL-2 dependent manner [133]. In the context of TCR signaling, OX40L/OX40 ligation increases the proliferation and effector functions of TCR-activated T-cells and can also contribute to memory T cell differentiation[121]. CD30 (TNFRSF8), is another co-stimulatory molecule whose expression is induced on activated CD4/CD8 T-cells and B-cells and its cognate ligand CD30L is expressed on activated CD4/CD8 T-cells, macrophages, and B-cells[134]. CD30L/CD30 interaction can modulate immune responses by regulating T-cell survival/apoptosis and proliferation[135]. CD30 signaling has been implicated in the thymic negative selection and CD30 deficient mice show impaired negative selection and suffer from autoimmunity[135]. CD27 is expressed on naïve CD4+ and CD8+ Tconv cells, a subset of Tregs, B-cells and NK cells under resting state and its expression is transiently increased upon TCR activation[136], whereas its cognate ligand CD70 is expressed on thymic epithelial cells[137] and activated DCs, T and B cells, NK cells and Tregs[136]. CD27-CD70 interaction is a strong co-stimuli inducing CD4+ and CD8+ T-cell proliferation and Teff cell differentiation[138]. HVEM is a unique TNFRSF member that can interact with TNF ligand LIGHT (Lymphotoxin-related Inducible ligand that competes with HSV Glycoprotein-D for Herpesvirus entry mediator on T-cells) to induce co-stimulatory signaling for T-cell activation [139]. It is expressed on both resting and activated T-cells, B-cells, NK cells, DCs [140]. LIGHT acts as a counter-regulator for HVEM-BTLA mediated co-inhibitory pathway[141, 142] and in addition to HVEM, LIGHT can signal through LT-bR[143] and DcR3[144] which are expressed on various non-hematopoietic cell types.

4. Regulation of Treg homeostasis by co-signaling receptors

Tregs exhibit their inhibitory functions through multiple mechanisms such as, by modulating APC functions, producing suppressive cytokines, nutrient deprivation, IL-2 exhaustion and cytolysis. Tregs constitutively express co-inhibitory receptor CTLA4 which compete with co-stimulatory receptor CD28 for binding to co-stimulatory ligands CD80/CD86 expressed on APCs. Ligation of CTLA4 to CD80/CD86 on APCs leads to trans-endocytosis of these co-stimulatory ligands resulting in the ablation of T-cell co-stimulation [46]. Tregs produce immunosuppressive cytokines like IL-10, TGF-b and IL-35 which can suppress Teff cell proliferation and pro-inflammatory cytokine production [145, 146] [79] [147]. IL-10 and IL-35 play critical roles in the prevention of experimental colitis, and Tregs deficient for IL-10 [145, 146] and IL-35 had impaired suppressive function and failed to protect against colitis and IBD [79]. In addition, interaction of co-inhibitory receptor TIGIT expressed on Tregs with dendritic cells induces the production of immunosuppressive cytokines such IL-10 and TGF-b which suppress Teff cells. Tregs express higher levels of two proteins namely, CD39 and CD73, which catalyze ATP degradation to AMP, and AMP to adenosine sequentially. Adenosine in turn can down-regulate NF-KB signaling upon binding to A2A receptors expressed on Teff cells and APCs, resulting in the inhibition of effector cytokine and chemokine production [148]. In addition, constitutive expression of high affinity CD25 (IL-2Ra) by Tregs allows for higher consumption of IL-2 relative to Teff cells and results in IL-2 exhaustion and reduced proliferation and survival of Teff cells [149]. Besides, Tregs can also induce apoptosis of target cells such as APCs, CD4+/CD8+T-cells and NK cells via granzyme-A/B and perforin-mediated cytolysis [150, 151] [152].

CD4+ Tregs can be broadly classified into two major types CD4+Foxp3+ Tregs and CD4+Foxp3IL-10+ Tr1 cells. In addition, CD8+Foxp3+ Tregs have been reported to constitute a minor proportion of Treg pool[153]. CD4+Foxp3+ Tregs can be further classified as natural Tregs (nTregs) and induced Tregs (iTregs) or adaptive Tregs, based on their site of differentiation. Regulation of Treg homeostasis by co-signaling receptors is depicted in Fig-2. The nTregs are generated in the thymus (tTregs) through a bi-phasic process. Thymocytes expressing TCRs with intermediate affinity for self-antigens differentiate into CD4+CD25+Foxp3 and CD4+CD25Foxp3lowTreg precursors in a TCR-dependent phase [31, 154, 155]. Next, in an initial TCR-independent phase these Treg precursors gain Foxp3 expression through IL-2-dependent STAT5 activation[156]. In general, thymus-derived nTregs represent phenotypically stable lineage-specific Tregs which prevent aberrant autoimmune response against self-tissues due to their higher TCR affinity for self-antigens. Studies involving neonatal thymectomy have revealed the critical importance of tTregs in preventing autoimmunity[157]. Furthermore, lack of cognate antigen expression in the thymus leads to impaired differentiation of antigen-specific Tregs and loss of peripheral tolerance to the corresponding antigen [158]. In addition, extra-thymic de novo differentiation of iTregs occurs in the periphery from CD4+CD25Foxp3 Tconv cells through signals emanating from TCR, IL-2R and TGF-βR activation. The iTregs function mainly to tame excessive inflammatory response elicited against non-self-antigens such as food and microbial antigens[5]. Studies on iTregs have shown their importance for graft and gut tolerance [159, 160]. However, there is no convincing marker yet identified to differentiate between nTregs and iTregs although Helios/Nrp1 expression has been used to distinguish them in naïve mice[161].

Figure-2: Regulation of Treg homeostasis by co-signaling receptors.

Figure-2:

CD28, OX40, GITR and TNFRII signaling facilitates selection of CD4+CD25Foxp3low and CD4+CD25+Foxp3 tTreg precursors and positively regulate IL-2 dependent STAT5 activation mediated maturation of tTreg precursors into matured CD4+CD25+Foxp3+ tTregs. They also facilitate the proliferation of mature Tregs in thymus and periphery upon thymic emigration. In addition, CD28 and PD-1 signaling positively regulate differentiation of peripherally induced (iTregs)Tregs from CD4+CD25Foxp3 Tconv cells in synergy with TCR-TGF-β and IL2-induced STAT5 signaling. OX40, GITR, 4-1BB, and TNFR-II signaling negatively regulate iTreg differentiation while promoting proliferation of iTregs upon differentiation.

The B7-CD28/CTLA-4 co-signaling pathway plays a vital role in thymic[31] and peripheral[162] Treg/Teff cell development and functions[163]. During T-cell development in the thymus, CD28 is highly expressed on CD4+CD8+ DP thymocytes and expressed at relatively low levels in CD4+ and CD8+ SP T-cells. B7.1 and B7.2 ligands are expressed at low levels in the thymic cortex and higher levels in medulla[164]. CD28−/− mice on both C57BL6[162] and NOD background[30] had significantly reduced CD4+CD25+Foxp3+ Tregs. Blockade of CD28 signaling followed by adoptive transfer of Tregs led to rapid loss of transferred Tregs indicating the critical role of CD28 signaling for the survival of Tregs in the periphery [165]. However, Treg specific CD28−/− mice had only a 25-30% reduction in their thymic Tregs and no significant reduction in the periphery, indicating a Treg extrinsic role for CD28 signaling to maintain Treg homeostasis. More importantly, CD28−/− Tregs were functionally compromised and Treg specific CD28−/− mice developed spontaneous skin and lung autoimmunity [166]. Collectively, these studies indicate that CD28 signaling is required for thymic Treg development and survival, and for the expansion of Tregs in the periphery, and is indispensable for their suppressive functions.

CTLA4 is constitutively expressed by Tregs and is one of the target genes of Foxp3 [167]. The autoimmune symptoms arising in both Foxp3 and CTLA4 deficient mice are similar in nature indicating the convergence of CTLA4 signaling with Treg functions[168]. Ctla4−/− mice developed severe lymphoproliferative disease and inflammation in multiple organs including severe myocarditis and pancreatitis and died by 3-4 weeks of age [169]. Though ligation of CTLA4 on Teff cells delivers co-inhibitory signal independent of CTLA4+ Tregs, Ctla4−/− Tregs showed impaired functional ability in suppressing wild-type Teff cells, indicating an inherent role for Ctla4 signaling in Treg functions [5, 170, 171]. In contrast to these germline CTLA4 deletion studies, recent studies using conditional deletion of CTLA4 during adulthood showed increased peripheral expansion of Tregs with intact functions. In a recent study, T-cell specificCTLA4 expression was deleted during adulthood by crossing Ctla4fl/fl mice to LckCr+e mice and the resultant Ctla4fl/fl.Lckcre mice were protected against experimental autoimmune encephalomyelitis (EAE) and did not develop autoimmune symptoms [172]. The increase in Treg expansion seen in CTLA4 deficient adult mice could be either due to enhanced CD28 signaling in Tregs or cell-intrinsic inhibitory effect of CTLA4 on cell cycle progression[173]. In another study, Ctla4fl/fl mice was crossed to tamoxifen-inducible Cre recombinase Rosa26Cre/ERT2+ mice and CTLA4 ablation was induced by tamoxifen treatment. Ablation of CTLA4 expression during adulthood increased Tregs and these mice were protected against peptide-induced EAE, but developed accelerated collagen-induced arthritis (CIA). In addition, these mice developed severe, but not fatal, autoimmunity despite increased Tregs [174]. The discrepancy between these two studies can be explained by the experimental approach used wherein the earlier study lead to complete deletion of Ctla4 gene and the later might cause a partial ablation of Ctla4 due to tamoxifen treatment. In addition, differences in MHC class II haplotype might also contribute to the differences in autoimmune susceptibility observed in the later study which had H-2q haplotype instead H-2b haplotype in the earlier study. Nevertheless, attenuated Treg functions and appearance of autoimmune symptoms in cancer patients treated with anti-CTLA4 blocking antibodies suggest a key role of CTLA4 signaling in human Treg functions [175]. Thus, it is pertinent that CTAL4 can contribute to Treg functions in a cell-extrinsic manner by modulating APCs while inhibiting their expansion in a cell-intrinsic manner.

PD-1 co-inhibitory signaling can regulate T-cell repertoire and peripheral tolerance mechanisms to control autoimmunity. The PD1 expression has been noted in CD4CD8 DN thymocytes, whereas PD-L1 and PD-L2 are widely expressed in the thymic cortex and medulla respectively [176]. Tregs express both PD1 and PD-L1 under resting as well as activated conditions[58]. Unlike CTLA4 which inhibits Treg expansion, PD-L1-Ig augments iTreg generation from naïve T-cells and enhances Foxp3 expression and suppressive functions [177]. Moreover, it has been demonstrated that PD-1 expression in Tregs is indispensable for their suppressive functions, and loss of PD1 expression in Tregs accelerates the generation of exFoxp3 Tregs (plastic phenotype of Tregs which lose Foxp3 expression and produce pro-inflammatory cytokines) and thereby flare autoimmunity[178]. In contrast, to previous reports of an obligatory role for PD-1/PD-L1 signaling on iTreg generation, sustained Foxp3 expression and Treg lineage stability[177], Wong et al. reported an inverse correlation between PD1 expression and Treg functions, and blockade of PD1 signaling in NZB/NZW.F1 mice resulted in enhanced CD4+PD1lowFoxp3hi and CD8+PD1lowFoxp3hi Treg survival/functions[179, 180]. This apparent discrepancy between the two studies may be attributable to either difference in mouse strains used; especially NZB/NZB.F1 mice which are prone to autoimmune disease.

LAG3 is preferentially over-expressed on natural Tregs and induced Tregs, and its expression is induced on activated CD4/CD8 cells and NK cells. LAG3 is highly expressed in type-1 regulatory (Tr1) cells and co-expression of CD49b along with LAG3 is used as markers to characterize Tr1 cells [108]. Moreover, CD4+CD25+ Tregs from LAG3−/− mice had a reduced suppressive capacity indicating its involvement in Treg functions [181]. TIM3 is expressed on a subset of Tregs and TIM3+Foxp3+ Tregs were superior in IL-10 production than TIM3Foxp3+ Tregs and were potent suppressors of IFN-γ and TNF-α production [182]. Similarly, TIGIT expression is confined to a subset of Tregs and TIGIT+Foxp3+ Tregs show increased demethylation of the Treg-specific demethylated region in Foxp3 gene loci compared to TIGITFoxp3+Tregs, and express higher levels of nTreg signature genes such as Ctla4, Cd25, and Foxp3, and possess superior suppressive capacity compared to TIGIT Tregs[183]. This could be explained by the fact that TIGIT is a target gene of Foxp3 [184] and perhaps, TIGIT signaling enhances Foxp3 expression and Foxp3-dependent Treg signature genes through a positive feedback mechanism.

Unlike CD28 co-stimulation which positively regulates both nTreg and iTreg differentiation, TNFRSF co-stimulatory molecules play a dual role in Treg differentiation (Fig-2). TNFRSF members like TNFRII, GITR and OX40 play a secondary role to CD28 signaling in facilitating thymic Treg differentiation. Mahmud et al. found that CD28−/− mice had reduced TNFR-II+/GITR+/OX40+ Tregs in their thymus. Additionally, co-stimulation through TNF-RII, GITR, and OX40, when coupled to TCR signal, increased thymic Treg precursor differentiation and also synergized with IL-2-induced STAT5 signaling to increase thymic Treg maturation. Furthermore, combined inhibition of these signaling pathways significantly impaired Treg generation in the thymus [31]. OX40−/− mice had reduced Tregs in the thymus, but not in the periphery. More recently, we have shown that soluble OX40L-Fc can induce TCR-independent proliferation of murine and human thymic Treg precursors and matured Tregs in an IL-2 dependent manner [185, 186]. Like OX40 other TNFRSF members like GITR [187], TNFRII [187, 188] and 4-1BB [189] can also drive the proliferation of Tregs.

In contrast to its critical role in thymic Treg differentiation, OX40 signaling has been shown to inhibit iTreg differentiation in the periphery by antagonizing TGF-β signaling[190]. We have observed a similar negative effect of other TNFSF ligands such as 4-1BBL, GITRL, and TNF-α (Unpublished observations BP) on iTreg differentiation. Given the significant overlap in signaling cascades activated by these TNFRSF members and their functional outcomes, it is not surprising to observe similar effects on iTreg induction. Though TNFRSF co-stimulation can inhibit Foxp3 induction in iTregs, there is no clear evidence on the molecular mechanism of direct inhibition of Foxp3 transcription or RNA/protein stability in nTregs. It is plausible that the TNFRSF co-stimulation of Tconv cells with TCR stimulation might produce proinflammatory cytokines like IL-6 which can antagonize iTreg differentiation; instead promote Th17 cell differentiation in combination with TGF-β [191, 192]. In addition, OX40, GITR, 4-1BB, and TNFRII are capable of activating PI3K-AKT signaling[193] which can suppress Foxp3 induction by down-modulating transcription factors driving Foxp3 expression such as Foxo1 and Foxo3a [194]. However, once iTreg differentiation occurs, TNFRSF signaling might promote their proliferation in synergy with IL-2 signaling [195, 196]. Another possible mechanism by which OX40 signaling inhibits Foxp3 expression could be the induction of IL-2 exhaustion by proliferating Tregs expanded by OX40 signaling. Such IL-2 deprivation might attenuate STAT5 activation required for Foxp3 transcription in Tregs. In line with this notion, a recent study demonstrated that OX40 signaling-induced negative effects on Foxp3 expression were due to exhaustion of IL-2 and addition of exogenous IL-2 drove Treg proliferation with sustained Foxp3 expression [197]. Several studies have shown that Tregs expanded by TNFRSF signaling have compromised suppressive functions[187, 198-200] and it is claimed that activation of these co-signaling receptors in Teff cells rendered them resistant to Treg mediated suppression[201, 202]. Conversely, other studies have shown that Tregs expanded using 4-1BBL, TNF-α, OX40L, and GITRL were functionally competent [185, 187-189]. However, there is no mechanistic proof available as to how TNFR signaling causes loss of suppressive functions in expanded Tregs or impart Treg unresponsiveness to Teff cells. Since Foxp3 is a transcriptional repressor for TCR-induced expression of inflammation-associated genes like Ifn-g, Il-2, and Zap70 in Tregs[167], it can be postulated that reduced Foxp3 expression due to IL-2 exhaustion by expanded Tregs may compromise its ability to repress inflammatory genes leading to a labile phenotype and attenuated suppressive functions. Altogether, the effect of TNFR signaling on Treg differentiation, expansion and functions is clearly dependent on the activation state of Tregs and the local cytokine milieu. In contrast, CD28 signaling is essential for both thymic and peripheral Treg differentiation and proliferation, and CTAL4/PD-1 signaling are indispensable for Treg functions, but not expansion.

5. Role of IgSF co-signaling pathways in autoimmunity

5.1. B7-CD28-CTLA4 co-signaling axis

The effects of IgSF co-signaling pathways in modulating autoimmunity in various experimental autoimmune disease models are summarized in Table-3. Perturbations in B7-CD28-CTLA4 signaling has been shown to be involved in the pathogenesis of many autoimmune diseases like Rheumatoid Arthritis (RA) , Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), and Type-1 Diabetes (T1D)[21]. For example, CD28−/− mice exhibited significant resistance to collagen-induced arthritis (CIA) even after increased Ag loading, indicating the crucial role of CD28 co-stimulatory signaling in self-reactive T-cell priming in RA[203]. Similarly, blockade of CD28-signaling with CTLA4-Ig either before, during or after the disease induction prevented the development of CIA and ameliorated ongoing disease [204, 205]. Consistent with murine studies, human CTLA4-Ig (Abatacept) was found to be effective in RA treatment in human clinical trials [206]. MRL-lpr mice which spontaneously develop lupus showed delayed and mild symptoms of autoimmune lupus upon CD28 knockout[207], and CTLA4-Ig treatment of lupus-prone NZB/NZW.F1 mice resulted in attenuated T-cell activation, B cell maturation and antibody production leading to increased survival[208]. Thus, negative regulation of CD28 signaling by CTLA4-Ig was beneficial in treating experimental lupus. However, Abatacept clinical trials in SLE patients have failed to meet the primary endpoint, despite being well tolerated and producing 20-30% reduction in lupus induced nephritis [209].

Table-3:

Summary of IgSF co-signaling in experimental autoimmune diseases

Pathway Blocking/genetic deficieny/
Activation/ overexpression
Effect on autoimmunity Reference
CD28 CD28−/− (C57BL6 mice) Resistant to CIA [203]
B7.1/B7.2−/− (C57BL6 mice) Resistant EAE [211]
CD28−/− (MRL/lpr mice) Reduced auto-Ab levels, nephritis and no arthritis/vasculitis [239]
CD28−/− and B7.1/B7.2−/− NOD mice Reduced Tregs and accelerated
diabetes
[30]
CTLA4 CTLA4−/− (Adult C57BL/10.Q mice) Exacerbated CIA [174]
Murine CTLA4-Ig Prevented and ameliorated CIA [204, 205]
CTLA4−/− (Adult C57BL/10.Q mice) Protected from EAE [212]
Human CTLA4-Ig (Lewis rats) Protected from EAE [214]
Murine CTLA4-Ig (NZB/NZW F1 mice) Protects from lupus [208]
CTLA4Ig transgenic mice Reduced Tregs and accelerated
diabetes
[240]
Anti-CTLA4 blockade (BALBc/SCID mice) Exacerbated colitis [241]
PD-1 PD-1 blockade with soluble PD-1-Fc (DBA/1 male mice) Exacerbated CIA [242]
PD-1−/−(C57BL6 mice) Exacerbated CIA [228]
PD-L1 Ig (CBA/1J male mice) Protected from CIA [229]
PD-L1−/−mice (129S4/SvJae strain) Induced EAE susceptibility in the resistant strain [226]
[236]
PD-1 blocking mAb (C57BL6) Increase severity of EAE [219]
PD-1−/− C57BL/6 mice Spontaneous lupus [231]
Anti-PD-L1 (NZB/W F1 mice) Exacerbated lupus [232]
PD-L1 Ig (NZB/W F1 mice) Protected from lupus [232]
PD-1−/−NOD mice Accelerated diabetes [220]
Anti-PD-L1 blockade Induced diabetes susceptibility in the resistant strain [238]
LAG 3 Lag3−/− NOD mice Accelerated diabetes [243]
Lag-3−/− B6.SJL mice. Increased mercury include autoimmune glomerulonephritis [244]
TIM3 Anti-TIM3 blockade (SJL mice) Increased EAE [90]
TIM3−/− ( TCR transgenic C3HeB/FeJ mice) TIM3 blockade (NOD) Increased EAE [245]
Galectin-9 administration Accelerated diabetes [246]
Gal9−/−(C57BL/6J) mice Ameliorated CIA [247]
Increased CIA [247]
TIGIT TIGIT−/− (C57BL6) mice Increased EAE [80]
Vstm3−/− (C57BL6) mice Increased EAE, CIA [248]
Soluble VSTM3-Ig Reduced CIA [248]
Vstm3 transgenic mice Reduced EAE [248]
Recombinant mouse CD155 (MRL/Ipr mice) Delayed SLE [249]

B7-CD28 signaling has been shown to play a crucial role in T-cell priming in experimental autoimmune encephalomyelitis (EAE). For example, CD28−/− mice, expressing Myelin Binding Protein (MBP) 1-17 reactive-TCR transgene, failed to develop EAE [210]. Although C57BL6 mice deficient for both B7.1/B7.2 were resistant to Myelin Oligodendrocyte Glycoprotein (MOG) 35-55-induced EAE[211], neither CD80−/− nor CD86−/− single knockout C57BL6 mice showed any resistance to MOG35-55 induced EAE indicating the redundant role of these two co-stimulatory ligands in EAE pathogenesis[211]. Interestingly, deletion of CTLA4 during adulthood increased Treg numbers and conferred resistance to EAE [212, 213], likely due to increased CD28 signaling in Tregs in the absence of CTLA4. While treatment with CTLA4-Ig produced mixed results in EAE models[214, 215], abatacept has been shown to restrict the activation of MBP-reactive T cells with limited success[216] in human trials. Ctla4 gene is associated with IDDM susceptibility in both mice and humans [217]. CTLA4-Ig treatment of 2-4 weeks old NOD mice resulted in impaired Treg development and rapid precipitation of T1D [30]. Similarly, CD28−/− and CD80/CD86 double-deficient NOD mice showed accelerated T1D development with a reduced number of Tregs[30]. However, abatacept showed moderate efficacy in normalizing the glycemic index of T1D patients [218]. Taken together, it is evident that despite its critical role in Treg generation in the thymus and periphery, activation of CD28 signaling might contribute to the pathogenesis of autoimmune diseases like RA, SLE, and MS due to its co-stimulatory effect on Teff cells. CTLA4 signaling in Tregs and Teff cells is indispensable for preventing autoimmune response and loss of CTLA4 function in Tregs and Teff cells might accelerate autoimmunity.

5.2. PD1/ PDL1/L2 co-signaling pathway

PD1 signaling came to light upon the observation that Pdcd1−/− C57BL6 mice developed spontaneous lupus-like disease[219] and Pdcd-1−/− NOD mice had accelerated autoimmune diabetes compared to wild-type controls[220]. PD-1 signaling can suppress antigen priming, activation, differentiation and expansion of T-cells [221, 222]. Polymorphisms in Pdcd1 gene were associated with disease susceptibility to many autoimmune diseases including SLE, RA,[223] MS and T1D[224]. While neither PD-L1−/− nor PD-L2−/− mice developed overt organ-specific autoimmune diseases, PD-L1−/− deficiency conferred EAE susceptibility to EAE resistant 129S4/SvJae strain, indicating a determinant role for PD1-PD-L1 signaling in autoimmunity[225, 226]. Wen et al. reported the higher expression of PD1 and PDL1 in synovial T cells and macrophages of RA patients who were not on the immune suppressive regimen. They also found a splice variant of PD1 (PD-1Δex3) which is a soluble form of PD1 in the synovial fluid of RA patients which could antagonize the co-inhibitory functions of membrane-bound PD1 [227]. Pdcd1−/− mice were more susceptible to CIA[228] and PD-L1-Ig pre-treatment inhibited CIA[229], indicating a key role of PD1 in RA pathogenesis.

Pdcd1−/− C57BL6 mice spontaneously developed lupus-like disease upon aging[219]. Elevated levels of anti-PD1 antibodies were noted in new-onset SLE patients with increased effector T-cell proliferation [230]. Nonetheless, anti-PDL1 blockade led to accelerated lupus nephritis [231] and PDL1-Ig treatment protected mice from SLE [232] indicating a protective role for PD1 signaling in SLE. PD1 is expressed on CNS infiltrating T-cells, and Pdcd1−− mice are susceptible to severe EAE[233]. PDL1/L2 are expressed on astrocytes, vascular endothelial cells and APCs in EAE mice [233, 234] and blockade of both PD-L1 [235] and PD-L2 [236] has been shown to augment EAE. Immunization of Pdcd1−/− mice, but not WT mice, with MOG peptide without pertussis toxin (PT, an adjuvant required to induce co-stimulatory signaling and pro-inflammatory cytokine production) induced severe EAE. Moreover, MOG+PT immunized WT mice had reduced PD-1 expression in their Tregs. Interestingly, in WT mice that did not develop EAE after MOG immunization without PT, there was an increase in Tregs which was not seen in Pdcd1−/− mice [237]. Thus, it is plausible that PD-1/Treg mediated co-inhibitory signaling might keep pathogenic co-stimulatory signals/Teff cell activation in check and loss of PD-1 signaling can lead to activation of these events and increased pro-inflammatory cytokine production which might in turn aggravate EAE. Furthermore, PD-1 expression was found to be involved in the generation of myelin-specific Tregs from naive Tconv cells and PD1 expression was reduced in Tregs of WT mice which developed severe EAE [237]. Thus, PD-1 signaling might restrain EAE pathogenesis by enhancing Treg differentiation and functions. Pdcd1−/− NOD female mice had accelerated diabetes onset at 5 weeks of age with 100% penetrance at 10 weeks, whereas NOD WT littermates developed diabetes at 17 weeks of age [220]. In addition, blockade of PDL-1 signaling with anti-PDL-1 antibody induced diabetes in NOD strains that are resistant to diabetes [238]. Thus, PD1/PDL1 signaling plays an indispensable role in the maintenance of tolerance and loss of PD-1 signaling can cause autoimmunity due to the loss of Treg dependent tolerance.

5.3. LAG3, TIM-3, and TIGIT co-signaling pathways

Co-inhibitory receptors like LAG3, TIM-3, and TIGIT constitute a second set of co-inhibitory receptors. Unlike CTLA4 or PD-1 deficiency which lead to the spontaneous development of autoimmunity in mice, there is no reported incidence of the spontaneous autoimmune disease in mice deficient in LAG3, TIM-3 or TIGIT [23]. Hence, these co-signaling molecules were not expected to play a primary role in the pathogenesis of autoimmune diseases. However, recently accumulating evidence from studies in which these signaling pathways were blocked suggests their potential impact on autoimmunity. LAG-3 signaling negatively regulates T-cell response through both Treg-dependent and independent mechanisms [108]. CD4+CD25+ Tregs from LAG3−/− mice had reduced suppressive capacity and forced expression of LAG3 on Teff cells resulted in reduced proliferation [181]. LAG3−/− NOD mice showed accelerated diabetes development and had severe invasive insulitis with increased infiltration of islet antigen-specific CD4+ and CD8+ T-cells[243]. Co-deficiency of LAG3 along with PD1 induced lethal myocarditis in BALB/c mice [250]. LAG3 deficient B6.SJL mice were more susceptible to mercury-induced autoimmunity and adoptive transfer of LAG3 sufficient CD4+T-cells partially protected LAG3−/− mice from mercury-induced autoimmune symptoms [244]. LAG3 upregulation on gut-derived CTLA4+TGF-β+ Intraepithelial lymphocytes (IELs) has been found to inhibit inflammation and adoptive transfer of these IELs ameliorated EAE[251].

TIM3+Foxp3+ Tregs were superior in IL-10 production than TIM3Foxp3+ Tregs and potently suppressed IFN-γ and TNF-α production. More importantly, RA patients had reduced frequency and functions of TIM3+Foxp3+ Tregs in their peripheral blood compared to healthy subjects [182] indicating a role for TIM3 signaling in Treg functions related to autoimmunity. While Galectin9 administration ameliorated CIA, Gal9−/− mice were more susceptible to CIA [247]. TIM3 blockade aggravated EAE by augmenting Th1 responses [90] and TIM3−/− mice were refractory to high dose tolerance against EAE [252]. T-cell clones from MS patients had reduced TIM3 expression and higher IFN-γ production[253], and ligation of galectin-9 with TIM3, inhibited Th1 mediated EAE in immunized SJL/J mice[90]. Overexpression of Galectin-9 in NOD resulted in significant protection against diabetes by attenuating Th1 response [90] while TIM3 blockade accelerated diabetes in these mice [246].

TIGIT signaling can negatively regulate Teff cell responses by modulating DCs, enhancing Treg functions and shifting the Th1/Th2/Th17 balance. TIGIT inhibits IL-12 and enhances IL-10 production from DCs via interaction with CD155 expressed on DCs [85]. Moreover, TIGIT can selectively restrain Th1 and Th17 pathogenic responses, but not Th2 responses, thereby shifting the Th1/Th2 or Th17/Th2 balance to negate Th1 and Th17 mediated autoimmune response [183]. Though TIGIT−/− mice had the lymphoproliferative disorder, they did not develop spontaneous autoimmunity but showed increased susceptibility to EAE [254]. Treatment with soluble VSTM3 (TIGIT) inhibited CIA and transgenic overexpression of VSTM3 ameliorated EAE [248]. TIGIT −/− (Vstm3−/−) mice exhibited severe autoimmune symptoms in CIA and MOG-induced EAE models [80, 255]. More recently, an agonistic anti-TIGIT monoclonal antibody has been shown to suppress autoimmune severity in EAE [256]. Activation of TIGIT signaling by CD155 administration delayed SLE development in MRL/lpr mice [249]. TIGIT expression was significantly decreased in NK cells from SLE patients and correlated with increased IFN-γ expressing NK cells[257]. Collectively, these studies suggest a potential role for these co-inhibitory molecules in Treg functions and autoimmunity. However, Future studies on the Treg specific role of these molecules will be required to define the precise role of these co-inhibitory signaling pathways on Tregs and their contribution to tolerance induction.

6. Role of TNFSF signaling in autoimmune diseases

6.1. TNFR-II

The effects of various TNFRSF members such as TNFR-II, OX40, GITR and 4-1BB signaling in modulating autoimmunity in various experimental autoimmune disease models are summarized in Table-4. Unlike TNFRI which is ubiquitously expressed on many cell types, TNFRII expression is predominantly restricted to lymphocytes. Dysregulated TNFRII signaling is implicated in several autoimmune diseases such as MS, RA, T1D, and colitis[117] (Refer Table-4). Polymorphisms in TNFRSF1B (TNFRII) genes were found to be associated with many autoimmune diseases such as RA, Crohn’s diseases, and ulcerative colitis[117]. Increased levels of soluble TNFR-II were observed in RA patient’s serum which might be associated with reduced TNFRII signaling these patients [258]. TNF-α exists in both membrane-bound and soluble forms and therefore, can exert systemic co-stimulation effects in addition to cell-to-cell communication. Earlier, TNF-α and TNFR-II agonist were shown to selectively induce cell death of self-reactive CD8+ T-cells (likely due to activation-induced cell death) while sparing normal T-cells in In vitro T-cell cultures of T1D patients[117, 259]. Intriguingly, anti-TNF blocking antibody (adalimumab) which was expected to inhibit TNF-α signaling has been shown to promote Treg cell expansion by facilitating interaction between membrane-bound TNF-α on monocytes and TNFRII on Tregs[260]. Moreover, TNFRII agonists where shown to expand Tregs. And TNFRII−/− nTregs had impaired suppressive functions while TNFRII−/− iTregs had normal suppressive functions indicating a dispensable role of TNFRII signaling in peripherally induced Treg functions [261].

Table-4:

Summary of TNFSF receptor co-signaling in experimental autoimmune diseases

Pathway Blocking/genetic deficieny/
Activation/ overexpression
Effect on autoimmunity Reference
TNFRI/II Anti-TNF-α-Ig (DBA/1 male mice) Reduced CIA [269]
Soluble TNFR-β Reduced CIA [269]
TNFR-I−/− mice Low incidence of CIA with mild symptoms
Exacerbated arthritis
[270]
TNFR-II−/− mice Pretreatment before disease induction was protective and post-treatment was not. [271]
Soluble TNFR-Ig (Female Lewis rats immunized with Myelin Binding Protein- MBP)
TNFR-I−/− mice (C57BL6 )
TNFR-II−/− mice (C57BL6)
NZB × Tnf0 deficient hemizygous mice
Resistant to EAE
Exacerbated EAE
Accelerated SLE
Accelerated nephritis
[272]
[273]
[273]
[274]
TNF-α administration to NZB/W mice at late stage
TNFR-I−/− mice (NOD mice)
TNFR-II−/− mice (NOD mice)
TNF-− treatment < 3 weeks
Anti-TNF-a treatment < 3 weeks
TNF-a treatment 4- 7 weeks
Increased Tregs and suppressed diabetes
Accelerated diabetes
Accelerated diabetes
Protected from diabetes
Protected from diabetes
[275]
[276]
[277]
[278]
[278]
[278]
GITR GITR−/− mice(Sv129 strain) Less severe CIA [262]
Agonistic-GITR-mAb Exacerbated CIA [264]
GITRL Exacerbated CIA [279]
GITRL overexpressing C57BL6 mice Delayed MOG-induced EAE [266]
Agonistic-GITR-mAb SJL female mice
Agonistic-GITR-mAb (NOD mice)
Exacerbated PLP139-151 induced EAE [263]
Anti-GITRL blocking Ab (BDC 2.5 transfer model of diabetes) Accelerated diabetes [265]
GITR−/− mice (129Sv/Ev strain) Protected from diabetes [265]
GITR-Fc (WT 129Sv/Ev mice) Protected from TNBS-induced colitis
Protected from TNBS-induced colitis
[280]
[280]
OX40 Blocking anti-OX40 (DBA/1 male mice)
Blocking anti-OX40 Fab’PEG
Ameliorated CIA
Ameliorated CIA
[281]
[282]
OX40L−/− (C57BL6 mice) immunized with MOG Reduced susceptibility to EAE [283]
OX40L overexpressing Tg mice Enhanced clinical disease [283]
Anti-OX40 agonist treatment at priming phase (PLP139-151/CFA-immunized SJL mice) Amelioration of EAE [284]
Anti-OX40 agonist treatment at onset phase(PLP139-151/CFA-immunized SJL
mice)
Increased severity of EAE [284]
Anti-OX40 agonist treatment of young (13 weeks) NZB/W F1 mice before onset of protenuria No effect on lupus nephritis [285]
Anti-OX40 agonist treatment of old (21-27 week) NZB/W F1 mice after onset of protenuria Exacerbated lupus nephritis [285]
Blockade of OX40 in NOD mice at 12
weeks
Reduced diabetes incidence [286]
OX40L-Fc treatment to NOD mice at 12 weeks of age Exacerbated disease [287]
Anti-OX40 agonist treatment o NOD mice Expansion of antigen-specific Tregs and [288]
at 6-weeks of age with antigen delayed diabetes onset
immunization Expansion of Tregs and delayed diabetes
OX40L-Fc treatment to NOD mice at 6 onset [185]
weeks of age
Anti-OX40L blocking antibody treated C.B-17 SCID mice were reconstituted with CD45RBhigh CD4+ T-cells
Inhibition of colitis [289]
RAG−/− mice transferred with CD45RBhigh CD4+ T-cells with OX40−/− Tregs Severe colitis [290]
4-1BB Agonistic 4-1BB antibody (DBA1/male mice) Inhibited CIA [291]
Agonistic 4-1BB antibody (MOG immunized C57BL6 mice) Reduced severity of EAE [292]
Agonistic 4-1BB antibody (Adoptive transfer mode of EAE) No effect [292]
4-1BB agonistic Ab treatment (PLP139-151/CFA-immunized SJL mice) Inhibited relapsing/remitting EAE [292]
4-1BB deficient MRL/lpr mice Increased SLE [293]
Agonistic 4-1BB antibody (MRL/lpr mice)
4-1BB deficient Tnfrsf9−/−.NOD mice
Ameliorated lupus [294]
Agonistic 4-1BB antibody (6-week-old
NOD mice)
Delayed onset of diabetes [295]
Delayed onset of diabetes [296]
Adoptive transfer colitis in SCID mice
with CD45RBhigh 4-1BB−/−CD4+ T-cells
Severe colitis [297]

6.2. GITR

GITR is one of the widely accepted Treg markers and a known nTreg specific signature molecule whose gene loci is demethylated in Tregs[167]. GITR is also expressed on CD4+ and CD8+ T-cells at low levels and its expression is increased upon TCR activation. GITR−/− mice were found to be protected from experimental CIA [262] and colitis [262]. Many lines of evidence have shown that agonistic anti-GITR antibody treatment could exacerbate autoimmunity in several autoimmune disease models including EAE[263], CIA[264] and T1D[265], and blockade of GITR signaling at the time of onset of diabetes was found to be protective against diabetes[265]. While activation of GITR signaling in Tregs by agonistic GITR antibody can lead to reduced suppressor function, activation of GITR in Teff cells rendered them less responsive to Treg mediated suppression [198, 262]. In contrast, another independent study with GITRL overexpressing mice using Foxp3 as Treg marker showed a parallel increase in both Tregs and Teff cells and these mice showed delayed onset of EAE compared to WT mice [266]. This apparent discrepancy could be due to use of CD25 as a Treg marker (before the identification of Foxp3 as a Treg lineage-specific marker) in the earlier studies [198, 262], which is also expressed on activated T-cells. In addition, given the absolute requirement of the multimeric assembly of TNFSF ligands for receptor clustering and signal transduction[20], it is not clear whether crosslinking of the agonistic antibody with GITR can recapitulate the downstream signaling provoked by GITRL. Moreover, CD4+CD25GITR+CTLA4+Foxp3 cells mark an anergic subset of T-cells producing IL-10 and TGF-β, that can suppress Teff cell proliferation and thus control mucosal inflammation in mice[267]. Similarly, CD4+CD25low/−FoxP3lowGITR+ T-cells were identified in SLE patients and frequency of this population inversely correlated with SLE Disease Activity Index indicating a role for GITR signaling in maintaining anergy and suppressing autoimmune response[268].

6.3. OX40

OX40, expressed on activated T-cells, was initially characterized as a Teff cell co-stimulatory molecule promoting their survival and proliferation, although it is constitutively expressed on Tregs[125]. Interestingly, OX40 co-stimulation has been shown to boost Treg proliferation by inducing IL-2 production from Teff cells under low inflammatory conditions[298].OX40 signaling has been implicated in many autoimmune diseases and disruption of OX40 signaling was found to be protective (Table-4). OX40 has been located in the susceptibility loci for human SLE [299]. OX40 stimulation had no effect on lupus nephritis when given to young 13-week-old mice before the onset of proteinuria, however, treatment after the onset of proteinuria at 21-27 weeks exacerbated lupus nephritis by mediating Tfh cell response [300]. OX40-OX40L interaction at inflamed joints was noted in CIA and disruption of OX40 signaling reduced auto-Ab levels and improved joint inflammation [282]. Earlier studies on EAE induction using OX40L transgenic mice showed that OX40L can exacerbate EAE severity with a delayed onset. However, OX40L overexpression in CD28−/− and CD40−/− mice failed to confer disease indicating a secondary role for OX40 signaling in EAE[283]. Moreover, OX40 agonist treatment during the antigen priming phase increased functional Tregs and protected mice from developing EAE, whereas treatment during the effector phase exacerbated the disease [301]. Similarly, NOD mice treated with OX40 agonist at 6 weeks of age showed expansion of antigen-specific Tregs and delayed the onset of diabetes [288], whereas treatment of NOD mice at 12-weeks of age expanded labile Tregs and precipitated the disease [196]. Interestingly, we have observed that co-treatment of 10-12 week old NOD mice with OX40L and another Notch family ligand Jagged-1, resulted in expansion of functional Tregs and delayed the onset of diabetes[302]. Thus, OX40 signaling can regulate both Teff cells and Treg cells based on their activation state, and exert differential effects on autoimmunity based on the treatment at antigen priming phase vs effector phase of the disease development.

6.4. 4-1BB

4-1BB (CD137) is transiently expressed on activated T-cells upon TCR stimulation. 4-1BB signaling has been implicated in CD8 T-cell responses as 4-1BBL−/− mice had reduced memory CD8+ T-cells in response to viral challenge [303]. Resting Tregs preferentially, but not constitutively, express 4-1BB, and 41BB co-stimulation was shown to expand functionally suppressive peripheral Tregs in vitro and in vivo[304]. Similarly, artificial APCs expressing 4-1BBL were shown to expand human umbilical cord blood Tregs with suppressive functions [305]. In line with these reports, agonistic 4-1BB antibody ameliorated EAE by modulating Th17/Treg balance [306]. Administration of agonistic 4-1BB antibody expanded Tregs and ameliorated inflammatory bowel disease in BALB/c mice [307]. Studies have identified 4-1BB as a candidate gene with diabetes susceptibility loci in NOD mice, and 4-1BB+ Tregs were found to be reduced in NOD mice and were shown to be functionally more potent than 4-1BB Tregs [296]. Treatment with anti-4-1BB agonist increased Tregs and protected mice from diabetes. More importantly, adoptive transfer of Tregs from anti-4-1BB agonist treated mice delayed onset of diabetes in a congenic NOD. SCID mice[296]. In contrast, 4-1BB−/−NOD mice had delayed onset of diabetes, likely due to loss of its co-stimulatory effect on diabetogenic T-cells[295]. Reduced 4-1BB+ Tregs and increased soluble 4-1BB levels in plasma of MS patients were observed further indicating a possible role for 4-1BB signaling in modulating Treg functions during autoimmunity[308]. In contrast, other reports have shown negative regulation of Tregs by 4-1BB signaling[199] and activation of 4-1BB signaling in Teff cells renders them unresponsive to Treg mediated suppression[309]. 4-1BB signaling has been shown to increase anti-tumor immunity by inhibiting Treg induction through down-modulation of IL-9 production [310]. Thus, despite exhibiting similar effects on Treg and Teff cell expansion, activation of signaling through same or different members of the TNFR family may or may not protect, or exacerbate autoimmunity under different contexts. Though activation of OX40, GITR, and 4-1BB signaling might not be the primary inducer of autoimmunity, they can exacerbate ongoing autoimmunity depending upon the cell type, the activation state of the target cell and local inflammatory milieu. More effort is needed to fully understand the contextual function of these molecules before targeting them for therapy.

7. Role of Tregs and co-inhibitory receptors in tumor immune evasion, and checkpoint blockade in tumor therapy

A growing body of evidence suggests that immune suppressive functions of Tregs favor tumor progression by dampening anti-tumor immunity[311]. Increased frequencies of Tregs were observed in the peripheral blood and tumor tissues in lung[312], head and neck[313], pancreatic[314], gastric and esophageal[315], liver[316], breast[317] cancers and melanoma[318]. Though few studies have indicated that infiltration of Tregs is associated with better prognosis in colorectal carcinoma (CRC)[319], recent reports have clarified the issue by noting accumulation of FOXP3hi suppressive Tregs in case of poor prognosis and FOXP3low non-suppressive Tregs in cases with better prognosis[320]. Expression of co-inhibitory molecules such as CTLA4, PD-1, TIGIT, LAG3 and TIM3 by cancer tissues, tumor infiltrating T-cells and Tregs was identified as a key mechanism promoting tumor immune evasion[321]. Since Tregs preferentially overexpress these co-inhibitory receptors, they represent a putative target for immune checkpoint inhibitors that can reverse immune suppression. The effect of different checkpoint inhibitors on Teff/Treg balance and tumor progression is summarized in Table-5. Iplimumab (anti-CTLA4 Ab) was approved by Food and Drug Administration (FDA) in 2011 for the treatment of unresectable metastatic melanoma[322]. Pembrolizumab (anti-PD-1 Ab) was approved for treating advanced or unresectable melanoma in 2014 and is currently used for the treatment of metastatic Non-Small Cell Lung Cancer (NSCLC) with PDL1 expression and recurrent Squamous Cell Carcinoma of the Head and Neck (SCCHN). Nivolumab, another PD1 blocking antibody was approved in 2014 for treating unresectable and metastatic melanoma with progression even after ipilimumab treatment, NSCLC and metastatic renal Cell Carcinoma (RCC). Atezolizumab (anti-PD-L1) antibody was approved in 2015 for treating NSCLC, and in 2016 for treating Urothelial carcinoma [323]. In addition, combination of ipilimumab and nivolumab was approved in 2015 for treating unresectable metastatic melanoma[322]. Studies using IgG2a isotype anti-CTLA4 antibodies found that tumor-infiltrating Tregs were selectively depleted through an Fc-dependent mechanism involving Fcγ-receptor expressing macrophages within TME, leading to increased CD8+ Teff/Treg ratio at tumor sites with a concomitant increase in peripheral blood Tregs [324, 325]. PD1/PD-L1 pathway has been found to be critical for Treg generation[326] and thus contribute significantly to tumor immune evasion[327]. Stage III/IV melanoma patients who were vaccinated with a cocktail of peptides including the glycoprotein 100 (gp100) 209-217 (210M) (gp100-2M) and MART-1 26-35 (27L) (MART 27L) heteroclitic peptide analogs showed increased PD-L1+ Tregs and PD-1+ CD8+ T-cells in their circulation. Interestingly, blockade of PD1 signaling in these patients reduced Foxp3 expression in Tregs and helped overcome Treg mediated inhibition of CD8+T-cell proliferation and effector cytokine production [328]. Moreover, combined blockade of CTLA4 and PD1/PD-L1 along with Flt3 vaccine increased Teff/Treg ratio in B16 melanoma tumors and doubled the tumor rejection efficiency of monotherapies[329], [330].

Table-5:

Effect of checkpoint inhibitors and TNFR agonists on tumors and Treg/Teff cells

Target and Approach Cancer model Effect on tumor Effect on Tregs and Teff cells Reference
Anti-CTLA4- antibody Subcutaneous MC38 and CT26 colon adenocarcinoma models Potent anti-tumor response Reduced intra-tumoral Tregs [331]
Anti-CTLA4 antibody B16 melanoma Potent anti-tumor response Reduced intra-tumoral Tregs, but increased Tregs in peripheral lymph nodes. [332]
Anti-CTLA4 antibody Colon26 Strong anti-tumor response Reduced intra tumor CD4+/CD8+ and Treg cells. Increased CD8/Treg ratio [333]
Anti-PDl and anti-PD1 antibodies + vaccine CT26 colon carcinoma Reduced tumor size and volume Reduced intratumoral Tregs [334]
Anti-CTLA4+ Anti-PD-1antibodies with Flt3 vaccine B16 melanoma Doubled tumor rejection efficiency of CTLA4 and PD-1 monotherapy Increased Teff/Treg ratio in tumor [329]
Anti-TIM3 and anti-PD-1 antibodies MC38 and CT26 colon carcinoma, B16 melanoma Overcome CD8 T-cell exhaustion and reduced tumor progression Reduced expression of Treg suppressive molecules and reversed Treg induced anergy in Teff cells [99]
Anti-LAG3 and anti-PD-1 antibodies Ovarian cancer IE9mp1 line implanted mice Delayed tumor growth Reduced Treg frequency in tumors and increased proliferation of CD8+ effector T-cells [335]
Anti-TIGIT and anti-PD-L1 antibodies CT26 colon carcinoma mice Enhanced tumor rejection Increased CD8+ T-cell response with no difference in tumor- infiltrating Treg frequency [336]
Anti-TIGIT and anti-PD1 antibodies MC38 colon carcinoma and GL261 glioblastoma mice Tumor regression and enhanced survival Increased IFN-γ, TNF-α and IL-2 secretion from effector T cells. [337]
TNFR2 blocking antibody + CpG ODN (TLR9 agonist) CT26 colon carcinoma mice Inhibition of tumor growth and increased tumor-free survival Reduced proportion of tumor infiltration of Tregs and increased IFN-γ secreting CD8+ T-cells [338]
Agonistic anti-GITR antibody (DTA-1) B16 melanoma mice Increased anti-tumor response Reduced Treg stability (decreased Foxp3 expression) and intratumoral Treg infiltration. [339]
Agonistic anti-OX40 antibody (0X86) Colon26 mice Increased tumor regression Depletion of intratumoral Tregs through FcγR dependent mechanism. [340]
Anti-4-1BB +anti-CTLA4 + Flt3 vaccine B16 melanoma Increased tumor rejection Increased CD4/Treg and CD8/Treg ratio in tumors. [341]
Agonistic anti-4-1BB antibody A20 lymphoma Increased tumor regression than anti-CTLA4, anti-GITR and anti-OX40 antibodies and conferred protection to rechallenge CD4+Foxp3+ Tregs inhibited anti-tumor response of anti-4-1BB agonist while NK-cells and CD8+ T-cells mediated anti-tumor response [342]

Though CTLA4 and PD-1 blockade has improved therapeutic efficacy in several types of solid tumors and melanoma, still many patients are refractory to this approach. This lead to the search for other co-inhibitory molecules which might compensate for the loss of CTLA4/PD1 signaling and thus might favor tumor immune evasion. Upregulation of another co-inhibitory molecule TIM3 was noted in murine lung adenocarcinoma treated with anti-PD1 antibody[343]. Lung cancer patients had increased TIM3 expression in tumor-infiltrating CD4+ and CD8+ T-cells, but not in peripheral blood. The frequency of IFN-γ+ T-cells was reduced within CD8+TIM3+ cells compared to CD8+TIM3 cells. More importantly, 70% of tumor- infiltrating CD4+TIM3+ cells were found to be FOXP3+ Tregs and correlated with poor prognosis[344]. In preclinical tumor models, TIM3 was expressed at low levels on peripheral blood Tregs but highly expressed on tumor-infiltrating Tregs[345] and were predominantly PD1+ as well. TIM3+ Tregs accumulated in the tumor before exhaustion of CD8+ T-cells and co-blockade of PD1 and TIM3 yielded improved efficacy than PD1 blockade alone[99]. Currently, humanized anti-TIM3 antibody (TSR-022) is being tested in patients with advanced solid tumors[321].

LAG3 is expressed on tumor infiltrating lymphocytes (TILs), especially on Tregs. CD4+CD25hiFoxp3+LAG3+ Tregs from PBMCs, tumor invaded lymph nodes and visceral metastatic tissues of melanoma and CRC patients showed preferential proliferation and produced higher amounts of immunosuppressive cytokines such as IL-10 and TGF-β, and also exerted potent suppressive activities in contact-dependent assays[346]. More interestingly, Scurr et al. have characterized CD4+CD25+Foxp3 LAG3+LAP+ intra-tumor T-cell population from CRC patients which can produce IL-10 and TGF-β and show 50% more suppressive activity than Foxp3+Tregs[347]. Combined inhibition of LAG3 and PD-1 using blocking antibodies resulted in potent antitumor response accompanied by reduced tumor infiltration of CD4+CD25+Foxp3+ Tregs and increased CD8+ Teff cell functions[335]. Humanized LAG-3 (BMS-986016) antibody is under phase I and phase II clinical trials in patients with colorectal, cervical, ovarian, renal cell carcinoma, head and neck, and squamous small cell carcinoma, gastric and hepatocellular cancers. In addition, anti-LAG3 antibody (MK-4280) is currently being tested as either monotherapy or in combination with an anti-PD1 antibody against advanced solid tumors[321].

TIGIT is co-expressed with PD1 on tumor infiltrating CD8+ T-cells and Tregs[336]. Emerging preclinical studies shed light on the inhibitory role of TIGIT on anti-tumor immunity [336, 348]. TIGIT signaling has been shown to predominantly regulate anti-tumor immune responses via Treg mediated suppression. Intra-tumoral TIGIT+ Tregs were found to be highly suppressive and they expressed other co-inhibitory molecules such as Pdcd-1, Lag3, Ctla4, and TIM-3, and suppressor cytokines IL-10 and TGF-β compared to TIGIT Tregs. Adoptive transfer of CD8+ T cells from WT or Tigit−/− mice mixed with WT CD4+Foxp3+ and Foxp3 T cells into Rag2−/− mice implanted with B16F10 tumor cells showed a dominant role of TIGIT+Foxp3+ Tregs than TIGIT+CD8+ T-cells in tumor evasion. It was also noted that TIGIT synergized with TIM-3 in restraining anti-tumor responses [348]. In contrast, another study observed no significant difference in the frequencies of tumor-infiltrating Tregs upon TIGIT and PD-L1 blockade despite increased CD8+ Teff functions and tumor regression. TIGIT expression correlated with CD8+ T-cell exhaustion and TIGIT blockade increased effector cytokine (IFN-γ and TNF-α) production from CD8+ T-cells in a Treg-independent manner [336]. This discrepancy could be attributed to the differences in the mouse models and experimental approaches used in these two independent studies. However, combined inhibition of TIGIT and PD1 improved tumor antigen-specific CD8+ T-cell response in melanoma patients [349]. Thus, TIGIT blockade may promote anti-tumor immunity through both Treg dependent and independent mechanisms. Currently, anti-TIGIT antibodies BMS-986207 and MTIG7192A are in clinical trials against advanced solid tumors and advanced metastatic tumors either alone in combination with anti-PD1 and anti-PD-L1 antibodies[321].

8. TNFSF members in tumor immunotherapy

In addition to overcoming T-cell suppression by targeting co-inhibitory receptors of IgSF members, activation of Teff cell through by co-stimulation through TNFRs, with concomitant suppression of Tregs, has also been beneficial in tumor immunotherapy[29]. Many TNFRSF agonists were tested in the past for their ability to induce anti-tumor immunity owing to the crucial role in T-cell activation and proliferation. Here, we restrict our focus to TNFRII, GITR, OX40, and 4-1BB which could target both Teff and Treg cells. TNFRII was found to be preferentially overexpressed on several tumor tissues and tumor-infiltrating Tregs which were highly suppressive[350]. In contrast to TNF-α signaling through TNFRI which causes cell death, alternative signaling through TNFRII can induce cell proliferation [119]. Therefore, TNFR-II agonism was shown to be associated with the proliferation of tumor cells and tumor-infiltrating Tregs, which synergistically promoted tumor progression[350]. Therefore, the selective targeting of TNFRII using TNFRII antagonist is proposed to be beneficial in cancer immunotherapy[350]. In a recent study, the anti-TNFR2 antagonistic antibody has been shown to induce cell death in OVCAR3 cancer cells which overexpress TNFR2. Moreover, TNFR2 antagonists also promoted cell death of Tregs isolated from ovarian cancer ascites more effectively than Tregs from healthy donor samples [351]. In addition, TNFR2 blocking antibody in combination with TLR9 agonist CpG ODN reduced tumor-infiltrating Tregs and increased IFN-γ+CD8+T-cell infiltration leading to tumor regression and tumor-free survival[352].

4-1BB co-stimulation can promote cytotoxic CD8+ T-cell survival/activity and protect tumor infiltrating T-cells from activation-induced cell death [353]. Consistent with this, 4-1BB agonistic antibody increased cytotoxic activity of TILs and showed anti-tumor activity in preclinical models (Table-5). However, in human clinical trials, 4-1BB agonists produced significant liver toxicity and the trial was halted although it showed some anti-tumor effects[354]. Currently, lower doses of 4-1BB agonists are being tested in combination with other agents such as anti-LAG3 antibodies[355]. On the other hand, 4-1BB intracellular domain is used in engineering CAR-T-cells with B-cell antigen CD19 and CD3-domains to enhance T-cell survival and effector functions, and CART19 cells have shown disease remission for more than 10 months in a chronic lymphocytic leukemia patient[356].

OX40 is highly expressed on TILs, especially on Tregs, in many cancers including melanoma, colon cancer, head and neck cancer, breast cancer and B-cell lymphoma[357] and used as a marker for tumor antigen-specific Tregs[358]. The agonistic anti-OX40 antibody has been shown to induce an anti-tumor immune response in several preclinical tumor models including B16 melanoma[359], fibrosarcoma[360], CT26 colon cancer[361] and GL261 glioma[362]. Consistent with the effect of OX40 signaling in memory T-cell formation, OX40 mediated antitumor response was driven through CD4+ and CD8+ memory T-cell responses, and OX40 agonist treated mice were found resistant to tumor re-challenge[363]. In addition, the agonistic OX40 antibody has also been shown to deplete intra-tumoral Tregs which express higher levels of OX40 through FcγR mediated ADCC caused by myeloid and NK cells present within the TME [340] which resulted in altered Teff/Treg ratio in tumors. However, in clinical trials murine anti-human OX40 agonistic antibody showed increased tumor infiltration of Tregs despite increased Teff cell proliferation [364] which could be due to Treg proliferation induced by constitutively expressed OX40 upon OX40L binding. Currently, humanized anti-OX40 agonistic antibody is in a phase II clinical trial in combination with stereotactic radiation and/or cyclophosphamide for many cancer types[29].

Similar to an OX40 agonistic antibody, GITR agonist DTA-1 has been shown to promote anti-tumor responses in several syngeneic murine tumor models including B16 melanoma [365], CT26 colon cancer [366], and fibrosarcoma [367]. Moreover, combination therapy with GITR+OX40 agonists further enhanced anti-tumor response [366]. GITR agonist increased infiltration of large numbers of IFN-γ secreting CD4+ and CD8+ T-cells into regressing tumors. Similar to OX40 agonist, GITR agonist also caused increased infiltration of Foxp3+ Tregs into tumors [367] and induced Treg depletion through FcγR mediated ADCC [368]. However, in contrast to co-stimulatory functions of murine GITRL on T-cells, human GITRL expression undermines NK cell immune-surveillance of acute myeloid leukemia. Several human AML cell lines and AML patients were found to express GITRL and GITRL signaling induced the production of immunosuppressive IL-10[369]. Thus, OX40 and GITR agonists are dependent on FcγR expressing cell types in the TME for the ADCC. However, in an immunosuppressive tumor micro-environment lacking these FcγR expressing cell types, OX40 and GITR agonists may induce Treg proliferation as evidenced by increased Tregs seen in the tumor sites in some clinical trials [364, 367].

9. Immune-related adverse events (IRAE) resulting from checkpoint blockade

Although immune checkpoint blockade using anti-CTLA4/anti-PD1/anti-PD-L1 can produce long-lasting anti-tumor responses, it can also lead to many IRAEs and most of them are autoimmune disorders due to the critical role played by these co-inhibitory signaling molecules on immune tolerance[370]. These autoimmune adverse events span a wide spectrum of immune dysfunctions including vitiligo, mucosal inflammation, colitis, endocrine disorders such as hypothyroidism, hypophysitis (reduced levels of pituitary hormones), diabetes mellitus, liver and kidney dysfunctions, polyarthritis, pancreatitis and hematologic syndromes [371]. Almost 60% of patients receiving anti-CTLA4 therapy develop autoimmune adverse events with colitis being the most common [372]. Thyroiditis and pneumonitis[373] are more common in anti-PD1/PDL-1 treated patients[374]. There are reports suggesting that patients who are more responsive to checkpoint blockade suffer from severe autoimmune adverse events compared to unresponsive patients, indicating the efficacy-toxicity relationship [375, 376]. However, other studies have failed to show a correlation between IRAE and efficacy[377]. Non-specific immunomodulation through checkpoint blockade can shift the balance of Teff and Treg cells, and perturb immune homeostasis resulting in autoimmune adverse events. Anti-CTLA4 antibodies can promote antitumor immunity by overcoming the intrinsic suppressive effects of CTLA4 signaling on Teff cell activation and expansion, and its extrinsic effect of CD80/CD86 trans-endocytosis which is mainly mediated by Tregs. In addition, constitutive expression of CTLA4 by Tregs allows targeting these suppressor cells for ADCC mediated by Fcγ receptor expressing macrophages present in the TME [324]. Thus, anti-CTLA4 blockade can abolish Treg numbers/functions. By adoptively transferring OVA- specific CTLA4−/− Tregs and OVA-TCR transgenic T-cells into β-cell-specific OVA-peptide expressing transgenic Rag2−/− mice, it was shown that CTLA4 expression in Tregs is necessary to confer antigen-specific tolerance [378]. Similarly, CTLA4 blockade used in cancer treatment can lead to inhibition of Tregs including antigen-specific Tregs which are essential for maintaining peripheral tolerance, resulting in autoimmune adverse events. Unlike CTLA4 which is constitutively expressed on most Tregs, the PD1 expression is confined to a subset of Tregs. Interestingly, in breast cancer patients, PD-L1 expression in tumor cells and FOXP3 expression in tumor-infiltrating Tregs have been shown to mutually upregulate each other’s expression[379]. Thus, in addition to the direct co-inhibitory effect on Teff cell activation, PD1/PD-L1 signaling can also promote tumor evasion by inducing Treg mediated immune suppression. In line with these findings, PD-1 blockade has been shown to attenuate FOXP3 expression, Treg suppressive function and induce antigen-specific CD8+ T-cell proliferation in melanoma patients [328]. Earlier, PD-1 expression in Tregs was found to be correlated with MOG-antigen specific EAE development and PD-1−/− mice developed severe EAE even in the absence of adjuvant [237]. Besides, PD-L1 signaling has been shown to augment adaptive Treg differentiation and regulate Treg lineage stability [177]. Therefore, in addition to promoting anti-tumor immunity, PD1/PD-L1 blockade can also attenuate Treg function and cause loss of self-antigen specific tolerance resulting in autoimmune adverse events.

IRAEs are more prevalent in CTLA4 blockade than PD-1[371] which correlates with constitutive expression of CTLA4 on all Tregs compared to the PD1 expression on a subset of Tregs. Moreover, CTLA4 expression in Tregs is one of the key determinants of Treg function and their blockade might severely affect their functions than PD1 blockade could. This is consistent with severe autoimmunity and lymphoproliferative disorder observed in CTLA4−/− mice [169] compared to moderate autoimmunity seen in PD1−/− mice [380]. Supporting this notion, a report on thoracic melanoma patients treated with anti-CTLA4 developed severe pan-colitis with significantly reduced peripheral blood Tregs, increased IFN-γ+/IL-17+ CD4+ T-cells and granzyme-B+ CD8+ T-cells[381]. Preclinical mouse models also showed a correlation between IRAEs and altered Treg/Teff ratio upon checkpoint blockade[382]. Moreover, prolonged or transient depletion of Tregs in tumorbearing Foxp3-DTR mice with diphtheria toxin (DT) administration showed IRAEs similar to that of patients treated with anti-CTLA4/anti-PD1 antibodies [383]. In addition, transient depletion of Tregs in tumor-bearing mice, followed by treatment with anti-CTLA4/anti-PD1/anti-4-1BB exacerbated IRAEs and anti-4-1BB agonist was the most toxic [383]. On the other hand, targeting TIM-3, LAG3 and TIGIT pathways in combination with PD1 which are predominantly expressed on intra-tumoral Tregs, but not constitutively on Tregs, may have reduced autoimmune adverse events compared to CTLA-4 blockade [371, 383, 384]. However, their efficacy and safety are yet to be determined in larger clinical trials. Though 4-1BB, OX40, and GITR agonists promote effective anti-tumor responses, they have been implicated in the exacerbation of autoimmune diseases in mice that are pre-disposed to spontaneous autoimmunity and experimentally induced autoimmune disease models. FcγR-mediated depletion of Tregs using anti-OX40 /GITR antibodies and activation Teff cells not only promotes anti-tumor response, but can also lead to loss of peripheral tolerance to tissue antigens. The effect of checkpoint inhibitors and anti-OX40/GITR agonists on Teff/Treg compartments to modulate antitumor immunity and consequential autoimmune adverse events are summarized in Fig-3. Earlier, the 4-1BB clinical trial was halted for safety concern and checkpoint blockade with anti-CTLA4 and anti-PD1/PDL1 has already been demonstrated to induce fatal autoimmune diseases in some patients [33]. Therefore, further investigations are needed to fully understand the potential benefits and undesirable side effects associated with the combination of anti-CTLA4/anti-PD-1/anti-PD-L1 with anti-GITR, anti-OX40 and anti-4-1BB antibodies for tumor immunotherapy. Moreover, relevant clinical studies using a larger cohort are required to draw definite conclusions on how Tregs are related to autoimmune complications arising from checkpoint blockade. Development of relevant animal models of IRAE might help to study the possible consequences of checkpoint blockade on immune homeostasis and identify the molecular leads that can be targeted to fine tune the balance to restore immune homeostasis while minimizing autoimmune adverse events.

Figure-3: Mechanism of autoimmune adverse events in tumor immunotherapy.

Figure-3:

Anti-CTLA4 antibody induces Fc-mediated ADCC killing of tumor infiltrating Tregs upon binding to FcγR expressing tumor associated macrophages (TAM ) present in the tumor micro-environement (left panel). Blockade of PD1/PD-L1 signaling in Tregs lead to reduced Foxp3 expression and iTreg generation (middle panel). Loss of intrinsic CTLA4/PD-1 co-inhibitory signalling in Teff cells (red arrow) as well as Treg mediated suppression can lead to enhanced Teff cell proliferation and function resulting in increased anti-tumor immune response. Similarly, anti-GITR/anti-OX40 antibodies can promote Fc-mediated ADCC killing of tumor infiltrating Tregs(right panel) while inducing Teff cell proliferation via co-stimulatory signaling (red arrow). However, nonspecific inhibition of CTLA4/PD1 signaling and activation of OX40/GITR in peripheral tissues other than tumor sites can lead to loss of self-antigen specific Tregs and expansion of self-reactive Teff cells resulting in autoimmune adverse events.

10. Conclusion

CTLA4 is constitutively expressed on Tregs and activated Teff cells and is indispensable for maintaining immune tolerance. Thus, blockade of this pathway for cancer treatment might not only overcome tumor-specific immune suppression but could also break physiological self-tolerance to cause autoimmunity. On the other hand, PD-1//PD-L1 are not constitutively expressed on Tregs, however, they play an essential role in tissue tolerance as noted by their expression on many non-immune cell types and the development of spontaneous autoimmunity in PD-1−/− mice[58]. Second line co-inhibitory receptors such as LAG3, TIM3, and TIGIT seem to offer an advantage as they are preferentially overexpressed on tumor-infiltrating Tregs and mice deficient in these co-inhibitory molecules do not develop spontaneous autoimmunity. However, more extensive studies are needed to determine their safety and efficacy. Though TNFSF members are potent stimulators of Teff cell response, their context-dependent dual role on Tregs should also be considered for effective targeting. Despite playing a secondary role to CD28 and CD40 signaling in the induction of autoimmunity, activation of 4-1BB, GITR, and OX40 signaling can aggravate ongoing autoimmune response. Therefore, pre-existence of IRAEs should be tested before using combinations of OX40, GITR and 4-1BB agonists with anti-CTLA4/PD1 blockade which might worsen existing autoimmune reactions. Moreover, murine and human immune systems exhibit several striking differences [186, 385] and relevant humanized mouse models must be developed to study human tumors and to ensure successful clinical translation. Currently, more suitable animal models are being developed to study the mechanism of pathogenesis of autoimmune adverse events arising from checkpoint blockade[382]. These animal models may help understand the molecular mechanisms affecting immune homeostasis upon ICB that can be potentially corrected to prevent IRAEs.

Highlights.

  1. The role Immunoglobulin and TNF superfamily co-stimulatory and co-inhibitory molecules on regulatory T-cell homeostasis are critically reviewed.

  2. Effect of loss and gain of co-signaling receptors on regulatory T-cells and its correlation to various autoimmune diseases is explained.

  3. The role of co-inhibitory receptors and regulatory T-cells on tumor evasion and use of immune checkpoint inhibitors are discussed.

  4. Association between immune checkpoint point blockade and resulting autoimmune adverse events are reviewed.

Acknowledgments

We thank National Institutes of Health for the grant R01 AI107516-01A1 and Juvenile Diabetes Research Foundation (JDRF) for the grant 2-SRA-2016-245-S-B to Dr. Prabhakar.

Footnotes

Disclosure of conflict of interest

The authors have no competing financial interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Schwartz RH. Acquisition of immunologic self-tolerance. Cell, 1989;57:1073–81. [DOI] [PubMed] [Google Scholar]
  • [2].Goodnow CC, Sprent J, Fazekas de St Groth B, Vinuesa CG. Cellular and genetic mechanisms of self tolerance and autoimmunity. Nature, 2005;435:590–7. [DOI] [PubMed] [Google Scholar]
  • [3].Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest, 2007;117:1137–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Jansa P. [A new hypothesis on the immune surveillance of tumors]. Acta Univ Palacki Olomuc Fac Med, 1984;107:139–42. [PubMed] [Google Scholar]
  • [5].Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T Cells: Mechanisms of Differentiation and Function. Annual Review of Immunology, Vol 30, 2012;30:531–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Jordan MS. Thymic selection of CD4(+)CD25(+) regulatory T cells induced by an agonist self-peptide (vol 2, pg 301, 2001). Nat Immunol, 2001;2:468-. [DOI] [PubMed] [Google Scholar]
  • [7].Lee HM, Bautista JL, Hsieh CS. Thymic and peripheral differentiation of regulatory T cells. Adv Immunol, 2011;112:25–71. [DOI] [PubMed] [Google Scholar]
  • [8].Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol, 2002;2:251–62. [DOI] [PubMed] [Google Scholar]
  • [9].Bai XF, Gao JX, Liu J, Wen J, Zheng P, Liu Y. On the site and mode of antigen presentation for the initiation of clonal expansion of CD8 T cells specific for a natural tumor antigen. Cancer Res, 2001;61:6860–7. [PubMed] [Google Scholar]
  • [10].Hirahara K, Nakayama T. CD4+ T-cell subsets in inflammatory diseases: beyond the Th1/Th2 paradigm. Int Immunol, 2016;28:163–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Dornmair K, Goebels N, Weltzien HU, Wekerle H, Hohlfeld R. T-cell-mediated autoimmunity: novel techniques to characterize autoreactive T-cell receptors. Am J Pathol, 2003;163:1215–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Danikowski KM, Jayaraman S, Prabhakar BS. Regulatory T cells in multiple sclerosis and myasthenia gravis. J Neuroinflammation, 2017;14:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Korn T, Reddy J, Gao W, Bettelli E, Awasthi A, Petersen TR et al. Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation. Nat Med, 2007;13:423–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol, 2006;6:295–307. [DOI] [PubMed] [Google Scholar]
  • [15].Hoffmann GW. A theory of regulation and self-nonself discrimination in an immune network. Eur J Immunol, 1975;5:638–47. [DOI] [PubMed] [Google Scholar]
  • [16].Bretscher P, Cohn M. A theory of self-nonself discrimination. Science, 1970;169:1042–9. [DOI] [PubMed] [Google Scholar]
  • [17].Mueller DL, Jenkins MK, Schwartz RH. Clonal expansion versus functional clonal inactivation: a costimulatory signalling pathway determines the outcome of T cell antigen receptor occupancy. Annu Rev Immunol, 1989;7:445–80. [DOI] [PubMed] [Google Scholar]
  • [18].Lafferty KJ, Cunningham AJ. A new analysis of allogeneic interactions. Aust J Exp Biol Med Sci, 1975;53:27–42. [DOI] [PubMed] [Google Scholar]
  • [19].Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol, 2013;13:227–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Wajant H. Principles of antibody-mediated TNF receptor activation. Cell Death Differ, 2015;22:1727–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Zhang Q, Vignali DA. Co-stimulatory and Co-inhibitory Pathways in Autoimmunity. Immunity, 2016;44:1034–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Bour-Jordan H, Bluestone JA. Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev, 2009;229:41–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity, 2016;44:989–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of Treg-mediated T cell suppression. Frontiers in Immunology, 2012;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med, 2014;211:781–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Belai EB, de Oliveira CE, Gasparoto TH, Ramos RN, Torres SA, Garlet GP et al. PD-1 blockage delays murine squamous cell carcinoma development. Carcinogenesis, 2014;35:424–31. [DOI] [PubMed] [Google Scholar]
  • [27].Butt AQ, Mills KHG. Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines. Oncogene, 2014;33:4623–31. [DOI] [PubMed] [Google Scholar]
  • [28].Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. P Natl Acad Sci USA, 2010;107:4275–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Schaer DA, Hirschhorn-Cymerman D, Wolchok JD. Targeting tumor-necrosis factor receptor pathways for tumor immunotherapy. J Immunother Cancer, 2014;2:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A et al. B7/CD28 costimulation is essential for the homeostasis of the CD4(+)CD25(+) immunoregulatory T cells that control autoimmune diabetes. Immunity, 2000;12:431–40. [DOI] [PubMed] [Google Scholar]
  • [31].Mahmud SA, Manlove LS, Schmitz HM, Xing Y, Wang Y, Owen DL et al. Costimulation via the tumor-necrosis factor receptor superfamily couples TCR signal strength to the thymic differentiation of regulatory T cells. Nat Immunol, 2014;15:473–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Kahler KC, Hassel JC, Heinzerling L, Loquai C, Moessner R, Ugurel S et al. Management of side effects of immune checkpoint blockade by anti-CTLA-4 and anti-PD-1 antibodies in metastatic melanoma. Journal Der Deutschen Dermatologischen Gesellschaft, 2016;14:662–81. [DOI] [PubMed] [Google Scholar]
  • [33].Michot JM, Bigenwald C, Champiat S, Collins M, Carbonnel F, Postel-Vinay S et al. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. European Journal of Cancer, 2016;54:139–48. [DOI] [PubMed] [Google Scholar]
  • [34].June CH, Ledbetter JA, Gillespie MM, Lindsten T, Thompson CB. T-Cell Proliferation Involving the Cd28 Pathway Is Associated with Cyclosporine-Resistant Interleukin-2 Gene-Expression. Mol Cell Biol, 1987;7:4472–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Linsley PS, Clark EA, Ledbetter JA. T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1. Proc Natl Acad Sci U S A, 1990;87:5031–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Gimmi CD, Freeman GJ, Gribben JG, Sugita K, Freedman AS, Morimoto C et al. B-Cell Surface Antigen-B7 Provides a Costimulatory Signal That Induces T-Cells to Proliferate and Secrete Interleukin-2. P Natl Acad Sci USA, 1991;88:6575–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Freeman GJ, Gribben JG, Boussiotis VA, Ng JW, Restivo VA, Lombard LA et al. Cloning of B7-2 - a Ctla-4 Counter-Receptor That Costimulates Human T-Cell Proliferation. Science, 1993;262:909–11. [DOI] [PubMed] [Google Scholar]
  • [38].Brunet JF, Denizot F, Luciani MF, Rouxdosseto M, Suzan M, Mattei MG et al. A New Member of the Immunoglobulin Superfamily - Ctla-4. Nature, 1987;328:267–70. [DOI] [PubMed] [Google Scholar]
  • [39].Harper K, Balzano C, Rouvier E, Mattei MG, Luciani MF, Golstein P. Ctla-4 and Cd28 Activated Lymphocyte Molecules Are Closely Related in Both Mouse and Human as to Sequence, Message Expression, Gene Structure, and Chromosomal Location. J Immunol, 1991;147:1037–44. [PubMed] [Google Scholar]
  • [40].Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM et al. Ctla-4 Can Function as a Negative Regulator of T-Cell Activation. Immunity, 1994;1:405–13. [DOI] [PubMed] [Google Scholar]
  • [41].Krummel MF, Allison JP. Cd28 and Ctla-4 Have Opposing Effects on the Response of T-Cells to Stimulation. Journal of Experimental Medicine, 1995;182:459–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Teft WA, Kirchhof MG, Madrenas J. A molecular perspective of CTLA-4 function. Annu Rev Immunol, 2006;24:65–97. [DOI] [PubMed] [Google Scholar]
  • [43].Walker LS, Sansom DM. Confusing signals: recent progress in CTLA-4 biology. Trends Immunol, 2015;36:63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev, 2009;229:12–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol, 2002;2:116–26. [DOI] [PubMed] [Google Scholar]
  • [46].Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science, 2011;332:600–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Schneider H, Rudd CE. Diverse mechanisms regulate the surface expression of immunotherapeutic target ctla-4. Front Immunol, 2014;5:619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Rudd CE, Schneider H. Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling. Nat Rev Immunol, 2003;3:544–56. [DOI] [PubMed] [Google Scholar]
  • [49].Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, Anagnostopoulos I et al. ICOS is an inducible T-cell costimulator structurally and functionally related to CD28. Nature, 1999;397:263–6. [DOI] [PubMed] [Google Scholar]
  • [50].Mages HW, Hutloff A, Heuck C, Buchner K, Himmelbauer H, Oliveri F et al. Molecular cloning and characterization of murine ICOS and identification of B7h as ICOS ligand. European Journal of Immunology, 2000;30:1040–7. [DOI] [PubMed] [Google Scholar]
  • [51].McAdam AJ, Chang TT, Lumelsky AE, Greenfield EA, Boussiotis VA, Duke-Cohan JS et al. Mouse inducible costimulatory molecule (ICOS) expression is enhanced by CD28 costimulation and regulates differentiation of CD4(+) T cells. J Immunol, 2000;165:5035–40. [DOI] [PubMed] [Google Scholar]
  • [52].Ling V, Wu PW, Finnerty HF, Bean KM, Spaulding V, Fouser LA et al. Cutting edge: Identification of GL50, a novel B7-like protein that functionally binds to ICOS receptor. J Immunol, 2000;164:1653–7. [DOI] [PubMed] [Google Scholar]
  • [53].Yao S, Zhu YW, Zhu GF, Augustine M, Zheng LH, Goode DJ et al. B7-H2 Is a Costimulatory Ligand for CD28 in Human. Immunity, 2011;34:729–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. P Natl Acad Sci USA, 2002;99:12293–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol, 2013;14:1212–8. [DOI] [PubMed] [Google Scholar]
  • [56].Freeman GJ, Long AJ, Iwai Y, Latchman Y, Bourque K, Brown JA et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7-family member leads to negative regulation of lymphocyte activation. Blood, 2000;96:810a–1a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Latchman Y, Wood C, Chemova T, Iwai Y, Malenkovich N, Long A et al. PD-L2, a novel B7 homologue, is a second ligand for PD-1 and inhibits T cell activation. Faseb J, 2001;15:A345–A. [Google Scholar]
  • [58].Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev, 2010;236:219–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. P Natl Acad Sci USA, 2001;98:13866–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. Febs Lett, 2004;574:37–41. [DOI] [PubMed] [Google Scholar]
  • [61].Patsoukis N, Li LQ, Sari D, Petkova V, Boussiotis VA. PD-1 Increases PTEN Phosphatase Activity While Decreasing PTEN Protein Stability by Inhibiting Casein Kinase 2. Mol Cell Biol, 2013;33:3091–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3 zeta signalosome and downstream signaling to PKC theta. Febs Lett, 2004;574:37–41. [DOI] [PubMed] [Google Scholar]
  • [63].Karwacz K, Bricogne C, MacDonald D, Arce F, Bennett CL, Collins M et al. PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8(+) T cells. Embo Mol Med, 2011;3:581–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Watanabe N, Gavrieli M, Sedy JR, Yang JF, Fallarino F, Loftin SK et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat Immunol, 2003;4:670–9. [DOI] [PubMed] [Google Scholar]
  • [65].Cheung TC, Humphreys IR, Potter KG, Norris PS, Shumway HM, Tran BR et al. Evolutionarily divergent herpesviruses modulate T cell activation by targeting the herpesvirus entry mediator cosignaling pathway. Proc Natl Acad Sci U S A, 2005;102:13218–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Steinberg MW, Huang YJ, Wang-Zhu Y, Ware CF, Cheroutre H, Kronenberg M. BTLA Interaction with HVEM Expressed on CD8(+) T Cells Promotes Survival and Memory Generation in Response to a Bacterial Infection. Plos One, 2013;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Kimzey SL, Liu P, Green JM. Requirement for CD28 in the effector phase of allergic airway inflammation. J Immunol, 2004;173:632–40. [DOI] [PubMed] [Google Scholar]
  • [68].Simpson TR, Quezada SA, Allison JP. Regulation of CD4 T cell activation and effector function by inducible costimulator (ICOS). Curr Opin Immunol, 2010;22:326–32. [DOI] [PubMed] [Google Scholar]
  • [69].Chan CJ, Andrews DM, Smyth MJ. Receptors that interact with nectin and nectin-like proteins in the immunosurveillance and immunotherapy of cancer. Current Opinion in Immunology, 2012;24:246–51. [DOI] [PubMed] [Google Scholar]
  • [70].Meinl E, Lengenfelder D, Blank N, Pirzer R, Barata L, Hivroz C. Differential requirement of ZAP-70 for CD2-mediated activation pathways of mature human T cells. J Immunol, 2000;165:3578–83. [DOI] [PubMed] [Google Scholar]
  • [71].Cannons JL, Tangye SG, Schwartzberg PL. SLAM family receptors and SAP adaptors in immunity. Annu Rev Immunol, 2011;29:665–705. [DOI] [PubMed] [Google Scholar]
  • [72].Baitsch L, Legat A, Barba L, Marraco SAF, Rivals JP, Baumgaertner P et al. Extended Co-Expression of Inhibitory Receptors by Human CD8 T-Cells Depending on Differentiation, Antigen-Specificity and Anatomical Localization. Plos One, 2012; 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Su EW, Lin JY, Kane LP. TIM-1 and TIM-3 proteins in immune regulation. Cytokine, 2008;44:9–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Xiao S, Brooks CR, Sobel RA, Kuchroo VK. Tim-1 Is Essential for Induction and Maintenance of IL-10 in Regulatory B Cells and Their Regulation of Tissue Inflammation. J Immunol, 2015;194:1602–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Kane LP. T cell Ig and mucin domain proteins and immunity (vol 184, pg 2743, 2010). J Immunol, 2012; 189:4695-. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Pauken KE, Wherry EJ. TIGIT and CD226: Tipping the Balance between Costimulatory and Coinhibitory Molecules to Augment the Cancer Immunotherapy Toolkit. Cancer Cell, 2014;26:785–7. [DOI] [PubMed] [Google Scholar]
  • [77].Lozano E, Dominguez-Villar M, Kuchroo V, Hafler DA. The TIGIT/CD226 axis regulates human T cell function. J Immunol, 2012;188:3869–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Liu J, Qian X, Chen Z, Xu X, Gao F, Zhang S et al. Crystal structure of cell adhesion molecule nectin-2/CD112 and its binding to immune receptor DNAM-1/CD226. J Immunol, 2012;188:5511–20. [DOI] [PubMed] [Google Scholar]
  • [79].Blake SJ, Dougall WC, Miles JJ, Teng MW, Smyth MJ. Molecular Pathways: Targeting CD96 and TIGIT for Cancer Immunotherapy. Clin Cancer Res, 2016;22:5183–8. [DOI] [PubMed] [Google Scholar]
  • [80].Joller N, Hafler JP, Brynedal B, Kassam N, Spoerl S, Levin SD et al. Cutting Edge: TIGIT Has T Cell-Intrinsic Inhibitory Functions. J Immunol, 2011;186:1338–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Joller N, Lozano E, Burkett PR, Patel B, Xiao S, Zhu C et al. Treg Cells Expressing the Coinhibitory Molecule TIGIT Selectively Inhibit Proinflammatory Th1 and Th17 Cell Responses. Immunity, 2014;40:569–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Boles KS, Vermi W, Facchetti F, Fuchs A, Wilson TJ, Diacovo TG et al. A novel molecular interaction for the adhesion of follicular CD4 T cells to follicular DC. European Journal of Immunology, 2009;39:695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. P Natl Acad Sci USA, 2009;106:17858–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Casado JG, Pawelec G, Morgado S, Sanchez-Correa B, Delgado E, Gayoso I et al. Expression of adhesion molecules and ligands for activating and costimulatory receptors involved in cell-mediated cytotoxicity in a large panel of human melanoma cell lines. Cancer Immunol Immun, 2009;58:1517–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Yu X, Harden K, Gonzalez LC, Francesco M, Chiang E, Irving B et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol, 2009;10:48–57. [DOI] [PubMed] [Google Scholar]
  • [86].Fuchs A, Cella M, Giurisato E, Shaw AS, Colonna M. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J Immunol, 2004;172:3994–8. [DOI] [PubMed] [Google Scholar]
  • [87].Zhu YW, Paniccia A, Schulick AC, Chen W, Koenig MR, Byers JT et al. Identification of CD112R as a novel checkpoint for human T cells. J Exp Med, 2016;213:167–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Mariat C, Degauque N, Balasubramanian S, Kenny J, DeKruyff RH, Umetsu DT et al. Tim-1 signaling substitutes for conventional signal 1 and requires costimulation to induce T cell proliferation. J Immunol, 2009;182:1379–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Rodriguez-Manzanet R, Meyers JH, Balasubramanian S, Slavik J, Kassam N, Dardalhon V et al. TIM-4 expressed on APCs induces T cell expansion and survival. J Immunol, 2008;180:4706–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature, 2002;415:536–41. [DOI] [PubMed] [Google Scholar]
  • [91].Chen TT, Li L, Chung DH, Allen CDC, Torti SV, Torti FM et al. TIM-2 is expressed on B cells and in liver and kidney and is a receptor for H-ferritin endocytosis. J Exp Med, 2005;202:955–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Balasubramanian S, Kota SK, Kuchroo VK, Humphreys BD, Strom TB. TIM Family Proteins Promote the Lysosomal Degradation of the Nuclear Receptor NUR77. Science Signaling, 2012;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Kaplan G, Totsuka A, Thompson P, Akatsuka T, Moritsugu Y, Feinstone SM. Identification of a surface glycoprotein on African green monkey kidney cells as a receptor for hepatitis A virus. Embo Journal, 1996;15:4282–96. [PMC free article] [PubMed] [Google Scholar]
  • [94].Mizui M, Shikina T, Arase H, Suzuki K, Yasui T, Rennert PD et al. Bimodal regulation of T cell-mediated immune responses by TIM-4. Int Immunol, 2008;20:695–708. [DOI] [PubMed] [Google Scholar]
  • [95].Xiao L, Fu ZR, Liu F, Zhang LD, Shi XM, Shen XY et al. Suppression of Allograft Rejection by Tim-1-Fc through Cross-Linking with a Novel Tim-1 Binding Partner on T Cells. Plos One, 2011;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Cao W, Ryan M, Buckley D, O'Connor R, Clarkson MR. Tim-4 inhibition of T-cell activation and T helper type 17 differentiation requires both the immunoglobulin V and mucin domains and occurs via the mitogen-activated protein kinase pathway. Immunology, 2011;133:179–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol, 2005;6:1245–52. [DOI] [PubMed] [Google Scholar]
  • [98].Chiba S, Baghdadi M, Akiba H, Yoshiyama H, Kinoshita I, Dosaka-Akita H et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat Immunol, 2012;13:832–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Sakuishi K, Ngiow SF, Sullivan JM, Teng MW, Kuchroo VK, Smyth MJ et al. TIM3(+)FOXP3(+) regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. Oncoimmunology, 2013;2:e23849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev, 2017;276:97–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Veillette A. SLAM-family receptors: immune regulators with or without SAP-family adaptors. Cold Spring Harb Perspect Biol, 2010;2:a002469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Chattopadhyay K, Lazar-Molnar E, Yan Q, Rubinstein R, Zhan C, Vigdorovich V et al. Sequence, structure, function, immunity: structural genomics of costimulation. Immunol Rev, 2009;229:356–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Brown MH, Boles K, van der Merwe PA, Kumar V, Mathew PA, Barclay AN. 2B4, the natural killer and T cell immunoglobulin superfamily surface protein, is a ligand for CD48. J Exp Med, 1998;188:2083–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Latchman Y, McKay PF, Reiser H. Cutting edge: Identification of the 2B4 molecule as a counter-receptor for CD48. J Immunol, 1998;161:5809–12. [PubMed] [Google Scholar]
  • [105].Bierer BE, Peterson A, Gorga JC, Herrmann SH, Burakoff SJ. Synergistic T cell activation via the physiological ligands for CD2 and the T cell receptor. J Exp Med, 1988;168:1145–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Kageyama R, Cannons JL, Zhao F, Yusuf I, Lao C, Locci M et al. The Receptor Ly108 Functions as a SAP Adaptor- Dependent On-Off Switch for T Cell Help to B Cells and NKT Cell Development. Immunity, 2012;36:986–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Bengsch B, Seigel B, Ruhl M, Timm J, Kuntz M, Blum HE et al. Coexpression of PD-1, 2B4, CD160 and KLRG1 on exhausted HCV-specific CD8+ T cells is linked to antigen recognition and T cell differentiation. PLoS Pathog, 2010;6:e1000947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Workman CJ, Vignali DA. Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223). J Immunol, 2005;174:688–95. [DOI] [PubMed] [Google Scholar]
  • [109].Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol, 1995;25:2718–21. [DOI] [PubMed] [Google Scholar]
  • [110].Xu F, Liu J, Liu D, Liu B, Wang M, Hu Z et al. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res, 2014;74:3418–28. [DOI] [PubMed] [Google Scholar]
  • [111].del Rio ML, Lucas CL, Buhler L, Rayat G, Rodriguez-Barbosa JI. HVEM/LIGHT/BTLA/CD160 cosignaling pathways as targets for immune regulation. J Leukocyte Biol, 2010;87:223–35. [DOI] [PubMed] [Google Scholar]
  • [112].Le Bouteiller P, Tabiasco J, Polgar B, Kozma N, Giustiniani J, Siewiera J et al. CD160: A unique activating NK cell receptor. Immunol Lett, 2011;138:93–6. [DOI] [PubMed] [Google Scholar]
  • [113].Ware CF. The TNF superfamily. Cytokine Growth Factor Rev, 2003;14:181–4. [DOI] [PubMed] [Google Scholar]
  • [114].Vanamee ES, Faustman DL. Structural principles of tumor necrosis factor superfamily signaling. Sci Signal, 2018;11. [DOI] [PubMed] [Google Scholar]
  • [115].Bodmer JL, Schneider P, Tschopp J. The molecular architecture of the TNF superfamily. Trends in Biochemical Sciences, 2002;27:19–26. [DOI] [PubMed] [Google Scholar]
  • [116].Etemadi N, Holien JK, Chau D, Dewson G, Murphy JM, Alexander WS et al. Lymphotoxin alpha induces apoptosis, necroptosis and inflammatory signals with the same potency as tumour necrosis factor. Febs Journal, 2013;280:5283–97. [DOI] [PubMed] [Google Scholar]
  • [117].Faustman DL, Davis M. TNF Receptor 2 and Disease: Autoimmunity and Regenerative Medicine. Front Immunol, 2013;4:478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].McKarns SC, Miller PG. Transmembrane TNF-TNFR2 Antagonizes T(H)17 Differentiation by Promoting II2 Expression. J Immunol, 2016;196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Ward-Kavanagh LK, Lin WW, Sedy JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity, 2016;44:1005–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Ephrem A, Epstein AL, Stephens GL, Thornton AM, Glass D, Shevach EM. Modulation of Treg cells/T effector function by GITR signaling is context-dependent. European Journal of Immunology, 2013;43:2421–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev, 2009;229:173–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Wang C, Lin GH, McPherson AJ, Watts TH. Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol Rev, 2009;229:192–215. [DOI] [PubMed] [Google Scholar]
  • [123].Hendriks J, Xiao YL, Rossen JWA, van der Sluijs KF, Sugamura K, Ishii N et al. During viral infection of the respiratory tract, CD27, 4-1BB, and OX40 collectively determine formation of CD8(+) memory T cells and their capacity for secondary expansion. J Immunol, 2005;175:1665–76. [DOI] [PubMed] [Google Scholar]
  • [124].Denoeud J, Moser M. Role of CD27/CD70 pathway of activation in immunity and tolerance. J Leukocyte Biol, 2011;89:195–203. [DOI] [PubMed] [Google Scholar]
  • [125].Croft M. Control of Immunity by the TNFR-Related Molecule OX40 (CD134). Annu Rev Immunol, 2010;28:57–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Zeiser R, Nguyen VH, Hou JZ, Beilhack A, Zambricki E, Buess M et al. Early CD30 signaling is critical for adoptively transferred CD4(+)CD25(+) regulatory T cells in prevention of acute graft-versus-host disease. Blood, 2007;109:2225–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Horwitz DA, Pan S, Ou JN, Wang J, Chen M, Gray JD et al. Therapeutic polyclonal human CD8+ CD25+ Fox3+ TNFR2+ PD-L1+ regulatory cells induced ex-vivo. Clin Immunol, 2013;149:450–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Kanamaru F, Youngnak P, Hashiguchi M, Nishioka T, Takahashi T, Sakaguchi S et al. Costimulation via glucocorticoid-induced TNF receptor in both conventional and CD25(+) regulatory CD4(+) T cells. J Immunol, 2004;172:7306–14. [DOI] [PubMed] [Google Scholar]
  • [129].Clouthier DL, Zhou AC, Watts TH. Anti-GITR agonist therapy intrinsically enhances CD8 T cell responses to chronic lymphocytic choriomeningitis virus (LCMV), thereby circumventing LCMV-induced downregulation of costimulatory GITR ligand on APC. J Immunol, 2014;193:5033–43. [DOI] [PubMed] [Google Scholar]
  • [130].Oh HS, Choi BK, Kim YH, Lee DG, Hwang S, Lee MJ et al. 4-1BB Signaling Enhances Primary and Secondary Population Expansion of CD8+ T Cells by Maximizing Autocrine IL-2/IL-2 Receptor Signaling. Plos One, 2015;10:e0126765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Bartkowiak T, Curran MA. 4-1BB Agonists: Multi-Potent Potentiators of Tumor Immunity. Front Oncol, 2015; 5:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Wortzman ME, Clouthier DL, McPherson AJ, Lin GH, Watts TH. The contextual role of TNFR family members in CD8(+) T-cell control of viral infections. Immunol Rev, 2013;255:125–48. [DOI] [PubMed] [Google Scholar]
  • [133].Bhattacharya P, Gopisetty A, Ganesh BB, Sheng JR, Prabhakar BS. GM-CSF-induced, bone-marrow-derived dendritic cells can expand natural Tregs and induce adaptive Tregs by different mechanisms. J Leukoc Biol, 2011;89:235–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Gilfillan MC, Noel PJ, Podack ER, Reiner SL, Thompson CB. Expression of the costimulatory receptor CD30 is regulated by both CD28 and cytokines. J Immunol, 1998;160:2180–7. [PubMed] [Google Scholar]
  • [135].Muta H, Boise LH, Fang L, Podack ER. CD30 signals integrate expression of cytotoxic effector molecules, lymphocyte trafficking signals, and signals for proliferation and apoptosis. J Immunol, 2000;165:5105–11. [DOI] [PubMed] [Google Scholar]
  • [136].van de Ven K, Borst J. Targeting the T-cell co-stimulatory CD27/CD70 pathway in cancer immunotherapy: rationale and potential. Immunotherapy, 2015;7:655–67. [DOI] [PubMed] [Google Scholar]
  • [137].Coquet JM, Ribot JC, Babala N, Middendorp S, van der Horst G, Xiao Y et al. Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27-CD70 pathway. J Exp Med, 2013;210:715–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Hendriks J, Gravestein LA, Tesselaar K, van Lier RAW, Schumacher TNM, Borst J. CD27 is required for generation and long-term maintenance of T cell immunity. Nat Immunol, 2000;1:433–40. [DOI] [PubMed] [Google Scholar]
  • [139].Mauri DN, Ebner R, Montgomery RI, Kochel KD, Cheung TC, Yu GL et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin alpha are ligands for herpesvirus entry mediator. Immunity, 1998;8:21–30. [DOI] [PubMed] [Google Scholar]
  • [140].Mortarini R, Scarito A, Nonaka D, Zanon M, Bersani I, Montaldi E et al. Constitutive expression and costimulatory function of LIGHT/TNFSF14 on human melanoma cells and melanoma-derived microvesicles. Cancer Res, 2005;65:3428–36. [DOI] [PubMed] [Google Scholar]
  • [141].Cheung TC, Oborne LM, Steinberg MW, Macauley MG, Fukuyama S, Sanjo H et al. T Cell Intrinsic Heterodimeric Complexes between HVEM and BTLA Determine Receptivity to the Surrounding Microenvironment. J Immunol, 2009;183:7286–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].del Rio ML, Fernandez-Renedo C, Scheu S, Pfeffer K, Shintani Y, Kronenberg M et al. Therapeutic Blockade of LIGHT Interaction With Herpesvirus Entry Mediator and Lymphotoxin beta Receptor Attenuates In Vivo Cytotoxic Allogeneic Responses. Transplantation, 2014;98:1165–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Granger SW, Butrovich KD, Houshmand P, Edwards WR, Ware CF. Genomic characterization of LIGHT reveals linkage to an immune response locus on chromosome 19p13.3 and distinct isoforms generated by alternate splicing or proteolysis. Journal of Leukocyte Biology, 2001:27-. [DOI] [PubMed] [Google Scholar]
  • [144].Yu KY, Kwon B, Ni J, Zhai YF, Ebner R, Kwon BS. A newly identified member of tumor necrosis factor receptor superfamily (TR6) suppresses LIGHT-mediated apoptosis. Journal of Biological Chemistry, 1999;274:13733–6. [DOI] [PubMed] [Google Scholar]
  • [145].Asseman C, Mauze S, Leach MW, Coffman RL, Powrie F. An essential role for interleukin 10 in the function of regulatory T cells that inhibit intestinal inflammation. J Exp Med, 1999;190:995–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Asseman C, Read S, Powrie F. Colitogenic Th1 cells are present in the antigen-experienced T cell pool in normal mice: Control by CD4(+) regulatory T cells and IL-10. J Immunol, 2003;171:971–8. [DOI] [PubMed] [Google Scholar]
  • [147].Andersson J, Tran DQ, Pesu M, Davidson TS, Ramsey H, O'Shea JJ et al. CD4+ FoxP3+ regulatory T cells confer infectious tolerance in a TGF-beta-dependent manner. J Exp Med, 2008;205:1975–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Romio M, Reinbeck B, Bongardt S, Huls S, Burghoff S, Schrader J. Extracellular purine metabolism and signaling of CD73-derived adenosine in murine Treg and Teff cells. Am J Physiol Cell Physiol, 2011;301:C530–9. [DOI] [PubMed] [Google Scholar]
  • [149].Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y et al. An essential role for the IL-2 receptor in Treg cell function. Nat Immunol, 2016;17:1322–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol, 2005;174:1783–6. [DOI] [PubMed] [Google Scholar]
  • [151].Zhao DM, Thornton AM, DiPaolo RJ, Shevach EM. Activated CD4+CD25+ T cells selectively kill B lymphocytes. Blood, 2006;107:3925–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR et al. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity, 2007;27:635–46. [DOI] [PubMed] [Google Scholar]
  • [153].Smith TR, Kumar V. Revival of CD8+ Treg-mediated suppression. Trends Immunol, 2008;29:337–42. [DOI] [PubMed] [Google Scholar]
  • [154].Lio CW, Hsieh CS. A two-step process for thymic regulatory T cell development. Immunity, 2008;28:100–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Tai X, Erman B, Alag A, Mu J, Kimura M, Katz G et al. Foxp3 Transcription Factor Is Proapoptotic and Lethal to Developing Regulatory T Cells unless Counterbalanced by Cytokine Survival Signals. Immunity, 2013;38:1116–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Mahmud SA, Manlove LS, Farrar MA. Interleukin-2 and STAT5 in regulatory T cell development and function. JAKSTAT, 2013;2:e23154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Bonomo A, Kehn PJ, Shevach EM. Post-thymectomy autoimmunity: abnormal T-cell homeostasis. Immunol Today, 1995;16:61–7. [DOI] [PubMed] [Google Scholar]
  • [158].Kieback E, Hilgenberg E, Stervbo U, Lampropoulou V, Shen P, Bunse M et al. Thymus-Derived Regulatory T Cells Are Positively Selected on Natural Self-Antigen through Cognate Interactions of High Functional Avidity. Immunity, 2016;44:1114–26. [DOI] [PubMed] [Google Scholar]
  • [159].Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity, 2011;35:109–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y et al. Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature, 2012;482:395–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Singh K, Hjort M, Thorvaldson L, Sandler S. Concomitant analysis of Helios and Neuropilin-1 as a marker to detect thymic derived regulatory T cells in naive mice. Sci Rep, 2015;5:7767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Guo F, Iclozan C, Suh WK, Anasetti C, Yu XZ. CD28 controls differentiation of regulatory T cells from naive CD4 T cells. J Immunol, 2008;181:2285–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Shahinian A, Pfeffer K, Lee KP, Kundig TM, Kishihara K, Wakeham A et al. Differential T cell costimulatory requirements in CD28-deficient mice. Science, 1993;261:609–12. [DOI] [PubMed] [Google Scholar]
  • [164].Williams JA, Hathcock KS, Klug D, Harada Y, Choudhury B, Allison JP et al. Regulated costimulation in the thymus is critical for T cell development: Dysregulated CD28 costimulation can bypass the pre-TCR checkpoint. J Immunol, 2005;175:4199–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Tang Q, Henriksen KJ, Boden EK, Tooley AJ, Ye J, Subudhi SK et al. Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells. J Immunol, 2003;171:3348–52. [DOI] [PubMed] [Google Scholar]
  • [166].Zhang R, Huynh A, Whitcher G, Chang J, Maltzman JS, Turka LA. An obligate cell-intrinsic function for CD28 in Tregs. J Clin Invest, 2013;123:580–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Ohkura N, Hamaguchi M, Morikawa H, Sugimura K, Tanaka A, Ito Y et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity, 2012;37:785–99. [DOI] [PubMed] [Google Scholar]
  • [168].Walker LS. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun, 2013;45:49–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity, 1995;3:541–7. [DOI] [PubMed] [Google Scholar]
  • [170].Deppong CM, Bricker TL, Rannals BD, Van Rooijen N, Hsieh CS, Green JM. CTLA4Ig Inhibits Effector T Cells through Regulatory T Cells and TGF-beta. J Immunol, 2013;191:3082–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med, 2000;192:303–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD et al. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med, 2015;212:1603–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Krummel MF, Allison JP. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J Exp Med, 1996;183:2533–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Klocke K, Sakaguchi S, Holmdahl R, Wing K. Induction of autoimmune disease by deletion of CTLA-4 in mice in adulthood. P Natl Acad Sci USA, 2016;113:E2383–E92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med, 2009;206:1717–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Nishimura H, Honjo T, Minato N. Facilitation of beta selection and modification of positive selection in the thymus of PD-1-deficient mice. J Exp Med, 2000;191:891–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med, 2009;206:3015–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Zhang B, Chikuma S, Hori S, Fagarasan S, Honjo T. Nonoverlapping roles of PD-1 and FoxP3 in maintaining immune tolerance in a novel autoimmune pancreatitis mouse model. Proc Natl Acad Sci U S A, 2016;113:8490–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Wong MD, La Cava A, Hahn BH. Blockade of Programmed Death-1 in Young (New Zealand Black x New Zealand White)F-1 Mice Promotes the Suppressive Capacity of CD4(+) Regulatory T Cells Protecting from Lupus-like Disease. J Immunol, 2013;190:5402–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Wong M, La Cava A, Singh RP, Hahn BH. Blockade of Programmed Death-1 in Young (New Zealand Black x New Zealand White)F-1 Mice Promotes the Activity of Suppressive CD8(+) T Cells That Protect from Lupus-Like Disease. J Immunol, 2010;185:6563–71. [DOI] [PubMed] [Google Scholar]
  • [181].Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G et al. Role of LAG-3 in regulatory T cells. Immunity, 2004;21:503–13. [DOI] [PubMed] [Google Scholar]
  • [182].Sun H, Gao W, Pan W, Zhang Q, Wang G, Feng D et al. Tim3(+) Foxp3 (+) Treg Cells Are Potent Inhibitors of Effector T Cells and Are Suppressed in Rheumatoid Arthritis. Inflammation, 2017;40:1342–50. [DOI] [PubMed] [Google Scholar]
  • [183].Joller N, Lozano E, Burkett PR, Patel B, Xiao S, Zhu C et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity, 2014;40:569–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Zhang Y, Maksimovic J, Naselli G, Qian J, Chopin M, Blewitt ME et al. Genome-wide DNA methylation analysis identifies hypomethylated genes regulated by FOXP3 in human regulatory T cells. Blood, 2013;122:2823–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Alharshawi K, Marinelarena A, Kumar P, El-Sayed O, Bhattacharya P, Sun ZM et al. PKC-theta is dispensable for OX40L-induced TCR-independent Treg proliferation but contributes by enabling IL-2 production from effector T-cells. Sci Rep-Uk, 2017;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Kumar P, Marinelarena A, Raghunathan D, Ragothaman VK, Saini S, Bhattacharya P et al. Critical role of OX40 signaling in the TCR-independent phase of human and murine thymic Treg generation. Cell Mol Immunol, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Liao G, Nayak S, Regueiro JR, Berger SB, Detre C, Romero X et al. GITR engagement preferentially enhances proliferation of functionally competent CD4+CD25+FoxP3+ regulatory T cells. Int Immunol, 2010;22:259–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Chen X, Baumel M, Mannel DN, Howard OM, Oppenheim JJ. Interaction of TNF with TNF receptor type 2 promotes expansion and function of mouse CD4+CD25+ T regulatory cells. J Immunol, 2007;179:154–61. [DOI] [PubMed] [Google Scholar]
  • [189].Zhang P, Gao F, Wang Q, Wang X, Zhu F, Ma C et al. Agonistic anti-4-1BB antibody promotes the expansion of natural regulatory T cells while maintaining Foxp3 expression. Scand J Immunol, 2007;66:435–40. [DOI] [PubMed] [Google Scholar]
  • [190].So T, Croft M. Cutting edge: OX40 inhibits TGF-beta- and antigen-driven conversion of naive CD4 T cells into CD25(+)Foxp3(+) T cells. Journal of Immunology, 2007;179:1427–30. [DOI] [PubMed] [Google Scholar]
  • [191].Korn T, Mitsdoerffer M, Croxford AL, Awasthi A, Dardalhon VA, Galileos G et al. IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells. Proc Natl Acad Sci U S A, 2008;105:18460–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Kumar P, Subramaniyam G. Molecular underpinnings of Th17 immune-regulation and their implications in autoimmune diabetes. Cytokine, 2015;71:366–76. [DOI] [PubMed] [Google Scholar]
  • [193].So T, Croft M. Regulation of PI-3-Kinase and Akt Signaling in T Lymphocytes and Other Cells by TNFR Family Molecules. Front Immunol, 2013; 4:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Merkenschlager M, von Boehmer H. PI3 kinase signalling blocks Foxp3 expression by sequestering Foxo factors. J Exp Med, 2010;207:1347–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].So T, Lee SW, Croft M. Immune regulation and control of regulatory T cells by OX40 and 4-1BB. Cytokine Growth Factor Rev, 2008;19:253–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Haddad CS, Bhattacharya P, Alharshawi K, Marinelarena A, Kumar P, El-Sayed O et al. Age-dependent divergent effects of OX40L treatment on the development of diabetes in NOD mice. Autoimmunity, 2016;49:298–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Xiao X, Gong W, Demirci G, Liu W, Spoerl S, Chu X et al. New insights on OX40 in the control of T cell immunity and immune tolerance in vivo. J Immunol, 2012;188:892–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S. Stimulation of CD25+CD4+regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol, 2002;3:135–42. [DOI] [PubMed] [Google Scholar]
  • [199].Choi BK, Bae JS, Choi EM, Kang WJ, Sakaguchi S, Vinay DS et al. 4-1BB-dependent inhibition of immunosuppression by activated CD4+CD25+ T cells. J Leukoc Biol, 2004;75:785–91. [DOI] [PubMed] [Google Scholar]
  • [200].Vu MD, Xiao X, Gao W, Degauque N, Chen M, Kroemer A et al. OX40 costimulation turns off Foxp3+ Tregs. Blood, 2007;110:2501–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Takeda I, Ine S, Killeen N, Ndhlovu LC, Murata K, Satomi S et al. Distinct roles for the OX40-OX40 ligand interaction in regulatory and nonregulatory T cells. J Immunol, 2004;172:3580–9. [DOI] [PubMed] [Google Scholar]
  • [202].Barsoumian HB, Yolcu ES, Shirwan H. 4-1BB Signaling in Conventional T Cells Drives IL-2 Production That Overcomes CD4+CD25+FoxP3+ T Regulatory Cell Suppression. PLoS One, 2016;11:e0153088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Tada Y, Nagasawa K, Ho A, Morito F, Ushiyama O, Suzuki N et al. CD28-deficient mice are highly resistant to collagen-induced arthritis. J Immunol, 1999;162:203–8. [PubMed] [Google Scholar]
  • [204].Quattrocchi E, Dallman MJ, Feldmann M. Adenovirus-mediated gene transfer of CTLA-4Ig fusion protein in the suppression of experimental autoimmune arthritis. Arthritis Rheum, 2000;43:1688–97. [DOI] [PubMed] [Google Scholar]
  • [205].Knoerzer DB, Karr RW, Schwartz BD, Mengle-Gaw LJ. Collagen-induced arthritis in the BB rat. Prevention of disease by treatment with CTLA-4-Ig. J Clin Invest, 1995;96:987–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Genovese MC, Becker JC, Schiff M, Luggen M, Sherrer Y, Kremer J et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med, 2005;353:1114–23. [DOI] [PubMed] [Google Scholar]
  • [207].Tada Y, Nagasawa K, Ho A, Morito F, Koarada S, Ushiyama O et al. Role of the costimulatory molecule CD28 in the development of lupus in MRL/lpr mice. J Immunol, 1999;163:3153–9. [PubMed] [Google Scholar]
  • [208].Mihara M, Tan I, Chuzhin Y, Reddy B, Budhai L, Holzer A et al. CTLA4Ig inhibits T cell-dependent B-cell maturation in murine systemic lupus erythematosus. Journal of Clinical Investigation, 2000;106:91–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Furie R, Nicholls K, Cheng TT, Houssiau F, Burgos-Vargas R, Chen SL et al. Efficacy and Safety of Abatacept in Lupus Nephritis A Twelve-Month, Randomized, Double-Blind Study. Arthritis & Rheumatology, 2014;66:379–89. [DOI] [PubMed] [Google Scholar]
  • [210].Oliveira-dos-Santos AJ, Ho A, Tada Y, Lafaille JJ, Tonegawa S, Mak TW et al. CD28 costimulation is crucial for the development of spontaneous autoimmune encephalomyelitis. J Immunol, 1999;162:4490–5. [PubMed] [Google Scholar]
  • [211].Chang TT, Jabs C, Sobel RA, Kuchroo VK, Sharpe AH. Studies in B7-deficient mice reveal a critical role for B7 costimulation in both induction and effector phases of experimental autoimmune encephalomyelitis. J Exp Med, 1999;190:733–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Klocke K, Sakaguchi S, Holmdahl R, Wing K. Induction of autoimmune disease by deletion of CTLA-4 in mice in adulthood. European Journal of Immunology, 2016;46:770-. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD et al. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med, 2015;212:1603–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Arima T, Rehman A, Hickey WF, Flye MW. Inhibition by CTLA4Ig of experimental allergic encephalomyelitis. J Immunol, 1996;156:4916–24. [PubMed] [Google Scholar]
  • [215].Croxford JL, O'Neill JK, Ali RR, Browne K, Byrnes AP, Dallman MJ et al. Local gene therapy with CTLA4-immunoglobulin fusion protein in experimental allergic encephalomyelitis. European Journal of Immunology, 1998;28:3904–16. [DOI] [PubMed] [Google Scholar]
  • [216].Viglietta V, Bourcier K, Buckle GJ, Healy B, Weiner HL, Hafler DA et al. CTLA4Ig treatment in patients with multiple sclerosis - An open-label, phase 1 clinical trial. Neurology, 2008;71:917–24. [DOI] [PubMed] [Google Scholar]
  • [217].Nistico L, Buzzetti R, Pritchard LE, VanderAuwera B, Giovannini C, Bosi E et al. The CTLA-4 gene region of chromosome 2q33 is linked to, and associated with, type 1 diabetes. Human Molecular Genetics, 1996;5:1075–80. [DOI] [PubMed] [Google Scholar]
  • [218].Orban T, Bundy B, Becker DJ, DiMeglio LA, Gitelman SE, Goland R et al. Costimulation Modulation With Abatacept in Patients With Recent-Onset Type 1 Diabetes: Follow-up 1 Year After Cessation of Treatment. Diabetes Care, 2014;37:1069–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity, 1999;11:141–51. [DOI] [PubMed] [Google Scholar]
  • [220].Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1(−/−) mice as an efficient animal model of type I diabetes. P Natl Acad Sci USA, 2005;102:11823–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med, 2006;203:883–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med, 2003;198:63–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].do Canto LM, Farias TD, Medeiros MD, Coelho CC, Sereia AF, de Carlos Back LK et al. Association of PDCD1 polymorphism to systemic lupus erythematosus and rheumatoid arthritis susceptibility. Rev Bras Reumatol Engl Ed, 2016;56:483–9. [DOI] [PubMed] [Google Scholar]
  • [224].Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol, 2007;19:813–24. [DOI] [PubMed] [Google Scholar]
  • [225].Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, Albacker LA et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med, 2006;203:883–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M et al. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. P Natl Acad Sci USA, 2004;101:10691–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Wan B, Nie H, Liu A, Feng G, He D, Xu R et al. Aberrant regulation of synovial T cell activation by soluble costimulatory molecules in rheumatoid arthritis. J Immunol, 2006;177:8844–50. [DOI] [PubMed] [Google Scholar]
  • [228].Raptopoulou AP, Bertsias G, Makrygiannakis D, Verginis P, Kritikos I, Tzardi M et al. The programmed death 1/programmed death ligand 1 inhibitory pathway is up-regulated in rheumatoid synovium and regulates peripheral T cell responses in human and murine arthritis. Arthritis Rheum, 2010;62:1870–80. [DOI] [PubMed] [Google Scholar]
  • [229].Wang G, Hu P, Yang J, Shen G, Wu X. The effects of PDL-Ig on collagen-induced arthritis. Rheumatol Int, 2011;31:513–9. [DOI] [PubMed] [Google Scholar]
  • [230].Shi H, Ye JN, Teng JL, Yin YF, Hu QY, Wu XY et al. Elevated serum autoantibodies against co-inhibitory PD-1 facilitate T cell proliferation and correlate with disease activity in new-onset systemic lupus erythematosus patients. Arthritis Res Ther, 2017;19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Kasagi S, Kawano S, Okazaki T, Honjo T, Morinobu A, Hatachi S et al. Anti-Programmed Cell Death 1 Antibody Reduces CD4(+)PD-1(+) T Cells and Relieves the Lupus-Like Nephritis of NZB/W F1 Mice. J Immunol, 2010;184:2337–47. [DOI] [PubMed] [Google Scholar]
  • [232].Liao WJ, Zheng H, Wu S, Zhang YM, Wang W, Zhang ZL et al. The Systemic Activation of Programmed Death 1-PD-L1 Axis Protects Systemic Lupus Erythematosus Model from Nephritis. Am J Nephrol, 2017;46:371–9. [DOI] [PubMed] [Google Scholar]
  • [233].Rui YX, Honjo T, Chikuma S. Programmed cell death 1 inhibits inflammatory helper T-cell development through controlling the innate immune response. P Natl Acad Sci USA, 2013;110:16073–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Cheng XD, Zhao Z, Ventura E, Gran B, Shindler KS, Rostami A. The PD-1/PD-L pathway is up-regulated during IL-12-induced suppression of EAE mediated by IFN-gamma. Journal of Neuroimmunology, 2007;185:75–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Schreiner B, Bailey SL, Shin T, Chen L, Miller SD. PD-1 ligands expressed on myeloid-derived APC in the CNS regulate T-cell responses in EAE. European Journal of Immunology, 2008;38:2706–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Salama AD, Chitnis T, Imitola J, Akiba H, Tushima F, Azuma M et al. Critical role of the programmed death-1 (PD-1) pathway in regulation of experimental autoimmune encephalomyelitis. J Exp Med, 2003;198:71–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Wang C, Li Y, Proctor TM, Vandenbark AA, Offner H. Down-modulation of programmed death 1 alters regulatory T cells and promotes experimental autoimmune encephalomyelitis. J Neurosci Res, 2010;88:7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Kochupurakkal NM, Kruger AJ, Tripathi S, Zhu B, Adams L, Rainbow DB et al. Blockade of the Programmed Death-1 (PD1) Pathway Undermines Potent Genetic Protection from Type 1 Diabetes. Plos One, 2014; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Tada Y, Nagasawa K, Ho A, Morito F, Koarada S, Ushiyama O et al. Role of the costimulatory molecule CD28 in the development of lupus in MRL/lpr mice. J Immunol, 1999;163:3153–9. [PubMed] [Google Scholar]
  • [240].Lenschow DJ, Herold KC, Rhee L, Patel B, Koons A, Qin HY et al. CD28/B7 regulation of Th1 and Th2 subsets in the development of autoimmune diabetes. Immunity, 1996;5:285–93. [DOI] [PubMed] [Google Scholar]
  • [241].Liu Z, Geboes K, Hellings P, Maerten P, Heremans H, Vandenberghe P et al. B7 interactions with CD28 and CTLA-4 control tolerance or induction of mucosal inflammation in chronic experimental colitis. J Immunol, 2001; 167:1830–8. [DOI] [PubMed] [Google Scholar]
  • [242].Liu CP, Jiang JA, Gao L, Wang XT, Hu XH, Wu M et al. Soluble PD-1 aggravates progression of collagen-induced arthritis through Th1 and Th17 pathways. Arthritis Res Ther, 2015; 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Bettini M, Szymczak-Workman AL, Forbes K, Castellaw AH, Selby M, Pan X et al. Cutting edge: accelerated autoimmune diabetes in the absence of LAG-3. J Immunol, 2011;187:3493–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Jha V, Workman CJ, McGaha TL, Li L, Vas J, Vignali DA et al. Lymphocyte Activation Gene-3 (LAG-3) negatively regulates environmentally-induced autoimmunity. Plos One, 2014;9:e104484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Lee SY, Goverman JM. The Influence of T Cell Ig Mucin-3 Signaling on Central Nervous System Autoimmune Disease Is Determined by the Effector Function of the Pathogenic T Cells. J Immunol, 2013;190:4991–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Sanchez-Fueyo A, Tian J, Picarella D, Domenig C, Zheng XX, Sabatos CA et al. Tim-3 inhibits T helper type 1- mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol, 2003;4:1093–101. [DOI] [PubMed] [Google Scholar]
  • [247].Seki M, Oomizu S, Sakata KM, Sakata A, Arikawa T, Watanabe K et al. Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol, 2008;127:78–88. [DOI] [PubMed] [Google Scholar]
  • [248].Levin SD, Taft DW, Brandt CS, Bucher C, Howard ED, Chadwick EM et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. European Journal of Immunology, 2011;41:902–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Mao L, Hou H, Wu S, Zhou Y, Wang J, Yu J et al. TIGIT signalling pathway negatively regulates CD4(+) T-cell responses in systemic lupus erythematosus. Immunology, 2017;151:280–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Okazaki T, Okazaki IM, Wang J, Sugiura D, Nakaki F, Yoshida T et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med, 2011;208:395–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Kadowaki A, Miyake S, Saga R, Chiba A, Mochizuki H, Yamamura T. Gut environment-induced intraepithelial autoreactive CD4(+) T cells suppress central nervous system autoimmunity via LAG-3. Nat Commun, 2016;7:11639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Sabatos CA, Chakravarti S, Cha E, Schubart A, Sanchez-Fueyo A, Zheng XX et al. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol, 2003;4:1102–10. [DOI] [PubMed] [Google Scholar]
  • [253].Koguchi K, Anderson DE, Yang L, O'Connor KC, Kuchroo VK, Hafler DA. Dysregulated T cell expression of TIM3 in multiple sclerosis. J Exp Med, 2006;203:1413–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Joller N, Hafler JP, Brynedal B, Kassam N, Spoerl S, Levin SD et al. Cutting edge: TIGIT has T cell-intrinsic inhibitory functions. J Immunol, 2011;186:1338–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Levin SD, Taft DW, Brandt CS, Bucher C, Howard ED, Chadwick EM et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur J Immunol, 2011;41:902–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Dixon KO, Schorer M, Nevin J, Etminan Y, Amoozgar Z, Kondo T et al. Functional Anti-TIGIT Antibodies Regulate Development of Autoimmunity and Antitumor Immunity. J Immunol, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Wang F, Hou H, Wu S, Tang Q, Liu W, Huang M et al. TIGIT expression levels on human NK cells correlate with functional heterogeneity among healthy individuals. Eur J Immunol, 2015;45:2886–97. [DOI] [PubMed] [Google Scholar]
  • [258].Cope AP, Aderka D, Doherty M, Engelmann H, Gibbons D, Jones AC et al. Increased levels of soluble tumor necrosis factor receptors in the sera and synovial fluid of patients with rheumatic diseases. Arthritis Rheum, 1992;35:1160–9. [DOI] [PubMed] [Google Scholar]
  • [259].Ban LQ, Zhang J, Wang LM, Kuhtreiber W, Burger D, Faustman DL. Selective death of autoreactive T cells in human diabetes by TNF or TNF receptor 2 agonism. P Natl Acad Sci USA, 2008;105:13644–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Nguyen DX, Ehrenstein MR. Anti-TNF drives regulatory T cell expansion by paradoxically promoting membrane TNF-TNF-RII binding in rheumatoid arthritis. J Exp Med, 2016;213:1241–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Housley WJ, Adams CO, Nichols FC, Puddington L, Lingenheld EG, Zhu L et al. Natural but Not Inducible Regulatory T Cells Require TNF-alpha Signaling for In Vivo Function. J Immunol, 2011;186:6779–87. [DOI] [PubMed] [Google Scholar]
  • [262].Cuzzocrea S, Ayroldi E, Di Paola R, Agostini M, Mazzon E, Bruscoli S et al. Role of glucocorticoid-induced TNF receptor family gene (GITR) in collagen-induced arthritis. Faseb J, 2005;19:1253–65. [DOI] [PubMed] [Google Scholar]
  • [263].Kohm AP, Williams JS, Miller SD. Cutting edge: Ligation of the glucocorticoid-induced TNF receptor enhances autoreactive CD4(+) T cell activation and experimental autoimmune encephalomyelitis. J Immunol, 2004;172:4686–90. [DOI] [PubMed] [Google Scholar]
  • [264].Patel M, Xu D, Kewin P, Choo-Kang B, McSharry C, Thomson NC et al. Glucocorticoid-induced TNFR family-related protein (GITR) activation exacerbates murine asthma and collagen-induced arthritis. European Journal of Immunology, 2005;35:3581–90. [DOI] [PubMed] [Google Scholar]
  • [265].You S, Poulton L, Cobbold S, Liu CP, Rosenzweig M, Ringler D et al. Key Role of the GITR/GITRLigand Pathway in the Development of Murine Autoimmune Diabetes: A Potential Therapeutic Target. Plos One, 2009;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].van Olffen RW, Koning N, van Gisbergen KPJM, Wensveen FM, Hoek RM, Boon L et al. GITR Triggering Induces Expansion of Both Effector and Regulatory CD4(+) T Cells In Vivo. J Immunol, 2009;182:7490–500. [DOI] [PubMed] [Google Scholar]
  • [267].Uraushihara K, Kanai T, Ko K, Totsuka T, Makita S, Iiyama R et al. Regulation of murine inflammatory bowel disease by CD25(+) and CD25(−) CD4(+) glucocorticoid-induced TNF receptor family-related gene(+) regulatory T cells. J Immunol, 2003;171:708–16. [DOI] [PubMed] [Google Scholar]
  • [268].Nocentini G, Alunno A, Petrillo MG, Bistoni O, Bartoloni E, Caterbi S et al. Expansion of regulatory GITR(+)CD25(low/−)CD4(+) T cells in systemic lupus erythematosus patients. Arthritis Res Ther, 2014;16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Piguet PF, Grau GE, Vesin C, Loetscher H, Gentz R, Lesslauer W. Evolution of Collagen Arthritis in Mice Is Arrested by Treatment with Antitumor Necrosis Factor (Tnf) Antibody or a Recombinant Soluble Tnf Receptor. Immunology, 1992;77:510–4. [PMC free article] [PubMed] [Google Scholar]
  • [270].Mori L, Iselin S, DeLibero G, Lesslauer W. Attenuation of collagen-induced arthritis in 55-kDa TNF receptor type 1 (TNFR1)-IgG1-treated and TNFR1-deficient mice. J Immunol, 1996;157:3178–82. [PubMed] [Google Scholar]
  • [271].Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kollias G. Impaired on/off regulation of TNF biosynthesis in mice lacking TNF AU-rich elements: Implications for joint and gut-associated immunopathologies. Immunity, 1999;10:387–98. [DOI] [PubMed] [Google Scholar]
  • [272].Korner H, Lemckert FA, Chaudhri G, Etteldorf S, Sedgwick JD. Tumor necrosis factor blockade in actively induced experimental autoimmune encephalomyelitis prevents clinical disease despite activated T cell infiltration to the central nervous system. European Journal of Immunology, 1997;27:1973–81. [DOI] [PubMed] [Google Scholar]
  • [273].Suvannavejh GC, Lee HO, Padilla J, Dal Canto MC, Barrett TA, Miller SD. Divergent roles for p55 and p75 tumor necrosis factor receptors in the pathogenesis of MOG(35-55)-induced experimental autoimmune encephalomyelitis. Cell Immunol, 2000;205:24–33. [DOI] [PubMed] [Google Scholar]
  • [274].Kontoyiannis D, Kollias G. Accelerated autoimmunity and lupus nephritis in NZB mice with an engineered heterozygous deficiency in tumor necrosis factor. European Journal of Immunology, 2000;30:2038–47. [DOI] [PubMed] [Google Scholar]
  • [275].Brennan DC, Yui MA, Wuthrich RP, Kelley VE. Tumor Necrosis Factor and Il-1 in New-Zealand Black-White Mice - Enhanced Gene-Expression and Acceleration of Renal Injury. J Immunol, 1989;143:3470–5. [PubMed] [Google Scholar]
  • [276].Chee J, Angstetra E, Mariana L, Graham KL, Carrington EM, Bluethmann H et al. TNF Receptor 1 Deficiency Increases Regulatory T Cell Function in Nonobese Diabetic Mice. J Immunol, 2011;187:1702–12. [DOI] [PubMed] [Google Scholar]
  • [277].Balasa B, Van Gunst K, Jung N, Balakrishna D, Santamaria P, Hanafusa T et al. Islet-specific expression of IL-10 promotes diabetes in nonobese diabetic mice independent of Fas, perforin, TNF receptor-1, and TNF receptor-2 molecules. J Immunol, 2000;165:2841–9. [DOI] [PubMed] [Google Scholar]
  • [278].Yang XD, Tisch R, Singer SM, Cao ZA, Liblau RS, Schreiber RD et al. Effect of Tumor-Necrosis-Factor-Alpha on Insulin-Dependent Diabetes-Mellitus in Nod Mice .1. The Early Development of Autoimmunity and the Diabetogenic Process. J Exp Med, 1994;180:995–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Tang XY, Tian J, Ma J, Wang JM, Qi C, Rui K et al. GITRL modulates the activities of p38 MAPK and STAT3 to promote Th17 cell differentiation in autoimmune arthritis. Oncotarget, 2016;7:8590–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Santucci L, Agostini M, Bruscoli S, Mencarelli A, Ronchetti S, Ayroldi E et al. GITR modulates innate and adaptive mucosal immunity during the development of experimental colitis in mice. Gut, 2007;56:52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Yoshioka T, Nakajima A, Akiba H, Ishiwata T, Asano G, Yoshino S et al. Contribution of OX40/OX40 ligand interaction to the pathogenesis of rheumatoid arthritis. Eur J Immunol, 2000;30:2815–23. [DOI] [PubMed] [Google Scholar]
  • [282].Findlay EG, Danks L, Madden J, Cavanagh MM, McNamee K, McCann F et al. OX40L blockade is therapeutic in arthritis, despite promoting osteoclastogenesis. P Natl Acad Sci USA, 2014;111:2289–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Ndhlovu LC, Ishii N, Murata K, Sato T, Sugamura K. Critical involvement of OX40 ligand signals in the T cell priming events during experimental autoimmune encephalomyelitis. J Immunol, 2001;167:2991–9. [DOI] [PubMed] [Google Scholar]
  • [284].Ruby CE, Yates MA, Hirschhorn-Cymerman D, Chlebeck P, Wolchok JD, Houghton AN et al. Cutting Edge: OX40 agonists can drive regulatory T cell expansion if the cytokine milieu is right. J Immunol, 2009;183:4853–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Sitrin J, Suto E, Wuster A, Eastham-Anderson J, Kim JM, Austin CD et al. The Ox40/Ox40 Ligand Pathway Promotes Pathogenic Th Cell Responses, Plasmablast Accumulation, and Lupus Nephritis in NZB/W F1 Mice. J Immunol, 2017;199:1238–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Pakala SV, Bansal-Pakala P, Halteman BS, Croft M. Prevention of diabetes in NOD mice at a late stage by targeting OX40/OX40 ligand interactions. Eur J Immunol, 2004;34:3039–46. [DOI] [PubMed] [Google Scholar]
  • [287].Haddad CS, Bhattacharya P, Alharshawi K, Marinelarena A, Kumar P, El-Sayed O et al. Age-dependent divergent effects of OX40L treatment on the development of diabetes in NOD mice. Autoimmunity, 2016:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [288].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;37:342–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [289].Malmstrom V, Shipton D, Singh B, Al-Shamkhani A, Puklavec MJ, Barclay AN et al. CD134L expression on dendritic cells in the mesenteric lymph nodes drives colitis in T cell-restored SCID mice. J Immunol, 2001;166:6972–81. [DOI] [PubMed] [Google Scholar]
  • [290].Griseri T, Asquith M, Thompson C, Powrie F. OX40 is required for regulatory T cell-mediated control of colitis. J Exp Med, 2010;207:699–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [291].Seo SK, Choi JH, Kim YH, Kang WJ, Park HY, Suh JH et al. 4-1BB-mediated immunotherapy of rheumatoid arthritis. Nat Med, 2004;10:1088–94. [DOI] [PubMed] [Google Scholar]
  • [292].Sun Y, Lin X, Chen HM, Wu Q, Subudhi SK, Chen L et al. Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis. J Immunol, 2002;168:1457–65. [DOI] [PubMed] [Google Scholar]
  • [293].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] [PMC free article] [PubMed] [Google Scholar]
  • [294].Sun Y, Chen HM, Subudhi SK, Chen J, Koka R, Chen L et al. Costimulatory molecule-targeted antibody therapy of a spontaneous autoimmune disease. Nat Med, 2002;8:1405–13. [DOI] [PubMed] [Google Scholar]
  • [295].Chen YG, Forsberg MH, Khaja S, Ciecko AE, Hessner MJ, Geurts AM. Gene Targeting in NOD Mouse Embryos Using Zinc-Finger Nucleases. Diabetes, 2014;63:68–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Irie J, Wu YH, Kachapati K, Mittler RS, Ridgway WM. Modulating protective and pathogenic CD4+ subsets via CD137 in type 1 diabetes. Diabetes, 2007;56:186–96. [DOI] [PubMed] [Google Scholar]
  • [297].Maerten P, Kwon BS, Shen C, De Hertogh G, Cadot P, Bullens DM et al. Involvement of 4-1BB (CD137)-4-1BBligand interaction in the modulation of CD4 T cell-mediated inflammatory colitis. Clin Exp Immunol, 2006;143:228–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Baeyens A, Saadoun D, Billiard F, Rouers A, Gregoire S, Zaragoza B et al. Effector T cells boost regulatory T cell expansion by IL-2, TNF, OX40, and plasmacytoid dendritic cells depending on the immune context. J Immunol, 2015;194:999–1010. [DOI] [PubMed] [Google Scholar]
  • [299].Manku H, Langefeld CD, Guerra SG, Malik TH, Alarcon-Riquelme M, Anaya JM et al. Trans-ancestral studies fine map the SLE-susceptibility locus TNFSF4. PLoS Genet, 2013;9:e1003554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Jacquemin C, Schmitt N, Contin-Bordes C, Liu Y, Narayanan P, Seneschal J et al. OX40 Ligand Contributes to Human Lupus Pathogenesis by Promoting T Follicular Helper Response. Immunity, 2015;42:1159–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Ruby CE, Yates MA, Hirschhorn-Cymerman D, Chlebeck P, Wolchok JD, Houghton AN et al. Cutting Edge: OX40 Agonists Can Drive Regulatory T Cell Expansion if the Cytokine Milieu Is Right. J Immunol, 2009;183:4853–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Kumar P, Alharshawi K, Bhattacharya P, Marinelarena A, Haddad C, Sun Z et al. Soluble OX40L and JAG1 Induce Selective Proliferation of Functional Regulatory T-Cells Independent of canonical TCR signaling. Sci Rep, 2017;7:39751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Humphreys IR, Lee SW, Jones M, Loewendorf A, Gostick E, Price DA et al. Biphasic role of 4-1BB in the regulation of mouse cytomegalovirus-specific CD8(+) T cells. Eur J Immunol, 2010;40:2762–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [304].Zheng G, Wang B, Chen A. The 4-1BB costimulation augments the proliferation of CD4+CD25+ regulatory T cells. J Immunol, 2004;173:2428–34. [DOI] [PubMed] [Google Scholar]
  • [305].Hippen KL, Harker-Murray P, Porter SB, Merkel SC, Londer A, Taylor DK et al. Umbilical cord blood regulatory T-cell expansion and functional effects of tumor necrosis factor receptor family members OX40 and 4-1BB expressed on artificial antigen-presenting cells. Blood, 2008;112:2847–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [306].Kim YH, Choi BK, Shin SM, Kim CH, Oh HS, Park SH et al. 4-1BB Triggering Ameliorates Experimental Autoimmune Encephalomyelitis by Modulating the Balance between Th17 and Regulatory T Cells. J Immunol, 2011;187:1120–8. [DOI] [PubMed] [Google Scholar]
  • [307].Lee J, Lee EN, Kim EY, Park HJ, Chang CY, Jung DY et al. Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of inflammatory bowel disease. Immunol Lett, 2005;101:210–6. [DOI] [PubMed] [Google Scholar]
  • [308].Liu GZ, Gomes AC, Fang LB, Gao XG, Hjelmstrom P. Decreased 4-1BB expression on CD4+CD25(high) regulatory T cells in peripheral blood of patients with multiple sclerosis. Clin Exp Immunol, 2008;154:22–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [309].Barsoumian HB, Yolcu ES, Shirwan H. 4-1BB Signaling in Conventional T Cells Drives IL-2 Production That Overcomes CD4(+)CD25(+)FoxP3(+) T Regulatory Cell Suppression. Plos One, 2016;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [310].Smith SE, Hoelzinger DB, Dominguez AL, Van Snick J, Lustgarten J. Signals through 4-1BB inhibit T regulatory cells by blocking IL-9 production enhancing antitumor responses. Cancer Immunol Immun, 2011;60:1775–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [311].von Boehmer H, Daniel C. Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer. Nat Rev Drug Discov, 2013;12:51–63. [DOI] [PubMed] [Google Scholar]
  • [312].Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck-Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res, 2003;9:606–12. [PubMed] [Google Scholar]
  • [313].Schaefer C, Kim GG, Albers A, Hoermann K, Myers EN, Whiteside TL. Characteristics of CD4+CD25+ regulatory T cells in the peripheral circulation of patients with head and neck cancer. Br J Cancer, 2005;92:913–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res, 2006;12:5423–34. [DOI] [PubMed] [Google Scholar]
  • [315].Ichihara F, Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin Cancer Res, 2003;9:4404–8. [PubMed] [Google Scholar]
  • [316].Ormandy LA, Hillemann T, Wedemeyer H, Manns MP, Greten TF, Korangy F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res, 2005;65:2457–64. [DOI] [PubMed] [Google Scholar]
  • [317].Syed Khaja AS, Toor SM, El Salhat H, Faour I, Ul Haq N, Ali BR et al. Preferential accumulation of regulatory T cells with highly immunosuppressive characteristics in breast tumor microenvironment. Oncotarget, 2017;8:33159–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [318].Fujii H, Josse J, Tanioka M, Miyachi Y, Husson F, Ono M. Regulatory T Cells in Melanoma Revisited by a Computational Clustering of FOXP3+ T Cell Subpopulations. J Immunol, 2016;196:2885–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [319].Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother, 2011;60:909–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [320].Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med, 2016;22:679–84. [DOI] [PubMed] [Google Scholar]
  • [321].Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunol Cell Biol, 2018;96:21–33. [DOI] [PubMed] [Google Scholar]
  • [322].Dine J, Gordon R, Shames Y, Kasler MK, Barton-Burke M. Immune Checkpoint Inhibitors: An Innovation in Immunotherapy for the Treatment and Management of Patients with Cancer. Asia Pac J Oncol Nurs, 2017;4:127–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [323].Abdin SM, Zaher DM, Arafa EA, Omar HA. Tackling Cancer Resistance by Immunotherapy: Updated Clinical Impact and Safety of PD-1/PD-L1 Inhibitors. Cancers (Basel), 2018;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [324].Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med, 2013;210:1695–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [325].Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res, 2013;1:32–42. [DOI] [PubMed] [Google Scholar]
  • [326].Chen X, Fosco D, Kline DE, Meng L, Nishi S, Savage PA et al. PD-1 regulates extrathymic regulatory T-cell differentiation. Eur J Immunol, 2014;44:2603–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Jacobs JF, Idema AJ, Bol KF, Nierkens S, Grauer OM, Wesseling P et al. Regulatory T cells and the PD-L1/PD-1 pathway mediate immune suppression in malignant human brain tumors. Neuro Oncol, 2009;11:394–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [328].Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol, 2009;21:1065–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [329].Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A, 2010; 107:4275–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [330].Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res, 2013;19:5300–9. [DOI] [PubMed] [Google Scholar]
  • [331].Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M et al. Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells. Cancer Immunol Res, 2013;1:32–42. [DOI] [PubMed] [Google Scholar]
  • [332].Simpson TR, Li FB, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med, 2013;210:1695–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [333].Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA et al. Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med, 2013;210:1685–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [334].Dyck L, Wilk MM, Raverdeau M, Misiak A, Boon L, Mills KHG. Anti-PD-1 inhibits Foxp3(+) Treg cell conversion and unleashes intratumoural effector T cells thereby enhancing the efficacy of a cancer vaccine in a mouse model. Cancer Immunol Immun, 2016;65:1491–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [335].Huang RY, Eppolito C, Lele S, Shrikant P, Matsuzaki J, Odunsi K. LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget, 2015;6:27359–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [336].Johnston RJ, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell, 2014;26:923–37. [DOI] [PubMed] [Google Scholar]
  • [337].Dixon KO, Schorer M, Nevin J, Etminan Y, Amoozgar Z, Kondo T et al. Functional Anti-TIGIT Antibodies Regulate Development of Autoimmunity and Antitumor Immunity. J Immunol, 2018;200:3000–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [338].Nie Y, He J, Shirota H, Trivett AL, Yang D, Klinman DM et al. Blockade of TNFR2 signaling enhances the immunotherapeutic effect of CpG ODN in a mouse model of colon cancer. Science Signaling, 2018;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [339].Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, Kim SC et al. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One, 2010;5:e10436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [340].Bulliard Y, Jolicoeur R, Zhang J, Dranoff G, Wilson NS, Brogdon JL. OX40 engagement depletes intratumoral Tregs via activating FcgammaRs, leading to antitumor efficacy. Immunol Cell Biol, 2014;92:475–80. [DOI] [PubMed] [Google Scholar]
  • [341].Curran MA, Kim M, Montalvo W, Al-Shamkhani A, Allison JP. Combination CTLA-4 blockade and 4-1BB activation enhances tumor rejection by increasing T-cell infiltration, proliferation, and cytokine production. Plos One, 2011;6:e19499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [342].Houot R, Goldstein MJ, Kohrt HE, Myklebust JH, Alizadeh AA, Lin JT et al. Therapeutic effect of CD137 immunomodulation in lymphoma and its enhancement by Treg depletion. Blood, 2009;114:3431–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [343].Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun, 2016;7:10501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [344].Gao X, Zhu Y, Li G, Huang H, Zhang G, Wang F et al. TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS One, 2012;7:e30676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [345].Bu M, Shen Y, Seeger WL, An S, Qi R, Sanderson JA et al. Ovarian carcinoma-infiltrating regulatory T cells were more potent suppressors of CD8(+) T cell inflammation than their peripheral counterparts, a function dependent on TIM3 expression. Tumour Biol, 2016;37:3949–56. [DOI] [PubMed] [Google Scholar]
  • [346].Camisaschi C, Casati C, Rini F, Perego M, De Filippo A, Triebel F et al. LAG-3 expression defines a subset of CD4(+)CD25(high)Foxp3(+) regulatory T cells that are expanded at tumor sites. J Immunol, 2010;184:6545–51. [DOI] [PubMed] [Google Scholar]
  • [347].Scurr M, Ladell K, Besneux M, Christian A, Hockey T, Smart K et al. Highly prevalent colorectal cancer-infiltrating LAP(+) Foxp3(−) T cells exhibit more potent immunosuppressive activity than Foxp3(+) regulatory T cells. Mucosal Immunol, 2014;7:428–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [348].Kurtulus S, Sakuishi K, Ngiow SF, Joller N, Tan DJ, Teng MW et al. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest, 2015;125:4053–62. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [349].Chauvin JM, Pagliano O, Fourcade J, Sun Z, Wang H, Sander C et al. TIGIT and PD-1 impair tumor antigen-specific CD8(+) T cells in melanoma patients. J Clin Invest, 2015;125:2046–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [350].Vanamee ES, Faustman DL. TNFR2: A Novel Target for Cancer Immunotherapy. Trends Mol Med, 2017;23:1037–46. [DOI] [PubMed] [Google Scholar]
  • [351].Torrey H, Butterworth J, Mera T, Okubo Y, Wang L, Baum D et al. Targeting TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated Tregs. Sci Signal, 2017;10. [DOI] [PubMed] [Google Scholar]
  • [352].Nie Y, He J, Shirota H, Trivett AL, Yang, Klinman DM et al. Blockade of TNFR2 signaling enhances the immunotherapeutic effect of CpG ODN in a mouse model of colon cancer. Sci Signal, 2018;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [353].Hernandez-Chacon JA, Li YF, Wu RC, Bernatchez C, Wang YJ, Weber JS et al. Costimulation Through the CD137/4-1BB Pathway Protects Human Melanoma Tumor-infiltrating Lymphocytes From Activation-induced Cell Death and Enhances Antitumor Effector Function. J Immunother, 2011;34:236–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [354].Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I. Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol, 2010;37:508–16. [DOI] [PubMed] [Google Scholar]
  • [355].Chester C, Sanmamed MF, Wang J, Melero I. Immunotherapy targeting 4-1BB: mechanistic rationale, clinical results, and future strategies. Blood, 2018;131:49–57. [DOI] [PubMed] [Google Scholar]
  • [356].Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med, 2011;365:725–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [357].Aspeslagh S, Postel-Vinay S, Rusakiewicz S, Soria JC, Zitvogel L, Marabelle A. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer, 2016;52:50–66. [DOI] [PubMed] [Google Scholar]
  • [358].Marabelle A, Kohrt H, Sagiv-Barfi I, Ajami B, Axtell RC, Zhou G et al. Depleting tumor-specific Tregs at a single site eradicates disseminated tumors. Journal of Clinical Investigation, 2013;123:2447–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [359].Weinberg AD, Rivera MM, Prell R, Morris A, Ramstad T, Vetto JT et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol, 2000;164:2160–9. [DOI] [PubMed] [Google Scholar]
  • [360].Pardee AD, McCurry D, Alber S, Hu P, Epstein AL, Storkus WJ. A Therapeutic OX40 Agonist Dynamically Alters Dendritic, Endothelial, and T Cell Subsets within the Established Tumor Microenvironment. Cancer Res, 2010;70:9041–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [361].Burocchi A, Pittoni P, Gorzanelli A, Colombo MP, Piconese S. Intratumor OX40 stimulation inhibits IRF1 expression and IL-10 production by Treg cells while enhancing CD40L expression by effector memory T cells. European Journal of Immunology, 2011;41:3615–26. [DOI] [PubMed] [Google Scholar]
  • [362].Murphy KA, Lechner MG, Popescu FE, Bedi J, Decker SA, Hu PS et al. An In Vivo Immunotherapy Screen of Costimulatory Molecules Identifies Fc-OX40L as a Potent Reagent for the Treatment of Established Murine Gliomas. Clin Cancer Res, 2012;18:4657–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [363].Gough MJ, Crittenden MR, Sarff M, Pang P, Seung SK, Vetto JT et al. Adjuvant Therapy With Agonistic Antibodies to CD134 (OX40) Increases Local Control After Surgical or Radiation Therapy of Cancer in Mice. J Immunother, 2010;33:798–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [364].Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K et al. OX40 Is a Potent Immune-Stimulating Target in Late-Stage Cancer Patients. Cancer Res, 2013;73:7189–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [365].Ramirez-Montagut T, Chow A, Hirschhorn-Cymerman D, Terwey TH, Kochman AA, Lu S et al. Glucocorticoid-induced TNF receptor family related gene activation overcomes tolerance/ignorance to melanoma differentiation antigens and enhances antitumor immunity. J Immunol, 2006;176:6434–42. [DOI] [PubMed] [Google Scholar]
  • [366].Leyland R, Watkins A, Mulgrew KA, Holoweckyj N, Bamber L, Tigue NJ et al. A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist. Clin Cancer Res, 2017;23:3416–27. [DOI] [PubMed] [Google Scholar]
  • [367].Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med, 2005;202:885–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [368].Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA et al. Activating Fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med, 2013;210:1685–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [369].Baessler T, Krusch M, Schmiedel BJ, Kloss M, Baltz KM, Wacker A et al. Glucocorticoid-induced tumor necrosis factor receptor-related protein ligand subverts immunosurveillance of acute myeloid leukemia in humans. Cancer Res, 2009;69:1037–45. [DOI] [PubMed] [Google Scholar]
  • [370].Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med, 2018;378:158–68. [DOI] [PubMed] [Google Scholar]
  • [371].Michot JM, Bigenwald C, Champiat S, Collins M, Carbonnel F, Postel-Vinay S et al. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer, 2016;54:139–48. [DOI] [PubMed] [Google Scholar]
  • [372].Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med, 2010;363:711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [373].Naidoo J, Wang X, Woo KM, Iyriboz T, Halpenny D, Cunningham J et al. Pneumonitis in Patients Treated With Anti-Programmed Death-1/Programmed Death Ligand 1 Therapy. J Clin Oncol, 2017;35:709-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [374].Abdel-Rahman O, ElHalawani H, Fouad M. Risk of endocrine complications in cancer patients treated with immune check point inhibitors: a meta-analysis. Future Oncol, 2016;12:413–25. [DOI] [PubMed] [Google Scholar]
  • [375].Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science, 2015;350:1079–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [376].Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med, 2014;371:2189–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [377].Horvat TZ, Adel NG, Dung TO, Momtaz P, Postow MA, Callahan MK et al. Immune-Related Adverse Events, Need for Systemic Immunosuppression, and Effects on Survival and Time to Treatment Failure in Patients With Melanoma Treated With Ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol, 2015;33:3193-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [378].Schmidt EM, Wang CJ, Ryan GA, Clough LE, Qureshi OS, Goodall M et al. Ctla-4 controls regulatory T cell peripheral homeostasis and is required for suppression of pancreatic islet autoimmunity. J Immunol, 2009;182:274–82. [DOI] [PubMed] [Google Scholar]
  • [379].Li Z, Dong P, Ren M, Song Y, Qian X, Yang Y et al. PD-L1 Expression Is Associated with Tumor FOXP3(+) Regulatory T-Cell Infiltration of Breast Cancer and Poor Prognosis of Patient. J Cancer, 2016;7:784–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [380].Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity, 1999;11:141–51. [DOI] [PubMed] [Google Scholar]
  • [381].Nancey S, Boschetti G, Cotte E, Ruel K, Almeras T, Chauvenet M et al. Blockade of cytotoxic T-lymphocyte antigen-4 by ipilimumab is associated with a profound long-lasting depletion of Foxp3+regulatory T cells: A mechanistic explanation for ipilimumab-induced severe enterocolitis? Inflamm Bowel Dis, 2012;18:E1598–E600. [DOI] [PubMed] [Google Scholar]
  • [382].Du X, Liu M, Su J, Zhang P, Tang F, Ye P et al. Uncoupling therapeutic from immunotherapy-related adverse effects for safer and effective anti-CTLA-4 antibodies in CTLA4 humanized mice. Cell Res, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [383].Liu J, Blake SJ, Harjunpaa H, Fairfax KA, Yong MCR, Allen S et al. Assessing Immune-Related Adverse Events of Efficacious Combination Immunotherapies in Preclinical Models of Cancer. Cancer Res, 2016;76:5288–301. [DOI] [PubMed] [Google Scholar]
  • [384].Bertrand A, Kostine M, Barnetche T, Truchetet ME, Schaeverbeke T. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med, 2015;13:211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [385].Caramalho I, Nunes-Cabaco H, Foxall RB, Sousa AE. Regulatory T-cell development in the human thymus. Frontiers in Immunology, 2015;6. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES